Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Kidney Int ; 96(4): 890-905, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31301888

RESUMEN

Fibroblast growth factor 23 (FGF23) regulates phosphate homeostasis, and its early rise in patients with chronic kidney disease is independently associated with all-cause mortality. Since inflammation is characteristic of chronic kidney disease and associates with increased plasma FGF23 we examined whether inflammation directly stimulates FGF23. In a population-based cohort, plasma tumor necrosis factor (TNF) was the only inflammatory cytokine that independently and positively correlated with plasma FGF23. Mouse models of chronic kidney disease showed signs of renal inflammation, renal FGF23 expression and elevated systemic FGF23 levels. Renal FGF23 expression coincided with expression of the orphan nuclear receptor Nurr1 regulating FGF23 in other organs. Antibody-mediated neutralization of TNF normalized plasma FGF23 and suppressed ectopic renal Fgf23 expression. Conversely, TNF administration to control mice increased plasma FGF23 without altering plasma phosphate. Moreover, in Il10-deficient mice with inflammatory bowel disease and normal kidney function, plasma FGF23 was elevated and normalized upon TNF neutralization. Thus, the inflammatory cytokine TNF contributes to elevated systemic FGF23 levels and also triggers ectopic renal Fgf23 expression in animal models of chronic kidney disease.


Asunto(s)
Factores de Crecimiento de Fibroblastos/sangre , Enfermedades Inflamatorias del Intestino/inmunología , Insuficiencia Renal Crónica/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Animales , Línea Celular , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/inmunología , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/sangre , Interleucina-10/deficiencia , Interleucina-10/genética , Riñón/inmunología , Riñón/patología , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Cultivo Primario de Células , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/patología , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/inmunología
2.
J Am Soc Nephrol ; 29(2): 518-531, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29021386

RESUMEN

microRNAs (miRNAs) are sequence-specific inhibitors of post-transcriptional gene expression. The physiologic function of these noncoding RNAs in postnatal renal tubules still remains unclear. Surprisingly, they appear to be dispensable for mammalian proximal tubule (PT) function. Here, we examined the effects of miRNA suppression in collecting ducts (CDs). To conclusively evaluate the role of miRNAs, we generated three mouse models with CD-specific inactivation of key miRNA pathway genes Dicer, Dgcr8, and the entire Argonaute gene family (Ago1, 2, 3, and 4). Characterization of these three mouse models revealed that inhibition of miRNAs in CDs spontaneously evokes a renal tubule injury-like response, which culminates in progressive tubulointerstitial fibrosis (TIF) and renal failure. Global miRNA profiling of microdissected renal tubules showed that miRNAs exhibit segmental distribution along the nephron and CDs. In particular, the expression of miR-200c is nearly 70-fold higher in CDs compared with PTs. Accordingly, miR-200s are downregulated in Dicer-KO CDs, its direct target genes Zeb1, Zeb2, and Snail2 are upregulated, and miRNA-depleted CDs undergo partial epithelial-to-mesenchymal transition (EMT). Thus, miRNAs are essential for CD homeostasis. Downregulation of CD-enriched miRNAs and the subsequent induction of partial EMT may be a new mechanism for TIF progression.


Asunto(s)
Epitelio/metabolismo , Epitelio/patología , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/patología , MicroARNs/genética , Animales , Proteínas Argonautas/genética , Línea Celular , ARN Helicasas DEAD-box/genética , Regulación hacia Abajo , Transición Epitelial-Mesenquimal/genética , Factores Eucarióticos de Iniciación/genética , Femenino , Fibrosis , Expresión Génica , Homeostasis/genética , Túbulos Renales Proximales/metabolismo , Masculino , Ratones , Ratones Noqueados , MicroARNs/antagonistas & inhibidores , Fenotipo , Proteínas de Unión al ARN/genética , Ribonucleasa III/genética , Factores de Transcripción de la Familia Snail/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
3.
Am J Physiol Renal Physiol ; 309(9): F779-90, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26336159

RESUMEN

The Rab GTPase-activating protein TBC1D4 (AS160) controls trafficking of the glucose transporter GLUT4 in adipocytes and skeletal muscle cells. TBC1D4 is also highly abundant in the renal distal tubule, although its role in this tubule is so far unknown. In vitro studies suggest that it is involved in the regulation of renal transporters and channels such as the epithelial sodium channel (ENaC), aquaporin-2 (AQP2), and the Na+-K+-ATPase. To assess the physiological role of TBC1D4 in the kidney, wild-type (TBC1D4+/+) and TBC1D4-deficient (TBC1D4-/-) mice were studied. Unexpectedly, neither under standard nor under challenging conditions (low Na+/high K+, water restriction) did TBC1D4-/- mice show any difference in urinary Na+ and K+ excretion, urine osmolarity, plasma ion and aldosterone levels, and blood pressure compared with TBC1D4+/+ mice. Also, immunoblotting did not reveal any change in the abundance of major renal sodium- and water-transporting proteins [Na-K-2Cl cotransporter (NKCC2) NKCC2, NaCl cotransporter (NCC), ENaC, AQP2, and the Na+-K+-ATPase]. However, the abundance of GLUT4, which colocalizes with TBC1D4 along the distal nephron of TBC1D4+/+ mice, was lower in whole kidney lysates of TBC1D4-/- mice than in TBC1D4+/+ mice. Likewise, primary thick ascending limb (TAL) cells isolated from TBC1D4-/- mice showed an increased basal glucose uptake and an abrogated insulin response compared with TAL cells from TBC1D4+/+ mice. Thus, TBC1D4 is dispensable for the regulation of renal Na+ and water transport, but may play a role for GLUT4-mediated basolateral glucose uptake in distal tubules. The latter may contribute to the known anaerobic glycolytic capacity of distal tubules during renal ischemia.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Riñón/metabolismo , Sodio/metabolismo , Equilibrio Hidroelectrolítico , Animales , Células Cultivadas , Proteínas Activadoras de GTPasa/deficiencia , Proteínas Activadoras de GTPasa/genética , Regulación de la Expresión Génica , Genotipo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Insulina/farmacología , Riñón/efectos de los fármacos , Túbulos Renales Distales/metabolismo , Asa de la Nefrona/metabolismo , Masculino , Ratones Noqueados , Parvalbúminas/genética , Fenotipo , Regiones Promotoras Genéticas , Sistema Renina-Angiotensina
4.
J Am Soc Nephrol ; 25(8): 1869-82, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24578125

RESUMEN

Uromodulin is expressed exclusively in the thick ascending limb and is the most abundant protein excreted in normal urine. Variants in UMOD, which encodes uromodulin, are associated with renal function, and urinary uromodulin levels may be a biomarker for kidney disease. However, the genetic factors regulating uromodulin excretion are unknown. We conducted a meta-analysis of urinary uromodulin levels to identify associated common genetic variants in the general population. We included 10,884 individuals of European descent from three genetic isolates and three urban cohorts. Each study measured uromodulin indexed to creatinine and conducted linear regression analysis of approximately 2.5 million single nucleotide polymorphisms using an additive model. We also tested whether variants in genes expressed in the thick ascending limb associate with uromodulin levels. rs12917707, located near UMOD and previously associated with renal function and CKD, had the strongest association with urinary uromodulin levels (P<0.001). In all cohorts, carriers of a G allele of this variant had higher uromodulin levels than noncarriers did (geometric means 10.24, 14.05, and 17.67 µg/g creatinine for zero, one, or two copies of the G allele). rs12446492 in the adjacent gene PDILT (protein disulfide isomerase-like, testis expressed) also reached genome-wide significance (P<0.001). Regarding genes expressed in the thick ascending limb, variants in KCNJ1, SORL1, and CAB39 associated with urinary uromodulin levels. These data indicate that common variants in the UMOD promoter region may influence urinary uromodulin levels. They also provide insights into uromodulin biology and the association of UMOD variants with renal function.


Asunto(s)
Variación Genética/genética , Uromodulina/orina , Población Blanca/genética , Creatinina/metabolismo , Humanos , Polimorfismo de Nucleótido Simple/genética , Uromodulina/genética
5.
Pflugers Arch ; 466(2): 343-56, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23887378

RESUMEN

The epithelial cells lining the thick ascending limb (TAL) of the loop of Henle perform essential transport processes and secrete uromodulin, the most abundant protein in normal urine. The lack of differentiated cell culture systems has hampered studies of TAL functions. Here, we report a method to generate differentiated primary cultures of TAL cells, developed from microdissected tubules obtained in mouse kidneys. The TAL tubules cultured on permeable filters formed polarized confluent monolayers in ∼12 days. The TAL cells remain differentiated and express functional markers such as uromodulin, NKCC2, and ROMK at the apical membrane. Electrophysiological measurements on primary TAL monolayers showed a lumen-positive transepithelial potential (+9.4 ± 0.8 mV/cm(2)) and transepithelial resistance similar to that recorded in vivo. The transepithelial potential is abolished by apical bumetanide and in primary cultures obtained from ROMK knockout mice. The processing, maturation and apical secretion of uromodulin by primary TAL cells is identical to that observed in vivo. The primary TAL cells respond appropriately to hypoxia, hypertonicity, and stimulation by desmopressin, and they can be transfected. The establishment of this primary culture system will allow the investigation of TAL cells obtained from genetically modified mouse models, providing a critical tool for understanding the role of that segment in health and disease.


Asunto(s)
Células Cultivadas , Asa de la Nefrona/citología , Uromodulina/biosíntesis , Animales , Ratones , Ratones Noqueados , Canales de Potasio de Rectificación Interna/biosíntesis , Miembro 1 de la Familia de Transportadores de Soluto 12/biosíntesis
6.
Nephrol Dial Transplant ; 29(1): 136-45, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24097801

RESUMEN

BACKGROUND: Uromodulin (Tamm-Horsfall protein) is the most abundant protein excreted in the urine under physiological conditions. It is exclusively produced in the kidney and secreted into the urine via proteolytic cleavage. The involvement of UMOD, the gene that encodes uromodulin, in rare autosomal dominant diseases, and its robust genome-wide association with the risk of chronic kidney disease suggest that the level of uromodulin in urine could represent a critical biomarker for kidney function. The structure of uromodulin is complex, with multiple disulfide bonds and typical domains of extracellular proteins. METHODS: Thus far, the conditions influencing stability and measurement of uromodulin in human urine have not been systematically investigated, giving inconsistent results. In this study, we used a robust, in-house ELISA to characterize the conditions of sampling and storage necessary to provide a faithful dosage of uromodulin in the urine. RESULTS: The levels of uromodulin in human urine were significantly affected by centrifugation and vortexing, as well as by the conditions and duration of storage. CONCLUSIONS: These results validate a simple, low-cost ELISA and document the optimal conditions of processing and storage for measuring uromodulin in human urine.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática/métodos , Riñón/metabolismo , Manejo de Especímenes , Uromodulina/orina , Adolescente , Adulto , Animales , Biomarcadores/orina , Centrifugación , Femenino , Gota/metabolismo , Humanos , Hiperuricemia/metabolismo , Enfermedades Renales/metabolismo , Masculino , Persona de Mediana Edad , Manejo de Especímenes/normas , Temperatura , Adulto Joven
7.
Nat Genet ; 37(5): 465-7, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15821734

RESUMEN

Feingold syndrome is characterized by variable combinations of esophageal and duodenal atresias, microcephaly, learning disability, syndactyly and cardiac defect. We show here that heterozygous mutations in the gene MYCN are present in Feingold syndrome. All mutations are predicted to disrupt both the full-length protein and a new shortened MYCN isoform, suggesting that multiple aspects of early embryogenesis and postnatal brain growth in humans are tightly regulated by MYCN dosage.


Asunto(s)
Encéfalo/anomalías , Heterocigoto , Atresia Intestinal/genética , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Análisis Mutacional de ADN , Femenino , Dosificación de Gen , Humanos , Masculino , Mutación , Proteína Proto-Oncogénica N-Myc , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Linaje , Análisis de Secuencia de ADN
8.
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
9.
J Biol Chem ; 285(1): 171-8, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19903818

RESUMEN

Mutations in the voltage-gated K(+) channel Kv1.1 have been linked with a mixed phenotype of episodic ataxia and/or myokymia. Recently, we presented autosomal dominant hypomagnesemia as a new phenotypic characteristic associated with a mutation in Kv1.1 (N255D) (Glaudemans, B., van der Wijst, J., Scola, R. H., Lorenzoni, P. J., Heister, A., van der Kemp, A. W., Knoers, N. V., Hoenderop, J. G., and Bindels, R. J. (2009) J. Clin. Invest. 119, 936-942). A conserved asparagine at position 255 in the third transmembrane segment was converted into an aspartic acid, resulting in a non-functional channel. In this study, we explored the functional consequence of this conserved residue by substitution with other hydrophobic, polar, or charged amino acids (N255E, N255Q, N255A, N255V, N255T, and N255H). Upon overexpression in human embryonic kidney (HEK293) cells, cell surface biotinylation revealed plasma membrane expression of all mutant channels. Next, we used the whole-cell patch clamp technique to demonstrate that the N255E and N255Q mutants were non-functional. Substitution of Asn-255 with other amino acids (N255A, N255V, N255T, and N255H) did not prevent ion conduction, and these mutant channels activated at more negative potentials when compared with wild-type channels, -41.5 +/- 1.6, -45.5 +/- 2.0, -50.5 +/- 1.9, and -33.8 +/- 1.3 mV to -29.4 +/- 1.1 mV, respectively. The time constant of activation was significantly faster for the two most hydrophobic mutations, N255A (6.2 +/- 0.2 ms) and N255V (5.2 +/- 0.3 ms), and the hydrophilic mutant N255T (9.8 +/- 0.4 ms) in comparison with wild type (13.0 +/- 0.9 ms). Furthermore, the voltage dependence of inactivation was shifted approximately 13 mV to more negative potentials in all mutant channels except for N255H. Taken together, our data showed that an asparagine at position 255 in Kv1.1 is required for normal voltage dependence and kinetics of channel gating.


Asunto(s)
Sustitución de Aminoácidos/genética , Genes Dominantes/genética , Predisposición Genética a la Enfermedad , Canal de Potasio Kv.1.1/genética , Mutación/genética , Defectos Congénitos del Transporte Tubular Renal/genética , Secuencia de Aminoácidos , Asparagina/metabolismo , Línea Celular , Humanos , Activación del Canal Iónico , Cinética , Canal de Potasio Kv.1.1/química , Canal de Potasio Kv.1.1/metabolismo , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Estructura Secundaria de Proteína , Propiedades de Superficie , Factores de Tiempo
10.
Kidney Int ; 77(1): 17-22, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19812536

RESUMEN

The renal distal convoluted tubule (DCT) has an essential role in maintaining systemic magnesium (Mg(2+)) concentration. The DCT is the final determinant of plasma Mg(2+) levels, as the more distal nephron segments are largely impermeable to Mg(2+). In the past decade, positional candidate strategies in families with inherited forms of hypomagnesemia have led to the identification of genes involved in Mg(2+) handling. A large fraction of this resides in the DCT, namely, (i) the transient receptor potential channel melastatin subtype 6 (TRPM6), a divalent cation-permeable channel located at the luminal membrane of the DCT, facilitates Mg(2+) entry from the pro-urine into the cell; (ii) the epidermal growth factor is a novel hormone regulating active Mg(2+) transport through TRPM6; (iii) the voltage-gated K(+) channel, Kv1.1, establishes a favorable luminal membrane potential for TRPM6-mediated Mg(2+) transport; (iv) the Na(+)/K(+)-ATPase gamma-subunit (gamma-Na(+)/K(+)-ATPase) was identified as mutated protein in a family with isolated dominant hypomagnesemia. The molecular mechanism by which gamma-Na(+)/K(+)-ATPase is involved in DCT Mg(2+) handling remains unknown; (v) a high percentage of patients with mutations in the renal transcription factor HNF1B (hepatocyte nuclear factor 1 homeobox B) gene develop hypomagnesemia; and (vi) Gitelman and EAST/SeSAME syndrome patients suffer from a similar tubulopathy due to mutations in NCC (NaCl cotransporter) and Kir4.1, respectively. In these patients, decreased expression of TRPM6 is proposed to cause hypomagnesemia. Insights into the molecular mechanisms of the identified genes, as well as the identification of novel genes, will further improve our knowledge about renal Mg(2+) handling.


Asunto(s)
Túbulos Renales Distales/metabolismo , Magnesio/metabolismo , Transporte Biológico , Predisposición Genética a la Enfermedad , Humanos , Deficiencia de Magnesio/genética
11.
Sci Rep ; 9(1): 19517, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31863061

RESUMEN

Uromodulin, the most abundant protein in normal urine, is produced by cells lining the thick ascending limb (TAL) of the loop of Henle. Uromodulin regulates the activity of the potassium channel ROMK in TAL cells. Common variants in KCNJ1, the gene encoding ROMK, are associated with urinary levels of uromodulin in population studies. Here, we investigated the functional link between ROMK and uromodulin in Kcnj1 knock-out mouse models, in primary cultures of mouse TAL (mTAL) cells, and in patients with Bartter syndrome due to KCNJ1 mutations. Both global and kidney-specific Kcnj1 knock-out mice showed reduced urinary levels of uromodulin paralleled by increased levels in the kidney, compared to wild-type controls. Pharmacological inhibition and genetic deletion of ROMK in mTAL cells caused a reduction in apical uromodulin excretion, reflected by cellular accumulation. In contrast, NKCC2 inhibition showed no effect on uromodulin processing. Patients with Bartter syndrome type 2 showed reduced urinary uromodulin levels compared to age and gender matched controls. These results demonstrate that ROMK directly regulates processing and release of uromodulin by TAL cells, independently from NKCC2. They support the functional link between transport activity and uromodulin in the TAL, relevant for blood pressure control and urinary concentrating ability.


Asunto(s)
Síndrome de Bartter/metabolismo , Síndrome de Bartter/orina , Canales de Potasio de Rectificación Interna/orina , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Uromodulina/metabolismo , Uromodulina/orina , Animales , Células Cultivadas , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Immunoblotting , Asa de la Nefrona/metabolismo , Masculino , Ratones , Ratones Noqueados , Mutación/genética
12.
Nat Med ; 19(12): 1655-60, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24185693

RESUMEN

Hypertension and chronic kidney disease (CKD) are complex traits representing major global health problems. Multiple genome-wide association studies have identified common variants in the promoter of the UMOD gene, which encodes uromodulin, the major protein secreted in normal urine, that cause independent susceptibility to CKD and hypertension. Despite compelling genetic evidence for the association between UMOD risk variants and disease susceptibility in the general population, the underlying biological mechanism is not understood. Here, we demonstrate that UMOD risk variants increased UMOD expression in vitro and in vivo. Uromodulin overexpression in transgenic mice led to salt-sensitive hypertension and to the presence of age-dependent renal lesions similar to those observed in elderly individuals homozygous for UMOD promoter risk variants. The link between uromodulin and hypertension is due to activation of the renal sodium cotransporter NKCC2. We demonstrated the relevance of this mechanism in humans by showing that pharmacological inhibition of NKCC2 was more effective in lowering blood pressure in hypertensive patients who are homozygous for UMOD promoter risk variants than in other hypertensive patients. Our findings link genetic susceptibility to hypertension and CKD to the level of uromodulin expression and uromodulin's effect on salt reabsorption in the kidney. These findings point to uromodulin as a therapeutic target for lowering blood pressure and preserving renal function.


Asunto(s)
Regulación de la Expresión Génica , Hipertensión/genética , Regiones Promotoras Genéticas/genética , Insuficiencia Renal Crónica/genética , Uromodulina/genética , Adulto , Anciano , Animales , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Hipertensión/inducido químicamente , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Sodio en la Dieta/efectos adversos , Sodio en la Dieta/farmacocinética , Regulación hacia Arriba , Adulto Joven
13.
Eur J Hum Genet ; 20(3): 263-70, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22009145

RESUMEN

Gitelman syndrome (GS) is an autosomal recessive disorder characterized by hypokalemic metabolic alkalosis in conjunction with significant hypomagnesemia and hypocalciuria. The GS phenotype is caused by mutations in the solute carrier family 12, member 3 (SLC12A3) gene that encodes the thiazide-sensitive NaCl cotransporter (NCC). We analyzed DNA samples of 163 patients with a clinical suspicion of GS by direct sequencing of all 26 exons of the SLC12A3 gene. In total, 114 different mutations were identified, 31 of which have not been reported before. These novel variants include 3 deletions, 18 missense, 6 splice site and 4 nonsense mutations. We selected seven missense mutations to investigate their effect on NCC activity and plasma membrane localization by using the Xenopus laevis oocyte expression system. The Thr392Ile mutant did not display transport activity (probably class 2 mutation), while the Asn442Ser and Gln1030Arg NCC mutants showed decreased plasma membrane localization and consequently function, likely due to impaired trafficking (class 3 mutation). Even though the NaCl uptake was hampered for NCC mutants Glu121Asp, Pro751Leu, Ser475Cys and Tyr489His, the transporters reached the plasma membrane (class 4 mutation), suggesting an effect on NCC regulation or ion affinity. The present study shows the identification of 38 novel mutations in the SLC12A3 gene and provides insight into the mechanisms that regulate NCC.


Asunto(s)
Síndrome de Gitelman/genética , Síndrome de Gitelman/metabolismo , Mutación , Receptores de Droga/genética , Receptores de Droga/metabolismo , Simportadores/genética , Simportadores/metabolismo , Secuencia de Aminoácidos , Animales , Membrana Celular/metabolismo , Estudios de Cohortes , Femenino , Eliminación de Gen , Expresión Génica , Humanos , Datos de Secuencia Molecular , Oocitos/metabolismo , Alineación de Secuencia , Miembro 3 de la Familia de Transportadores de Soluto 12 , Xenopus laevis
14.
J Clin Invest ; 119(4): 936-42, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19307729

RESUMEN

Primary hypomagnesemia is a heterogeneous group of disorders characterized by renal or intestinal magnesium (Mg2+) wasting, resulting in tetany, cardiac arrhythmias, and seizures. The kidney plays an essential role in maintaining blood Mg2+ levels, with a prominent function for the Mg2+-transporting channel transient receptor potential cation channel, subfamily M, member 6 (TRPM6) in the distal convoluted tubule (DCT). In the DCT, Mg2+ reabsorption is an active transport process primarily driven by the negative potential across the luminal membrane. Here, we studied a family with isolated autosomal dominant hypomagnesemia and used a positional cloning approach to identify an N255D mutation in KCNA1, a gene encoding the voltage-gated potassium (K+) channel Kv1.1. Kv1.1 was found to be expressed in the kidney, where it colocalized with TRPM6 along the luminal membrane of the DCT. Upon overexpression in a human kidney cell line, patch clamp analysis revealed that the KCNA1 N255D mutation resulted in a nonfunctional channel, with a dominant negative effect on wild-type Kv1.1 channel function. These data suggest that Kv1.1 is a renal K+ channel that establishes a favorable luminal membrane potential in DCT cells to control TRPM6-mediated Mg2+ reabsorption.


Asunto(s)
Canal de Potasio Kv.1.1/genética , Deficiencia de Magnesio/genética , Mutación Missense , Secuencia de Aminoácidos , Secuencia de Bases , Brasil , Línea Celular , Mapeo Cromosómico , Análisis Mutacional de ADN , Femenino , Genes Dominantes , Humanos , Riñón/metabolismo , Canal de Potasio Kv.1.1/química , Canal de Potasio Kv.1.1/metabolismo , Deficiencia de Magnesio/metabolismo , Masculino , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Linaje , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Canales Catiónicos TRPM/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA