Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Pflugers Arch ; 476(4): 533-543, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38110744

RESUMEN

Pendrin (SLC26A4) is an anion exchanger from the SLC26 transporter family which is mutated in human patients affected by Pendred syndrome, an autosomal recessive disease characterized by sensoneurinal deafness and hypothyroidism. Pendrin is also expressed in the kidney where it mediates the exchange of internal HCO3- for external Cl- at the apical surface of renal type B and non-A non-B-intercalated cells. Studies using pendrin knockout mice have first revealed that pendrin is essential for renal base excretion. However, subsequent studies have demonstrated that pendrin also controls chloride absorption by the distal nephron and that this mechanism is critical for renal NaCl balance. Furthermore, pendrin has been shown to control vascular volume and ultimately blood pressure. This review summarizes the current knowledge about how pendrin is linking renal acid-base regulation to blood pressure control.


Asunto(s)
Riñón , Nefronas , Animales , Ratones , Humanos , Presión Sanguínea/fisiología , Transportadores de Sulfato , Riñón/metabolismo , Nefronas/metabolismo , Cloruro de Sodio , Cloruros/metabolismo , Proteínas de Transporte de Anión/genética
2.
J Med Genet ; 59(11): 1035-1043, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35115415

RESUMEN

BACKGROUND: Nephrolithiasis (NL) is a complex multifactorial disease affecting up to 10%-20% of the human population and causing a significant burden on public health systems worldwide. It results from a combination of environmental and genetic factors. Hyperoxaluria is a major risk factor for NL. METHODS: We used a whole exome-based approach in a patient with calcium oxalate NL. The effects of the mutation were characterised using cell culture and in silico analyses. RESULTS: We identified a rare heterozygous missense mutation (c.1519C>T/p.R507W) in the SLC26A6 gene that encodes a secretory oxalate transporter. This mutation cosegregated with hyperoxaluria in the family. In vitro characterisation of mutant SLC26A6 demonstrated that Cl--dependent oxalate transport was dramatically reduced because the mutation affects both SLC26A6 transport activity and membrane surface expression. Cotransfection studies demonstrated strong dominant-negative effects of the mutant on the wild-type protein indicating that the phenotype of patients heterozygous for this mutation may be more severe than predicted by haploinsufficiency alone. CONCLUSION: Our study is in line with previous observations made in the mouse showing that SLC26A6 inactivation can cause inherited enteric hyperoxaluria with calcium oxalate NL. Consistent with an enteric form of hyperoxaluria, we observed a beneficial effect of increasing calcium in the patient's diet to reduce urinary oxalate excretion.


Asunto(s)
Antiportadores , Hiperoxaluria , Nefrolitiasis , Transportadores de Sulfato , Humanos , Antiportadores/genética , Calcio/metabolismo , Oxalato de Calcio/metabolismo , Hiperoxaluria/complicaciones , Hiperoxaluria/genética , Mutación , Nefrolitiasis/genética , Nefrolitiasis/complicaciones , Nefrolitiasis/metabolismo , Oxalatos/metabolismo , Transportadores de Sulfato/genética
3.
J Am Soc Nephrol ; 29(6): 1706-1719, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29773687

RESUMEN

BackgroundNedd4-2 is an E3 ubiquitin-protein ligase that associates with transport proteins, causing their ubiquitylation, and then internalization and degradation. Previous research has suggested a correlation between Nedd4-2 and BP. In this study, we explored the effect of intercalated cell (IC) Nedd4-2 gene ablation on IC transporter abundance and function and on BP.Methods We generated IC Nedd4-2 knockout mice using Cre-lox technology and produced global pendrin/Nedd4-2 null mice by breeding global Nedd4-2 null (Nedd4-2-/- ) mice with global pendrin null (Slc26a4-/- ) mice. Mice ate a diet with 1%-4% NaCl; BP was measured by tail cuff and radiotelemetry. We measured transepithelial transport of Cl- and total CO2 and transepithelial voltage in cortical collecting ducts perfused in vitro Transporter abundance was detected with immunoblots, immunohistochemistry, and immunogold cytochemistry.Results IC Nedd4-2 gene ablation markedly increased electroneutral Cl-/HCO3- exchange in the cortical collecting duct, although benzamil-, thiazide-, and bafilomycin-sensitive ion flux changed very little. IC Nedd4-2 gene ablation did not increase the abundance of type B IC transporters, such as AE4 (Slc4a9), H+-ATPase, barttin, or the Na+-dependent Cl-/HCO3- exchanger (Slc4a8). However, IC Nedd4-2 gene ablation increased CIC-5 total protein abundance, apical plasma membrane pendrin abundance, and the ratio of pendrin expression on the apical membrane to the cytoplasm. IC Nedd4-2 gene ablation increased BP by approximately 10 mm Hg. Moreover, pendrin gene ablation eliminated the increase in BP observed in global Nedd4-2 knockout mice.Conclusions IC Nedd4-2 regulates Cl-/HCO3- exchange in ICs., Nedd4-2 gene ablation increases BP in part through its action in these cells.


Asunto(s)
Presión Sanguínea/genética , Canales Epiteliales de Sodio/metabolismo , Transporte Iónico/genética , Ubiquitina-Proteína Ligasas Nedd4/genética , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacología , Animales , Bicarbonatos/metabolismo , Membrana Celular/metabolismo , Canales de Cloruro/metabolismo , Antiportadores de Cloruro-Bicarbonato/metabolismo , Cloruros/metabolismo , Intercambio Iónico , Túbulos Renales Colectores/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , ATPasas de Translocación de Protón/metabolismo , Protones , Reabsorción Renal/efectos de los fármacos , Simportadores de Sodio-Bicarbonato/metabolismo , Transportadores de Sulfato/genética , Transportadores de Sulfato/metabolismo , Tiazidas/farmacología
5.
Am J Physiol Renal Physiol ; 315(3): F429-F444, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29993276

RESUMEN

The vacuolar-type H+-ATPase B1 subunit is heavily expressed in the intercalated cells of the collecting system, where it contributes to H+ transport, but has also been described in other segments of the renal tubule. This study aimed to determine the localization of the B1 subunit of the vacuolar-type H+-ATPase in the early distal nephron, encompassing thick ascending limbs (TAL) and distal convoluted tubules (DCT), in human kidney and determine whether the localization differs between rodents and humans. Antibodies directed against the H+-ATPase B1 subunit were used to determine its localization in paraffin-embedded formalin-fixed mouse, rat, and human kidneys by light microscopy and in sections of Lowicryl-embedded rat kidneys by electron microscopy. Abundant H+-ATPase B1 subunit immunoreactivity was observed in the human kidney. As expected, intercalated cells showed the strongest signal, but significant signal was also observed in apical membrane domains of the distal nephron, including TAL, macula densa, and DCT. In mouse and rat, H+-ATPase B1 subunit expression could also be detected in apical membrane domains of these segments. In rat, electron microscopy revealed that the H+-ATPase B1 subunit was located in the apical membrane. Furthermore, the H+-ATPase B1 subunit colocalized with other H+-ATPase subunits in the TAL and DCT. In conclusion, the B1 subunit is expressed in the early distal nephron. The physiological importance of H+-ATPase expression in these segments remains to be delineated in detail. The phenotype of disease-causing mutations in the B1 subunit may also relate to its presence in the TAL and DCT.


Asunto(s)
Túbulos Renales Distales/enzimología , ATPasas de Translocación de Protón Vacuolares/metabolismo , Animales , Polaridad Celular , Humanos , Inmunohistoquímica , Túbulos Renales Distales/ultraestructura , Ratones Noqueados , Microscopía Electrónica de Transmisión , Especificidad de la Especie , ATPasas de Translocación de Protón Vacuolares/deficiencia , ATPasas de Translocación de Protón Vacuolares/genética
6.
Kidney Int ; 94(3): 514-523, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30146013

RESUMEN

Pseudohypoaldosteronism type II (PHAII) is a genetic disease characterized by association of hyperkalemia, hyperchloremic metabolic acidosis, hypertension, low renin, and high sensitivity to thiazide diuretics. It is caused by mutations in the WNK1, WNK4, KLHL3 or CUL3 gene. There is strong evidence that excessive sodium chloride reabsorption by the sodium chloride cotransporter NCC in the distal convoluted tubule is involved. WNK4 is expressed not only in distal convoluted tubule cells but also in ß-intercalated cells of the cortical collecting duct. These latter cells exchange intracellular bicarbonate for external chloride through pendrin, and therefore, account for renal base excretion. However, these cells can also mediate thiazide-sensitive sodium chloride absorption when the pendrin-dependent apical chloride influx is coupled to apical sodium influx by the sodium-driven chloride/bicarbonate exchanger. Here we determine whether this system is involved in the pathogenesis of PHAII. Renal pendrin activity was markedly increased in a mouse model carrying a WNK4 missense mutation (Q562E) previously identified in patients with PHAII. The upregulation of pendrin led to an increase in thiazide-sensitive sodium chloride absorption by the cortical collecting duct, and it caused metabolic acidosis. The function of apical potassium channels was altered in this model, and hyperkalemia was fully corrected by pendrin genetic ablation. Thus, we demonstrate an important contribution of pendrin in renal regulation of sodium chloride, potassium and acid-base homeostasis and in the pathophysiology of PHAII. Furthermore, we identify renal distal bicarbonate secretion as a novel mechanism of renal tubular acidosis.


Asunto(s)
Acidosis Tubular Renal/fisiopatología , Túbulos Renales Colectores/fisiopatología , Proteínas Serina-Treonina Quinasas/genética , Seudohipoaldosteronismo/complicaciones , Transportadores de Sulfato/metabolismo , Acidosis Tubular Renal/sangre , Acidosis Tubular Renal/etiología , Animales , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Humanos , Túbulos Renales Colectores/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense , Potasio/sangre , Potasio/metabolismo , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/fisiopatología , Eliminación Renal , Cloruro de Sodio/metabolismo , Simportadores de Sodio-Bicarbonato/metabolismo , Transportadores de Sulfato/genética , Regulación hacia Arriba
7.
J Am Soc Nephrol ; 28(1): 130-139, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27151921

RESUMEN

We recently described a novel thiazide-sensitive electroneutral NaCl transport mechanism resulting from the parallel operation of the Cl-/HCO3- exchanger pendrin and the Na+-driven Cl-/2HCO3- exchanger (NDCBE) in ß-intercalated cells of the collecting duct. Although a role for pendrin in maintaining Na+ balance, intravascular volume, and BP is well supported, there is no in vivo evidence for the role of NDCBE in maintaining Na+ balance. Here, we show that deletion of NDCBE in mice caused only subtle perturbations of Na+ homeostasis and provide evidence that the Na+/Cl- cotransporter (NCC) compensated for the inactivation of NDCBE. To unmask the role of NDCBE, we generated Ndcbe/Ncc double-knockout (dKO) mice. On a normal salt diet, dKO and single-knockout mice exhibited similar activation of the renin-angiotensin-aldosterone system, whereas only dKO mice displayed a lower blood K+ concentration. Furthermore, dKO mice displayed upregulation of the epithelial sodium channel (ENaC) and the Ca2+-activated K+ channel BKCa. During NaCl depletion, only dKO mice developed marked intravascular volume contraction, despite dramatically increased renin activity. Notably, the increase in aldosterone levels expected on NaCl depletion was attenuated in dKO mice, and single-knockout and dKO mice had similar blood K+ concentrations under this condition. In conclusion, NDCBE is necessary for maintaining sodium balance and intravascular volume during salt depletion or NCC inactivation in mice. Furthermore, NDCBE has an important role in the prevention of hypokalemia. Because NCC and NDCBE are both thiazide targets, the combined inhibition of NCC and the NDCBE/pendrin system may explain thiazide-induced hypokalemia in some patients.


Asunto(s)
Volumen Sanguíneo , Antiportadores de Cloruro-Bicarbonato/fisiología , Hipopotasemia/etiología , Animales , Ratones , Ratones Noqueados , Regulación hacia Arriba
8.
J Am Soc Nephrol ; 28(1): 209-217, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27335120

RESUMEN

Chloride transport by the renal tubule is critical for blood pressure (BP), acid-base, and potassium homeostasis. Chloride uptake from the urinary fluid is mediated by various apical transporters, whereas basolateral chloride exit is thought to be mediated by ClC-Ka/K1 and ClC-Kb/K2, two chloride channels from the ClC family, or by KCl cotransporters from the SLC12 gene family. Nevertheless, the localization and role of ClC-K channels is not fully resolved. Because inactivating mutations in ClC-Kb/K2 cause Bartter syndrome, a disease that mimics the effects of the loop diuretic furosemide, ClC-Kb/K2 is assumed to have a critical role in salt handling by the thick ascending limb. To dissect the role of this channel in detail, we generated a mouse model with a targeted disruption of the murine ortholog ClC-K2. Mutant mice developed a Bartter syndrome phenotype, characterized by renal salt loss, marked hypokalemia, and metabolic alkalosis. Patch-clamp analysis of tubules isolated from knockout (KO) mice suggested that ClC-K2 is the main basolateral chloride channel in the thick ascending limb and in the aldosterone-sensitive distal nephron. Accordingly, ClC-K2 KO mice did not exhibit the natriuretic response to furosemide and exhibited a severely blunted response to thiazide. We conclude that ClC-Kb/K2 is critical for salt absorption not only by the thick ascending limb, but also by the distal convoluted tubule.


Asunto(s)
Proteínas de Transporte de Anión/fisiología , Canales de Cloruro/fisiología , Nefronas/metabolismo , Cloruro de Sodio/metabolismo , Animales , Diuréticos/farmacología , Furosemida/farmacología , Ratones , Ratones Noqueados , Nefronas/efectos de los fármacos , Inhibidores de los Simportadores del Cloruro de Sodio/farmacología
9.
Nephrol Dial Transplant ; 32(7): 1137-1145, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28064162

RESUMEN

BACKGROUND: Pendrin, the chloride/bicarbonate exchanger of ß-intercalated cells of the renal connecting tubule and the collecting duct, plays a key role in NaCl reabsorption by the distal nephron. Therefore, pendrin may be important for the control of extracellular fluid volume and blood pressure. METHODS: Here, we have used a genetic mouse model in which the expression of pendrin can be switched-on in vivo by the administration of doxycycline. Pendrin can also be rapidly removed when doxycycline administration is discontinued. Therefore, our genetic strategy allows us to test selectively the acute effects of loss of pendrin function. RESULTS: We show that acute loss of pendrin leads to a significant decrease of blood pressure. In addition, acute ablation of pendrin did not alter significantly the acid-base status or blood K + concentration. CONCLUSION: By using a transgenic mouse model, avoiding off-target effects related to pharmacological compounds, this study suggests that pendrin could be a novel target to treat hypertension.


Asunto(s)
Proteínas de Transporte de Anión/fisiología , Presión Sanguínea/fisiología , Hipertensión/etiología , Animales , Hipertensión/metabolismo , Hipertensión/patología , Masculino , Ratones , Ratones Transgénicos , Transportadores de Sulfato
10.
J Am Soc Nephrol ; 27(12): 3511-3520, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27468975

RESUMEN

Metabolic acidosis is associated with increased urinary calcium excretion and related sequelae, including nephrocalcinosis and nephrolithiasis. The increased urinary calcium excretion induced by metabolic acidosis predominantly results from increased mobilization of calcium out of bone and inhibition of calcium transport processes within the renal tubule. The mechanisms whereby acid alters the integrity and stability of bone have been examined extensively in the published literature. Here, after briefly reviewing this literature, we consider the effects of acid on calcium transport in the renal tubule and then discuss why not all gene defects that cause renal tubular acidosis are associated with hypercalciuria and nephrocalcinosis.


Asunto(s)
Acidosis/genética , Acidosis/orina , Calcio/orina , Túbulos Renales , Desequilibrio Ácido-Base/complicaciones , Desequilibrio Ácido-Base/etiología , Desequilibrio Ácido-Base/metabolismo , Acidosis/clasificación , Enfermedades Óseas/etiología , Calcio/metabolismo , Humanos , Hipercalciuria/etiología , Túbulos Renales/metabolismo , Nefrocalcinosis/etiología
11.
J Am Soc Nephrol ; 27(11): 3320-3330, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27044666

RESUMEN

ATPase H+-transporting lysosomal accessory protein 2 (Atp6ap2), also known as the (pro)renin receptor, is a type 1 transmembrane protein and an accessory subunit of the vacuolar H+-ATPase (V-ATPase) that may also function within the renin-angiotensin system. However, the contribution of Atp6ap2 to renin-angiotensin-dependent functions remains unconfirmed. Using mice with an inducible conditional deletion of Atp6ap2 in mouse renal epithelial cells, we found that decreased V-ATPase expression and activity in the intercalated cells of the collecting duct impaired acid-base regulation by the kidney. In addition, these mice suffered from marked polyuria resistant to desmopressin administration. Immunoblotting revealed downregulation of the medullary Na+-K+-2Cl- cotransporter NKCC2 in these mice compared with wild-type mice, an effect accompanied by a hypotonic medullary interstitium and impaired countercurrent multiplication. This phenotype correlated with strong autophagic defects in epithelial cells of medullary tubules. Notably, cells with high accumulation of the autophagosomal substrate p62 displayed the strongest reduction of NKCC2 expression. Finally, nephron-specific Atp6ap2 depletion did not affect angiotensin II production, angiotensin II-dependent BP regulation, or sodium handling in the kidney. Taken together, our results show that nephron-specific deletion of Atp6ap2 does not affect the renin-angiotensin system but causes a combination of renal concentration defects and distal renal tubular acidosis as a result of impaired V-ATPase activity.


Asunto(s)
Riñón/enzimología , ATPasas de Translocación de Protón/fisiología , Receptores de Superficie Celular/fisiología , Sistema Renina-Angiotensina/fisiología , ATPasas de Translocación de Protón Vacuolares/fisiología , Animales , Femenino , Masculino , Ratones
12.
J Physiol ; 594(17): 4945-66, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27068441

RESUMEN

KEY POINTS: STE20 (Sterile 20)/SPS-1 related proline/alanine-rich kinase (SPAK) and oxidative stress-response kinase-1 (OSR1) phosphorylate and activate the renal Na(+) -K(+) -2Cl(-) cotransporter 2 (NKCC2) and Na(+) Cl(-) cotransporter (NCC). Mouse models suggest that OSR1 mainly activates NKCC2-mediated sodium transport along the thick ascending limb, while SPAK mainly activates NCC along the distal convoluted tubule, but the kinases may compensate for each other. We hypothesized that disruption of both kinases would lead to polyuria and severe salt-wasting, and generated SPAK/OSR1 double knockout mice to test this. Despite a lack of SPAK and OSR1, phosphorylated NKCC2 abundance was still high, suggesting the existence of an alternative activating kinase. Compensatory changes in SPAK/OSR1-independent phosphorylation sites on both NKCC2 and NCC and changes in sodium transport along the collecting duct were also observed. Potassium restriction revealed that SPAK and OSR1 play essential roles in the emerging model that NCC activation is central to sensing changes in plasma [K(+) ]. ABSTRACT: STE20 (Sterile 20)/SPS-1 related proline/alanine-rich kinase (SPAK) and oxidative stress-response kinase-1 (OSR1) activate the renal cation cotransporters Na(+) -K(+) -2Cl(-) cotransporter (NKCC2) and Na(+) -Cl(-) cotransporter (NCC) via phosphorylation. Knockout mouse models suggest that OSR1 mainly activates NKCC2, while SPAK mainly activates NCC, with possible cross-compensation. We tested the hypothesis that disrupting both kinases causes severe polyuria and salt-wasting by generating SPAK/OSR1 double knockout (DKO) mice. DKO mice displayed lower systolic blood pressure compared with SPAK knockout (SPAK-KO) mice, but displayed no severe phenotype even after dietary salt restriction. Phosphorylation of NKCC2 at SPAK/OSR1-dependent sites was lower than in SPAK-KO mice, but still significantly greater than in wild type mice. In the renal medulla, there was significant phosphorylation of NKCC2 at SPAK/OSR1-dependent sites despite a complete absence of SPAK and OSR1, suggesting the existence of an alternative activating kinase. The distal convoluted tubule has been proposed to sense plasma [K(+) ], with NCC activation serving as the primary effector pathway that modulates K(+) secretion, by metering sodium delivery to the collecting duct. Abundance of phosphorylated NCC (pNCC) is dramatically lower in SPAK-KO mice than in wild type mice, and the additional disruption of OSR1 further reduced pNCC. SPAK-KO and kidney-specific OSR1 single knockout mice maintained plasma [K(+) ] following dietary potassium restriction, but DKO mice developed severe hypokalaemia. Unlike mice lacking SPAK or OSR1 alone, DKO mice displayed an inability to phosphorylate NCC under these conditions. These data suggest that SPAK and OSR1 are essential components of the effector pathway that maintains plasma [K(+) ].


Asunto(s)
Túbulos Renales Distales/metabolismo , Potasio/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Presión Sanguínea , Homeostasis , Túbulos Renales Distales/fisiología , Masculino , Ratones , Ratones Noqueados , Potasio/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo
13.
Proc Natl Acad Sci U S A ; 110(19): 7928-33, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23610411

RESUMEN

The Na(+) concentration of the intracellular milieu is very low compared with the extracellular medium. Transport of Na(+) along this gradient is used to fuel secondary transport of many solutes, and thus plays a major role for most cell functions including the control of cell volume and resting membrane potential. Because of a continuous leak, Na(+) has to be permanently removed from the intracellular milieu, a process that is thought to be exclusively mediated by the Na(+)/K(+)-ATPase in animal cells. Here, we show that intercalated cells of the mouse kidney are an exception to this general rule. By an approach combining two-photon imaging of isolated renal tubules, physiological studies, and genetically engineered animals, we demonstrate that inhibition of the H(+) vacuolar-type ATPase (V-ATPase) caused drastic cell swelling and depolarization, and also inhibited the NaCl absorption pathway that we recently discovered in intercalated cells. In contrast, pharmacological blockade of the Na(+)/K(+)-ATPase had no effects. Basolateral NaCl exit from ß-intercalated cells was independent of the Na(+)/K(+)-ATPase but critically relied on the presence of the basolateral ion transporter anion exchanger 4. We conclude that not all animal cells critically rely on the sodium pump as the unique bioenergizer, but can be replaced by the H(+) V-ATPase in renal intercalated cells. This concept is likely to apply to other animal cell types characterized by plasma membrane expression of the H(+) V-ATPase.


Asunto(s)
Riñón/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/fisiología , Sodio/metabolismo , Absorción , Animales , Membrana Celular/metabolismo , Células Cultivadas , Antiportadores de Cloruro-Bicarbonato/genética , Inmunohistoquímica , Iones , Potenciales de la Membrana , Ratones , Ratones Noqueados , Perfusión , Bombas de Protones/fisiología , Cloruro de Sodio/farmacología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo
14.
PLoS Genet ; 9(7): e1003641, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874234

RESUMEN

Mutations of SLC26A4 are a common cause of human hearing loss associated with enlargement of the vestibular aqueduct. SLC26A4 encodes pendrin, an anion exchanger expressed in a variety of epithelial cells in the cochlea, the vestibular labyrinth and the endolymphatic sac. Slc26a4 (Δ/Δ) mice are devoid of pendrin and develop a severe enlargement of the membranous labyrinth, fail to acquire hearing and balance, and thereby provide a model for the human phenotype. Here, we generated a transgenic mouse line that expresses human SLC26A4 controlled by the promoter of ATP6V1B1. Crossing this transgene into the Slc26a4 (Δ/Δ) line restored protein expression of pendrin in the endolymphatic sac without inducing detectable expression in the cochlea or the vestibular sensory organs. The transgene prevented abnormal enlargement of the membranous labyrinth, restored a normal endocochlear potential, normal pH gradients between endolymph and perilymph in the cochlea, normal otoconia formation in the vestibular labyrinth and normal sensory functions of hearing and balance. Our study demonstrates that restoration of pendrin to the endolymphatic sac is sufficient to restore normal inner ear function. This finding in conjunction with our previous report that pendrin expression is required for embryonic development but not for the maintenance of hearing opens the prospect that a spatially and temporally limited therapy will restore normal hearing in human patients carrying a variety of mutations of SLC26A4.


Asunto(s)
Oído Interno/metabolismo , Saco Endolinfático/metabolismo , Pérdida Auditiva/genética , Proteínas de Transporte de Membrana/genética , Animales , Proteínas de Transporte de Anión/metabolismo , Oído Interno/patología , Endolinfa/metabolismo , Saco Endolinfático/patología , Femenino , Pérdida Auditiva/patología , Humanos , Ratones , Ratones Transgénicos , Mutación , Embarazo , Transportadores de Sulfato , ATPasas de Translocación de Protón Vacuolares/genética , Acueducto Vestibular/metabolismo , Acueducto Vestibular/fisiopatología
15.
Annu Rev Physiol ; 74: 325-49, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21888509

RESUMEN

The distal nephron plays a critical role in the renal control of homeostasis. Until very recently most studies focused on the control of Na(+), K(+), and water balance by principal cells of the collecting duct and the regulation of solute and water by hormones from the renin-angiotensin-aldosterone system and by antidiuretic hormone. However, recent studies have revealed the unexpected importance of renal intercalated cells, a subtype of cells present in the connecting tubule and collecting ducts. Such cells were thought initially to be involved exclusively in acid-base regulation. However, it is clear now that intercalated cells absorb NaCl and K(+) and hence may participate in the regulation of blood pressure and potassium balance. The second paradigm-challenging concept we highlight is the emerging importance of local paracrine factors that play a critical role in the renal control of water and electrolyte balance.


Asunto(s)
Electrólitos/metabolismo , Túbulos Renales Distales/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Transporte Biológico/fisiología , Presión Sanguínea/fisiología , Cloruros/metabolismo , Colon/metabolismo , Diuréticos/farmacología , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Homeostasis/fisiología , Humanos , Calicreínas/metabolismo , Calicreínas/fisiología , Túbulos Renales Distales/citología , Potasio/metabolismo , Sistema Renina-Angiotensina/fisiología , Canales de Sodio/fisiología , Cloruro de Sodio/metabolismo , Tiazidas/farmacología
16.
Curr Hypertens Rep ; 17(4): 538, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25794953

RESUMEN

The kidney continuously adapts daily renal excretion of NaCl to match dietary intakes in order to maintain the NaCl content of the body, and keep vascular volume constant. Any situation that leads to NaCl retention favors a rise in blood pressure. The aldosterone-sensitive distal nephron, which contains two main types of cells, principal (PC) and intercalated (IC) cells, is an important site for the final regulation of urinary Na(+) excretion. Research over the past 20 years established a paradigm in which PCs are the exclusive site of Na(+) absorption while ICs are solely dedicated to acid-base transport. Recent studies have revealed the unexpected importance of ICs for NaCl reabsorption. Here, we review the mechanisms of Na(+) and Cl(-) transport in the aldosterone-sensitive distal nephron, with emphasis on the role of ICs in maintaining NaCl balance and normal blood pressure.


Asunto(s)
Presión Sanguínea , Sodio/metabolismo , Aldosterona/metabolismo , Animales , Transporte Biológico , Humanos , Hipertensión/metabolismo , Hipertensión/fisiopatología , Riñón/fisiología , Cloruro de Sodio/metabolismo
17.
Cell Mol Life Sci ; 71(15): 2879-95, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24556999

RESUMEN

Sodium absorption by the distal part of the nephron, i.e., the distal convoluted tubule, the connecting tubule, and the collecting duct, plays a major role in the control of homeostasis by the kidney. In this part of the nephron, sodium transport can either be electroneutral or electrogenic. The study of electrogenic Na(+) absorption, which is mediated by the epithelial sodium channel (ENaC), has been the focus of considerable interest because of its implication in sodium, potassium, and acid-base homeostasis. However, recent studies have highlighted the crucial role played by electroneutral NaCl absorption in the regulation of the body content of sodium chloride, which in turn controls extracellular fluid volume and blood pressure. Here, we review the identification and characterization of the NaCl cotransporter (NCC), the molecule accounting for the main part of electroneutral NaCl absorption in the distal nephron, and its regulators. We also discuss recent work describing the identification of a novel "NCC-like" transport system mediated by pendrin and the sodium-driven chloride/bicarbonate exchanger (NDCBE) in the ß-intercalated cells of the collecting system.


Asunto(s)
Aldosterona/metabolismo , Electrólitos/metabolismo , Homeostasis , Nefronas/metabolismo , Cloruro de Sodio/metabolismo , Animales , Transporte Biológico , Presión Sanguínea , Humanos , Sistema Renina-Angiotensina
18.
J Am Soc Nephrol ; 24(7): 1104-13, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23766534

RESUMEN

Inherited and acquired disorders that enhance the activity of transporters mediating renal tubular Na(+) reabsorption are well established causes of hypertension. It is unclear, however, whether primary activation of an Na(+)-independent chloride transporter in the kidney can also play a pathogenic role in this disease. Here, mice overexpressing the chloride transporter pendrin in intercalated cells of the distal nephron (Tg(B1-hPDS) mice) displayed increased renal absorption of chloride. Compared with normal mice, these transgenic mice exhibited a delayed increase in urinary NaCl and ultimately, developed hypertension when exposed to a high-salt diet. Administering the same sodium intake as NaHCO3 instead of NaCl did not significantly alter BP, indicating that the hypertension in the transgenic mice was chloride-sensitive. Moreover, excessive chloride absorption by pendrin drove parallel absorption of sodium through the epithelial sodium channel ENaC and the sodium-driven chloride/bicarbonate exchanger (Ndcbe), despite an appropriate downregulation of these sodium transporters in response to the expanded vascular volume and hypertension. In summary, chloride transport in the distal nephron can play a primary role in driving NaCl transport in this part of the kidney, and a primary abnormality in renal chloride transport can provoke arterial hypertension. Thus, we conclude that the chloride/bicarbonate exchanger pendrin plays a major role in controlling net NaCl absorption, thereby influencing BP under conditions of high salt intake.


Asunto(s)
Presión Sanguínea/fisiología , Antiportadores de Cloruro-Bicarbonato/metabolismo , Cloruros/metabolismo , Hipertensión/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Nefronas/metabolismo , Cloruro de Sodio/metabolismo , Animales , Humanos , Inmunohistoquímica , Transporte Iónico , Ratones , Ratones Transgénicos , Transportadores de Sulfato
19.
Kidney Int ; 84(6): 1271-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23760292

RESUMEN

Inactivation of the tissue kallikrein gene in mice impairs renal handling of potassium due to enhanced H, K-ATPase activity, and induces hyperkalemia. We investigated whether the R53H loss-of-function polymorphism of the human tissue kallikrein gene affects renal potassium handling. In a crossover study, 30 R53R homozygous and 10 R53H heterozygous healthy males were randomly assigned to a low-sodium/high-potassium or a high-sodium/low-potassium diet to modulate tissue kallikrein synthesis. On the seventh day of each diet, participants were studied before and during a 2-h infusion of furosemide to stimulate distal potassium secretion. Urinary kallikrein activity was significantly lower in R53H than in R53R subjects on the low-sodium/high-potassium diet and was similarly reduced in both genotypes on high-sodium/low-potassium. Plasma potassium and renal potassium reabsorption were similar in both genotypes on an ad libitum sodium/potassium diet or after 7 days of a high-sodium/low-potassium diet. However, the median plasma potassium was significantly higher after 7 days of low-sodium/high-potassium diet in R53H than in R53R individuals. Urine potassium excretion and plasma aldosterone concentrations were similar. On the low-sodium/high-potassium diet, furosemide-induced decrease in plasma potassium was significantly larger in R53H than in R53R subjects. Thus, impaired tissue kallikrein stimulation by a low-sodium/high-potassium diet in R53H subjects with partial tissue kallikrein deficiency highlights an inappropriate renal adaptation to potassium load, consistent with experimental data in mice.


Asunto(s)
Riñón/metabolismo , Potasio en la Dieta/metabolismo , Calicreínas de Tejido/genética , Adaptación Fisiológica , Adulto , Aldosterona/sangre , Estudios Cruzados , Dieta Hiposódica , Furosemida/administración & dosificación , Voluntarios Sanos , Heterocigoto , Homocigoto , Humanos , Infusiones Intravenosas , Riñón/efectos de los fármacos , Masculino , Fenotipo , Potasio en la Dieta/administración & dosificación , Potasio en la Dieta/sangre , Potasio en la Dieta/orina , Cloruro de Sodio Dietético/metabolismo , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/administración & dosificación , Factores de Tiempo , Calicreínas de Tejido/deficiencia
20.
Proc Natl Acad Sci U S A ; 107(30): 13526-31, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20624970

RESUMEN

Tissue kallikrein (TK) is a serine protease synthetized in renal tubular cells located upstream from the collecting duct where renal potassium balance is regulated. Because secretion of TK is promoted by K+ intake, we hypothesized that this enzyme might regulate plasma K+ concentration ([K+]). We showed in wild-type mice that renal K+ and TK excretion increase in parallel after a single meal, representing an acute K+ load, whereas aldosterone secretion is not modified. Using aldosterone synthase-deficient mice, we confirmed that the control of TK secretion is aldosterone-independent. Mice with TK gene disruption (TK-/-) were used to assess the impact of the enzyme on plasma [K+]. A single large feeding did not lead to any significant change in plasma [K+] in TK+/+, whereas TK-/- mice became hyperkalemic. We next examined the impact of TK disruption on K+ transport in isolated cortical collecting ducts (CCDs) microperfused in vitro. We found that CCDs isolated from TK-/- mice exhibit net transepithelial K+ absorption because of abnormal activation of the colonic H+,K+-ATPase in the intercalated cells. Finally, in CCDs isolated from TK-/- mice and microperfused in vitro, the addition of TK to the perfusate but not to the peritubular bath caused a 70% inhibition of H+,K+-ATPase activity. In conclusion, we have identified the serine protease TK as a unique kalliuretic factor that protects against hyperkalemia after a dietary K+ load.


Asunto(s)
Adaptación Fisiológica/fisiología , Riñón/fisiología , Potasio/metabolismo , Calicreínas de Tejido/metabolismo , Adaptación Fisiológica/efectos de los fármacos , Aldosterona/metabolismo , Aldosterona/orina , Animales , Transporte Biológico , Citocromo P-450 CYP11B2/deficiencia , Citocromo P-450 CYP11B2/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Riñón/metabolismo , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/fisiología , Ratones , Ratones Noqueados , Potasio/sangre , Potasio/orina , Potasio en la Dieta/administración & dosificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sodio/metabolismo , Calicreínas de Tejido/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA