Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Appl Biochem Biotechnol ; 191(2): 657-665, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31845192

RESUMEN

This study describes the development of a new methodology based on a new integrated equation which allows the determination of the kinetic parameters for two mutually non-exclusive inhibitors when one of which is produced during the time-course reaction. Alkaline phosphatase simultaneously inhibited by phosphate and urea is used to illustrate this methodology, including the evaluation of interaction effects between them. Data analyses were carried out using two integrated velocity equations: exclusive linear mixed inhibition (EMI) and non-exclusive linear mixed inhibition (NEMI). Kinetic parameters are estimated using non-linear regression and results show that (i) the interaction between enzyme and the inhibitors urea and phosphate exhibit a mutually non-exclusive behavior; (ii) more specifically, these inhibitors are non-exclusive only in free enzyme (E) species; (iii) the inhibitors also show an interaction with enzyme classified as facilitation; (iv) phosphate is a competitive inhibitor and urea a mixed inhibitor; (v) the inhibition constant for phosphate is much lower than that determined for urea. In addition, a functional Excel Spreadsheet which can be adapted to any kinetic study is also included as a supplement.


Asunto(s)
Fosfatasa Alcalina/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Unión Competitiva , Interacciones Farmacológicas , Cinética , Modelos Químicos , Fosfatos/antagonistas & inhibidores , Urea/antagonistas & inhibidores
2.
Arch Physiol Biochem ; 126(1): 89-93, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30081678

RESUMEN

This study was conducted to determine the effect of astaxanthin (ASX) treatment on alleviation of renal damage in high fructose induced nephrotoxicity in rats. Treatments were arranged in a 2 × 2 factorial fashion: administrations of fructose (30%, via drinking water) and ASX (1 mg/kg/day, within 0.2 ml olive oil) for 8 weeks. Data were analyzed by two-way ANOVA. The ASX treatment decreased serum urea (p < .01) and blood urea-N concentrations (p < .02) at a lower extent in rats receiving fructose than those not receiving fructose. Moreover, the ASX treatment reversed the increases in malondialdehyde (MDA) (p < .0001) and nuclear factor kappa B (NF-κB) (p < .0003) levels and the decreases in superoxide dismutase (SOD) activity (p < .0001) and sirtuin-1 (SIRT1) level (p < .0004), in the kidney upon high fructose consumption. The data suggest that ASX supplementation alleviates renal damage induced by high fructose consumption through modulating NF-κB/SIRT1 pathway and mitigating oxidative stress.


Asunto(s)
Antioxidantes/farmacología , Fructosa/efectos adversos , Riñón/efectos de los fármacos , FN-kappa B/genética , Sirtuina 1/genética , Animales , Nitrógeno de la Urea Sanguínea , Dieta/efectos adversos , Regulación de la Expresión Génica , Riñón/metabolismo , Riñón/patología , Masculino , Malondialdehído/antagonistas & inhibidores , Malondialdehído/sangre , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Transducción de Señal , Sirtuina 1/metabolismo , Superóxido Dismutasa/sangre , Urea/antagonistas & inhibidores , Urea/sangre , Xantófilas/farmacología
3.
Int Endod J ; 51(12): 1398-1409, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29858522

RESUMEN

AIM: To investigate the role of rostral ventromedial medulla orexin-1 receptors in the modulation of orofacial nociception as well as nociception-induced learning and memory impairment in adult male rats. METHODOLOGY: Pulpal nociception was induced by intradental application of capsaicin (100 µg) into the incisors of rats. Orexin-1 receptors agonist (orexin-A, 10, 25 and 50 pmol L-1  rat-1 ) and antagonist (SB-334867-A, 40 and 80 nmol L-1  rat-1 ) were microinjected into the rostral ventromedial medulla prior to capsaicin administration. Total time spent on nocifensive behaviour was recorded by direct visualization of freely moving rats whilst learning and memory were evaluated by the Morris water maze test. One-way analysis of variance and repeated-measures were used for the statistical analysis. RESULTS: Capsaicin-treated rats had a significant increase of nocifensive behaviours (P < 0.001), as well as learning and memory impairment (P < 0.001). However, intraventromedial medulla prior micro-injection of orexin-A (50 pmol L-1  rat-1 ) significantly reduced the nociceptive behaviour (P < 0.001). This effect was blocked by pre-treatment with SB334867-A (80 nmol L-1  rat-1 ). Orexin-A (50 pmol L-1  rat-1 ) also inhibited nociception-induced learning and memory deficits. Moreover, administration of SB-334867-A (80 nmol L-1  rat-1 ) plus orexin-A (50 pmol L-1  rat-1 ) had no effect on learning and memory deficits induced by capsaicin. CONCLUSIONS: The data suggest that rostral ventromedial medulla orexin-A receptors are involved in pulpal nociceptive modulation and improvement of learning and memory deficits induced by intradental application of capsaicin.


Asunto(s)
Capsaicina/farmacología , Pulpa Dental/efectos de los fármacos , Bulbo Raquídeo/efectos de los fármacos , Nocicepción/efectos de los fármacos , Receptores de Orexina/metabolismo , Aprendizaje Espacial/efectos de los fármacos , Memoria Espacial/efectos de los fármacos , Experimentación Animal , Animales , Benzoxazoles/antagonistas & inhibidores , Capsaicina/administración & dosificación , Relación Dosis-Respuesta a Droga , Masculino , Naftiridinas , Antagonistas de los Receptores de Orexina/farmacología , Orexinas , Ratas , Ratas Wistar , Fármacos del Sistema Sensorial/administración & dosificación , Fármacos del Sistema Sensorial/farmacología , Urea/análogos & derivados , Urea/antagonistas & inhibidores
4.
J Phys Chem B ; 119(30): 9820-34, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26115143

RESUMEN

To provide the underlying mechanism of the inhibiting effect of trehalose on the urea denatured protein, we perform classical molecular dynamics simulations of N-methylacetamide (NMA) in aqueous urea and/or trehalose solution. The site-site radial distribution functions and hydrogen bond properties indicate in binary urea solution the replacement of NMA-water hydrogen bonds by NMA-urea hydrogen bonds. On the other hand, in ternary urea and trehalose solution, trehalose does not replace the NMA-urea hydrogen bonds significantly; rather, it forms hydrogen bonds with the NMA molecule. The calculation of a preferential interaction parameter shows that, at the NMA surface, trehalose molecules are preferred and the preference for urea decreases slightly in ternary solution with respect to the binary solution. The exclusion of urea molecules in the ternary urea-NMA-trehalose system causes alleviation in van der Waals interaction energy between urea and NMA molecules. Our findings also reveal the following: (a) trehalose and urea induced second shell collapse of water structure, (b) a reduction in the mean trehalose cluster size in ternary solution, and (c) slowing down of translational motion of solution species in the presence of osmolytes. Implications of these results for the molecular explanations of the counteracting mechanism of trehalose on urea induced protein denaturation are discussed.


Asunto(s)
Simulación de Dinámica Molecular , Desnaturalización Proteica/efectos de los fármacos , Trehalosa/química , Trehalosa/farmacología , Urea/antagonistas & inhibidores , Urea/farmacología , Acetamidas/química , Difusión , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Conformación Molecular , Urea/química , Agua/química
5.
Clin Sci (Lond) ; 125(7): 349-59, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23611540

RESUMEN

We have shown previously that inhibition of sEH (soluble epoxide hydrolase) increased EETs (epoxyeicosatrienoic acids) levels and reduced renal injury in diabetic mice and these changes were associated with induction of HO (haem oxygenase)-1. The present study determines whether the inhibition of HO negates the renoprotective effect of sEH inhibition in diabetic SHR (spontaneously hypertensive rats). After 6 weeks of induction of diabetes with streptozotocin, SHR were divided into the following groups: untreated, treated with the sEH inhibitor t-AUCB {trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid}, treated with the HO inhibitor SnMP (stannous mesoporphyrin), and treated with both inhibitors for 4 more weeks; non-diabetic SHR served as a control group. Induction of diabetes significantly increased renal sEH expression and decreased the renal EETs/DHETEs (dihydroxyeicosatrienoic acid) ratio without affecting HO-1 activity or expression in SHR. Inhibition of sEH with t-AUCB increased the renal EETs/DHETEs ratio and HO-1 activity in diabetic SHR; however, it did not significantly alter systolic blood pressure. Treatment of diabetic SHR with t-AUCB significantly reduced the elevation in urinary albumin and nephrin excretion, whereas co-administration of the HO inhibitor SnMP with t-AUCB prevented these changes. Immunohistochemical analysis revealed elevations in renal fibrosis as indicated by increased renal TGF-ß (transforming growth factor ß) levels and fibronectin expression in diabetic SHR and these changes were reduced with sEH inhibition. Co-administration of SnMP with t-AUCB prevented its ability to reduce renal fibrosis in diabetic SHR. In addition, SnMP treatment also prevented t-AUCB-induced decreases in renal macrophage infiltration, IL-17 expression and MCP-1 levels in diabetic SHR. These findings suggest that HO-1 induction is involved in the protective effect of sEH inhibition against diabetic renal injury.


