Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 155
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Phys Ther Sci ; 27(6): 1645-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26180289

RESUMEN

[Purpose] This study aimed to evaluate the anti-inflammatory and analgesic effects of intraoral application of low-level laser therapy (660 nm) to control pain, swelling and interincisal opening following the extraction of mandibular third molars. [Subjects and Methods] Ten patients underwent removal of lower third molars using the same surgical protocol and pharmacological approach. In the postoperative period, all patients received four consecutive daily sessions of low-level laser therapy, beginning 24 hours after the surgery. Intraoral applications using the diode laser with 660 nm wavelength in the continuous scan mode were performed covering the entire surgical area, which was divided into four quadrants, each of 1 cm(2) area at a distance of 1 cm. The energy applied at each point was 5 J/cm(2) during 8 seconds. [Results] The swelling and interincisal opening returned to normal 24 hours after the first low-level laser therapy application (Friedman test). Moreover, the pain intensity was reduced on the third postoperative day, according to the Friedman test. [Conclusion] Low-level laser therapy (660 nm), at the dosimetry used in this study, was effective in reducing postoperative pain and swelling following oral surgery.

2.
Kidney Int ; 70(1): 51-9, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16710355

RESUMEN

Type II Bartter's syndrome is a hereditary hypokalemic renal salt-wasting disorder caused by mutations in the ROMK channel (Kir1.1; Kcnj1), mediating potassium recycling in the thick ascending limb of Henle's loop (TAL) and potassium secretion in the distal tubule and cortical collecting duct (CCT). Newborns with Type II Bartter are transiently hyperkalemic, consistent with loss of ROMK channel function in potassium secretion in distal convoluted tubule and CCT. Yet, these infants rapidly develop persistent hypokalemia owing to increased renal potassium excretion mediated by unknown mechanisms. Here, we used free-flow micropuncture and stationary microperfusion of the late distal tubule to explore the mechanism of renal potassium wasting in the Romk-deficient, Type II Bartter's mouse. We show that potassium absorption in the loop of Henle is reduced in Romk-deficient mice and can account for a significant fraction of renal potassium loss. In addition, we show that iberiotoxin (IBTX)-sensitive, flow-stimulated maxi-K channels account for sustained potassium secretion in the late distal tubule, despite loss of ROMK function. IBTX-sensitive potassium secretion is also increased in high-potassium-adapted wild-type mice. Thus, renal potassium wasting in Type II Bartter is due to both reduced reabsorption in the TAL and K secretion by max-K channels in the late distal tubule.


Asunto(s)
Síndrome de Bartter/metabolismo , Hipopotasemia/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Canales de Potasio de Rectificación Interna/deficiencia , Potasio/orina , Adaptación Fisiológica , Animales , Síndrome de Bartter/genética , Transporte Biológico , Dieta , Modelos Animales de Enfermedad , Hipopotasemia/genética , Túbulos Renales Distales/efectos de los fármacos , Túbulos Renales Distales/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Asa de la Nefrona/efectos de los fármacos , Asa de la Nefrona/fisiología , Ratones , Ratones Mutantes , Péptidos/farmacología , Potasio/metabolismo , Canales de Potasio de Rectificación Interna/genética , Potasio en la Dieta/administración & dosificación
3.
Proc Natl Acad Sci U S A ; 99(14): 9231-6, 2002 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-12093923

RESUMEN

To determine whether calcium polyvalent cation-sensing receptors (CaRs) are salinity sensors in fish, we used a homology-based cloning strategy to isolate a 4.1-kb cDNA encoding a 1,027-aa dogfish shark (Squalus acanthias) kidney CaR. Expression studies in human embryonic kidney cells reveal that shark kidney senses combinations of Ca(2+), Mg(2+), and Na(+) ions at concentrations present in seawater and kidney tubules. Shark kidney is expressed in multiple shark osmoregulatory organs, including specific tubules of the kidney, rectal gland, stomach, intestine, olfactory lamellae, gill, and brain. Reverse transcriptase-PCR amplification using specific primers in two teleost fish, winter flounder (Pleuronectes americanus) and Atlantic salmon (Salmo salar), reveals a similar pattern of CaR tissue expression. Exposure of the lumen of winter flounder urinary bladder to the CaR agonists, Gd(3+) and neomycin, reversibly inhibit volume transport, which is important for euryhaline teleost survival in seawater. Within 24-72 hr after transfer of freshwater-adapted Atlantic salmon to seawater, there are increases in their plasma Ca(2+), Mg(2+), and Na(+) that likely serve as a signal for internal CaRs, i.e., brain, to sense alterations in salinity in the surrounding water. We conclude that CaRs act as salinity sensors in both teleost and elasmobranch fish. Their tissue expression patterns in fish provide insights into CaR functions in terrestrial animals including humans.


Asunto(s)
Calcio/metabolismo , Peces/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , ADN Complementario/genética , Cazón/genética , Cazón/metabolismo , Peces/genética , Lenguado/genética , Lenguado/metabolismo , Humanos , Riñón/metabolismo , Magnesio/metabolismo , Datos de Secuencia Molecular , Estructura Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Sensibles al Calcio , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Salmo salar/genética , Salmo salar/metabolismo , Agua de Mar , Homología de Secuencia de Aminoácido , Sodio/metabolismo , Cloruro de Sodio , Distribución Tisular , Transfección , Vejiga Urinaria/metabolismo
4.
Am J Physiol Renal Physiol ; 281(6): F1021-7, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11704552

RESUMEN

The organization of Na(+) and Ca(2+) transport pathways along the mouse distal nephron is incompletely known. We revealed by immunohistochemistry a set of Ca(2+) and Na(+) transport proteins along the mouse distal convolution. The thiazide-sensitive Na(+)-Cl(-) cotransporter (NCC) characterized the distal convoluted tubule (DCT). The amiloride-sensitive epithelial Na(+) channel (ENaC) colocalized with NCC in late DCT (DCT2) and extended to the downstream connecting tubule (CNT) and collecting duct (CD). In early DCT (DCT1), the basolateral Ca(2+)-extruding proteins [Na(+)/Ca(2+) exchanger (NCX), plasma membrane Ca(2+)-ATPase (PCMA)] and the cytoplasmic Ca(2+)-binding protein calbindin D(28K) (CB) were found at very low levels, whereas the cytoplasmic Ca(2+)/Mg(2+)-binding protein parvalbumin was highly abundant. NCX, PMCA, and CB prevailed in DCT2 and CNT, where we located the apical epithelial Ca(2+) channel (ECaC1). Its subcellular localization changed from apical in DCT2 to exclusively cytoplasmic at the end of CNT. NCX and PMCA decreased in parallel with the fading of ECaC1 in the apical membrane. All three of them were undetectable in CD. These findings disclose DCT2 and CNT as major sites for transcellular Ca(2+) transport in the mouse distal nephron. Cellular colocalization of Ca(2+) and Na(+) transport pathways suggests their mutual interactions in transport regulation.


Asunto(s)
Calcio/metabolismo , Proteínas Portadoras/análisis , Túbulos Renales Distales/metabolismo , Receptores de Droga , Sodio/metabolismo , Simportadores , Animales , Calbindinas , Canales de Calcio/análisis , Canales de Calcio/inmunología , ATPasas Transportadoras de Calcio/análisis , ATPasas Transportadoras de Calcio/inmunología , Proteínas Portadoras/inmunología , Proteínas de Transporte de Catión , Canales Epiteliales de Sodio , Femenino , Inmunohistoquímica , Transporte Iónico , Túbulos Renales Distales/química , Ratones , Modelos Biológicos , Parvalbúminas/análisis , Parvalbúminas/inmunología , ATPasas Transportadoras de Calcio de la Membrana Plasmática , Proteína G de Unión al Calcio S100/análisis , Proteína G de Unión al Calcio S100/inmunología , Canales de Sodio/análisis , Canales de Sodio/inmunología , Simportadores del Cloruro de Sodio , Intercambiador de Sodio-Calcio/análisis , Intercambiador de Sodio-Calcio/inmunología , Miembro 3 de la Familia de Transportadores de Soluto 12 , Canales Catiónicos TRPV
5.
J Biol Chem ; 276(47): 44347-53, 2001 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-11567030

RESUMEN

ATP-regulated (K(ATP)) channels are formed by an inward rectifier pore-forming subunit (Kir) and a sulfonylurea (glibenclamide)-binding protein, a member of the ATP binding cassette family (sulfonylurea receptor (SUR) or cystic fibrosis transmembrane conductance regulator). The latter is required to confer glibenclamide sensitivity to K(ATP) channels. In the mammalian kidney ROMK1-3 are components of K(ATP) channels that mediate K(+) secretion into urine. ROMK1 and ROMK3 splice variants share the core polypeptide of ROMK2 but also have distinct NH(2)-terminal extensions of 19 and 26 amino acids, respectively. The SUR2B is also expressed in rat kidney tubules and may combine with Kir.1 to form renal K(ATP) channels. Our previous studies showed that co-expression of ROMK2, but not ROMK1 or ROMK3, with rat SUR2B in oocytes generated glibenclamide-sensitive K(+) currents. These data suggest that the NH(2)-terminal extensions in both ROMK1 and ROMK3 block ROMK-SUR2B interaction. Seven amino acids in the NH(2)-terminal extensions of ROMK1 and ROMK3 are identical (amino acids 13-19 in ROMK1 and 20-26 in ROMK3) and may determine ROMK-SUR2B interaction. We constructed a series of hemagglutinin-tagged ROMK1 NH(2)-terminal deletion and substitution mutants and examined glibenclamide-sensitive K(+) currents in oocytes when co-expressed with SUR2B. These studies identified an amino acid triplet "IRA" within the conserved segment in the NH(2) terminus of ROMK1 and ROMK3 that blocks the ability of SUR2B to confer glibenclamide sensitivity to the expressed K(+) currents. The position of this triplet in the ROMK1 NH(2)-terminal extension is also important for the ROMK-SUR2B interactions. In vitro co-translation and immunoprecipitation studies with hemagglutinin-tagged ROMK mutants and SUR2B indicted that direct interaction between these two proteins is required for glibenclamide sensitivity of induced K(+) currents in oocytes. These results suggest that the IRA triplet in the NH(2)-terminal extensions of both ROMK1 and ROMK3 plays a key role in subunit assembly of the renal secretary K(ATP) channel.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Canales de Potasio de Rectificación Interna , Canales de Potasio/metabolismo , Receptores de Droga/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cartilla de ADN , Datos de Secuencia Molecular , Canales de Potasio/química , Canales de Potasio/genética , Pruebas de Precipitina , Biosíntesis de Proteínas , Ratas , Receptores de Droga/genética , Receptores de Sulfonilureas , Xenopus laevis
6.
Biochem Biophys Res Commun ; 286(2): 376-80, 2001 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-11500048

RESUMEN

Biophysical properties of ROMK2 channel were investigated at physiological temperature, after reexpression of the recombinant ROMK2 protein in a mammalian cell expression system (COS-7). We observed that ROMK2 induced an inwardly rectifying K(+) current whether polyvalent cations were present or not. Above +10 mV, ROMK2-induced current exhibited a voltage- and time-dependent decay, consistent with an inactivation process. Inactivation of ROMK2-induced current was also seen in inside out patch from ROMK2-expressing Xenopus oocyte. In COS-7 cells, inactivation was found to account for most of the inward rectification. Mg(2+) and spermine modulated rectification by accelerating inactivation kinetics independently of membrane potential. These results establish for the first time ROMK2 properties in a mammalian cell expression system.


Asunto(s)
Canales de Potasio de Rectificación Interna , Canales de Potasio/fisiología , Animales , Células COS , Conductividad Eléctrica , Cinética , Magnesio/farmacología , Potenciales de la Membrana , Oocitos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/genética , Proteínas Recombinantes/metabolismo , Espermidina/farmacología , Transfección , Xenopus
7.
Am J Physiol Renal Physiol ; 280(4): F574-82, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11249848

RESUMEN

In the absence of vasopressin, medullary thick ascending limb cells express a K(+)-independent, furosemide-sensitive Na(+)-Cl(-) cotransporter that is inhibited by hypertonicity. The murine renal specific Na(+)-K(+)-2 Cl(-) cotransporter gene (SLC12A1) gives rise to six alternatively spliced isoforms. Three feature a long COOH-terminal domain that encodes the butmetanide-sensitive Na(+)-K(+)-2 Cl(-) cotransporter (BSC1-9/NKCC2), and three with a short COOH-terminal domain, known as mBSC1-A4, B4, or F4 (19). Here we have determined the functional characteristics of mBSC1-A4, as expressed in Xenopus laevis oocytes. When incubated at normal oocyte osmolarity (approximately 200 mosmol/kgH(2)O), mBSC1-4-injected oocytes do not express significant Na(+) uptake over H(2)O-injected controls, and immunohistochemical analysis shows that the majority of mBSC1-4 protein is in the oocyte cytoplasm and not at the plasma membrane. In contrast, when mBSC1-4 oocytes are exposed to hypotonicity (approximately 100 mosmol/kgH(2)O), a significant increase in Na(+) uptake but not in (86)Rb(+) uptake is observed. The increased Na(+) uptake is Cl(-) dependent, furosemide sensitive, and cAMP sensitive but K(+) independent. Sodium uptake increases with decreasing osmolarity between 120 and 70 mosmol/kgH(2)O (r = 0.95, P < 0.01). Immunohistochemical analysis shows that in hypotonic conditions mBSC1-A4 protein is expressed in the plasma membrane. These studies indicate that the mBSC1-A4 isoform of the SLC12A1 gene encodes a hypotonically activated, cAMP- and furosemide-sensitive Na(+)-Cl(-) cotransporter. Thus it is possible that alternative splicing of the BSC1 gene could provide the molecular mechanism enabling the Na(+)-Cl(-)-to-Na(+)-K(+)-2Cl(-) switching in thick ascending limb cells.


Asunto(s)
Empalme Alternativo/fisiología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Diuréticos/farmacología , Furosemida/farmacología , Sulfonamidas , 1-Metil-3-Isobutilxantina/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Bumetanida/farmacología , Proteínas Portadoras/química , Membrana Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citosol/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Expresión Génica/fisiología , Soluciones Hipotónicas/farmacología , Isomerismo , Isoquinolinas/farmacología , Asa de la Nefrona/enzimología , Mamíferos , Oocitos/fisiología , Concentración Osmolar , Inhibidores de Fosfodiesterasa/farmacología , Radioisótopos de Sodio/farmacocinética , Simportadores de Cloruro de Sodio-Potasio , Tritio , Xenopus laevis
8.
J Biol Chem ; 276(10): 7156-63, 2001 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-11114300

RESUMEN

We have used the two-electrode voltage clamp technique and the patch clamp technique to investigate the regulation of ROMK1 channels by protein-tyrosine phosphatase (PTP) and protein-tyrosine kinase (PTK) in oocytes coexpressing ROMK1 and cSrc. Western blot analysis detected the presence of the endogenous PTP-1D isoform in the oocytes. Addition of phenylarsine oxide (PAO), an inhibitor of PTP, reversibly reduced K(+) current by 55% in oocytes coinjected with ROMK1 and cSrc. In contrast, PAO had no significant effect on K(+) current in oocytes injected with ROMK1 alone. Moreover, application of herbimycin A, an inhibitor of PTK, increased K(+) current by 120% and completely abolished the effect of PAO in oocytes coexpressing ROMK1 and cSrc. The effects of herbimycin A and PAO were absent in oocytes expressing the ROMK1 mutant R1Y337A in which the tyrosine residue at position 337 was mutated to alanine. However, addition of exogenous cSrc had no significant effect on the activity of ROMK1 channels in inside-out patches. Moreover, the effect of PAO was completely abolished by treatment of oocytes with 20% sucrose and 250 microg/ml concanavalin A, agents that inhibit the endocytosis of ROMK1 channels. Furthermore, the effect of herbimycin A is absent in the oocytes pretreated with either colchicine, an inhibitor of microtubules, or taxol, an agent that freezes microtubules. We conclude that PTP and PTK play an important role in regulating ROMK1 channels. Inhibiting PTP increases the internalization of ROMK1 channels, whereas blocking PTK stimulates the insertion of ROMK1 channels.


Asunto(s)
Canales de Potasio de Rectificación Interna , Canales de Potasio/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Alanina/química , Animales , Benzoquinonas , Western Blotting , Colchicina/farmacología , Concanavalina A/farmacología , Inhibidores Enzimáticos/farmacología , Lactamas Macrocíclicas , Microscopía Fluorescente , Microtúbulos/metabolismo , Modelos Biológicos , Mutación , Oocitos/metabolismo , Paclitaxel/farmacología , Técnicas de Placa-Clamp , Potasio/metabolismo , Quinonas/farmacología , ARN Complementario/metabolismo , Rifabutina/análogos & derivados , Sacarosa/farmacología , Factores de Tiempo , Tirosina/química , Xenopus
9.
Am J Physiol Renal Physiol ; 279(5): F919-26, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11053053

RESUMEN

Close similarity between the rat native low-conductance K(+) channel in the apical membrane of renal cortical collecting duct principal cells and the cloned rat ROMK channel strongly suggest that the two are identical. Prominent features of ROMK regulation are a steep pH dependence and activation by protein kinase A (PKA)-dependent phosphorylation. In this study, we investigated the pH dependence of cloned renal K(+) channel (ROMK2), wild-type (R2-WT), and PKA site mutant channels (R2-S25A, R2-S200A, and R2-S294A). Ba(2+)-sensitive outward whole cell currents (holding voltage -50 mV) were measured in two-electrode voltage-clamp experiments in Xenopus laevis oocytes expressing either R2-WT or mutant channels. Intracellular pH (pH(i)) was measured with pH-sensitive microelectrodes in a different group of oocytes from the same batch on the same day. Resting pH(i) of R2-WT and PKA site mutants was the same: 7.32 +/- 0.02 (n = 22). The oocytes were acidified by adding 3 mM Na butyrate with external pH (pH(o)) adjusted to 7.4, 6.9, 6.4, or 5.4. At pH(o) 7.4, butyrate led to a rapid (tau: 163 +/- 14 s, where tau means time constant, n = 4) and stable acidification of the oocytes (DeltapH(i) 0.13 +/- 0. 02 pH units, where Delta means change, n = 12). Intracellular acidification reversibly inhibited ROMK2-dependent whole cell current. The effective acidic dissociation constant (pK(a)) value of R2-WT was 6.92 +/- 0.03 (n = 8). Similarly, the effective pK(a) value of the N-terminal PKA site mutant R2-S25A was 6.99 +/- 0.02 (n = 6). The effective pK(a) values of the two COOH-terminal PKA site mutant channels, however, were significantly shifted to alkaline values; i.e., 7.15 +/- 0.06 (n = 5) for R2-S200A and 7.16 +/- 0.03 (n = 8) for R2-S294A. The apparent DeltapH shift between the R2-WT and the R2-S294A mutant was 0.24 pH units. In excised inside-out patches, alkaline pH 8.5 activated R2-S294A channel current by 32 +/- 6.7%, whereas in R2-WT channel patches alkalinzation only marginally increased current by 6.5 +/- 1% (n = 5). These results suggest that channel phosphorylation may substantially influence the pH sensitivity of ROMK2 channel. Our data are consistent with the hypothesis that in the native channel PKA activation involves a shift of the pK(a) value of ROMK channels to more acidic values, thus relieving a H(+)-mediated inhibition of ROMK channels.


Asunto(s)
Equilibrio Ácido-Base/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Mutagénesis Sitio-Dirigida , Canales de Potasio de Rectificación Interna , Canales de Potasio/genética , Canales de Potasio/metabolismo , Álcalis/metabolismo , Álcalis/farmacología , Animales , Sitios de Unión/genética , Ácido Butírico/metabolismo , Ácido Butírico/farmacología , Células Cultivadas , Concentración de Iones de Hidrógeno/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Oocitos/citología , Oocitos/metabolismo , Técnicas de Placa-Clamp , Fosforilación/efectos de los fármacos , Ratas , Relación Estructura-Actividad , Xenopus laevis
10.
J Biol Chem ; 275(43): 33548-53, 2000 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-10942780

RESUMEN

To assess whether glucocorticoids regulate rBSC1, the apical Na(+)-K(+)(NH(4)(+))-2Cl(-) cotransporter of kidney medullary thick ascending limb (MTAL), studies were performed in normal rats, adrenalectomized (ADX) rats, and ADX rats infused with dexamethasone for 6 days. The effects of dexamethasone on rBSC1 were also studied in vitro using isolated rat MTAL segments. Cotransport activity was estimated by intracellular pH measurements; rBSC1 protein was quantified in MTAL crude membranes by immunoblotting analysis, and mRNA was quantified by quantitative reverse transcription-polymerase chain reaction. The abundance of rBSC1 protein and mRNA increased in ADX rats infused with dexamethasone compared with ADX rats (p < 0. 04). In addition, application of dexamethasone for 1-3 h to MTALs caused rBSC1 protein and mRNA abundance and cotransport activity to significantly increase in a hyperosmotic medium (450 mosmol/kg of H(2)O) containing 0.7 nm arginine vasopressin, which is an in vitro experimental condition that resembles the in vivo MTAL environment. Results obtained in various media and with 8-bromo-cAMP indicated that stimulation of rBSC1 expression by glucocorticoids required interactions between glucocorticoid receptor- and cAMP-dependent factors. Up to 100 nm d-aldosterone had no effect on cotransport activity in vitro. Thus glucocorticoids directly stimulate MTAL rBSC1 expression and activity, which contributes to glucocorticoid-dependent effects on the renal regulation of acid-base balance and urinary concentrating ability.


Asunto(s)
Proteínas Portadoras/biosíntesis , Dexametasona/farmacología , Médula Renal/metabolismo , Túbulos Renales/metabolismo , Adrenalectomía , Animales , Arginina Vasopresina/farmacología , Proteínas Portadoras/genética , Masculino , Compuestos de Amonio Cuaternario/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Simportadores de Cloruro de Sodio-Potasio
11.
Proc Natl Acad Sci U S A ; 97(14): 8157-62, 2000 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-10884438

RESUMEN

Transforming growth factor beta (TGF-beta) has been shown to participate in the pathophysiology of diabetic complications. As shown most recently, TGF-beta stimulates the expression of a distinct serine/threonine kinase (hSGK) which had previously been cloned as an early gene transcriptionally regulated by cell volume alterations. The present study was performed to elucidate transcription and function of hSGK in diabetic nephropathy. As shown by Northern blotting, an increase of extracellular glucose concentration increased hSGK mRNA levels in cultured cells, an effect qualitatively mimicked by osmotic cell shrinkage or treatment with TGF-beta (2 microgram/liter), phorbol 12,13-didecanoate (1 microM), or the Ca(2+) ionophore ionomycin (1 microM) and blunted by high concentrations of nifedipine (10 and 100 microM). In situ hybridization revealed that hSGK transcription was markedly enhanced in diabetic nephropathy, with particularly high expression in mesangial cells, interstitial cells, and cells in thick ascending limbs of Henle's loop and distal tubules. According to voltage clamp and tracer flux studies in Xenopus oocytes expressing the renal epithelial Na(+) channel ENaC or the mouse thick ascending limb Na(+),K(+),2Cl(-) cotransporter BSC-1, coexpression with hSGK stimulated ENaC and BSC-1 11-fold and 6-fold, respectively, effects reversed by kinase inhibitors staurosporine (1 microM) and chelerythrine (1 microM) and not elicited by inactive hSGK. In conclusion, excessive extracellular glucose concentrations enhance hSGK transcription, which in turn stimulates renal tubular Na(+) transport. These observations disclose an additional element in the pathophysiology of diabetic nephropathy.


Asunto(s)
Nefropatías Diabéticas/genética , Proteínas Serina-Treonina Quinasas/genética , Células 3T3 , Animales , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Tamaño de la Célula , Canales Epiteliales de Sodio , Regulación de la Expresión Génica , Humanos , Hibridación in Situ , Riñón/patología , Ratones , Técnicas de Placa-Clamp , Proteínas Serina-Treonina Quinasas/biosíntesis , Ratas , Proteínas Recombinantes/biosíntesis , Sodio/metabolismo , Canales de Sodio/biosíntesis , Canales de Sodio/genética , Simportadores de Cloruro de Sodio-Potasio , Distribución Tisular , Transcripción Genética
12.
Am J Physiol Renal Physiol ; 279(1): F161-9, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10894798

RESUMEN

The thiazide-sensitive Na(+)-Cl(-) cotransporter (TSC) is the major pathway for salt reabsorption in the apical membrane of the mammalian distal convoluted tubule. When expressed in Xenopus laevis oocytes, rat TSC exhibits high affinity for both cotransported ions, with the Michaelis-Menten constant (K(m)) for Na(+) of 7.6 +/- 1.6 mM and for Cl(-) of 6.3 +/- 1.1 mM, and Hill coefficients for Na(+) and Cl(-) consistent with electroneutrality. The affinities of both Na(+) and Cl(-) were increased by increasing concentration of the counterion. The IC(50) values for thiazides were affected by both extracellular Na(+) and Cl(-). The higher the Na(+) or Cl(-) concentration, the lower the inhibitory effect of thiazides. Finally, rTSC function is affected by extracellular osmolarity. We propose a transport model featuring a random order of binding in which the binding of each ion facilitates the binding of the counterion. Both ion binding sites alter thiazide-mediated inhibition of transport, indicating that the thiazide-binding site is either shared or modified by both Na(+) and Cl(-).


Asunto(s)
Proteínas Portadoras/metabolismo , Receptores de Droga/metabolismo , Inhibidores de los Simportadores del Cloruro de Sodio/metabolismo , Inhibidores de los Simportadores del Cloruro de Sodio/farmacología , Sodio/metabolismo , Simportadores , Animales , Bendroflumetiazida/metabolismo , Bendroflumetiazida/farmacología , Sitios de Unión/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Proteínas Portadoras/genética , Cloruros/metabolismo , Cloruros/farmacología , Diuréticos , Hidroclorotiazida/metabolismo , Hidroclorotiazida/farmacología , Concentración de Iones de Hidrógeno , Concentración 50 Inhibidora , Cinética , Metolazona/metabolismo , Metolazona/farmacología , Microinyecciones , Modelos Biológicos , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Concentración Osmolar , Politiazida/metabolismo , Politiazida/farmacología , Ratas , Receptores de Droga/genética , Sodio/farmacología , Simportadores del Cloruro de Sodio , Miembro 3 de la Familia de Transportadores de Soluto 12 , Xenopus
13.
Am J Physiol Renal Physiol ; 278(4): F530-9, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10751213

RESUMEN

The amiloride-sensitive epithelial sodium channel (ENaC) and the vasopressin-dependent water channel aquaporin-2 (AQP2) mediate mineralocorticoid-regulated sodium- and vasopressin-regulated water reabsorption, respectively. Distributions of ENaC and AQP2 have been shown by immunohistochemistry in rats. Functional data from rabbits suggest a different distribution pattern of these channels than in rats. We studied, by immunohistochemistry in the rabbit kidney cortex, the distributions of ENaC and AQP2, in conjunction with marker proteins for distal segments. In rabbit cortex ENaC is restricted to the connecting tubule (CNT) cells and cortical collecting duct (CCD) cells. The intracellular distribution of ENaC shifts from the apical membrane in the most upstream CNT cells to a cytoplasmic location further downstream in the CNT and in the CCD cells. AQP2 is detected in the CCD cells exclusively. The anatomic subdivisions in the rabbit distal nephron coincide exactly with distributions of apical transport systems. The differences between rabbits and rats in the distribution patterns of ENaC and AQP2 may explain functional differences in renal salt and water handling between these species.


Asunto(s)
Acuaporinas/metabolismo , Proteínas Portadoras/efectos de los fármacos , Corteza Renal/metabolismo , Canales de Sodio/metabolismo , Animales , Acuaporina 2 , Acuaporina 6 , Benzotiadiazinas , Bumetanida/farmacología , Calbindinas , Proteínas Portadoras/metabolismo , Diuréticos/farmacología , Canales Epiteliales de Sodio , Femenino , Corteza Renal/anatomía & histología , Masculino , ATPasas de Translocación de Protón/metabolismo , Conejos , Ratas , Proteína G de Unión al Calcio S100/metabolismo , Inhibidores de los Simportadores del Cloruro de Sodio/farmacología , Simportadores de Cloruro de Sodio-Potasio , Distribución Tisular
14.
Am J Physiol Renal Physiol ; 278(4): F659-66, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10751228

RESUMEN

Recent studies showed that coexpression of Kir6.1 or Kir6.2 with the sulfonylurea receptor (SUR1, SUR2A, or SUR2B) reconstituted an inwardly rectifying, ATP-sensitive K(+) channel that was inhibited by glibenclamide (2, 15-17). Here we report the isolation of a rat homolog of mouse SUR2B (denoted rSUR2B) from a rat kidney cDNA library. The rSUR2B sequence contains a 4,635-bp open reading frame that encodes a 1,545-amino acid polypeptide, showing 67% shared identity with SUR1 (a pancreatic beta-cell isoform) and 98% with both SUR2A (a brain isoform) and SUR2B (a vascular smooth muscle isoform). Consistent with the predicted structures of other members of the ATP-binding cassette (ABC) superfamily, the sequence of rSUR2B contains 17 putative membrane-spanning segments. Also, predicted Walker A and B consensus binding motifs, present in other ABC members, are conserved in the rSUR2B sequence. RT-PCR revealed that rSUR2B is widely expressed in various rat tissues including brain, colon, heart, kidney, liver, skeletal muscle, and spleen. The intrarenal distribution of the rSUR2B transcript was investigated using RT-PCR and Southern blot of microdissected tubules. The rSUR2B transcript was detected in proximal tubule, cortical thick ascending limb, distal collecting tubule, cortical collecting duct, and outer medullary collecting duct, but not medullary thick ascending limb. This distal distribution overlaps with that of ROMK. Coexpression of rSUR2B with ROMK2 cRNA (in 1:10 ratio) in Xenopus laevis oocytes resulted in whole cell Ba(2+)-sensitive K(+) currents that were inhibited by glibenclamide (50% inhibition with 0.2 mM glibenclamide). In contrast, rSUR2B did not confer significant glibenclamide sensitivity to oocytes coinjected with ROMK1 or ROMK3. The interaction between ROMK2 and rSUR2B was further studied by coimmunoprecipitation of in vitro translated rSUR2B and ROMK2. In agreement with the functional data, the rSUR2B protein was coimmunoprecipitated with ROMK2 in the ROMK2-rSUR2B cotranslated samples. Our data demonstrate that ROMK2, but not ROMK1 and ROMK3, can interact with rSUR2B to confer a sulfonylurea-sensitive K(+) channel, implicating SUR proteins in forming and regulating renal ATP-sensitive K(+) channels. The ROMK isoform specificity of glibenclamide effects suggests that the NH(2) terminus of the ROMK protein mediates rSUR2B-ROMK2 interactions.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Gliburida/farmacología , Canales de Potasio de Rectificación Interna , Canales de Potasio/genética , Canales de Potasio/metabolismo , Canales de Potasio/fisiología , Receptores de Droga/genética , Receptores de Droga/fisiología , Secuencia de Aminoácidos , Animales , Femenino , Riñón/metabolismo , Datos de Secuencia Molecular , Oocitos/metabolismo , Isoformas de Proteínas/metabolismo , Ratas , Homología de Secuencia de Aminoácido , Receptores de Sulfonilureas , Transcripción Genética , Xenopus laevis
15.
Pflugers Arch ; 441(2-3): 379-87, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11211126

RESUMEN

Dietary phosphate (Pi) intake and parathyroid hormone (PTH) are essential regulators of proximal tubular (PT) Pi reabsorption; both factors are associated with adaptive changes in PT apical brush border membrane (BBM) Na/Pi-cotransport activity and specific transporter protein (NaPi-2) content. Urinary Pi excretion is also inversely correlated with luminal Ca2+ concentration ([Ca2+]) both in a PTH-dependent and -independent fashion. A cell-surface, Ca2+(/polyvalent cation)-sensing receptor (CaR) has been localized to the PT BBM with unknown function. To investigate whether PTH and/or dietary Pi intake could affect the distribution or the expression of the CaR, we evaluated their effects on rat kidney CaR and the NaPi-2 expression by Western blot analysis and immunofluorescence microscopy. A chronic high-Pi (1.2%) versus low-Pi (0.1%) diet and acute PTH (1-34) infusion significantly reduced the PT BBM expression of both NaPi-2 and CaR proteins. CaR-specific immunoreactivity in nephron segments other than the PT was not affected by PTH or Pi intake. These results suggest that reduced renal PT CaR expression by a high-Pi diet and by increased circulating PTH levels could contribute to the local control of PT handling of Ca2+ and Pi.


Asunto(s)
Proteínas Portadoras/análisis , Túbulos Renales Proximales/química , Hormona Paratiroidea/farmacología , Fosfatos/farmacología , Receptores de Superficie Celular/análisis , Sodio/farmacología , Animales , Western Blotting , Dieta , Técnica del Anticuerpo Fluorescente , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Masculino , Microscopía Fluorescente , Hormona Paratiroidea/sangre , Proteínas de Unión a Fosfato , Fosfatos/administración & dosificación , Fosfatos/metabolismo , Fosfatos/orina , Ratas , Ratas Wistar , Receptores Sensibles al Calcio
16.
Semin Nephrol ; 19(6): 504-23, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10598539

RESUMEN

The clinically useful and potent distal acting diuretics enhance urinary NaCl excretion by specific inhibition of distinct sodium transport processes in the loop of Henle and distal nephron. When these compounds were first used as diuretics little was known about their cellular mechanisms of action. Physiological investigations over the past 25 years, however, have shown that each class of diuretics inhibits a specific ion transport system in the kidney. Over the past few years, the molecular cloning of the distal diuretic-sensitive Na+ transporters has significantly enhanced our understanding of the mechanism of action of each class of diuretics and has clearly defined the specific protein (and its gene) that is the target for each of these diuretics. The identification of mutations in the genes encoding these transporters in inherited disorders characterized by alterations in salt balance has provided unequivocal evidence for roles of the cloned diuretic-sensitive transporters in sodium homeostasis. Many laboratories are actively engaged in defining the structural sites for ion transport and diuretic binding, and the molecular mechanisms of transport regulation. This information may enable the design of new diuretics and provide the basis for improved use of diuretics. This review will focus on this recent molecular information.


Asunto(s)
Túbulos Renales Distales/enzimología , Túbulos Renales Proximales/enzimología , Cloruro de Potasio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Benzotiadiazinas , Transporte Biológico , Diuréticos , Humanos , Túbulos Renales Distales/efectos de los fármacos , Túbulos Renales Proximales/efectos de los fármacos , Ratones , Biología Molecular , Conejos , Ratas , Sensibilidad y Especificidad , Inhibidores de los Simportadores del Cloruro de Sodio/farmacología , Inhibidores de los Simportadores del Cloruro de Sodio/uso terapéutico , ATPasa Intercambiadora de Sodio-Potasio/efectos de los fármacos
17.
Am J Physiol ; 276(3): F347-58, 1999 03.
Artículo en Inglés | MEDLINE | ID: mdl-10070158

RESUMEN

We have identified several alternatively spliced cDNAs encoding mBSC1, an apical bumetanide-sensitive Na+-K+-2Cl- cotransporter from mouse kidney. Two full-length clones were isolated, designated C4 and C9, predicting proteins of 770 and 1,095 amino acids, respectively. The C4 isoforms are generated by utilization of an alternative polyadenylation site located within the intron between exons 16 and 17 of the mBSC1 gene on chromosome 2; the resultant transcripts predict a truncated COOH terminus ending in a unique 55 amino acid sequence. The predicted C4 and C9 COOH termini differ in the distribution of putative phosphorylation sites for both protein kinase A and C. Independent splicing events involve three previously described cassette exons, which are predicted to encode most of the second transmembrane domain. A total of six different isoforms are expressed, generated by the combinatorial association of three cassette exons and two alternative 3' ends. C9-specific and C4-specific antibodies detect proteins of approximately 150 and 120 kDa, respectively, in mouse kidney. Immunofluorescence and immunohistochemistry indicate expression of both COOH-terminal isoforms within the thick ascending limb of the loop of Henle (TAL). However, staining with the C4 antibody is more heterogeneous, with a decreased proportion of positive cells in the cortical TAL. Functional expression in Xenopus oocytes indicates a dominant negative function for C4 isoforms [companion study, C. Plata, D. B. Mount, V. Rubio, S. C. Hebert, and G. Gamba. Am. J. Physiol. 276 (Renal Physiol. 45): F347-F358, 1999], and the differential expression of these isoforms may contribute to functional heterogeneity of Na+-K+-2Cl- cotransport in mouse TAL.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Asa de la Nefrona/metabolismo , Secuencia de Aminoácidos/genética , Animales , Mapeo Cromosómico , ADN Complementario/genética , Genoma , Isomerismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Datos de Secuencia Molecular , Simportadores de Cloruro de Sodio-Potasio , Distribución Tisular , Transcripción Genética/fisiología
18.
Am J Physiol ; 276(3): F359-66, 1999 03.
Artículo en Inglés | MEDLINE | ID: mdl-10070159

RESUMEN

The functional properties of alternatively spliced isoforms of the mouse apical Na+-K+-2Cl- cotransporter (mBSC1) were examined, using expression in Xenopus oocytes and measurement of 22Na+ or 86Rb+ uptake. A total of six isoforms, generated by the combinatorial association of three 5' exon cassettes (A, B, and F) with two alternative 3' ends, are expressed in mouse thick ascending limb (TAL) [see companion article, D. B. Mount, A. Baekgaard, A. E. Hall, C. Plata, J. Xu, D. R. Beier, G. Gamba, and S. C. Hebert. Am. J. Physiol. 276 (Renal Physiol. 45): F347-F358, 1999]. The two 3' ends predict COOH-terminal cytoplasmic domains of 129 amino acids (the C4 COOH terminus) and 457 amino acids (the C9 terminus). The three C9 isoforms (mBSC1-A9/F9/B9) all express Na+-K+-2Cl- cotransport activity, whereas C4 isoforms are nonfunctional in Xenopus oocytes. Activation or inhibition of protein kinase A (PKA) does not affect the activity of the C9 isoforms. The coinjection of mBSC1-A4 with mBSC1-F9 reduces tracer uptake, compared with mBSC1-F9 alone, an effect of C4 isoforms that is partially reversed by the addition of cAMP-IBMX to the uptake medium. The inhibitory effect of C4 isoforms is a dose-dependent function of the alternatively spliced COOH terminus. Isoforms with a C4 COOH terminus thus exert a dominant negative effect on Na+-K+-2Cl- cotransport, a property that is reversed by the activation of PKA. This interaction between coexpressed COOH-terminal isoforms of mBSC1 may account for the regulation of Na+-K+-2Cl- cotransport in the mouse TAL by hormones that generate cAMP.


Asunto(s)
Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/metabolismo , Asa de la Nefrona/metabolismo , Sulfonamidas , 1-Metil-3-Isobutilxantina/farmacología , Animales , Bumetanida/farmacología , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Femenino , Isomerismo , Isoquinolinas/farmacología , Ratones , Oocitos/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Simportadores de Cloruro de Sodio-Potasio , Xenopus laevis
20.
Gastroenterology ; 116(1): 118-26, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9869609

RESUMEN

BACKGROUND & AIMS: Circulating levels of Ca2+ can influence secretory functions and myoelectrical properties of the stomach. A Ca2+-sensing receptor (CaR) has recently been identified in tissues that regulate systemic Ca2+ homeostasis. The aim of this study was to evaluate expression of CaR in the stomach of the rat. METHODS: In forestomach and glandular stomach, reverse-transcription polymerase chain reaction was used to amplify a 380-base pair product, which is 99% homologous with transcripts obtained in parathyroid and kidney. RESULTS: Northern analysis of gastric mucosal polyA+ RNA revealed 7. 5- and 4.1-kilobase transcripts, similar to those obtained in rat parathyroid and kidney. Immunohistochemistry revealed CaR expression in regions of the submucosal plexus and myenteric neurons. In sections of intact tissue, preparations of primary culture surface cells and surgically dissected gastric glands, staining was observed consistently in epithelial cells of the gastric glands and in gastric surface cells. In parietal cells in isolated gastric glands, intracellular levels of Ca2+ responded to conditions that are known to activate CaR. CONCLUSIONS: These are the first reported observations that CaR is expressed in different epithelial cells of mammalian gastric mucosa and its enteric nerve regions. The effects of extracellular Ca2+ on gastric function may be attributable to activation of CaR.


Asunto(s)
Calcio/metabolismo , Espacio Extracelular/metabolismo , Mucosa Gástrica/metabolismo , Receptores de Superficie Celular/biosíntesis , Animales , Secuencia de Bases , Northern Blotting , Células Cultivadas , ADN Complementario/genética , Mucosa Gástrica/citología , Inmunohistoquímica , Datos de Secuencia Molecular , Músculo Liso/citología , Músculo Liso/metabolismo , Células Parietales Gástricas/metabolismo , ARN/biosíntesis , ARN/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Receptores Sensibles al Calcio , Receptores de Superficie Celular/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estómago/citología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA