Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 298(6): 102004, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35504352

RESUMEN

The epithelial sodium channel (ENaC) is a heterotrimer consisting of α-, ß-, and γ-subunits. Channel activation requires proteolytic release of inhibitory tracts from the extracellular domains of α-ENaC and γ-ENaC; however, the proteases involved in the removal of the γ-inhibitory tract remain unclear. In several epithelial tissues, ENaC is coexpressed with the transmembrane serine protease 2 (TMPRSS2). Here, we explored the effect of human TMPRSS2 on human αßγ-ENaC heterologously expressed in Xenopus laevis oocytes. We found that coexpression of TMPRSS2 stimulated ENaC-mediated whole-cell currents by approximately threefold, likely because of an increase in average channel open probability. Furthermore, TMPRSS2-dependent ENaC stimulation was not observed using a catalytically inactive TMPRSS2 mutant and was associated with fully cleaved γ-ENaC in the intracellular and cell surface protein fractions. This stimulatory effect of TMPRSS2 on ENaC was partially preserved when inhibiting its proteolytic activity at the cell surface using aprotinin but was abolished when the γ-inhibitory tract remained attached to its binding site following introduction of two cysteine residues (S155C-Q426C) to form a disulfide bridge. In addition, computer simulations and site-directed mutagenesis experiments indicated that TMPRSS2 can cleave γ-ENaC at sites both proximal and distal to the γ-inhibitory tract. This suggests a dual role of TMPRSS2 in the proteolytic release of the γ-inhibitory tract. Finally, we demonstrated that TMPRSS2 knockdown in cultured human airway epithelial cells (H441) reduced baseline proteolytic activation of endogenously expressed ENaC. Thus, we conclude that TMPRSS2 is likely to contribute to proteolytic ENaC activation in epithelial tissues in vivo.


Asunto(s)
Canales Epiteliales de Sodio , Oocitos , Serina Endopeptidasas , Animales , Canales Epiteliales de Sodio/metabolismo , Humanos , Transporte Iónico/fisiología , Oocitos/metabolismo , Péptido Hidrolasas/metabolismo , Proteolisis , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Xenopus laevis/genética
2.
Am J Physiol Renal Physiol ; 322(1): F42-F54, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34843658

RESUMEN

The renal outer medullary K+ channel (ROMK) is colocalized with the epithelial Na+ channel (ENaC) in the late distal convoluted tubule (DCT2), connecting tubule (CNT), and cortical collecting duct (CCD). ENaC-mediated Na+ absorption generates the electrical driving force for ROMK-mediated tubular K+ secretion, which is critically important for maintaining renal K+ homeostasis. ENaC activity is aldosterone dependent in the late CNT and early CCD (CNT/CCD) but aldosterone independent in the DCT2 and early CNT (DCT2/CNT). This suggests that under baseline conditions with low plasma aldosterone, ROMK-mediated K+ secretion mainly occurs in the DCT2/CNT. Therefore, we hypothesized that baseline ROMK activity is higher in the DCT2/CNT than in the CNT/CCD. To test this hypothesis, patch-clamp experiments were performed in the DCT2/CNT and CNT/CCD microdissected from mice maintained on a standard diet. In single-channel recordings from outside-out patches, we detected typical ROMK channel activity in both the DCT2/CNT and CNT/CCD and confirmed that ROMK is the predominant K+ channel in the apical membrane. Amiloride-sensitive and tertiapin-sensitive whole-cell currents were determined to assess ENaC and ROMK activity, respectively. As expected, baseline amiloride-sensitive current was high in the DCT2/CNT (∼370 pA) but low in the CNT/CCD (∼60 pA). Importantly, tertiapin-sensitive current was significantly higher in the DCT2/CNT than in the CNT/CCD (∼810 vs. ∼350 pA). We conclude that high ROMK activity in the DCT2/CNT is critical for aldosterone-independent renal K+ secretion under baseline conditions. A low-K+ diet significantly reduced ENaC but not ROMK activity in the DCT2/CNT. This suggests that modifying ENaC activity in the DCT2/CNT plays a key regulatory role in adjusting renal K+ excretion to dietary K+ intake.NEW & NOTEWORTHY ROMK-mediated renal K+ secretion is essential for maintaining K+ balance and requires a lumen negative transepithelial potential critically dependent on ENaC activity. Using microdissected distal mouse tubules, we demonstrated that baseline apical ROMK activity is high in the DCT2/CNT. Aldosterone-independent baseline ENaC activity is also high in the DCT2/CNT and downregulated by a low-K+ diet, which highlights the important role of the DCT2/CNT in regulating K+ secretion in an aldosterone-independent manner.


Asunto(s)
Aldosterona/farmacología , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Distales/efectos de los fármacos , Canales de Potasio de Rectificación Interna/metabolismo , Potasio/metabolismo , Eliminación Renal/efectos de los fármacos , Animales , Canales Epiteliales de Sodio/metabolismo , Femenino , Túbulos Renales Colectores/metabolismo , Túbulos Renales Distales/metabolismo , Masculino , Potenciales de la Membrana , Ratones Endogámicos C57BL , Potasio en la Dieta/metabolismo
3.
Am J Physiol Renal Physiol ; 321(3): F257-F268, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34251271

RESUMEN

The epithelial Na+ channel (ENaC) constitutes the rate-limiting step for Na+ absorption in the aldosterone-sensitive distal nephron (ASDN) comprising the late distal convoluted tubule (DCT2), connecting tubule (CNT), and collecting duct (CD). Previously, we demonstrated that ENaC activity in the DCT2/CNT transition zone is constitutively high and independent of aldosterone, in contrast to its aldosterone dependence in the late CNT/initial cortical CD (CCD). The mineralocorticoid receptor (MR) is expressed in the entire ASDN. Its activation by glucocorticoids is prevented through 11ß-hydroxysteroid dehydrogenase 2 (11ß-HSD2) abundantly expressed in the late but probably not early part of the ASDN. We hypothesized that ENaC function in the early part of the ASDN is aldosterone independent but may depend on MR activated by glucocorticoids due to low 11ß-HSD2 abundance. To test this hypothesis, we used doxycycline-inducible nephron-specific MR-deficient [MR knockout (KO)] mice. Whole cell ENaC currents were investigated in isolated nephron fragments from the DCT2/CNT or CNT/CCD transition zones using the patch-clamp technique. ENaC activity was detectable in the CNT/CCD of control mice but absent or barely detectable in the majority of CNT/CCD preparations from MR KO mice. Importantly, ENaC currents in the DCT2/CNT were greatly reduced in MR KO mice compared with ENaC currents in the DCT2/CNT of control mice. Immunofluorescence for 11ß-HSD2 was abundant in the CCD, less prominent in the CNT, and very low in the DCT2. We conclude that MR is critically important for maintaining aldosterone-independent ENaC activity in the DCT2/CNT. Aldosterone-independent MR activation is probably mediated by glucocorticoids due to low expression of 11ß-HSD2.NEW & NOTEWORTHY Using a mouse model with inducible nephron-specific mineralocorticoid receptor (MR) deficiency, we demonstrated that MR is not only critical for maintaining aldosterone-dependent ENaC activity in CNT/CCD but also for aldosterone-independent ENaC activity in DCT2/CNT. Furthermore, we demonstrated that cells of this latter nephron segment express little 11ß-HSD2, which probably allows glucocorticoids to stimulate MR, resulting in aldosterone-independent ENaC activity in DCT2/CNT. This site-specific ENaC regulation has physiologically relevant implications for renal sodium and potassium homeostasis.


Asunto(s)
Aldosterona/farmacocinética , Túbulos Renales Colectores/metabolismo , Potasio/metabolismo , Receptores de Mineralocorticoides/efectos de los fármacos , Receptores de Mineralocorticoides/metabolismo , Aldosterona/metabolismo , Animales , Canales Epiteliales de Sodio/metabolismo , Ratones , Nefronas/metabolismo , Sodio/metabolismo , Sodio en la Dieta/metabolismo
4.
J Gen Physiol ; 152(8)2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32442241

RESUMEN

Prostaglandin E2 (PGE2) is the most abundant prostanoid in the kidney, affecting a wide range of renal functions. Conflicting data have been reported regarding the effects of PGE2 on tubular water and ion transport. The amiloride-sensitive epithelial sodium channel (ENaC) is rate limiting for transepithelial sodium transport in the aldosterone-sensitive distal nephron. The aim of the present study was to explore a potential role of PGE2 in regulating ENaC in cortical collecting duct (CCD) cells. Short-circuit current (ISC) measurements were performed using the murine mCCDcl1 cell line known to express characteristic properties of CCD principal cells and to be responsive to physiological concentrations of aldosterone and vasopressin. PGE2 stimulated amiloride-sensitive ISC via basolateral prostaglandin E receptors type 4 (EP4) with an EC50 of ∼7.1 nM. The rapid stimulatory effect of PGE2 on ISC resembled that of vasopressin. A maximum response was reached within minutes, coinciding with an increased abundance of ß-ENaC at the apical plasma membrane and elevated cytosolic cAMP levels. The effects of PGE2 and vasopressin were nonadditive, indicating similar signaling cascades. Exposing mCCDcl1 cells to aldosterone caused a much slower (∼2 h) increase of the amiloride-sensitive ISC. Interestingly, the rapid effect of PGE2 was preserved even after aldosterone stimulation. Furthermore, application of arachidonic acid also increased the amiloride-sensitive ISC involving basolateral EP4 receptors. Exposure to arachidonic acid resulted in elevated PGE2 in the basolateral medium in a cyclooxygenase 1 (COX-1)-dependent manner. These data suggest that in the cortical collecting duct, locally produced and secreted PGE2 can stimulate ENaC-mediated transepithelial sodium transport.


Asunto(s)
Dinoprostona/farmacología , Canales Epiteliales de Sodio , Túbulos Renales Colectores , Animales , Línea Celular , Agonistas del Canal de Sodio Epitelial , Canales Epiteliales de Sodio/fisiología , Transporte Iónico , Túbulos Renales Colectores/citología , Ratones
5.
Nat Commun ; 11(1): 1995, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32332732

RESUMEN

Gut microbial dysbiosis is associated with the development of autoimmune disease, but the mechanisms by which microbial dysbiosis affects the transition from asymptomatic autoimmunity to inflammatory disease are incompletely characterized. Here, we identify intestinal barrier integrity as an important checkpoint in translating autoimmunity to inflammation. Zonulin family peptide (zonulin), a potent regulator for intestinal tight junctions, is highly expressed in autoimmune mice and humans and can be used to predict transition from autoimmunity to inflammatory arthritis. Increased serum zonulin levels are accompanied by a leaky intestinal barrier, dysbiosis and inflammation. Restoration of the intestinal barrier in the pre-phase of arthritis using butyrate or a cannabinoid type 1 receptor agonist inhibits the development of arthritis. Moreover, treatment with the zonulin antagonist larazotide acetate, which specifically increases intestinal barrier integrity, effectively reduces arthritis onset. These data identify a preventive approach for the onset of autoimmune disease by specifically targeting impaired intestinal barrier function.


Asunto(s)
Artritis Reumatoide/prevención & control , Permeabilidad de la Membrana Celular/efectos de los fármacos , Disbiosis/complicaciones , Haptoglobinas/antagonistas & inhibidores , Mucosa Intestinal/efectos de los fármacos , Oligopéptidos/administración & dosificación , Precursores de Proteínas/antagonistas & inhibidores , Adulto , Animales , Artritis Experimental/sangre , Artritis Experimental/inmunología , Artritis Experimental/microbiología , Artritis Experimental/prevención & control , Artritis Reumatoide/sangre , Artritis Reumatoide/inmunología , Artritis Reumatoide/microbiología , Traslocación Bacteriana/efectos de los fármacos , Traslocación Bacteriana/inmunología , Células CACO-2 , Permeabilidad de la Membrana Celular/inmunología , Estudios de Cohortes , Estudios Transversales , Disbiosis/inmunología , Disbiosis/microbiología , Femenino , Microbioma Gastrointestinal/inmunología , Haptoglobinas/metabolismo , Voluntarios Sanos , Humanos , Íleon/citología , Íleon/efectos de los fármacos , Íleon/microbiología , Íleon/patología , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Ratones , Persona de Mediana Edad , Precursores de Proteínas/sangre , Precursores de Proteínas/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo
6.
Am J Physiol Renal Physiol ; 318(5): F1113-F1121, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32174140

RESUMEN

Ubiquitination of the epithelial Na+ channel (ENaC) in epithelial cells may influence trafficking and hormonal regulation of the channels. We assessed ENaC ubiquitination (ub-ENaC) in mouse and rat kidneys using affinity beads to capture ubiquitinated proteins from tissue homogenates and Western blot analysis with anti-ENaC antibodies. Ub-αENaC was observed primarily as a series of proteins of apparent molecular mass of 40-70 kDa, consistent with the addition of variable numbers of ubiquitin molecules primarily to the NH2-terminal cleaved fragment (~30 kDa) of the subunit. No significant Ub-ßENaC was detected, indicating that ubiquitination of this subunit is minimal. For γENaC, the protein eluted from the affinity beads had the same apparent molecular mass as the cleaved COOH-terminal fragment of the subunit (~65 kDa). This suggests that the ubiquitinated NH2 terminus remains attached to the COOH-terminal moiety during isolation through disulfide bonds. Consistent with this, under nonreducing conditions, eluates contained material with increased molecular mass (90-150 kDa). In mice with a Liddle syndrome mutation (ß566X) deleting a putative binding site for the ubiquitin ligase neural precursor cell expressed developmentally downregulated 4-2, the amount of ub-γENaC was reduced as expected. To assess aldosterone dependence of ubiquitination, we fed rats either control or low-Na+ diets for 7 days before kidney harvest. Na+ depletion increased the amounts of ub-αENaC and ub-γENaC by three- to fivefold, probably reflecting increased amounts of fully cleaved ENaC. We conclude that ubiquitination occurs after complete proteolytic processing of the subunits, contributing to retrieval and/or disposal of channels expressed at the cell surface. Diminished ubiquitination does not appear to be a major factor in aldosterone-dependent ENaC upregulation.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Riñón/metabolismo , Síndrome de Liddle/metabolismo , Ubiquitinación , Aldosterona/sangre , Animales , Modelos Animales de Enfermedad , Canales Epiteliales de Sodio/genética , Femenino , Síndrome de Liddle/genética , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Proteolisis , Ratas Sprague-Dawley
7.
Pflugers Arch ; 470(2): 295-304, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28861610

RESUMEN

The epithelial sodium channel (ENaC) marks the tightly regulated, rate-limiting step of sodium re-absorption in the aldosterone-sensitive distal nephron (ASDN). Stimulation of ENaC activity by aldosterone involves the serum and glucocorticoid-induced kinase 1 (SGK1) and is mediated via complex mechanisms including inhibition of channel retrieval. Retrieved channels may be recycled or degraded, e.g. by the proteasomal pathway. The aim of the present study was to investigate whether inhibitors of the proteasome affect ENaC activity and surface expression, and to explore a possible involvement of SGK1. Short circuit current (I SC) measurements were performed on confluent mCCDcl1 murine cortical collecting duct cells to investigate the effect of two distinct proteasomal inhibitors, MG132 and bortezomib, on amiloride-sensitive ENaC-mediated I SC. Both inhibitors robustly stimulated amiloride-sensitive I SC. The time course and magnitude of the stimulatory effect of the proteasomal inhibitors on I SC were similar to those of aldosterone. Both, MG132 and aldosterone, significantly increased the abundance of ß-ENaC at the cell surface. SGK1 activity was assessed by monitoring the phosphorylation of a downstream target, NDRG1, and was found to be increased by MG132. Importantly, inhibiting SGK1 activity prevented not only the stimulatory effect of aldosterone but also that of proteasomal inhibition. In conclusion, these data suggest that ENaC stimulation following proteasomal inhibition is due to an accumulation of active SGK1 resulting in increased expression of ENaC at the cell surface. Thus, inhibition of the proteasome mimics SGK1-dependent stimulation of ENaC by aldosterone.


Asunto(s)
Aldosterona/farmacología , Canales Epiteliales de Sodio/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Inhibidores de Proteasoma/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Amilorida/farmacología , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Bloqueadores del Canal de Sodio Epitelial/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/metabolismo , Ratones
8.
Kidney Int ; 93(1): 159-172, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29042083

RESUMEN

Volume retention in nephrotic syndrome has been linked to activation of the epithelial sodium channel (ENaC) by proteolysis of its γ-subunit following urinary excretion of serine proteases such as plasmin. Here we tested whether pharmacological inhibition of urinary serine protease activity might protect from ENaC activation and volume retention in nephrotic syndrome. Urine from both nephrotic mice (induced by doxorubicin injection) and nephrotic patients exhibited high aprotinin-sensitive serine protease activity. Treatment of nephrotic mice with the serine protease inhibitor aprotinin by means of subcutaneous sustained-release pellets normalized urinary serine protease activity and prevented sodium retention, as did treatment with the ENaC inhibitor amiloride. In the kidney cortex from nephrotic mice, immunofluorescence revealed increased apical γ-ENaC staining, normalized by aprotinin treatment. In Xenopus laevis oocytes heterologously expressing murine ENaC, aprotinin had no direct inhibitory effect on channel activity but prevented proteolytic channel activation. Thus, our study shows that volume retention in experimental nephrotic syndrome is related to proteolytic ENaC activation by proteasuria and can be prevented by treatment with aprotinin. Hence, inhibition of urinary serine protease activity might become a therapeutic approach to treat patients with nephrotic-range proteinuria.


Asunto(s)
Aprotinina/farmacología , Edema/tratamiento farmacológico , Canales Epiteliales de Sodio/efectos de los fármacos , Riñón/efectos de los fármacos , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/enzimología , Serina Proteasas/orina , Inhibidores de Serina Proteinasa/farmacología , Equilibrio Hidroelectrolítico/efectos de los fármacos , Animales , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Doxorrubicina , Edema/enzimología , Edema/etiología , Edema/fisiopatología , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Humanos , Activación del Canal Iónico/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Ratones de la Cepa 129 , Síndrome Nefrótico/inducido químicamente , Síndrome Nefrótico/fisiopatología , Proteolisis , Transducción de Señal/efectos de los fármacos , Xenopus laevis
9.
Hypertension ; 67(6): 1256-62, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27170740

RESUMEN

The epithelial sodium channel (ENaC) is rate limiting for Na(+) absorption in the aldosterone-sensitive distal nephron comprising the late distal convoluted tubule (DCT2), the connecting tubule (CNT), and the entire collecting duct. Liddle syndrome (pseudohyperaldosteronism), a severe form of salt-sensitive hypertension, is caused by gain-of-function mutations of ENaC, but the precise tubular site of increased ENaC function is unknown. In the cortical collecting duct (CCD), ENaC is known to be regulated by aldosterone. In contrast, we recently reported aldosterone-independent ENaC regulation in the early part of the aldosterone-sensitive distal nephron. Here, we investigated ENaC function in the transition zone of DCT2/CNT or CNT/CCD microdissected from mice homozygous for Liddle syndrome mutation or from wild-type control mice. Whole-cell patch-clamp recordings were used to measure amiloride-sensitive ENaC currents in nephron fragments from mice maintained on different sodium diets to vary plasma aldosterone levels. Our data indicate that in mice with Liddle syndrome, the primary site of increased Na(+) reabsorption is the DCT2/CNT. In addition, increased aldosterone responsiveness of ENaC in CNT/CCD may contribute to salt-sensitive hypertension in Liddle syndrome. Single channel properties of ENaC were similar in Liddle syndrome mutation and wild-type mice, but ENaC expression at the apical membrane was increased in Liddle syndrome mutation when compared with wild-type mice, in particular, in animals maintained on a high salt diet. Our findings highlight the importance of ENaC function and regulation in the early part of the aldosterone-sensitive distal nephron for the maintenance of sodium balance and blood pressure control.


Asunto(s)
Aldosterona/sangre , Canales Epiteliales de Sodio/metabolismo , Síndrome de Liddle/genética , Sodio en la Dieta/farmacología , Animales , Modelos Animales de Enfermedad , Canales Epiteliales de Sodio/genética , Hipertensión/genética , Hipertensión/fisiopatología , Túbulos Renales Colectores/metabolismo , Síndrome de Liddle/fisiopatología , Ratones , Ratones Endogámicos , Mutación , Nefronas/metabolismo , Sensibilidad y Especificidad
10.
J Clin Invest ; 126(5): 1773-82, 2016 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-27043284

RESUMEN

The mTOR pathway orchestrates cellular homeostasis. The rapamycin-sensitive mTOR complex (mTORC1) in the kidney has been widely studied; however, mTORC2 function in renal tubules is poorly characterized. Here, we generated mice lacking mTORC2 in the distal tubule (Rictorfl/fl Ksp-Cre mice), which were viable and had no obvious phenotype, except for a 2.5-fold increase in plasma aldosterone. Challenged with a low-Na+ diet, these mice adequately reduced Na+ excretion; however, Rictorfl/fl Ksp-Cre mice rapidly developed hyperkalemia on a high-K+ diet, despite a 10-fold increase in serum aldosterone levels, implying that mTORC2 regulates kaliuresis. Phosphorylation of serum- and glucocorticoid-inducible kinase 1 (SGK1) and PKC-α was absent in Rictorfl/fl Ksp-Cre mice, indicating a functional block in K+ secretion activation via ROMK channels. Indeed, patch-clamp experiments on split-open tubular segments from the transition zone of the late connecting tubule and early cortical collecting duct demonstrated that Ba2+-sensitive apical K+ currents were barely detectable in the majority of Rictorfl/fl Ksp-Cre mice. Conversely, epithelial sodium channel (ENaC) activity was largely preserved, suggesting that the reduced ability to maintain K+ homeostasis is the result of impaired apical K+ conductance and not a reduced electrical driving force for K+ secretion. Thus, these data unravel a vital and nonredundant role of mTORC2 for distal tubular K+ handling.


Asunto(s)
Túbulos Renales/metabolismo , Complejos Multiproteicos/metabolismo , Potasio/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Equilibrio Hidroelectrolítico/fisiología , Animales , Hiperpotasemia/genética , Hiperpotasemia/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Transgénicos , Complejos Multiproteicos/genética , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Serina-Treonina Quinasas TOR/genética
11.
Clin J Am Soc Nephrol ; 11(5): 761-769, 2016 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-26933188

RESUMEN

BACKGROUND AND OBJECTIVES: Hypervolemia is a common feature of patients with CKD and associated with hypertension. Recent work has shown stimulation of sodium retention by urinary plasmin during nephrotic syndrome. However, it is unclear whether plasminuria plays a role in patients with stable CKD and non-nephrotic proteinuria. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: In this cross-sectional study, we analyzed the fluid status of 171 patients with CKD consecutively presenting to our outpatient clinic from 2012 to 2013 using bioimpedance spectroscopy (Body Composition Monitor [BCM]; Fresenius Medical Care, Germany) and its associations to the urinary excretion of plasminogen and plasmin from a spot urine sample. Two-electrode voltage clamp measurements were performed in Xenopus laevis oocytes expressing human epithelial sodium channel to investigate whether plasmin in concentrations found in urine can activate the channel. RESULTS: Overhydration >5% and overhydration >10% of the extracellular volume were found in 29% and 17% of the patients, respectively, and overhydration was associated with edema, hypertension, higher stages of CKD, and proteinuria. Proteinuria was the strongest independent predictor for overhydration (+0.58 L/1.73 m(2) per 10-fold increase; P<0.001). Urinary excretion of plasmin(ogen) quantified by ELISA correlated strongly with proteinuria (r=0.87) and overhydration (r=0.47). Using a chromogenic substrate, active plasmin was found in 44% of patients and correlated with proteinuria and overhydration. Estimated urinary plasmin concentrations were in a range sufficient to activate epithelial sodium channel currents in vitro. In multivariable analysis, urinary excretion of plasmin(ogen) was associated with overhydration similar to proteinuria. CONCLUSIONS: Hypervolemia in patients with CKD is strongly associated with proteinuria, even in the non-nephrotic range. Protein-rich urine contains high amounts of plasminogen and active plasmin, rendering plasminuria as a possible link between proteinuria and hypervolemia.


Asunto(s)
Edema/fisiopatología , Líquido Extracelular , Fibrinolisina/orina , Líquido Intracelular , Insuficiencia Renal Crónica/fisiopatología , Adulto , Composición Corporal , Índice de Masa Corporal , Estudios Transversales , Edema/complicaciones , Impedancia Eléctrica , Canales Epiteliales de Sodio/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estado de Hidratación del Organismo , Plasminógeno/orina , Proteinuria/etiología , Proteinuria/fisiopatología , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/orina , Factores Sexuales
12.
FASEB J ; 30(1): 45-53, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26324851

RESUMEN

Recently, a novel feedforward activation of the endothelial epithelial sodium channel (ENaC) [endothelial sodium channel (EnNaC)] by sodium was reported that counteracts ENaC function in kidney. In the absence of aldosterone, a rise in extracellular sodium (>145 mM) increases EnNaC surface abundance, thereby stiffening the cortex of vascular endothelial cells (ECs) in vitro. The latter reduces the release of NO-the hallmark of endothelial dysfunction. Here, we test whether high extracellular sodium per se increases EnNaC expression and cortical stiffness in an aldosterone synthase (Cyp11b2)-deficient (AS(-/-)) mouse model. Therefore, we employed in situ ECs of ex vivo aorta preparations from wild-type (WT) and AS(-/-). EnNaC surface expression (-16%) and cortical stiffness (-22%) were reduced in AS(-/-), compared with WT, whereas NO secretion was exclusively detectable in AS(-/-). EnNaC inhibition with benzamil decreased stiffness in both, while mineralocorticoid receptor antagonism diminished stiffness only in the WT. In the absence of aldosterone, high sodium (150 mM) increased EnNaC surface expression ex vivo (plus 19%) and cortical stiffness ex vivo (plus 41%) and in vivo (plus 44%). Application of aldosterone adjusted the stiffness of AS(-/-) to the WT level. We conclude that high sodium per se determines EnNaC expression and consequently endothelial cortical nanomechanics, thus likely contributing to endothelial dysfunction.


Asunto(s)
Citocromo P-450 CYP11B2/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Óxido Nítrico/metabolismo , Sodio/metabolismo , Aldosterona/farmacología , Amilorida/análogos & derivados , Amilorida/farmacología , Animales , Células Cultivadas , Citocromo P-450 CYP11B2/deficiencia , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/metabolismo , Canales Epiteliales de Sodio/metabolismo , Femenino , Masculino , Ratones , Ratones Noqueados , Antagonistas de Receptores de Mineralocorticoides/farmacología , Modelos Animales , Regulación hacia Arriba/efectos de los fármacos
13.
Am J Physiol Renal Physiol ; 308(5): F450-8, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25520009

RESUMEN

There is good evidence for a causal link between excessive sympathetic drive to the kidney and hypertension. We hypothesized that sympathetic regulation of tubular Na(+) absorption may occur in the aldosterone-sensitive distal nephron, where the fine tuning of renal Na(+) excretion takes place. Here, the appropriate regulation of transepithelial Na(+) transport, mediated by the amiloride-sensitive epithelial Na(+) channel (ENaC), is critical for blood pressure control. To explore a possible effect of the sympathetic transmitter norepinephrine on ENaC-mediated Na(+) transport, we performed short-circuit current (Isc) measurements on confluent mCCDcl1 murine cortical collecting duct cells. Norepinephrine caused a complex Isc response with a sustained increase of amiloride-sensitive Isc by ∼44%. This effect was concentration dependent and mediated via basolateral α2-adrenoceptors. In cells pretreated with aldosterone, the stimulatory effect of norepinephrine was reduced. Finally, we demonstrated that noradrenergic nerve fibers are present in close proximity to ENaC-expressing cells in murine kidney slices. We conclude that the sustained stimulatory effect of locally elevated norepinephrine on ENaC-mediated Na(+) absorption may contribute to the hypertensive effect of increased renal sympathetic activity.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Túbulos Renales Colectores/metabolismo , Norepinefrina/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Sodio/metabolismo , Aldosterona , Animales , Línea Celular , Ratones Endogámicos C57BL
14.
J Biol Chem ; 289(27): 19067-78, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24841206

RESUMEN

Proteolytic activation is a unique feature of the epithelial sodium channel (ENaC). However, the underlying molecular mechanisms and the physiologically relevant proteases remain to be identified. The serine protease trypsin I can activate ENaC in vitro but is unlikely to be the physiologically relevant activating protease in ENaC-expressing tissues in vivo. Herein, we investigated whether human trypsin IV, a form of trypsin that is co-expressed in several extrapancreatic epithelial cells with ENaC, can activate human ENaC. In Xenopus laevis oocytes, we monitored proteolytic activation of ENaC currents and the appearance of γENaC cleavage products at the cell surface. We demonstrated that trypsin IV and trypsin I can stimulate ENaC heterologously expressed in oocytes. ENaC cleavage and activation by trypsin IV but not by trypsin I required a critical cleavage site (Lys-189) in the extracellular domain of the γ-subunit. In contrast, channel activation by trypsin I was prevented by mutating three putative cleavage sites (Lys-168, Lys-170, and Arg-172) in addition to mutating previously described prostasin (RKRK(178)), plasmin (Lys-189), and neutrophil elastase (Val-182 and Val-193) sites. Moreover, we found that trypsin IV is expressed in human renal epithelial cells and can increase ENaC-mediated sodium transport in cultured human airway epithelial cells. Thus, trypsin IV may regulate ENaC function in epithelial tissues. Our results show, for the first time, that trypsin IV can stimulate ENaC and that trypsin IV and trypsin I activate ENaC by cleavage at distinct sites. The presence of distinct cleavage sites may be important for ENaC regulation by tissue-specific proteases.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Proteolisis , Tripsina/metabolismo , Secuencia de Aminoácidos , Animales , Azetidinas/farmacología , Bencilaminas/farmacología , Sitios de Unión , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/genética , Espacio Extracelular/metabolismo , Humanos , Riñón/metabolismo , Datos de Secuencia Molecular , Mutación , Oocitos/metabolismo , Estructura Terciaria de Proteína , Proteolisis/efectos de los fármacos , Xenopus laevis/genética
15.
Am J Physiol Renal Physiol ; 303(9): F1289-99, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22933298

RESUMEN

Aldosterone is thought to be the main hormone to stimulate the epithelial sodium channel (ENaC) in the aldosterone-sensitive distal nephron (ASDN) comprising the late distal convoluted tubule (DCT2), the connecting tubule (CNT) and the entire collecting duct (CD). There is immunohistochemical evidence for an axial gradient of ENaC expression along the ASDN with highest expression in the DCT2 and CNT. However, most of our knowledge about renal ENaC function stems from studies in the cortical collecting duct (CCD). Here we investigated ENaC function in the transition zone of DCT2/CNT or CNT/CCD microdissected from mice maintained on different sodium diets to vary plasma aldosterone levels. Single-channel recordings demonstrated amiloride-sensitive Na(+) channels in DCT2/CNT with biophysical properties typical for ENaC previously described in CNT/CCD. In animals maintained on a standard salt diet, the average ENaC-mediated whole cell current (ΔI(ami)) was higher in DCT2/CNT than in CNT/CCD. A low salt diet increased ΔI(ami) in CNT/CCD but had little effect on ΔI(ami) in DCT2/CNT. To investigate whether aldosterone is necessary for ENaC activity in the DCT2/CNT, we used aldosterone synthase knockout (AS(-/-)) mice that lack aldosterone. In CNT/CCD of AS(-/-) mice, ΔI(ami) was lower than that in wild-type (WT) animals and was not stimulated by a low salt diet. In contrast, in DCT2/CNT of AS(-/-) mice, ΔI(ami) was similar to that in DCT2/CNT of WT animals both on a standard and on a low salt diet. We conclude that ENaC function in the DCT2/CNT is largely independent of aldosterone which is in contrast to its known aldosterone sensitivity in CNT/CCD.


Asunto(s)
Aldosterona/fisiología , Canales Epiteliales de Sodio/fisiología , Túbulos Renales Distales/fisiología , Nefronas/fisiología , Animales , Citocromo P-450 CYP11B2/deficiencia , Citocromo P-450 CYP11B2/genética , Citocromo P-450 CYP11B2/fisiología , Canales Epiteliales de Sodio/efectos de los fármacos , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/fisiología , Túbulos Renales Distales/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Nefronas/efectos de los fármacos , Técnicas de Placa-Clamp , Sodio en la Dieta/farmacología
16.
Pflugers Arch ; 464(4): 353-65, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22864553

RESUMEN

Proteolytic processing of the amiloride-sensitive epithelial sodium channel (ENaC) by serine proteases is known to be important for channel activation. Inappropriate ENaC activation by proteases may contribute to the pathophysiology of cystic fibrosis and could be involved in sodium retention and the pathogenesis of arterial hypertension in the context of renal disease. We hypothesized that in addition to serine proteases, cathepsin proteases may activate ENaC. Cathepsin proteases belong to the group of cysteine proteases and play a pathophysiological role in inflammatory diseases. Under pathophysiological conditions, cathepsin-S (Cat-S) may reach ENaC in the apical membrane of epithelial cells. The aim of this study was to investigate the effect of purified Cat-S on human ENaC heterologously expressed in Xenopus laevis oocytes and on ENaC-mediated sodium transport in cultured M-1 mouse renal collecting duct cells. We demonstrated that Cat-S activates amiloride-sensitive whole-cell currents in ENaC-expressing oocytes. The stimulatory effect of Cat-S was preserved at pH 5. ENaC stimulation by Cat-S was associated with the appearance of a γENaC cleavage fragment at the plasma membrane indicating proteolytic channel activation. Mutating two valine residues (V182 and V193) in the critical region of γENaC prevented proteolytic activation of ENaC by Cat-S. Pre-incubation of the oocytes with the Cat-S inhibitor morpholinurea-leucine-homophenylalanine-vinylsulfone-phenyl (LHVS) prevented the stimulatory effect of Cat-S on ENaC. In contrast, LHVS had no effect on ENaC activation by the prototypical serine proteases trypsin and chymotrypsin. Cat-S also stimulated ENaC in differentiated renal epithelial cells. These findings demonstrate that the cysteine protease Cat-S can activate ENaC which may be relevant under pathophysiological conditions.


Asunto(s)
Catepsinas/metabolismo , Agonistas del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/metabolismo , Amilorida/farmacología , Secuencia de Aminoácidos , Animales , Catepsinas/antagonistas & inhibidores , Membrana Celular/metabolismo , Quimotripsina/metabolismo , Dipéptidos/farmacología , Bloqueadores del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/química , Canales Epiteliales de Sodio/genética , Humanos , Transporte Iónico , Ratones , Datos de Secuencia Molecular , Mutación , Proteolisis , Sodio/metabolismo , Sulfonas/farmacología , Tripsina/metabolismo , Valina/genética , Xenopus
17.
Am J Pathol ; 174(5): 1663-74, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19349364

RESUMEN

Hypoxia-inducible transcription factors (HIFs) play important roles in the response of the kidney to systemic and regional hypoxia. Degradation of HIFs is mediated by three oxygen-dependent HIF-prolyl hydroxylases (PHDs), which have partially overlapping characteristics. Although PHD inhibitors, which can induce HIFs in the presence of oxygen, are already in clinical development, little is known about the expression and regulation of these enzymes in the kidney. Therefore, we investigated the expression levels of the three PHDs in both isolated tubular cells and rat kidneys. All three PHDs were present in the kidney and were expressed predominantly in three different cell populations: (a) in distal convoluted tubules and collecting ducts (PHD1,2,3), (b) in glomerular podocytes (PHD1,3), and (c) in interstitial fibroblasts (PHD1,3). Higher levels of PHDs were found in tubular segments of the inner medulla where oxygen tensions are known to be physiologically low. PHD expression levels were unchanged in HIF-positive tubular and interstitial cells after induction by systemic hypoxia. In rat models of acute renal injury, changes in PHD expression levels were variable; while cisplatin and ischemia/reperfusion led to significant decreases in PHD2 and 3 expression levels, no changes were seen in a model of contrast media-induced nephropathy. These results implicate the non-uniform expression of HIF-regulating enzymes that modify the hypoxic response in the kidney under both regional and temporal conditions.


Asunto(s)
Lesión Renal Aguda/enzimología , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Riñón/enzimología , Procolágeno-Prolina Dioxigenasa/metabolismo , Animales , Antineoplásicos/toxicidad , Western Blotting , Cisplatino/toxicidad , Medios de Contraste/farmacología , Proteínas de Unión al ADN/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Proteínas Inmediatas-Precoces/genética , Técnicas para Inmunoenzimas , Isquemia/metabolismo , Isquemia/patología , Riñón/efectos de los fármacos , Riñón/lesiones , Médula Renal/efectos de los fármacos , Médula Renal/enzimología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/enzimología , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Podocitos/efectos de los fármacos , Podocitos/enzimología , Procolágeno-Prolina Dioxigenasa/genética , Pronóstico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
J Am Soc Nephrol ; 20(2): 299-310, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19073825

RESUMEN

Proteinuria and increased renal reabsorption of NaCl characterize the nephrotic syndrome. Here, we show that protein-rich urine from nephrotic rats and from patients with nephrotic syndrome activate the epithelial sodium channel (ENaC) in cultured M-1 mouse collecting duct cells and in Xenopus laevis oocytes heterologously expressing ENaC. The activation depended on urinary serine protease activity. We identified plasmin as a urinary serine protease by matrix-assisted laser desorption/ionization time of-flight mass spectrometry. Purified plasmin activated ENaC currents, and inhibitors of plasmin abolished urinary protease activity and the ability to activate ENaC. In nephrotic syndrome, tubular urokinase-type plasminogen activator likely converts filtered plasminogen to plasmin. Consistent with this, the combined application of urokinase-type plasminogen activator and plasminogen stimulated amiloride-sensitive transepithelial sodium transport in M-1 cells and increased amiloride-sensitive whole-cell currents in Xenopus laevis oocytes heterologously expressing ENaC. Activation of ENaC by plasmin involved cleavage and release of an inhibitory peptide from the ENaC gamma subunit ectodomain. These data suggest that a defective glomerular filtration barrier allows passage of proteolytic enzymes that have the ability to activate ENaC.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Fibrinolisina/orina , Nefrosis/orina , Amilorida/farmacología , Animales , Humanos , Riñón/metabolismo , Ratones , Oocitos/metabolismo , Técnicas de Placa-Clamp , Péptido Hidrolasas/metabolismo , Plasminógeno/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Xenopus laevis
19.
Am J Physiol Renal Physiol ; 295(4): F1052-62, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18653483

RESUMEN

Proteases are involved in the processing and activation of the epithelial sodium channel (ENaC). The aim of the present study was to investigate whether the prototypical serine protease trypsin can activate ENaC in microdissected, split-open mouse renal distal tubules. Whole-cell patch-clamp recordings from principal cells of connecting tubules (CNT) or cortical collecting ducts (CCD) demonstrated that addition of trypsin (20 microg/ml) to the bath solution increased the ENaC-mediated amiloride-sensitive whole cell current (DeltaIAmi) in the majority of cells. In contrast, trypsin applied in the presence of an excess of soybean trypsin inhibitor had no stimulatory effect. The DeltaIAmi response to trypsin was variable, ranging from no apparent effect to a twofold increase in DeltaI(Ami) with an average stimulatory effect of 31 or 37% in mice on low-Na+ or standard Na+ diet, respectively. In cultured M-1 mouse collecting duct cells, a robust stimulatory effect of trypsin on DeltaIAmi was only observed in cells pretreated with protease inhibitors. This suggests that endogenous proteases contribute to ENaC activation in renal tubular cells and that the degree of ENaC prestimulation by endogenous proteases determines the magnitude of the stimulatory response to exogenous trypsin. In conclusion, we provide electrophysiological evidence that trypsin can stimulate ENaC activity in native renal mouse tubules. Thus, in the kidney, ENaC stimulation by extracellular proteases may be a relevant regulatory mechanism in vivo.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Túbulos Renales Distales/metabolismo , Nefronas/metabolismo , Tripsina/metabolismo , Amilorida/farmacología , Animales , Aprotinina/farmacología , Células Cultivadas , Canales Epiteliales de Sodio/efectos de los fármacos , Túbulos Renales Distales/citología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Microdisección , Técnicas de Placa-Clamp , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Sodio/metabolismo , Bloqueadores de los Canales de Sodio/farmacología , Sodio en la Dieta/farmacología , Tripsina/farmacología , Inhibidores de Tripsina/farmacología
20.
J Physiol ; 586(2): 459-75, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18006588

RESUMEN

Liddle's syndrome is an autosomal dominant form of human hypertension, caused by gain-of-function mutations of the epithelial sodium channel (ENaC) which is expressed in aldosterone target tissues including the distal colon. We used a mouse model for Liddle's syndrome to investigate ENaC-mediated Na+ transport in late distal colon by measuring the amiloride-sensitive transepithelial short circuit current (Delta I SC-Ami) ex vivo. In Liddle mice maintained on a standard salt diet, Delta I SC-Ami was only slightly increased but plasma aldosterone (P Aldo) was severely suppressed. Liddle mice responded to a low or a high salt diet by increasing or decreasing, respectively, their P Aldo and Delta I SC-Ami. However, less aldosterone was required in Liddle animals to achieve similar or even higher Na+ transport rates than wild-type animals. Indeed, the ability of aldosterone to stimulate Delta I SC-Ami was about threefold higher in Liddle animals than in the wild-type controls. Application of aldosterone to colon tissue in vitro confirmed that ENaC stimulation by aldosterone was not only preserved but enhanced in Liddle mice. Aldosterone-induced transcriptional up-regulation of the channel's beta- and gamma-subunit (beta ENaC and gamma ENaC) and of the serum- and glucocorticoid-inducible kinase 1 (SGK1) was similar in colon tissue from Liddle and wild-type animals, while aldosterone had no transcriptional effect on the alpha-subunit (alpha ENaC). Moreover, Na+ feedback regulation was largely preserved in colon tissue of Liddle animals. In conclusion, we have demonstrated that in the colon of Liddle mice, ENaC-mediated Na+ transport is enhanced with an increased responsiveness to aldosterone. This may be pathophysiologically relevant in patients with Liddle's syndrome, in particular on a high salt diet, when suppression of P Aldo is likely to be insufficient to reduce Na+ absorption to an appropriate level.


Asunto(s)
Aldosterona/sangre , Colon/metabolismo , Canales Epiteliales de Sodio/metabolismo , Hipertensión/metabolismo , Hipopotasemia/metabolismo , Renina/sangre , Sodio en la Dieta/farmacología , Aldosterona/fisiología , Animales , Modelos Animales de Enfermedad , Canales Epiteliales de Sodio/genética , Femenino , Hipertensión/genética , Hipopotasemia/genética , Proteínas Inmediatas-Precoces/metabolismo , Riñón/metabolismo , Pulmón/metabolismo , Masculino , Ratones , Ratones Mutantes , Proteínas Serina-Treonina Quinasas/metabolismo , Renina/genética , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...