Asunto(s)
Benzoatos/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Nefropatías Diabéticas/prevención & control , Epóxido Hidrolasas/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/fisiología , Urea/análogos & derivados , Actinas/metabolismo , Albuminuria/prevención & control , Animales , Benzoatos/antagonistas & inhibidores , Benzoatos/farmacología , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Creatinina/orina , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/fisiopatología , Nefropatías Diabéticas/enzimología , Nefropatías Diabéticas/metabolismo , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Epóxido Hidrolasas/metabolismo , Epóxido Hidrolasas/fisiología , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/fisiología , Corteza Renal/metabolismo , Masculino , Proteínas de la Membrana/orina , Metaloporfirinas/farmacología , Ratas , Ratas Endogámicas SHR , Urea/antagonistas & inhibidores , Urea/farmacología , Urea/uso terapéutico
6.
Protein Pept Lett ; 20(1): 61-70, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22670764

RESUMEN

Kidney cells of animals including human and marine invertebrates contain high amount of the protein denaturant, urea. Methylamine osmolytes are generally believed to offset the harmful effects of urea on proteins in vitro and in vivo. In this study we have investigated the possibility of glycine to counteract the effects of urea on three proteins by measuring thermodynamic stability, ΔGD° and functional activity parameters (K(m) and k(cat)). We discovered that glycine does not counteract the effects of urea in terms of both protein stability and functional activity. We also observed that the glycine alone is compatible with enzymes function and increases protein stability in terms of T(m) (midpoint of thermal denaturation) to a great extent. Our study indicates that a most probable reason for the absence of a stabilizing osmolyte, glycine in the urea-rich cells is due to the fact that this osmolyte is non-protective to macromolecules against the hostile effects of urea, and hence is not chosen by evolutionary selection pressure.


Asunto(s)
Glicina/farmacología , Desnaturalización Proteica/efectos de los fármacos , Urea/antagonistas & inhibidores , Animales , Humanos , Lactalbúmina/efectos de los fármacos , Metilaminas/farmacología , Muramidasa/efectos de los fármacos , Ósmosis , Estabilidad Proteica , Estructura Secundaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Ribonucleasa Pancreática/efectos de los fármacos , Termodinámica , Urea/farmacología
7.
Eur J Pharmacol ; 641(2-3): 135-41, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20534382

RESUMEN

The transient receptor potential vanilloid 1 (TRPV1) receptor is activated by noxious heat, various endogenous mediators and exogenous irritants. The aim of the present study was to compare three TRPV1 receptor antagonists (SB705498, BCTC and AMG9810) in rat models of heat hyperalgesia. The behavioural noxious heat threshold, defined as the lowest temperature evoking nocifensive reaction, was measured with an increasing-temperature water bath. The effects of TRPV1 receptor antagonists were assessed in thermal hyperalgesia induced by the TRPV1 agonist resiniferatoxin (RTX), mild heat injury (51 degrees C, 20s) or plantar incision in rats. The control heat threshold was 43.2+/-0.4 degrees C. RTX induced an 8-10 degrees C decrease in heat threshold which was dose-dependently inhibited by oral pre-treatment with any of the TRPV1 receptor antagonists with a minimum effective dose of 1mg/kg. The mild heat injury-evoked 7-8 degrees C heat threshold drop was significantly reversed by all three antagonists injected i.p. as post-treatment. The minimum effective doses were as follows: SB705498 10, BCTC 3 and AMG9810 1mg/kg. Plantar incision-induced heat threshold drop (7-8 degrees C) was dose-dependently diminished by an oral post-treatment with any of the antagonists with minimum effective doses of 10, 3 and 3mg/kg, respectively. Assessment of RTX hyperalgesia by measurement of the paw withdrawal latency with a plantar test apparatus yielded 30 mg/kg minimum effective dose for each antagonist. In conclusion, measurement of the noxious heat threshold with the increasing-temperature water bath is suitable to sensitively detect the effects of TRPV1 receptor antagonists in thermal hyperalgesia models.


Asunto(s)
Acrilamidas/antagonistas & inhibidores , Compuestos Bicíclicos Heterocíclicos con Puentes/antagonistas & inhibidores , Calor/efectos adversos , Hiperalgesia/tratamiento farmacológico , Pirazinas/antagonistas & inhibidores , Piridinas/antagonistas & inhibidores , Canales Catiónicos TRPV/antagonistas & inhibidores , Animales , Frío , Modelos Animales de Enfermedad , Diterpenos/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Hiperalgesia/inducido químicamente , Dolor/tratamiento farmacológico , Pirrolidinas/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Urea/análogos & derivados , Urea/antagonistas & inhibidores
8.
Eur J Oral Sci ; 116(5): 424-30, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18821984

RESUMEN

Thymosin beta4 (Tbeta(4)) is a naturally occurring, ubiquitous, non-toxic protein with documented wound-healing, anti-inflammatory, anti-apoptotic, and tissue-repair properties in skin, the ocular surface, and the heart. The ability of Tbeta(4) to demonstrate similar protective properties in cells of the oral cavity was analyzed using an in vitro model of cultured human gingival fibroblasts. Thymosin beta 4 significantly suppressed the secretion of interleukin-8 (IL-8) following stimulation with tumor necrosis factoralpha (TNF-alpha), suggesting that it may suppress the inflammatory response initiated by pro-inflammatory cytokines. By contrast, Tbeta(4) was not effective in protecting fibroblasts from challenge with lipopolysaccharide purified from Porphyromonas gingivalis or Escherichia coli. Thymosin beta 4 was able to protect gingival fibroblasts against the known cytotoxic effects of chlorhexidine digluconate, a mouthrinse containing chlorhexidine digluconate, and carbamide peroxide. Additionally, Tbeta(4) was able to protect gingival fibroblasts from the apoptosis that is induced by stimulation with TNF-alpha or by exposure to chlorhexidine. Because of its multifunctional roles in protecting cells against damage, Tbeta(4) may have significant potential for use as an oral heathcare aid with combined antimicrobial, anti-inflammatory, anti-apoptotic, and cytoprotective properties.


Asunto(s)
Citoprotección , Encía/efectos de los fármacos , Interleucina-8/antagonistas & inhibidores , Timosina/farmacología , Antiinfecciosos Locales/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Peróxido de Carbamida , Células Cultivadas , Clorhexidina/análogos & derivados , Clorhexidina/antagonistas & inhibidores , Combinación de Medicamentos , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Encía/citología , Encía/inmunología , Humanos , Lipopolisacáridos/farmacología , Antisépticos Bucales , Oxidantes/antagonistas & inhibidores , Peróxidos/antagonistas & inhibidores , Pruebas de Toxicidad , Factor de Necrosis Tumoral alfa/farmacología , Urea/análogos & derivados , Urea/antagonistas & inhibidores
9.
J Agric Food Chem ; 55(6): 2268-75, 2007 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-17311398

RESUMEN

White bean (Phaseolus vulgaris L.) was used to study the antagonism caused by Na-bentazon on the phytotoxic action of the sulfonylurea (SU) herbicide tritosulfuron. After 168 h, uptake and translocation of [14C]tritosulfuron were reduced by 60 and 89%, respectively, when Na-bentazon was added to the mixture. Addition of (NH4)2SO4 or replacement of Na-bentazon with NH4-bentazon completely eliminated the negative effects on [14C]tritosulfuron uptake but not on its translocation. Scanning electron microscopy revealed that a mixture of Na-bentazon plus tritosulfuron plus DASH HC (0.156%) formed a rough layer of grain-like crystals on the leaf surface, whereas the addition of (NH4)2SO4 or replacement of Na-bentazon with NH4-bentazon resulted in amorphous deposits that may be more easily absorbed. The antagonism of tritosulfuron's phytotoxicity by Na-bentazon involves two separate processes, chemical (uptake effect) and biochemical (translocation effect).


Asunto(s)
Benzotiadiazinas/farmacología , Herbicidas/antagonistas & inhibidores , Hidrocarburos Fluorados/antagonistas & inhibidores , Phaseolus/efectos de los fármacos , Triazinas/antagonistas & inhibidores , Urea/análogos & derivados , Radioisótopos de Carbono , Herbicidas/farmacología , Hidrocarburos Fluorados/farmacología , Microscopía Electrónica de Transmisión , Triazinas/farmacología , Urea/antagonistas & inhibidores , Urea/farmacología
10.
Chem Biol Drug Des ; 69(1): 5-13, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17313452

RESUMEN

Multiscale simulations (coarse-grained Brownian dynamics simulations and all-atom molecular dynamics simulations in implicit solvent) were applied to reveal the binding processes of ligands as they enter the binding site of the HIV-1 protease. The initial structures used for the molecular dynamics simulations were generated based on the Brownian dynamics trajectories, and this is the first molecular dynamics simulation of modeling the association of a ligand with the protease. We found that a protease substrate successfully binds to the protein when the flaps are fully open. Surprisingly, a smaller cyclic urea inhibitor (XK263) can reach the binding site when the flaps are not fully open. However, if the flaps are nearly closed, the inhibitor must rearrange or binding can fail because the inhibitor cannot attain proper conformations to enter the binding site. Both the peptide substrate and XK263 can also affect the protein's internal motion, which may help the flaps to open. Simulations allow us to efficiently study the ligand binding processes and may help those who study drug discovery to find optimal association pathways and to design those ligands with the best binding kinetics.


Asunto(s)
Simulación por Computador , Diseño de Fármacos , Inhibidores de la Proteasa del VIH/farmacología , Proteasa del VIH/química , Proteasa del VIH/metabolismo , Azepinas/antagonistas & inhibidores , Azepinas/química , Azepinas/metabolismo , Sitios de Unión , Cinética , Ligandos , Conformación Proteica , Especificidad por Sustrato , Urea/análogos & derivados , Urea/antagonistas & inhibidores , Urea/metabolismo
11.
J Am Chem Soc ; 128(9): 2812-3, 2006 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-16506755

RESUMEN

We report unrestrained, all-atom molecular dynamics simulations of HIV-1 protease (HIV-PR) with a continuum solvent model that reproducibly sample closing of the active site flaps following manual placement of a cyclic urea inhibitor into the substrate binding site of the open protease. The open form was obtained from the unbound, semi-open HIV-PR crystal structure, which we recently reported (Hornak, V.; et al. Proc. Natl. Acad. Sci. U.S.A. 2006, 103, 915-920.) to have spontaneously opened during unrestrained dynamics. In those simulations, the transiently open flaps always returned to the semi-open form that is observed in all crystal structures of the free protease. Here, we show that manual docking of the inhibitor reproducibly induces spontaneous conversion to the closed form as seen in all inhibitor-bound HIV-PR crystal structures. These simulations reproduced not only the greater degree of flap closure, but also the striking difference in flap "handedness" between bound and free enzyme. In most of the simulations, the final structures were highly accurate. Root-mean-square deviations (RMSD) from the crystal structure of the complex were approximately 1.5 A (averaged over the last 100 ps) for the inhibitor and each flap despite initial RMSD of 2-5 A for the inhibitors and 6-11 A for the flaps. Key hydrogen bonds were formed between the flap tips and between flaps and inhibitor that match those seen in the crystal structure. The results demonstrate that all-atom simulations have the ability to significantly improve poorly docked ligand conformations and reproduce large-scale receptor conformational changes that occur upon binding.


Asunto(s)
Inhibidores de la Proteasa del VIH/química , Proteasa del VIH/química , VIH-1/enzimología , Azepinas/química , Azepinas/metabolismo , Azepinas/farmacología , Sitios de Unión , Simulación por Computador , Cristalografía por Rayos X , Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/farmacología , Modelos Químicos , Modelos Moleculares , Pliegue de Proteína , Urea/antagonistas & inhibidores , Urea/química , Urea/metabolismo , Agua/química , Agua/metabolismo
12.
Am J Physiol Lung Cell Mol Physiol ; 287(1): L60-8, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-14977627

RESUMEN

Nitric oxide (NO) is produced by NO synthase (NOS) from L-arginine (L-Arg). Alternatively, L-Arg can be metabolized by arginase to produce L-ornithine and urea. Arginase (AR) exists in two isoforms, ARI and ARII. We hypothesized that inhibiting AR with L-valine (L-Val) would increase NO production in bovine pulmonary arterial endothelial cells (bPAEC). bPAEC were grown to confluence in either regular medium (EGM; control) or EGM with lipopolysaccharide and tumor necrosis factor-alpha (L/T) added. Treatment of bPAEC with L/T resulted in greater ARI protein expression and ARII mRNA expression than in control bPAEC. Addition of L-Val to the medium led to a concentration-dependent decrease in urea production and a concentration-dependent increase in NO production in both control and L/T-treated bPAEC. In a second set of experiments, control and L/T bPAEC were grown in EGM, EGM with 30 mM L-Val, EGM with 10 mM L-Arg, or EGM with both 10 mM L-Arg and 30 mM L-Val. In both control and L/T bPAEC, treatment with L-Val decreased urea production and increased NO production. Treatment with L-Arg increased both urea and NO production. The addition of the combination L-Arg and L-Val decreased urea production compared with the addition of L-Arg alone and increased NO production compared with L-Val alone. These data suggest that competition for intracellular L-Arg by AR may be involved in the regulation of NOS activity in control bPAEC and in response to L/T treatment.


Asunto(s)
Arginasa/antagonistas & inhibidores , Endotelio Vascular/metabolismo , Óxido Nítrico/biosíntesis , Arteria Pulmonar/metabolismo , Valina/farmacología , Animales , Arginasa/genética , Arginasa/metabolismo , Arginina/farmacocinética , Arginina/farmacología , Bovinos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Endotelio Vascular/citología , Isoenzimas/genética , Isoenzimas/metabolismo , Lipopolisacáridos/farmacología , Nitratos/metabolismo , Nitritos/metabolismo , Concentración Osmolar , Arteria Pulmonar/citología , ARN Mensajero/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/farmacología , Urea/antagonistas & inhibidores
13.
Can J Anaesth ; 50(8): 830-4, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14525825

RESUMEN

BACKGROUND: Clinical use of high dose beta-blocker therapy is limited by excessive negative inotropic effects. Previous studies suggest that milrinone may be of utility in limiting the inotropic but not the chronotropic effects of beta blockers. We examined the hemodynamic effects of co-administration of a new potent selective beta(1) blocker, landiolol, and milrinone in halothane-anesthetized dogs. METHODS: Eighteen adult mongrel dogs were anesthetized with 1.2 MAC halothane. Hemodynamic measurements were made at baseline, 30 min after starting the milrinone (0.5 micro g x kg(-1) x min(-1)) or normal saline infusion (n = 9 in each), then 30 min after each change in the dose of landiolol infusion. The tested doses of landiolol were 10, 100, and 1000 micro g x kg(-1) x min(-1). RESULTS: Landiolol (>/= 10 micro g x kg(-1) x min(-1)) has significant and comparable negative chronotropic effects in both groups of dogs. While it also has significant negative inotropic effects in both groups, such effects are significantly attenuated in the dogs treated with milrinone. CONCLUSION: Milrinone is effective to attenuate the negative inotropic effects of landiolol in halothane-anesthetized dogs.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Anestesia por Inhalación , Anestésicos por Inhalación , Cardiotónicos/farmacología , Halotano , Milrinona/farmacología , Morfolinas/antagonistas & inhibidores , Morfolinas/farmacología , Urea/análogos & derivados , Urea/antagonistas & inhibidores , Urea/farmacología , Animales , Perros , Frecuencia Cardíaca/efectos de los fármacos , Contracción Miocárdica/efectos de los fármacos
14.
Biosens Bioelectron ; 18(4): 345-51, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12604251

RESUMEN

An urea biosensor based on urease-BSA (bovine serum albumin) membrane immobilised on the surface of an ion-sensitive field effect transistor (ISFET) has been studied in a mix buffer solution composed of potassium phosphate, Tris, citric acid and sodium tetraborate. In this mix buffer, the biosensor showed a dynamic larger than the one observed in a phosphate or Tris buffer. Investigation of the individual effect of each component of the buffer solution on the biosensor response has shown that tetraborate anion acts as a strong competitive inhibitor for the hydrolysis reaction of urea catalysed by urease. The biosensor response was investigated in a phosphate buffer with different concentrations of tetraborate anion. The results showed that the apparent constant of Michaelis-Menten, K(m(app)), increases from 4.3 to 79.3 mM, for experiments realised without and with 0.5 mM sodium tetraborate, respectively. The mean value, determined graphically, for the inhibition constant, K(i), was 29 microM. The graphical representation of biosensor calibration curves in semilogarithmic co-ordinates showed that the linear range of the biosensor can be extended up to three orders of magnitude, allowing an urea detection in a concentration range 0-100 mM.


Asunto(s)
Técnicas Biosensibles/instrumentación , Boratos/química , Electroquímica/instrumentación , Membranas Artificiales , Albúmina Sérica Bovina/química , Urea/análisis , Ureasa/química , Técnicas Biosensibles/métodos , Materiales Biocompatibles Revestidos/síntesis química , Electroquímica/métodos , Activación Enzimática , Enzimas Inmovilizadas , Diseño de Equipo , Concentración de Iones de Hidrógeno , Iones , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Transistores Electrónicos , Urea/antagonistas & inhibidores , Urea/química
15.
Proc Natl Acad Sci U S A ; 96(11): 6517-22, 1999 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-10339620

RESUMEN

The concentration of urea in renal medullary cells is high enough to affect enzymes seriously by reducing Vmax or raising Km, yet the cells survive and function. The usual explanation is that the methylamines found in the renal medulla, namely glycerophosphocholine and betaine, have actions opposite to those of urea and thus counteract its effects. However, urea and methylamines have the similar (not counteracting) effects of reducing both the Km and Vmax of aldose reductase (EC 1.1.1.21), an enzyme whose function is important in renal medullas. Therefore, we examined factors that might determine whether counteraction occurs, namely different combinations of assay conditions (pH and salt concentration), methylamines (glycerophosphocholine, betaine, and trimethylamine N-oxide), substrates (DL-glyceraldehyde and D-xylose), and a mutation in recombinant aldose reductase protein (C298A). We find that Vmax of both wild-type and C298A mutant generally is reduced by urea and/or the methylamines. However, the effects on Km are much more complex, varying widely with the combination of conditions. At one extreme, we find a reduction of Km of wild-type enzyme by urea and/or methylamines that is partially additive, whereas at the other extreme we find that urea raises Km for D-xylose of the C298A mutant, betaine lowers the Km, and the two counteract in a classical fashion so that at a 2:1 molar ratio of betaine to urea there is no net effect. We conclude that counteraction of urea effects on enzymes by methylamines can depend on ion concentration, pH, the specific methylamine and substrate, and identity of even a single amino acid in the enzyme.


Asunto(s)
Aldehído Reductasa/metabolismo , Metilaminas/farmacología , Urea/farmacología , Aldehído Reductasa/antagonistas & inhibidores , Gliceraldehído/metabolismo , Humanos , Médula Renal/fisiología , Cinética , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Urea/antagonistas & inhibidores , Xilosa/farmacología
16.
Protein Sci ; 8(1): 180-95, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10210196

RESUMEN

The aspartyl dyad of free HIV-1 protease has apparent pK(a)s of approximately 3 and approximately 6, but recent NMR studies indicate that the aspartyl dyad is fixed in the doubly protonated form over a wide pH range when cyclic urea inhibitors are bound, and in the monoprotonated form when the inhibitor KNI-272 is bound. We present computations and measurements related to these changes in protonation and to the thermodynamic linkage between protonation and inhibition. The Poisson-Boltzmann model of electrostatics is used to compute the apparent pK(a)s of the aspartyl dyad in the free enzyme and in complexes with four different inhibitors. The calculations are done with two parameter sets. One assigns epsilon = 4 to the solute interior and uses a detailed model of ionization; the other uses epsilon = 20 for the solute interior and a simplified representation of ionization. For the free enzyme, both parameter sets agree well with previously measured apparent pK(a)s of approximately 3 and approximately 6. However, the calculations with an internal dielectric constant of 4 reproduce the large pKa shifts upon binding of inhibitors, but the calculations with an internal dielectric constant of 20 do not. This observation has implications for the accurate calculation of pK(a)s in complex protein environments. Because binding of a cyclic urea inhibitor shifts the pK(a)s of the aspartyl dyad, changing the pH is expected to change its apparent binding affinity. However, we find experimentally that the affinity is independent of pH from 5.5 to 7.0. Possible explanations for this discrepancy are discussed.


Asunto(s)
Inhibidores de la Proteasa del VIH/química , Protones , Concentración de Iones de Hidrógeno , Cinética , Espectroscopía de Resonancia Magnética , Oligopéptidos/farmacología , Termodinámica , Urea/antagonistas & inhibidores
17.
Am J Physiol ; 276(2): R622-6, 1999 02.
Artículo en Inglés | MEDLINE | ID: mdl-9950946

RESUMEN

Marine elasmobranch fishes retain relatively high levels of urea to balance the osmotic stress of living in seawater. To maintain osmotic balance and reduce the energetic costs of making urea, it is important for these animals to minimize urea excretion to the environment. We have isolated a novel 2.2-kb cDNA from Squalus acanthias (spiny dogfish shark) kidney encoding a 380-amino acid hydrophobic protein (ShUT) with 66% identity to the rat facilitated urea transporter protein UT-A2. Injection of ShUT cRNA into Xenopus oocytes induced a 10-fold increase in 14C-labeled urea uptake, inhibitable by phloretin (0.35 mM). ShUT mRNA is expressed in kidney and brain. Related mRNA species are found in liver, blood, kidney, gill, intestine, muscle, and rectal gland. This is the first facilitated urea transporter to be identified in a marine fish. We propose that the ShUT protein is involved in urea reabsorption by the renal tubules of the dogfish shark, which in turn minimizes urea loss in the urine.


Asunto(s)
Proteínas Portadoras/genética , Cazón/genética , Glicoproteínas de Membrana/genética , Proteínas de Transporte de Membrana , Secuencia de Aminoácidos/genética , Animales , Proteínas Portadoras/metabolismo , ADN Complementario/genética , Femenino , Glicoproteínas de Membrana/metabolismo , Datos de Secuencia Molecular , Oocitos/metabolismo , Floretina/farmacología , ARN Mensajero/metabolismo , Distribución Tisular , Urea/antagonistas & inhibidores , Urea/metabolismo , Xenopus , Transportadores de Urea
18.
Biochemistry ; 37(43): 15042-9, 1998 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-9790666

RESUMEN

The long-term therapeutic benefit of HIV antiretroviral therapy is still threatened by drug-resistant variants. Mutations in the S1 subsite of the protease are the primary cause for the loss of sensitivity toward many HIV protease inhibitors, including our first-generation cyclic urea-based inhibitors DMP323 and DMP450. We now report the structures of the three active-site mutant proteases V82F, I84V, and V82F/I84V in complex with XV638 and SD146, two P2 analogues of DMP323 that are 8-fold more potent against the wild type and are able to inhibit a broad panel of drug-resistant variants [Jadhav, P. K., et al. (1997) J. Med. Chem. 40, 181-191]. The increased efficacy of XV638 and SD146 is due primarily to an increase in P2-S2 interactions: 30-40% more van der Waals contacts and two to four additional hydrogen bonds. Furthermore, because these new interactions do not perturb other subsites in the protease, it appears that the large complementary surface areas of their P2 substituents compensate for the loss of P1-S1 interactions and reduce the probability of selecting for drug-resistant variants.


Asunto(s)
Inhibidores de la Proteasa del VIH/química , Proteasa del VIH/química , Proteasa del VIH/genética , VIH-1/enzimología , Urea/análogos & derivados , Sustitución de Aminoácidos/genética , Azepinas , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Farmacorresistencia Microbiana/genética , Proteasa del VIH/farmacología , Inhibidores de la Proteasa del VIH/antagonistas & inhibidores , Inhibidores de la Proteasa del VIH/farmacología , VIH-1/efectos de los fármacos , Humanos , Sustancias Macromoleculares , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Especificidad por Sustrato , Urea/antagonistas & inhibidores , Urea/química , Urea/farmacología
19.
Am J Physiol ; 274(4): F762-5, 1998 04.
Artículo en Inglés | MEDLINE | ID: mdl-9575901

RESUMEN

Urea in renal medullas is sufficiently high to perturb macromolecules, yet the cells survive and function. The counteracting osmolytes hypothesis holds that methylamines, such as glycine betaine (betaine) and glycerophosphocholine (GPC) in renal medullas, stabilize macromolecules and oppose the effects of urea. Although betaine counteracts effects of urea on macromolecules in vitro and protects renal cells from urea in tissue culture, renal cells accumulate GPC rather than betaine in response to high urea both in vivo and in tissue culture. A proposed explanation is that GPC counteracts urea more effectively than betaine. However, we previously found GPC slightly less effective than betaine in counteracting inhibition of pyruvate kinase activity by urea. To test another macromolecule, we now compare GPC and betaine in counteracting reduction of the thermal stability of Rnase A by urea. We find that urea decreases the thermal transition temperature and that betaine and GPC increase it, counteracting urea approximately equally. Therefore, the preference for GPC in response to high urea presumably has some other basis, such as a lower metabolic cost of GPC accumulation.


Asunto(s)
Betaína/farmacología , Glicerilfosforilcolina/farmacología , Calor , Ribonucleasa Pancreática/química , Animales , Bovinos , Combinación de Medicamentos , Estabilidad de Medicamentos , Metilaminas/farmacología , Ribonucleasa Pancreática/efectos de los fármacos , Urea/antagonistas & inhibidores , Urea/farmacología
20.
Am J Physiol ; 273(2 Pt 2): R669-76, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9277553

RESUMEN

The natural osmolyte trimethylamine N-oxide (TMAO) is one of the methylamine compounds often accumulated by diverse organisms in response to osmotic stress and/or to compensate for the deleterious effects of urea. Tubulin polymerization is promoted by TMAO. At 1 M TMAO, tubulin polymers are produced with properties expected of normal steady-state microtubules (MT): polymerization is reversed by exposure to cold or the antimitotic drug podophyllotoxin, a critical concentration for polymerization at 30 degrees C of 1.5 microM is found, and the morphology of the polymers in electron micrographs is typical of MT and ribbons, or open MT. At 2 M TMAO, polymerization is very rapid and hyperstable polymers are formed. These are resistant to cold-induced depolymerization although still sensitive to podophyllotoxin inhibition. A lower critical concentration of 0.7 microM is observed, and electron micrographs reveal MT, ribbons, and other polymer forms not usually stable, such as splayed protofilaments. Inhibition of tubulin polymerization by low concentrations of urea (Sackett, D. L., B. Bhattacharyya, and J. wolff. Biochemistry 33: 12868-12878, 1994) is largely reversed by the presence of TMAO at one-half the molarity of urea, the physiological ratio observed in cartilaginous fishes. Other methylamines, including betaine, dimethylglycine, glycine, and sarcosine, failed to stimulate MT polymerization or protect against urea inhibition. Trimethylamine, taurine and glycylglycine inhibit polymerization. TMAO did not interfere with binding of MT-associated proteins (MAP) and protected both tubulin assembly and MAP binding from urea.


Asunto(s)
Metilaminas/farmacología , Polímeros/metabolismo , Tubulina (Proteína)/metabolismo , Urea/metabolismo , Animales , Frío , Microscopía Electrónica , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Concentración Osmolar , Ratas , Tubulina (Proteína)/efectos de los fármacos , Tubulina (Proteína)/ultraestructura , Urea/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA