Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nephrol Dial Transplant ; 28(4): 879-89, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23136218

RESUMEN

BACKGROUND: Cisplatin is an effective anti-neoplastic drug, but its clinical use is limited due to dose-dependent nephrotoxicity. The majority of cisplatin-treated patients develop hypomagnesaemia, often associated with a reduced glomerular filtration rate (GFR), polyuria and other electrolyte disturbances. The aim of this study is to unravel the molecular mechanism responsible for these particular electrolyte disturbances. METHODS: Two groups of 10 mice were injected intraperitoneally three times, once every 4 days, with cisplatin (5 mg/kg body weight,) or vehicle. Serum and urine electrolyte concentrations were determined. Next, renal mRNA levels of distal convoluted tubule (DCT) genes epithelial Mg(2+) channel TRPM6, the Na(+)-Cl(-) cotransporter (NCC), and parvalbumin (PV), as well as marker genes for other tubular segments were measured by real-time qPCR. Subsequently, renal protein levels of NCC, PV, aquaporin 1 and aquaporin 2 were determined using immunoblotting and immunohistochemistry (IHC). RESULTS: The cisplatin-treated mice developed significant polyuria (2.5 ± 0.3 and 0.9 ± 0.1 mL/24 h, cisplatin versus control, P < 0.05), reduced creatinine clearance rate (CCr) (0.18 ± 0.02 and 0.26 ± 0.02 mL/min, cisplatin versus control, P < 0.05) and a substantially reduced serum level of Mg(2+) (1.23 ± 0.03 and 1.58 ± 0.03 mmol/L, cisplatin versus control, P < 0.05), whereas serum Ca(2+), Na(+) and K(+) values were not altered. Measurements of 24 h urinary excretion demonstrated markedly increased Mg(2+), Ca(2+), Na(+) and K(+) levels in the cisplatin-treated group, whereas Pi levels were not changed. The mRNA levels of TRPM6, NCC and PV were significantly reduced in the cisplatin group. The expression levels of the marker genes for other tubular segments were unaltered, except for claudin-16, which was significantly up-regulated by the cisplatin treatment. The observed DCT-specific down-regulation was confirmed at the protein level. CONCLUSIONS: The present study identified the DCT as an important cisplatin-affected renal segment, explaining the high prevalence of hypomagnesaemia following treatment.


Asunto(s)
Antineoplásicos/toxicidad , Biomarcadores/metabolismo , Cisplatino/toxicidad , Enfermedades Renales/complicaciones , Túbulos Renales Distales/efectos de los fármacos , Deficiencia de Magnesio/etiología , Animales , Acuaporina 2/genética , Acuaporina 2/metabolismo , Western Blotting , Electrólitos/metabolismo , Femenino , Tasa de Filtración Glomerular , Técnicas para Inmunoenzimas , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/patología , Túbulos Renales Distales/lesiones , Deficiencia de Magnesio/diagnóstico , Deficiencia de Magnesio/metabolismo , Ratones , Ratones Endogámicos C57BL , Parvalbúminas/genética , Parvalbúminas/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Droga/genética , Receptores de Droga/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Miembro 3 de la Familia de Transportadores de Soluto 12 , Simportadores/genética , Simportadores/metabolismo , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo
2.
Nephrol Dial Transplant ; 27(11): 4061-8, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22778178

RESUMEN

BACKGROUND: Klotho(-/-) mice display disturbed Ca(2+) and vitamin D homeostasis. Renal cytochrome p450 27b1 (Cyp27b1), the enzyme that catalyzes the hydrolysis to 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)), is increased in klotho(-/-) mice, and a 1,25(OH)(2)D(3)-deficient diet partially normalized Ca(2+) homeostasis in these klotho(-/-) mice. The aim of the present study was to further delineate the interplay between 1,25(OH)(2)D(3) and klotho and their relative contribution to the Ca(2+) homeostasis of klotho(-/-) mice. METHODS: Double-klotho(-/-)/Cyp27b1(-/-) mice were generated and mice aged 8-12 weeks were housed in metabolic cages to collect 24-h urine. Blood samples were taken and the animals were sacrificed, and the kidney and duodenum tissues were sampled for RNA extraction. The bone was fixed in 10% v/v formalin and analysed by microcomputed tomography (µCT) scans. RESULTS: Klotho(-/-)/Cyp27b1(-/-) mice, like Cyp27b1(-/-) mice, displayed significantly decreased serum total calcium concentrations compared with wild-type mice (1.44 ± 0.03 and 2.25 ± 0.02 mM) along with normal urinary total calcium excretion. Hyperphosphataemia of klotho(-/-) mice normalized to wild-type levels in klotho(-/-)/Cyp27b1(-/-) mice. The mRNA levels of duodenal transient receptor potential vanilloid subtype 6 (TRPV6) and calcium-binding protein-D(9K), and renal calbindin-D(28K) and NCX1 were significantly reduced in the double knockouts compared with wild-type or klotho(-/-) mice. Elevated TRPV5 protein levels in klotho(-/-) mice normalized to wild type in klotho(-/-)/Cyp27b1(-/-) mice, but were decreased in Cyp27b1(-/-) mice. µCT scans showed that klotho(-/-)/Cyp27b1(-/-) mice, as Cyp27b1(-/-) mice, display significant bone hypomineralization and severely decreased bone mass. Klotho(-/-) mice show a reduced bone mass and increased trabecular numbers. CONCLUSIONS: Klotho(-/-)/Cyp27b1(-/-) mice resemble Cyp27b1(-/-) mice. Since 1,25(OH)(2)D(3) is absent in these mice, our results imply that Ca(2+) homeostasis in klotho(-/-) mice is affected by their excessive 1,25(OH)(2)D(3) levels.


Asunto(s)
Huesos/metabolismo , Calcio/sangre , Glucuronidasa/metabolismo , Fosfatos/sangre , Deficiencia de Vitamina D/sangre , Vitamina D/análogos & derivados , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , Animales , Western Blotting , Calcio/orina , Duodeno/metabolismo , Glucuronidasa/genética , Homeostasis , Inmunohistoquímica , Proteínas Klotho , Ratones , Ratones Noqueados , Fosfatos/orina , Reacción en Cadena en Tiempo Real de la Polimerasa , Canales Catiónicos TRPV/metabolismo , Vitamina D/sangre , Deficiencia de Vitamina D/orina
3.
Pflugers Arch ; 457(1): 91-101, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18392847

RESUMEN

The transient receptor potential vanilloid channels 5 and 6 (TRPV5/6) are the most Ca(2+)-selective channels within the TRP superfamily of ion channels. These epithelial Ca(2+) channels are regulated at different intra- and extracellular sites by the feedback response of Ca(2+) itself, calciotropic hormones, and by TRPV5/6-associated proteins. In the present study, bioinformatics was used to search for novel TRPV5/6-associated genes. By including pull-down assays and functional analysis, Nipsnap1-a hitherto functionally uncharacterized globular protein-was identified as a novel factor involved in the regulation of TRPV6. Electrophysiological recordings revealed that Nipsnap1 abolishes TRPV6 currents. Subsequent biotinylation assays showed that TRPV6 plasma membrane expression did not change in the presence of Nipsnap1, suggesting that TRPV6 inhibition by Nipsnap1 is independently regulated from reduced cell surface channel expression. In addition, semi-quantitative reverse transcriptase PCR and immunohistochemical labeling of Nipsnap1 indicated that Nipsnap1 is expressed in mouse intestinal tissues-where TRPV6 is predominantly expressed-but that it does not co-localize with TRPV5 in the kidney. In conclusion, this study presents the first physiological function of Nipsnap1 as an associated protein inhibiting TRPV6 activity that possibly exerts its effect directly at the plasma membrane.


Asunto(s)
Canales de Calcio/fisiología , Proteínas/fisiología , Canales Catiónicos TRPV/fisiología , Secuencia de Aminoácidos , Canales de Calcio/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Biología Computacional , Electrofisiología , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Unión Proteica , Proteínas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales Catiónicos TRPV/genética , Distribución Tisular
4.
Am J Physiol Renal Physiol ; 291(6): F1177-83, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16705151

RESUMEN

Aging is associated with alterations in Ca2+ homeostasis, which predisposes elder people to hyperparathyroidism and osteoporosis. Intestinal Ca2+ absorption decreases with aging and, in particular, active transport of Ca2+ by the duodenum. In addition, there are age-related changes in renal Ca2+ handling. To examine age-related changes in expression of the renal and intestinal epithelial Ca2+ channels, control (TRPV5+/+) and TRPV5 knockout (TRPV5-/-) mice aged 10, 30, and 52 wk were studied. Aging of TRPV5(+/+) mice resulted in a tendency toward increased renal Ca2+ excretion and significantly decreased intestinal Ca2+ absorption, which was accompanied by reduced expression of TRPV5 and TRPV6, respectively, despite increased serum 1,25(OH)2D3 levels. Similarly, in TRPV5-/- mice the existing renal Ca2+ loss was more pronounced in elder animals, whereas the compensatory intestinal Ca2+ absorption and TRPV6 expression declined with aging. In both mice strains, aging resulted in a resistance to 1,25(OH)2D3 and diminished renal vitamin D receptor mRNA levels, whereas serum Ca2+ levels remained constant. Furthermore, 52-wk-old TRPV5-/- mice showed severe hyperparathyroidism, whereas PTH levels in elder TRPV5+/+ mice remained normal. In 52-wk-old TRPV5-/- mice, serum osteocalcin levels were increased in accordance with the elevated PTH levels, suggesting an increased bone turnover in these mice. In conclusion, downregulation of TRPV5 and TRPV6 is likely involved in the impaired Ca2+ (re)absorption during aging. Moreover, TRPV5-/- mice likely develop age-related hyperparathyroidism and osteoporotic characteristics before TRPV5+/+ mice, demonstrating the importance of the epithelial Ca2+ channels in Ca2+ homeostasis.


Asunto(s)
Envejecimiento/fisiología , Canales de Calcio/metabolismo , Calcio/sangre , Osteoporosis/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Calcio/orina , Canales de Calcio/genética , Duodeno/metabolismo , Células Epiteliales/metabolismo , Homeostasis/fisiología , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo/fisiopatología , Riñón/metabolismo , Ratones , Ratones Mutantes , Osteocalcina/sangre , Osteoporosis/fisiopatología , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Canales Catiónicos TRPV/genética
5.
Pflugers Arch ; 452(4): 407-17, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16565876

RESUMEN

The epithelial Ca(2+) channels TRPV5 and TRPV6 constitute the apical Ca(2+) influx pathway in epithelial Ca(2+) transport. PDZ proteins have been demonstrated to play a crucial role in the targeting or anchoring of ion channels and transporters in the apical domain of the cell. In this study, we describe the identification of NHERF4 (Na-P(i) Cap2/IKEPP/PDZK2) as a novel TRPV5- and TRPV6-associated PDZ protein. NHERF4 was identified using two separate yeast two-hybrid screens with the carboxyl termini of TRPV5 and TRPV6 as bait. Binding of the carboxyl termini of TRPV5 and TRPV6 with NHERF4 was confirmed by GST pull-down assays using in-vitro-translated NHERF4 or lysates of Xenopus laevis oocytes expressing NHERF4. Furthermore, the interaction was confirmed by GST pull-down and co-immunoprecipitation assays using in-vitro-translated full-length TRPV5 and Xenopus oocytes or HEK293 cells co-expressing NHERF4 and TRPV5/TRPV6, respectively. The fourth PDZ domain of NHERF4 was sufficient for the interaction, although PDZ domain 1 also contributed to the binding. The binding site for NHERF4 localized in a conserved region in the carboxyl terminus of TRPV5 and was distinct from the binding site of the PDZ protein NHERF2. NHERF4 predominantly localized at the plasma membrane of X. laevis oocytes and HeLa cells. This localization was independent of the presence of TRPV5. Therefore, we hypothesize a role for this novel PDZ protein as a putative plasma membrane scaffold for the epithelial Ca(2+) channels.


Asunto(s)
Canales de Calcio/metabolismo , Membrana Celular/metabolismo , Mucosa Intestinal/metabolismo , Riñón/metabolismo , Oocitos/fisiología , Canales Catiónicos TRPV/metabolismo , Animales , Canales de Calcio/fisiología , Células Cultivadas , Activación del Canal Iónico , Ratones , Fosfoproteínas , Mapeo de Interacción de Proteínas , Intercambiadores de Sodio-Hidrógeno , Xenopus laevis
6.
J Am Soc Nephrol ; 17(1): 26-30, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16380433

RESUMEN

Transient receptor potential vallinoid 5 (TRPV5) and TRPV6 are the most Ca2+-selective members of the TRP superfamily and are essential for active Ca2+ (re)absorption in epithelia. However, little is known about intracellular proteins that regulate the activity of these channels. This study identified BSPRY (B-box and SPRY-domain containing protein) as a novel factor involved in the control of TRPV5. The interaction between BSPRY and TRPV5 by GST pull-down and co-immunoprecipitation assays was demonstrated. BSPRY showed co-localization with TRPV5 in mouse kidney. Expression of BSPRY resulted in a significant reduction of the Ca2+ influx in Madin-Darby Canine Kidney cells that stably express TRPV5 without affecting channel cell-surface abundance. Finally, BSPRY expression in kidney was increased in 25-hydroxyvitamin D3-1alpha-hydroxylase knockout mice, suggesting an inverse regulation by vitamin D3. Together, these results demonstrate the physiologic role of the novel protein BSPRY in the regulation of epithelial Ca2+ transport via negative modulation of TRPV5 activity.


Asunto(s)
Proteínas/fisiología , Canales Catiónicos TRPV/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Calcitriol/fisiología , Calcio/metabolismo , Canales de Calcio/análisis , Canales de Calcio/fisiología , Línea Celular , Datos de Secuencia Molecular , Proteínas/análisis , Proteínas/química , Canales Catiónicos TRPV/análisis , Canales Catiónicos TRPV/fisiología , Xenopus
7.
Kidney Int ; 68(4): 1708-21, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16164647

RESUMEN

BACKGROUND: The kidney is one of the affected organs involved in the clinical symptoms of parathyroid hormone (PTH)-related disorders, like primary hyperparathyroidism and familial hypocalciuric hypercalcemia. The molecular mechanism(s) underlying alterations in renal Ca(2+) handling in these disorders is poorly understood. METHODS: Parathyroidectomized and PTH-supplemented rats and mice infused with the calcimimetic compound NPS R-467 were used to study the in vivo effect of PTH on the expression of renal transcellular Ca(2+) transport proteins, including the epithelial Ca(2+) channel transient receptor potential, vanilloid, member 5 (TRPV5), calbindins, and the Na(+)/Ca(2+)-exchanger (NCX1). In addition, the effect of PTH on transepithelial Ca(2+) transport in rabbit connecting tubule/cortical collecting duct (CNT/CCD) primary cultures was determined. RESULTS: Decreased PTH levels in parathyroidectomized rats or NPS R-467-infused mice, resulted in reduced expression of these proteins, which is consistent with diminished Ca(2+) reabsorption, causing the development of the observed hypocalcemia. PTH supplementation of parathyroidectomized rats restored the expression of the renal Ca(2+) transport machinery and serum Ca(2+) levels, independent of serum 1,25-dihydroxyvitamin D(3) levels and renal vitamin D or Ca(2+)-sensing receptor mRNA abundance. Inhibition of the PTH-stimulated transepithelial Ca(2+) transport by the TRPV5-specific inhibitor ruthenium red reduced the PTH-stimulated expression of calbindin-D(28K) and NCX1 in rabbit CNT/CCD primary cultures. CONCLUSION: PTH stimulates renal Ca(2+) reabsorption through the coordinated expression of renal transcellular Ca(2+) transport proteins. Moreover, the PTH-induced stimulation is enhanced by the magnitude of the Ca(2+) influx through the gatekeeper TRPV5, which in turn facilitates the expression of the downstream Ca(2+) transport proteins. Therefore, the renal transcellular Ca(2+) transport proteins, including TRPV5, could contribute to the pathogenesis of PTH-related disorders.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Riñón/fisiología , Hormona Paratiroidea/fisiología , Canales Catiónicos TRPV/metabolismo , Compuestos de Anilina/farmacología , Animales , Calbindinas , Calcio/agonistas , Canales de Calcio/genética , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Expresión Génica , Hipocalcemia/tratamiento farmacológico , Hipocalcemia/metabolismo , Hipocalcemia/fisiopatología , Riñón/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Hormona Paratiroidea/sangre , Hormona Paratiroidea/farmacología , Paratiroidectomía , Conejos , Ratas , Ratas Endogámicas , Receptores Sensibles al Calcio/genética , Receptores Sensibles al Calcio/metabolismo , Proteína G de Unión al Calcio S100/genética , Proteína G de Unión al Calcio S100/metabolismo , Intercambiador de Sodio-Calcio/genética , Intercambiador de Sodio-Calcio/metabolismo , Canales Catiónicos TRPV/genética
8.
J Am Soc Nephrol ; 16(11): 3188-95, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16148038

RESUMEN

Vitamin D plays an important role in Ca(2+) homeostasis by controlling Ca(2+) (re)absorption in intestine, kidney, and bone. The epithelial Ca(2+) channel TRPV5 mediates the Ca(2+) entry step in active Ca(2+) reabsorption. TRPV5 knockout (TRPV5(-/-)) mice show impaired Ca(2+) reabsorption, hypercalciuria, hypervitaminosis D, and intestinal hyperabsorption of Ca(2+). Moreover, these mice demonstrate upregulation of intestinal TRPV6 and calbindin-D(9K) expression compared with wild-type mice. For addressing the role of the observed hypervitaminosis D in the maintenance of Ca(2+) homeostasis and the regulation of expression levels of the Ca(2+) transport proteins in kidney and intestine, TRPV5/25-hydroxyvitamin-D(3)-1alpha-hydroxylase double knockout (TRPV5(-/-)/1alpha-OHase(-/-)) mice, which show undetectable serum 1,25(OH)(2)D(3) levels, were generated. TRPV5(-/-)/1alpha-OHase(-/-) mice displayed a significant hypocalcemia compared with wild-type mice (1.10 +/- 0.02 and 2.54 +/- 0.01 mM, respectively; P < 0.05). mRNA levels of renal calbindin-D(28K) (7 +/- 2%), calbindin-D(9K) (32 +/- 4%), Na(+)/Ca(2+) exchanger (12 +/- 2%), and intestinal TRPV6 (40 +/- 8%) and calbindin-D(9K) (26 +/- 4%) expression levels were decreased compared with wild-type mice. Hyperparathyroidism and rickets were present in TRPV5(-/-)/1alpha-OHase(-/-) mice, more pronounced than observed in single TRPV5 or 1alpha-OHase knockout mice. It is interesting that a renal Ca(2+) leak, as demonstrated in TRPV5(-/-) mice, persisted in TRPV5(-/-)/1alpha-OHase(-/-) mice, but a compensatory upregulation of intestinal Ca(2+) transporters was abolished. In conclusion, the elevation of serum 1,25(OH)(2)D(3) levels in TRPV5(-/-) mice is responsible for the upregulation of intestinal Ca(2+) transporters and Ca(2+) hyperabsorption. Hypervitaminosis D, therefore, is of crucial importance to maintain normocalcemia in impaired Ca(2+) reabsorption in TRPV5(-/-) mice.


Asunto(s)
Canales de Calcio/deficiencia , Canales de Calcio/fisiología , Calcio/metabolismo , Enfermedades Metabólicas/metabolismo , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/fisiología , Vitamina D/metabolismo , Animales , Transporte Biológico , Calcio/sangre , Calcio/orina , Canales de Calcio/genética , Cruzamientos Genéticos , Femenino , Homeostasis , Riñón/fisiología , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Canales Catiónicos TRPV/genética
9.
J Clin Invest ; 115(6): 1651-8, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15902302

RESUMEN

Thiazide diuretics enhance renal Na+ excretion by blocking the Na+-Cl- cotransporter (NCC), and mutations in NCC result in Gitelman syndrome. The mechanisms underlying the accompanying hypocalciuria and hypomagnesemia remain debated. Here, we show that enhanced passive Ca2+ transport in the proximal tubule rather than active Ca2+ transport in distal convolution explains thiazide-induced hypocalciuria. First, micropuncture experiments in mice demonstrated increased reabsorption of Na+ and Ca2+ in the proximal tubule during chronic hydrochlorothiazide (HCTZ) treatment, whereas Ca2+ reabsorption in distal convolution appeared unaffected. Second, HCTZ administration still induced hypocalciuria in transient receptor potential channel subfamily V, member 5-knockout (Trpv5-knockout) mice, in which active distal Ca2+ reabsorption is abolished due to inactivation of the epithelial Ca2+ channel Trpv5. Third, HCTZ upregulated the Na+/H+ exchanger, responsible for the majority of Na+ and, consequently, Ca2+ reabsorption in the proximal tubule, while the expression of proteins involved in active Ca2+ transport was unaltered. Fourth, experiments addressing the time-dependent effect of a single dose of HCTZ showed that the development of hypocalciuria parallels a compensatory increase in Na+ reabsorption secondary to an initial natriuresis. Hypomagnesemia developed during chronic HCTZ administration and in NCC-knockout mice, an animal model of Gitelman syndrome, accompanied by downregulation of the epithelial Mg2+ channel transient receptor potential channel subfamily M, member 6 (Trpm6). Thus, Trpm6 downregulation may represent a general mechanism involved in the pathogenesis of hypomagnesemia accompanying NCC inhibition or inactivation.


Asunto(s)
Calcio/metabolismo , Túbulos Renales Proximales/metabolismo , Magnesio/metabolismo , Defectos Congénitos del Transporte Tubular Renal/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Desequilibrio Hidroelectrolítico/metabolismo , Animales , Benzotiadiazinas , Canales de Calcio/genética , Canales de Calcio/metabolismo , Trastornos del Metabolismo del Calcio/inducido químicamente , Trastornos del Metabolismo del Calcio/metabolismo , Trastornos del Metabolismo del Calcio/patología , Modelos Animales de Enfermedad , Diuréticos , Regulación hacia Abajo , Humanos , Transporte Iónico/genética , Túbulos Renales Proximales/patología , Ratones , Ratones Noqueados , Defectos Congénitos del Transporte Tubular Renal/genética , Defectos Congénitos del Transporte Tubular Renal/patología , Inhibidores de los Simportadores del Cloruro de Sodio/toxicidad , Intercambiadores de Sodio-Hidrógeno/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo , Simportadores de Cloruro de Sodio-Potasio/genética , Canales Catiónicos TRPV , Regulación hacia Arriba , Desequilibrio Hidroelectrolítico/inducido químicamente , Desequilibrio Hidroelectrolítico/patología
10.
Kidney Int ; 66(3): 1082-9, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15327402

RESUMEN

BACKGROUND: Vitamin D compounds are used clinically to control secondary hyperparathyroidism (SHPT) due to renal failure. Newer vitamin D compounds retain the suppressive action of 1,25(OH)(2)D(3) on the parathyroid glands and may have less Ca(2+)-mobilizing activity, offering potentially safer therapies. METHODS: This study investigated the effect of a single dose of compound (1,25(OH)(2)D(3), 1,24(OH)(2)D(2), or 1alpha(OH)D(2)) on renal and intestinal Ca(2+) transport proteins, including TRPV5 and TRPV6, and serum Ca(2+), in a novel SHPT model, the 25-OH-D(3)-1alpha-hydroxylase knockout mouse, which lacks endogenous 1,25(OH)(2)D(3) and is severely hypocalcemic. Animals were injected intraperitoneally with compound (100 ng/mouse). RESULTS: Serum levels of 1,25(OH)(2)D(3) and 1,24(OH)(2)D(2) peaked at four hours post-injection (pi), then declined rapidly. 1,25(OH)(2)D(2) generated from 1alpha(OH)D(2) peaked at 12 hours pi and then remained stable. Serum Ca(2+) was increased to near-normal within four hours by 1,25(OH)(2)D(3) and 1,24(OH)(2)D(2), and within 12 hours by 1alpha(OH)D(2). 1,25(OH)(2)D(3) and 1,24(OH)(2)D(2) up-regulated duodenal TRPV5 and TRPV6 mRNA to a similar degree within four hours; mRNA levels decreased by 12 hours after 1,24(OH)(2)D(2) treatment, and by 24 hours after 1,25(OH)(2)D(3) treatment. 1,25(OH)(2)D(3) increased kidney levels of TRPV5, calbindin-D(28K), and calbindin-D(9K) mRNA within four hours; 1,24(OH)(2)D(2) did not change kidney TRPV5 levels and modestly increased calbindin D(9K) by 48 hours. 1alpha(OH)D(2) produced later-onset effects, increasing duodenal TRPV6 and calbindin-D(9K) mRNA levels by 12 hours and TRPV5 by 48 hours. CONCLUSION: In kidney, 1alpha(OH)D(2) increased TRPV5, calbindin-D(28K), and calbindin-D(9K) mRNA levels by 12 hours. This study indicates that Ca(2+) transport proteins, including TRPV5 and TRPV6, are differentially up-regulated by vitamin D compounds.


Asunto(s)
25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , Calcitriol/farmacología , Agonistas de los Canales de Calcio/farmacología , Mucosa Intestinal/metabolismo , Riñón/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Calbindinas , Calcio/sangre , Canales de Calcio/genética , Canales de Calcio/metabolismo , Ergocalciferoles/farmacología , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Proteína G de Unión al Calcio S100/genética , Proteína G de Unión al Calcio S100/metabolismo , Canales Catiónicos TRPV , Regulación hacia Arriba/efectos de los fármacos , Vitamina D/metabolismo
11.
J Biol Chem ; 279(1): 19-25, 2004 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-14576148

RESUMEN

Mg2+ is an essential ion involved in a multitude of physiological and biochemical processes and a major constituent of bone tissue. Mg2+ homeostasis in mammals depends on the equilibrium between intestinal Mg2+ absorption and renal Mg2+ excretion, but little is known about the molecular nature of the proteins involved in the transepithelial transport of Mg2+ in these organs. Recently, it was shown that patients with mutations in TRPM6, a member of the transient receptor potential family of cation channels, suffer from hypomagnesemia with secondary hypocalcemia (HSH) as a result of impaired renal and/or intestinal Mg2+ handling. Here, we show that TRPM6 is specifically localized along the apical membrane of the renal distal convoluted tubule and the brush-border membrane of the small intestine, epithelia particularly associated with active Mg2+ (re)absorption. In kidney, parvalbumin and calbindin-D28K, two divalent-binding proteins, are co-expressed with TRPM6 and might function as intracellular Mg2+ buffers in the distal convoluted tubule. Heterologous expression of wild-type TRPM6 but not TRPM6 mutants identified in HSH patients induces a Mg2+- and Ca2+-permeable cation channel tightly regulated by intracellular Mg2+ levels. The TRPM6-induced channel displays strong outward rectification, has a 5-fold higher affinity for Mg2+ than for Ca2+, and is blocked in a voltage-dependent manner by ruthenium red. Our data indicate that TRPM6 comprises all or part of the apical Mg2+ channel of Mg2+-absorbing epithelia.


Asunto(s)
Absorción Intestinal , Canales Iónicos/metabolismo , Riñón/fisiología , Deficiencia de Magnesio/genética , Magnesio/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico , Línea Celular , Membrana Celular/metabolismo , Humanos , Hipocalcemia/genética , Mucosa Intestinal/metabolismo , Canales Iónicos/química , Túbulos Renales Distales/metabolismo , Ratones , Ratones Endogámicos C57BL , Microvellosidades/metabolismo , Datos de Secuencia Molecular , Nefronas/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Proteínas Recombinantes/metabolismo , Canales Catiónicos TRPM , Transfección
12.
J Clin Invest ; 112(12): 1906-14, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14679186

RESUMEN

Ca2+ ions play a fundamental role in many cellular processes, and the extracellular concentration of Ca2+ is kept under strict control to allow the proper physiological functions to take place. The kidney, small intestine, and bone determine the Ca2+ flux to the extracellular Ca2+ pool in a concerted fashion. Transient receptor potential (TRP) cation channel subfamily V, members 5 and 6 (TRPV5 and TRPV6) have recently been postulated to be the molecular gatekeepers facilitating Ca2+ influx in these tissues and are members of the TRP family, which mediates diverse biological effects ranging from pain perception to male aggression. Genetic ablation of TRPV5 in the mouse allowed us to investigate the function of this novel Ca2+ channel in maintaining the Ca2+ balance. Here, we demonstrate that mice lacking TRPV5 display diminished active Ca2+ reabsorption despite enhanced vitamin D levels, causing severe hypercalciuria. In vivo micropuncture experiments demonstrated that Ca2+ reabsorption was malfunctioning within the early part of the distal convolution, exactly where TRPV5 is localized. In addition, compensatory hyperabsorption of dietary Ca2+ was measured in TRPV5 knockout mice. Furthermore, the knockout mice exhibited significant disturbances in bone structure, including reduced trabecular and cortical bone thickness. These data demonstrate the key function of TRPV5 in active Ca2+ reabsorption and its essential role in the Ca2+ homeostasis.


Asunto(s)
Huesos/fisiología , Canales de Calcio/genética , Canales de Calcio/metabolismo , Canales de Calcio/fisiología , Calcio/metabolismo , Riñón/metabolismo , Absorción , Animales , Señalización del Calcio , Cationes , Femenino , Fémur/metabolismo , Biblioteca de Genes , Genotipo , Inmunohistoquímica , Iones , Enfermedades Renales/metabolismo , Masculino , Ratones , Ratones Noqueados , Modelos Genéticos , Osteoporosis , Fenotipo , Potasio/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sodio/metabolismo , Canales Catiónicos TRPV , Factores de Tiempo
13.
J Am Soc Nephrol ; 14(11): 2731-40, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14569082

RESUMEN

The family of epithelial Ca(2+) channels consists of two highly homologues members, TRPV5 and TRPV6, which constitute the apical Ca(2+) entry mechanism in active Ca(2+) (re)absorption in kidney and small intestine. In kidney, TRPV5 expression has been extensively studied, whereas TRPV6 localization and regulation has been largely confined to the small intestine. The present study investigated the renal distribution of TRPV6 and regulation by 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)). In mouse kidney, TRPV6 was detected by immunohistochemistry at the apical domain of the distal convoluted tubules (DCT2), connecting tubules (CNT), and cortical and medullary collecting ducts (CD). Furthermore, several putative vitamin D-responsive elements were detected upstream of the mouse TRPV6 start codon, and 1,25(OH)(2)D(3) treatment significantly increased renal TRPV6 mRNA and protein expression. In DCT2 and CNT, TRPV6 co-localizes with the other known Ca(2+) transport proteins, including TRPV5 and calbindin-D(28K). Together, these data suggest a role for TRPV6 in 1,25(OH)(2)D(3)-stimulated Ca(2+) reabsorption in these segments. Interestingly, distribution of TRPV6 extended to the CD, where it localized to the apical domain of principal and intercalated cells, which are not generally implicated in active Ca(2+) reabsorption. In addition, TRPV6 mRNA levels were quantified in a large set of tissues, and in the order of decreasing expression level were detected: prostate > stomach, brain > lung > duodenum, kidney, bone, cecum, heart > colon > skeletal muscle > pancreas. Therefore, additional physiologic functions for TRPV6 are feasible. In conclusion, TRPV6 is expressed along the apical domain of DCT2, CNT, and CD, where TRPV6 expression is positively regulated by 1,25(OH)(2)D(3).


Asunto(s)
Calcitriol/farmacología , Agonistas de los Canales de Calcio/farmacología , Canales de Calcio/metabolismo , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Animales , Canales de Calcio/genética , Ratones , Ratones Endogámicos C57BL , Modelos Animales , ARN Mensajero/genética , Fracciones Subcelulares/metabolismo , Canales Catiónicos TRPV
14.
Kidney Int ; 64(2): 555-64, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12846750

RESUMEN

INTRODUCTION: Thiazide diuretics have the unique characteristic of increasing renal Na+ excretion, while decreasing Ca2+ excretion. However, the molecular mechanism responsible for this thiazide-induced hypocalciuria remains unclear. The present study investigates the effect of thiazides on the expression of the proteins involved in active Ca2+ transport as well as the role of extracellular volume (ECV) status. METHODS: Hydrochlorothiazide (HCTZ), 12 mg/24 hours, was administered during 7 days to Wistar rats by osmotic minipumps. In addition, ECV contraction was either prevented by Na+ repletion or induced by a low-salt diet. Expression levels of the proteins involved in active Ca2+ transport [i.e., epithelial Ca2+ channel (TRPV5/ECaC1), calbindin-D28K, Na+/Ca2+ exchanger (NCX1)], as well as the thiazide-sensitive Na+ Cl- cotransporter (NCC) were determined by real-time quantitative polymerase chain reaction (PCR) and semiquantitative immunohistochemistry. RESULTS: HCTZ significantly reduced urinary Ca2+ excretion (22%+/- 5% relative to controls). Hematocrit was significantly increased, confirming ECV contraction. In addition, Na+ depletion virtually abolished Ca2+ excretion (8%+/- 1%), while Na+ repletion during HCTZ treatment prevented both ECV contraction and hypocalciuria. HCTZ significantly decreased mRNA expression of TRPV5 (71%+/- 6%), calbindin-D28K (53%+/- 6%), NCX1 (51%+/- 8%) and NCC (50%+/- 11%), regardless of ECV status or calciuresis. Immunohistochemistry revealed reduced TRPV5 (43%+/- 2%), calbindin-D28K (59%+/- 1%) and NCC (56%+/- 4%) abundance. Furthermore, during HCTZ treatment, the subset of tubules coexpressing NCC and calbindin-D28K was significantly reduced (43%+/- 5%) and a disturbed cellular localization of NCC was observed. CONCLUSION: These data suggest that ECV contraction is a critical determinant of the thiazide-induced hypocalciuria, which is accompanied by a decreased expression of Ca2+ transport proteins.


Asunto(s)
Canales de Calcio/genética , Calcio/orina , Hidroclorotiazida/farmacología , Inhibidores de los Simportadores del Cloruro de Sodio/farmacología , Intercambiador de Sodio-Calcio/genética , Animales , Canales de Calcio/metabolismo , Diuréticos , Electrólitos/sangre , Electrólitos/orina , Immunoblotting , Inmunohistoquímica , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Reacción en Cadena de la Polimerasa , Ratas , Ratas Wistar , Sodio/orina , Simportadores del Cloruro de Sodio , Intercambiador de Sodio-Calcio/metabolismo , Simportadores/genética , Simportadores/metabolismo , Canales Catiónicos TRPV
15.
Am J Physiol Gastrointest Liver Physiol ; 285(1): G78-85, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12620887

RESUMEN

The epithelial Ca2+ channels TRPV5 and TRPV6 are localized to the brush border membrane of intestinal cells and constitute the postulated rate-limiting entry step of active Ca2+ absorption. The aim of the present study was to investigate the hormonal regulation of these channels. To this end, the effect of 17beta-estradiol (17beta-E2), 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], and dietary Ca2+ on the expression of the duodenal Ca2+ transport proteins was investigated in vivo and analyzed using realtime quantitative PCR. Supplementation with 17beta-E2 increased duodenal gene expression of TRPV5 and TRPV6 but also calbindin-D9K and plasma membrane Ca2+-ATPase (PMCA1b) in ovariectomized rats. 25-Hydroxyvitamin D3-1alpha-hydroxylase (1alpha-OHase) knockout mice are characterized by hyperparathyroidism, rickets, hypocalcemia, and undetectable levels of 1,25(OH)2D3 and were used to study the 1,25(OH)2D3-dependency of the stimulatory effects of 17beta-E2. Treatment with 17beta-E2 upregulated mRNA levels of duodenal TRPV6 in these 1alpha-OHase knockout mice, which was accompanied by increased serum Ca2+ concentrations from 1.69 +/- 0.10 to 2.03 +/- 0.12 mM (P < 0.05). In addition, high dietary Ca2+ intake normalized serum Ca2+ in these mice and upregulated expression of genes encoding the duodenal Ca2+ transport proteins except for PMCA1b. Supplementation with 1,25(OH)2D3 resulted in increased expression of TRPV6, calbindin-D9K, and PMCA1b and normalization of serum Ca2+. Expression levels of duodenal TRPV5 mRNA are below detection limits in these 1alpha-OHase knockout mice, but supplementation with 1,25(OH)2D3 upregulated the expression to significant levels. In conclusion, TRPV5 and TRPV6 are regulated by 17beta-E2 and 1,25(OH)2D3, whereas dietary Ca2+ is positively involved in the regulation of TRPV6 only.


Asunto(s)
Calcitriol/genética , Canales de Calcio/genética , Canales de Calcio/metabolismo , Mucosa Intestinal/metabolismo , Animales , Calcitriol/farmacología , Calcio/metabolismo , Calcio de la Dieta/farmacología , Duodeno/citología , Duodeno/metabolismo , Estradiol/sangre , Estradiol/farmacología , Femenino , Expresión Génica/fisiología , Mucosa Intestinal/citología , Ratones , Ratones Noqueados , Ovariectomía , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , Ratas , Ratas Wistar , Canales Catiónicos TRPV
16.
FASEB J ; 16(11): 1398-406, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12205031

RESUMEN

Pseudovitamin D-deficiency rickets (PDDR) is an autosomal disease characterized by hyperparathyroidism, rickets, and undetectable levels of 1,25-dihydroxyvitaminD3 (1,25(OH)2D3). Mice in which the 25-hydroxyvitamin D3-1alpha-hydroxylase (1alpha-OHase) gene was inactivated presented the same clinical phenotype as patients with PDDR and were used to study renal expression of the epithelial Ca2+ channel (ECaC1), the calbindins, Na+/Ca2+ exchanger (NCX1), and Ca2+-ATPase (PMCA1b). Serum Ca2+ (1.20+/-0.05 mM) and mRNA/protein expression of ECaC1 (41+/-3%), calbindin-D28K (31+/-2%), calbindin-D9K (58+/-7%), NCX1 (10+/-2%), PMCA1b (96+/-4%) were decreased in 1alpha-OHase-/- mice compared with 1alpha-OHase+/- littermates. Feeding these mice a Ca2+-enriched diet normalized serum Ca2+ levels and expression of Ca2+ proteins except for calbindin-D9K expression. 1,25(OH)2D3 repletion resulted in increased expression of Ca2+ transport proteins and normalization of serum Ca2+ levels. Localization of Ca2+ transport proteins was clearly polarized in which ECaC1 was localized along the apical membrane, calbindin-D28K in the cytoplasm, and calbindin-D9K along the apical and basolateral membranes, resulting in a comprehensive mechanism facilitating renal transcellular Ca2+ transport. This study demonstrated that high dietary Ca2+ intake is an important regulator of the renal Ca2+ transport proteins in 1,25(OH)2D3-deficient status and thus contributes to the normalization of blood Ca2+ levels.


Asunto(s)
25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , Calcitriol/farmacología , Proteínas de Unión al Calcio/biosíntesis , Calcio/farmacología , Riñón/metabolismo , Administración Oral , Animales , Calbindina 1 , Calbindinas , Calcitriol/administración & dosificación , Calcio/administración & dosificación , Calcio/sangre , Canales de Calcio/análisis , Proteínas de Unión al Calcio/análisis , Proteínas de Unión al Calcio/genética , ATPasas Transportadoras de Calcio/biosíntesis , ATPasas Transportadoras de Calcio/genética , Proteínas de Transporte de Catión , Riñón/química , Ratones , Ratones Noqueados , ATPasas Transportadoras de Calcio de la Membrana Plasmática , ARN Mensajero/biosíntesis , Raquitismo/genética , Raquitismo/metabolismo , Proteína G de Unión al Calcio S100/biosíntesis , Proteína G de Unión al Calcio S100/genética , Intercambiador de Sodio-Calcio/biosíntesis , Intercambiador de Sodio-Calcio/genética , Canales Catiónicos TRPV , Regulación hacia Arriba , Deficiencia de Vitamina D/genética , Deficiencia de Vitamina D/metabolismo
17.
Pflugers Arch ; 443(3): 466-72, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11810218

RESUMEN

Bartter syndrome is an autosomal recessive heterogeneous renal tubular disorder affecting NaCl reabsorption in the thick ascending limb of Henle's loop (TAL). The aim of this study was to elucidate the functional implications of mutations in the predominant human ROMK isoform in TAL, hROMK2, involved in Bartter syndrome type II. cRNA of flag-tagged hROMK2 and eight mutants identified in seven non-related patients was expressed in Xenopus laevis oocytes. hROMK2 activity was measured by two-electrode voltage-clamp analysis and defined as the Ba2+ -sensitive current at a holding potential of -75 mV. The subcellular localization of hROMK2 in oocytes was studied by immunocytochemistry. Injection of 25 pg hROMK2 cRNA resulted in an inwardly rectifying Ba2+ -sensitive current of 522+/-43 nA ( n=22). The mutants could be divided into three distinct groups. First, at 25 pg injection mutants W80C, V103E and T313/350X exhibited no significant currents and could only be detected intracellularly. Upon 8 ng injection, plasma membrane presence was observed as well as currents up to 60% of wild-type current. Second, mutants V53E and V296G exhibited no Ba2+ -sensitive current, but were present in the plasma membrane at 0.1 ng and 8 ng injection levels. Third, mutants P91L and A179T were detectable on the plasma membrane (0.1 ng) and yielded currents of 98% and 80% of wild-type, respectively, at 25 pg injection. S294C yielded currents that were 45% of wild-type and were detected both on and just below the plasma membrane at 0.1 ng injection. This study has unraveled three distinct mechanisms by which mutations in hROMK2 could impair channel function in Bartter syndrome. Future experiments on kidney epithelial cell lines will have to confirm this classification, after which specific pharmacological treatments could be considered for each group of mutations.


Asunto(s)
Síndrome de Bartter/fisiopatología , Médula Renal/fisiopatología , Canales de Potasio de Rectificación Interna , Canales de Potasio/genética , Canales de Potasio/metabolismo , Animales , Síndrome de Bartter/genética , Humanos , Inmunohistoquímica , Potenciales de la Membrana/fisiología , Mutagénesis/fisiología , Oocitos/fisiología , Técnicas de Placa-Clamp , Canales de Potasio/química , Estructura Terciaria de Proteína , Equilibrio Hidroelectrolítico/fisiología , Xenopus laevis
18.
J Am Soc Nephrol ; 12(7): 1342-1349, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11423563

RESUMEN

The recently cloned epithelial Ca2+ channel (ECaC), which is expressed primarily in 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3))-responsive Ca2+ -transporting epithelia, is postulated to constitute the rate-limiting step in active Ca2+ reabsorption. In the present study, the effect of 1,25(OH)(2)D(3) was investigated on ECaC mRNA and protein levels in kidneys of rats that were raised on a vitamin D-depleting diet. This diet decreased the serum 1,25(OH)(2)D(3) concentration significantly, which was accompanied by a marked drop in serum Ca2+ level. Both 1,25(OH)(2)D(3) and Ca2+ levels were normalized within 48 h after 1,25(OH)(2)D(3) administration. In 1,25(OH)(2)D(3)-deficient rats, ECaC mRNA and protein levels of the kidney cortex were significantly decreased compared with the repleted animals, suggesting that 1,25(OH)(2)D(3) exerts its stimulatory effect on Ca2+ reabsorption via increased ECaC expression. In agreement with this observation, the elucidated human ECaC promoter contains several consensus vitamin D-responsive elements. ECaC was restricted to the apical membrane of the distal part of the distal convoluted and the connecting tubule. This conclusion was based on only minor overlap with the localization of the thiazide-sensitive NaCl co-transporter and complete co-localization with the 1,25(OH)(2)D(3)-dependent Ca2+ binding protein, calbindin-D(28K). In conclusion, ECaC, present in the distal part of the nephron, is an important target for 1,25(OH)(2)D(3)-mediated Ca2+ reabsorption.


Asunto(s)
Calcitriol/fisiología , Agonistas de los Canales de Calcio/metabolismo , Canales de Calcio/efectos de los fármacos , Canales de Calcio/metabolismo , Riñón/metabolismo , Animales , Secuencia de Bases/genética , Calbindinas , Calcitriol/farmacología , Calcio/metabolismo , Agonistas de los Canales de Calcio/farmacología , Canales de Calcio/genética , Dieta , Homeostasis/fisiología , Riñón/efectos de los fármacos , Corteza Renal/metabolismo , Masculino , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Ratas , Ratas Wistar , Proteína G de Unión al Calcio S100/metabolismo , Canales Catiónicos TRPV , Distribución Tisular , Deficiencia de Vitamina D
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA