Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 2747, 2022 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-35585053

RESUMEN

Cation-chloride cotransporters (CCCs) NKCC1 and NKCC2 catalyze electroneutral symport of 1 Na+, 1 K+, and 2 Cl- across cell membranes. NKCC1 mediates trans-epithelial Cl- secretion and regulates excitability of some neurons and NKCC2 is critical to renal salt reabsorption. Both transporters are inhibited by the so-called loop diuretics including bumetanide, and these drugs are a mainstay for treating edema and hypertension. Here, our single-particle electron cryo-microscopy structures supported by functional studies reveal an outward-facing conformation of NKCC1, showing bumetanide wedged into a pocket in the extracellular ion translocation pathway. Based on these and the previously published inward-facing structures, we define the translocation pathway and the conformational changes necessary for ion translocation. We also identify an NKCC1 dimer with separated transmembrane domains and extensive transmembrane and C-terminal domain interactions. We further define an N-terminal phosphoregulatory domain that interacts with the C-terminal domain, suggesting a mechanism whereby (de)phosphorylation regulates NKCC1 by tuning the strength of this domain association.


Asunto(s)
Bumetanida , Simportadores , Bumetanida/farmacología , Cationes/metabolismo , Cloruros/metabolismo , Diuréticos/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12
2.
Commun Biol ; 4(1): 226, 2021 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-33597714

RESUMEN

NKCC and KCC transporters mediate coupled transport of Na++K++Cl- and K++Cl- across the plasma membrane, thus regulating cell Cl- concentration and cell volume and playing critical roles in transepithelial salt and water transport and in neuronal excitability. The function of these transporters has been intensively studied, but a mechanistic understanding has awaited structural studies of the transporters. Here, we present the cryo-electron microscopy (cryo-EM) structures of the two neuronal cation-chloride cotransporters human NKCC1 (SLC12A2) and mouse KCC2 (SLC12A5), along with computational analysis and functional characterization. These structures highlight essential residues in ion transport and allow us to propose mechanisms by which phosphorylation regulates transport activity.


Asunto(s)
Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Simportadores/metabolismo , Animales , Aniones , Sitios de Unión , Cationes , Microscopía por Crioelectrón , Células HEK293 , Humanos , Activación del Canal Iónico , Transporte Iónico , Simulación de Dinámica Molecular , Fosforilación , Unión Proteica , Conformación Proteica , Células Sf9 , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/ultraestructura , Relación Estructura-Actividad , Simportadores/genética , Simportadores/ultraestructura , Cotransportadores de K Cl
3.
Photochem Photobiol Sci ; 15(5): 604-8, 2016 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-27050155

RESUMEN

The photolysis quantum yield, Qp, of 1-(2-nitrophenyl)ethyl phosphate (caged Pi) measured in the near-UV (342 nm peak with 60 nm half-bandwidth) is 0.53 and is based on results reported in 1978 (Biochemistry, 17, 1929-1935). This article amplifies methodology for determining that Qp in view of different recent estimates. Some general principles together with other examples relating to measurement of Qp values are discussed together with their relevance to biological research.


Asunto(s)
Organofosfatos/química , Fotólisis , Espectrofotometría Ultravioleta , Rayos Ultravioleta
5.
J Biol Chem ; 289(11): 7569-79, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24451383

RESUMEN

We examined the relationship between transmembrane domain (TM) 10 and TM11/12 in NKCC1, testing homology models based on the structure of AdiC in the same transporter superfamily. We hypothesized that introduced cysteine pairs would be close enough for disulfide formation and would alter transport function: indeed, evidence for cross-link formation with low micromolar concentrations of copper phenanthroline or iodine was found in 3 of 8 initially tested pairs and in 1 of 26 additionally tested pairs. Inhibition of transport was observed with copper phenanthroline and iodine treatment of P676C/A734C and I677C/A734C, consistent with the proximity of these residues and with movement of TM10 during the occlusion step of ion transport. We also found Cu(2+) inhibition of the single-cysteine mutant A675C, suggesting that this residue and Met(382) of TM3 are involved in a Cu(2+)-binding site. Surprisingly, cross-linking of P676C/I730C was found to prevent rapid deactivation of the transporter while not affecting the dephosphorylation rate, thus uncoupling the phosphorylation and activation steps. Consistent with this, (a) cross-linking of P676C/I730C was dependent on activation state, and (b) mutants lacking the phosphoregulatory domain could still be activated by cross-linking. These results suggest a model of NKCC activation that involves movement of TM12 relative to TM10, which is likely tied to movement of the large C terminus, a process somehow triggered by phosphorylation of the regulatory domain in the N terminus.


Asunto(s)
Transporte Iónico , Simportadores de Cloruro de Sodio-Potasio/química , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular , Cloruros/química , Cobre/química , Reactivos de Enlaces Cruzados/química , Disulfuros/química , Homeostasis , Humanos , Iones , Cinética , Microscopía Confocal , Microscopía Fluorescente , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Fenantrolinas/química , Fosforilación , Estructura Terciaria de Proteína , Radioisótopos de Rubidio/química , Homología de Secuencia de Aminoácido , Miembro 2 de la Familia de Transportadores de Soluto 12/química , Miembro 2 de la Familia de Transportadores de Soluto 12/genética
6.
PLoS One ; 8(12): e82060, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24339991

RESUMEN

The Na-K-Cl cotransporter (NKCC) couples the movement of Na(+), K(+), and Cl(-) ions across the plasma membrane of most animal cells and thus plays a central role in cellular homeostasis and human physiology. In order to study the structure, function, and regulation of NKCC1 we have engineered a synthetic cDNA encoding the transporter with 30 unique silent restriction sites throughout the open reading frame, and with N-terminal 3xFlag and YFP tags. We show that the novel cDNA is appropriately expressed in HEK-293 cells and that the YFP-tag does not alter the transport function of the protein. Utilizing the Cl(-) -sensing capability of YFP, we demonstrate a sensitive assay of Na-K-Cl cotransport activity that measures normal cotransport activity in a fully activated transporter. In addition we present three newly developed epitope tags for NKCC1 all of which can be detected from outside of the cell, one of which is very efficiently delivered to the plasma membrane. Finally, we have characterized cysteine mutants of NKCC1 and found that whereas many useful combinations of cysteine mutations are tolerated by the biosynthetic machinery, the fully "cys-less" NKCC1 is retained in the endoplasmic reticulum. Together these advances are expected to greatly assist future studies of NKCC1.


Asunto(s)
Cisteína , ADN Complementario/metabolismo , Epítopos/biosíntesis , Miembro 2 de la Familia de Transportadores de Soluto 12/biosíntesis , ADN Complementario/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Epítopos/genética , Células HEK293 , Humanos , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/genética
7.
J Biol Chem ; 287(21): 17308-17317, 2012 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-22437837

RESUMEN

The Na-K-Cl cotransporter (NKCC) plays central roles in cellular chloride homeostasis and in epithelial salt transport, but to date little is known about the mechanism by which the transporter moves ions across the membrane. We examined the functional role of transmembrane helix 3 (TM3) in NKCC1 using cysteine- and tryptophan-scanning mutagenesis and analyzed our results in the context of a structural homology model based on an alignment of NKCC1 with other amino acid polyamine organocation superfamily members, AdiC and ApcT. Mutations of residues along one face of TM3 (Tyr-383, Met-382, Ala-379, Asn-376, Ala-375, Phe-372, Gly-369, and Ile-368) had large effects on translocation rate, apparent ion affinities, and loop diuretic affinity, consistent with a proposed role of TM3 in the translocation pathway. The prediction that Met-382 is part of an extracellular gate that closes to form an occluded state is strongly supported by conformational sensitivity of this residue to 2-(trimethylammonium)ethyl methanethiosulfonate, and the bumetanide insensitivity of M382W is consistent with tryptophan blocking entry of bumetanide into the cavity. Substitution effects on residues at the intracellular end of TM3 suggest that this region is also involved in ion coordination and may be part of the translocation pathway in an inward-open conformation. Mutations of predicted pore residues had large effects on binding of bumetanide and furosemide, consistent with the hypothesis that loop diuretic drugs bind within the translocation cavity. The results presented here strongly support predictions of homology models of NKCC1 and demonstrate important roles for TM3 residues in ion translocation and loop diuretic inhibition.


Asunto(s)
Bumetanida/farmacología , Diuréticos/farmacología , Mutagénesis , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Sitios de Unión , Bumetanida/farmacocinética , Diuréticos/farmacocinética , Células HEK293 , Humanos , Transporte Iónico/efectos de los fármacos , Mesilatos/farmacología , Mutación Missense , Estructura Secundaria de Proteína , Simportadores de Cloruro de Sodio-Potasio/genética , Miembro 2 de la Familia de Transportadores de Soluto 12 , Reactivos de Sulfhidrilo/farmacología
8.
J Biol Chem ; 287(3): 2210-20, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22121194

RESUMEN

The Na-K-Cl cotransporter (NKCC1) is expressed in most vertebrate cells and is crucial in the regulation of cell volume and intracellular chloride concentration. To study the structure and function of NKCC1, we tagged the transporter with cyan (CFP) and yellow (YFP) fluorescent proteins at two sites within the C terminus and measured fluorescence resonance energy transfer (FRET) in stably expressing human embryonic kidney cell lines. Both singly and doubly tagged NKCC1s were appropriately produced, trafficked to the plasma membrane, and exhibited (86)Rb transport activity. When both fluorescent probes were placed within the same C terminus of an NKCC1 transporter, we recorded an 11% FRET decrease upon activation of the transporter. This result clearly demonstrates movement of the C terminus during the regulatory response to phosphorylation of the N terminus. When we introduced CFP and YFP separately in different NKCC1 constructs and cotransfected these in HEK cells, we observed FRET between dimer pairs, and the fractional FRET decrease upon transporter activation was 46%. Quantitatively, this indicates that the largest FRET-signaled movement is between dimer pairs, an observation supported by further experiments in which the doubly tagged construct was cotransfectionally diluted with untagged NKCC1. Our results demonstrate that regulation of NKCC1 is accompanied by a large movement between two positions in the C termini of a dimeric cotransporter. We suggest that the NKCC1 C terminus is involved in transport regulation and that dimerization may play a key structural role in the regulatory process. It is anticipated that when combined with structural information, our findings will provide a model for understanding the conformational changes that bring about NKCC1 regulation.


Asunto(s)
Proteínas de Peces/química , Modelos Moleculares , Multimerización de Proteína , Simportadores de Cloruro de Sodio-Potasio/química , Animales , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Movimiento , Tiburones , Simportadores de Cloruro de Sodio-Potasio/genética , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12
9.
Am J Physiol Renal Physiol ; 300(4): F840-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21209010

RESUMEN

The Na-K-Cl cotransporter (NKCC2) is the major salt transport pathway in the thick ascending limb of Henle's loop and is part of the molecular mechanism for blood pressure regulation. Recent screening of ∼3,000 members of the Framingham Heart Study identified nine rare independent mutations in the gene encoding NKCC2 (SLC12A1) associated with clinically reduced blood pressure and protection from hypertension (Ji WZ, Foo JN, O'Roak BJ, Zhao H, Larson MG, Simon DB, Newton-Cheh C, State M, Levy D, Lifton RP. Nat Genet 40: 592-599, 2008). To investigate their functional consequences, we introduced the nine mutations in human NKCC2A and examined protein function, expression, localization, regulation, and ion transport kinetics using heterologous expression in Xenopus laevis oocytes and HEK-293 cells. When expressed in oocytes, four of the mutants (T235M, R302W, L505V, and P569H) exhibited reduced transport function compared with wild-type. In HEK-293 cells, the same four mutants exhibited reduced function, and in addition N399S and P1083A had significantly lower activity than wild-type. The two most functionally impaired mutants (R302W and L505V) exhibited dramatically diminished production of complex-glycosylated protein and a decrease in or absence of plasma membrane localization, indicative of a processing defect. All of the functional human (h) NKCC2A variants were regulated by changes in oocyte volume and intracellular chloride in HEK cells, but P254A and N399S exhibited a lower constitutive activity in HEK cells. The P569H mutant exhibited a 50% reduction in sodium affinity compared with wild-type, predicting lower transport activity at lower intratubular salt concentrations, while the P254A mutant exhibited a 35% increase in rubidium affinity. We conclude that defects in NKCC2 processing, transport turnover rate, regulation, and ion affinity contribute to impaired transport function in six of the nine identified mutants, providing support for the predictive approach of Ji et al. to identify functionally important residues by sequence conservation. Such mutations in hNKCC2A are likely to reduce renal salt reabsorption, providing a mechanism for lower blood pressure.


Asunto(s)
Riñón/metabolismo , Potasio/metabolismo , Simportadores de Cloruro de Sodio-Potasio/genética , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Sodio/metabolismo , Análisis de Varianza , Animales , Transporte Biológico/genética , Western Blotting , Células HEK293 , Humanos , Mutación , Oocitos , Miembro 1 de la Familia de Transportadores de Soluto 12 , Xenopus
10.
Glia ; 59(2): 320-32, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21125654

RESUMEN

The nervous system is protected by blood barriers that use multiple systems to control extracellular solute composition, osmotic pressure, and fluid volume. In the human nervous system, misregulation of the extracellular volume poses serious health threats. Here, we show that the glial cells that form the Drosophila blood-nerve barrier have a conserved molecular mechanism that regulates extracellular volume: the Serine/Threonine kinase Fray, which we previously showed is an ortholog of mammalian PASK/SPAK; and the Na-K-Cl cotransporter Ncc69, which we show is an ortholog of human NKCC1. In mammals, PASK/SPAK binds to NKCC1 and regulates its activity. In Drosophila, larvae mutant for Ncc69 develop a peripheral neuropathy, where fluid accumulates between glia and axons. The accumulation of fluid has no detectable impact on action potential conduction, suggesting that the role of Ncc69 is to maintain volume or osmotic homeostasis. Drosophila Ncc69 has kinetics similar to human NKCC1, and NKCC1 can rescue Ncc69, suggesting that they function in a conserved physiological mechanism. We show that fray and Ncc69 are coexpressed in nerve glia, interact in a yeast-two-hybrid assay, and have an essentially identical bulging nerve phenotype. We propose that normally functioning nerves generate extracellular solutes that are removed by Ncc69 under the control of Fray. This mechanism may perform a similar role in humans, given that NKCC1 is expressed at the blood-brain barrier.


Asunto(s)
Barrera Hematonerviosa/citología , Proteínas de Drosophila/metabolismo , Espacio Extracelular/fisiología , Neuroglía/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Simportadores/metabolismo , Potenciales de Acción/fisiología , Animales , Animales Modificados Genéticamente , Células Cultivadas , Drosophila/anatomía & histología , Proteínas de Drosophila/genética , Humanos , Técnicas In Vitro , Larva , Microscopía Electrónica de Transmisión/métodos , Modelos Biológicos , Mutación/genética , Conducción Nerviosa/genética , Neuronas/fisiología , Nervios Periféricos/citología , Proteínas Serina-Treonina Quinasas/genética , Simportadores/genética , Técnicas del Sistema de Dos Híbridos , Cotransportadores de K Cl
11.
Mol Biol Cell ; 21(22): 3985-97, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20861303

RESUMEN

The renal-specific Na+-K+-2Cl- cotransporter (NKCC2) is the major salt transport pathway of the apical membrane of the mammalian thick ascending limb of Henle's loop. Here, we analyze the role of the tetraspan protein myelin and lymphocytes-associated protein (MAL)/VIP17 in the regulation of NKCC2. We demonstrated that 1) NKCC2 and MAL/VIP17 colocalize and coimmunoprecipitate in Lilly Laboratories cell porcine kidney cells (LLC-PK1) as well as in rat kidney medullae, 2) a 150-amino acid stretch of NKCC2 C-terminal tail is involved in the interaction with MAL/VIP17, 3) MAL/VIP17 increases the cell surface retention of NKCC2 by attenuating its internalization, and 4) this coincides with an increase in cotransporter phosphorylation. Interestingly, overexpression of MAL/VIP17 in the kidney of transgenic mice results in cysts formation in distal nephron structures consistent with the hypothesis that MAL/VIP17 plays an important role in apical sorting or in maintaining the stability of the apical membrane. The NKCC2 expressed in these mice was highly glycosylated and phosphorylated, suggesting that MAL/VIP17 also is involved in the stabilization of NKCC2 at the apical membrane in vivo. Thus, the involvement of MAL/VIP17 in the activation and surface expression of NKCC2 could play an important role in the regulated absorption of Na+ and Cl- in the kidney.


Asunto(s)
Células Epiteliales/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas de la Mielina/metabolismo , Proteolípidos/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Animales , Western Blotting , Línea Celular , Endocitosis , Humanos , Inmunoprecipitación , Riñón/citología , Células LLC-PK1 , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Transgénicos , Proteínas de la Mielina/genética , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito , Fosforilación , Unión Proteica , Proteolípidos/genética , Interferencia de ARN , Ratas , Ratas Endogámicas WKY , Simportadores de Cloruro de Sodio-Potasio/genética , Miembro 1 de la Familia de Transportadores de Soluto 12 , Porcinos
12.
J Exp Biol ; 213(Pt 9): 1558-66, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20400641

RESUMEN

Euryhaline teleosts such as Atlantic killifish (Fundulus heteroclitus) are able to acclimate to changing environmental salinity by tightly regulating NaCl absorption and secretion across their gills. Many studies have examined the mechanisms responsible for long-term (days) salinity acclimation; however, much remains unknown about the mechanisms of acute (hours) salinity acclimation. In this study, we tested the hypotheses that phosphorylation of the Na(+)-K(+)-Cl(-) cotransporter (NKCC1) located in the basolateral membrane of the gill plays a role in acute salinity acclimation and that changes in NKCC1 phosphorylation are mediated by a cAMP-protein kinase A (cAMP-PKA) pathway. Using a phospho-specific antibody, we determined the time course of changes in total and phosphorylated NKCC1 protein during acclimation to water of various salinities. Long-term (>or=14 days) acclimation of killifish to seawater (SW) and 2x SW resulted in 4- to 6-fold and 5- to 8-fold increases, respectively, in total gill NKCC1 protein relative to fish maintained in freshwater (FW). NKCC1 was found to be between 20% and 70% activated in fish, with lower average activation in fish acclimated to SW and 2x SW compared with FW fish. Increases and decreases in the fractional level of NKCC1 phosphorylation were seen within 1 h of transfer of fish to water of higher and lower salinity, respectively, consistent with a regulatory role of phosphorylation prior to an increase in the biosynthesis of NKCC1; large changes in protein expression of NKCC1 were observed over periods of hours to days. We found that NKCC1 phosphorylation is acutely regulated in the killifish gill in response to changing environmental salinity and that phosphorylation in excised gills increases in response to forskolin stimulation of the cAMP-PKA pathway. The role of phosphorylation is further underscored by the observation that mRNA expression of sterile 20 (Ste20)-related proline-alanine-rich kinase (SPAK) changes with salinity acclimation, being 2.7-fold greater in SW-acclimated killifish relative to FW fish. Overall, these results demonstrate an important role of NKCC1 phosphorylation in the gill of Atlantic killifish during acute salinity acclimation.


Asunto(s)
Aclimatación , Fundulidae/fisiología , Branquias/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Animales , Regulación hacia Abajo , Fundulidae/metabolismo , Fosforilación , ARN Mensajero/genética , Salinidad , Simportadores de Cloruro de Sodio-Potasio/genética , Regulación hacia Arriba
13.
Cell ; 138(3): 525-36, 2009 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-19665974

RESUMEN

Modulation of intracellular chloride concentration ([Cl(-)](i)) plays a fundamental role in cell volume regulation and neuronal response to GABA. Cl(-) exit via K-Cl cotransporters (KCCs) is a major determinant of [Cl(-)](I); however, mechanisms governing KCC activities are poorly understood. We identified two sites in KCC3 that are rapidly dephosphorylated in hypotonic conditions in cultured cells and human red blood cells in parallel with increased transport activity. Alanine substitutions at these sites result in constitutively active cotransport. These sites are highly phosphorylated in plasma membrane KCC3 in isotonic conditions, suggesting that dephosphorylation increases KCC3's intrinsic transport activity. Reduction of WNK1 expression via RNA interference reduces phosphorylation at these sites. Homologous sites are phosphorylated in all human KCCs. KCC2 is partially phosphorylated in neonatal mouse brain and dephosphorylated in parallel with KCC2 activation. These findings provide insight into regulation of [Cl(-)](i) and have implications for control of cell volume and neuronal function.


Asunto(s)
Simportadores/química , Simportadores/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Humanos , Ratones , Datos de Secuencia Molecular , Fosforilación , Alineación de Secuencia , Cotransportadores de K Cl
14.
J Neurosci ; 29(32): 9943-54, 2009 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-19675228

RESUMEN

Chloride influx through GABA-gated chloride channels, the primary mechanism by which neural activity is inhibited in the adult mammalian brain, depends on chloride gradients established by the potassium chloride cotransporter KCC2. We used a genetic screen to identify genes important for inhibition of the hermaphrodite-specific motor neurons (HSNs) that stimulate Caenorhabditis elegans egg-laying behavior and discovered mutations in a potassium chloride cotransporter, kcc-2. Functional analysis indicates that, like mammalian KCCs, C. elegans KCC-2 transports chloride, is activated by hypotonic conditions, and is inhibited by the loop diuretic furosemide. KCC-2 appears to establish chloride gradients required for the inhibitory effects of GABA-gated and serotonin-gated chloride channels on C. elegans behavior. In the absence of KCC-2, chloride gradients appear to be altered in neurons and muscles such that normally inhibitory signals become excitatory. kcc-2 is transcriptionally upregulated in the HSN neurons during synapse development. Loss of KCC-2 produces a decrease in the synaptic vesicle population within mature HSN synapses, which apparently compensates for a lack of HSN inhibition, resulting in normal egg-laying behavior. Thus, KCC-2 coordinates the development of inhibitory neurotransmission with synapse maturation to produce mature neural circuits with appropriate activity levels.


Asunto(s)
Caenorhabditis elegans/fisiología , Simportadores/metabolismo , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Animales , Proteínas de Caenorhabditis elegans/metabolismo , Cloruros/metabolismo , Furosemida/farmacología , Soluciones Hipotónicas , Neuronas Motoras/fisiología , Músculos/fisiología , Mutación , Receptores Acoplados a Proteínas G/metabolismo , Homología de Secuencia , Conducta Sexual Animal/fisiología , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Simportadores/antagonistas & inhibidores , Simportadores/genética , Vesículas Sinápticas/fisiología , Regulación hacia Arriba , Cotransportadores de K Cl
15.
Mol Biol Cell ; 19(10): 4341-51, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18667527

RESUMEN

The renal Na-K-Cl cotransporter (NKCC2) is selectively expressed in the apical membranes of cells of the mammalian kidney, where it is the target of the clinically important loop diuretics. In contrast, the "secretory" NKCC1 cotransporter is localized in the basolateral membranes of many epithelia. To identify the sorting signal(s) that direct trafficking of NKCCs, we generated chimeras between the two isoforms and expressed these constructs in polarized renal epithelial cell lines. This analysis revealed an amino acid stretch in NKCC2 containing apical sorting information. The NKCC1 C terminus contains a dileucine motif that constitutes the smallest essential component of its basolateral sorting signal. NKCC1 lacking this motif behaves as an apical protein. Examination of the NKCC gene structure reveals that this dileucine motif is encoded by an additional exon in NKCC1 absent in NKCC2. Phylogenetic analysis of this exon suggests that the evolutionary loss of this exon from the gene encoding the basolateral NKCC1 constitutes a novel mechanism that accounts for the apical sorting of the protein encoded by the NKCC2 gene.


Asunto(s)
Células Epiteliales/metabolismo , Exones , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Secuencias de Aminoácidos , Animales , Biotinilación , Línea Celular , Perros , Humanos , Riñón/metabolismo , Microscopía Confocal , Modelos Biológicos , Isoformas de Proteínas , Estructura Terciaria de Proteína , Transfección
16.
J Biol Chem ; 283(5): 2663-74, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18045874

RESUMEN

To examine the structure and function of the Na-K-Cl cotransporter, NKCC1, we tagged the transporter with cyan (CFP) and yellow (YFP) fluorescent proteins and measured fluorescence resonance energy transfer (FRET) in stably expressing human embryonic kidney cell lines. Fluorescent protein tags were added at the N-terminal residue between the regulatory domain and the membrane domain and within a poorly conserved region of the C terminus. Both singly and doubly tagged NKCC1s were appropriately trafficked to the cell membrane and were fully functional; regulation was normal except when YFP was inserted near the regulatory domain, in which case activation occurred only upon incubation with calyculin A. Quenching of YFP fluorescence by Cl(-) provided a ratiometric indicator of intracellular [Cl(-)]. All of the CFP/YFP NKCC pairs exhibited some level of FRET, demonstrating the presence of dimers or higher multimers in functioning NKCC1. With YFP near the regulatory domain and CFP in the C terminus, we recorded a 6% FRET change signaling the regulatory phosphorylation event. On the other hand, when the probe was placed at the extreme N terminus, such changes were not seen, presumably due to the length and predicted flexibility of the N terminus. Substantial FRET changes were observed contemporaneous with cell volume changes, possibly reflective of an increase in molecular crowding upon cell shrinkage.


Asunto(s)
Simportadores de Cloruro de Sodio-Potasio/química , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Tamaño de la Célula , Cloruros/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Modelos Moleculares , Fosforilación , Conformación Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Tiburones/genética , Tiburones/metabolismo , Simportadores de Cloruro de Sodio-Potasio/genética , Miembro 1 de la Familia de Transportadores de Soluto 12 , Transfección
17.
J Biol Chem ; 282(9): 6540-7, 2007 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-17186942

RESUMEN

Three alternatively spliced variants of the renal Na-K-Cl cotransporter (NKCC2) are found in distinct regions of the thick ascending limb of the mammalian kidney; these variants mediate Na(+)K(+)2Cl(-) transport with different ion affinities. Here, we examine the specific residues involved in the variant-specific affinity differences, utilizing a mutagenic approach to change the NKCC2B variant into the A or F variant, with functional expression in Xenopus oocytes. The splice region contains the second transmembrane domain (TM2) and the putative intracellular loop (ICL1) connecting TM2 and TM3. It is found that the B variant is functionally changed to the F variant by replacement of six residues, half of the effect brought about by three TM2 residues and half by three ICL1 residues. The involvement of the ICL1 residues strongly suggests that this region of ICL1 may actually be part of a membrane-embedded domain. Changing six residues is also sufficient to bring about the smaller functional change from the B to the A variant; three residues in TM2 appear to be primarily responsible, two of which correspond to residues involved in the B-to-F changes. A B-variant mutation reported in a mild case of Bartter disease was found to render the cotransporter inactive. These results identify the combination of amino acid variations responsible for the differences among the three splice variants of NKCC2, and they support a model in which a reentrant loop following TM2 contributes to the chloride binding and translocation domains.


Asunto(s)
Riñón/química , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Sustitución de Aminoácidos , Animales , Transporte Biológico , Cloro/metabolismo , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Isoformas de Proteínas , Estructura Terciaria de Proteína , Simportadores de Cloruro de Sodio-Potasio/genética , Miembro 1 de la Familia de Transportadores de Soluto 12
18.
Am J Physiol Cell Physiol ; 290(2): C492-8, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16192298

RESUMEN

Serum- and glucocorticoid-induced kinase 1 (SGK1) is thought to be an important regulator of Na(+) reabsorption in the kidney. It has been proposed that SGK1 mediates the effects of aldosterone on transepithelial Na(+) transport. Previous studies have shown that SGK1 increases Na(+) transport and epithelial Na(+) channel (ENaC) activity in the apical membrane of renal epithelial cells. SGK1 has also been implicated in the modulation of Na(+)-K(+)-ATPase activity, the transporter responsible for basolateral Na(+) efflux, although this observation has not been confirmed in renal epithelial cells. We examined Na(+)-K(+)-ATPase function in an A6 renal epithelial cell line that expresses SGK1 under the control of a tetracycline-inducible promoter. The results showed that expression of a constitutively active mutant of SGK1 (SGK1(T)(S425D)) increased the transport activity of Na(+)-K(+)-ATPase 2.5-fold. The increase in activity was a direct consequence of activation of the pump itself. The onset of Na(+)-K(+)-ATPase activation was observed between 6 and 24 h after induction of SGK1 expression, a delay that is significantly longer than that required for activation of ENaC in the same cell line (1 h). SGK1 and aldosterone stimulated the Na(+) pump synergistically, indicating that the pathways mediated by these molecules operate independently. This observation was confirmed by demonstrating that aldosterone, but not SGK1(T)(S425D), induced an approximately 2.5-fold increase in total protein and plasma membrane Na(+)-K(+)-ATPase alpha(1)-subunit abundance. We conclude that aldosterone increases the abundance of Na(+)-K(+)-ATPase, whereas SGK1 may activate existing pumps in the membrane in response to chronic or slowly acting stimuli.


Asunto(s)
Células Epiteliales/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Túbulos Renales Distales/citología , Proteínas Serina-Treonina Quinasas/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Aldosterona/metabolismo , Animales , Calnexina/metabolismo , Línea Celular , Membrana Celular/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/metabolismo , Células Epiteliales/citología , Proteínas Inmediatas-Precoces/genética , Ouabaína/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Radioisótopos de Rubidio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , Xenopus laevis
19.
Am J Physiol Cell Physiol ; 289(6): C1492-501, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16079189

RESUMEN

Brain edema that forms during the early stages of stroke involves increased transport of Na+ and Cl- across an intact blood-brain barrier (BBB). Our previous studies have shown that a luminal BBB Na+-K+-Cl- cotransporter is stimulated by conditions present during ischemia and that inhibition of the cotransporter by intravenous bumetanide greatly reduces edema formation in the rat middle cerebral artery occlusion model of stroke. The present study focused on investigating the effects of hypoxia, which develops rapidly in the brain during ischemia, on the activity and expression of the BBB Na+-K+-Cl- cotransporter, as well as on Na+-K+-ATPase activity, cell ATP content, and intracellular volume. Cerebral microvascular endothelial cells (CMECs) were assessed for Na+-K+-Cl- cotransporter and Na+-K+-ATPase activities as bumetanide-sensitive and ouabain-sensitive 86Rb influxes, respectively. ATP content was assessed by luciferase assay and intracellular volume by [3H]-3-O-methyl-D-glucose and [14C]-sucrose equilibration. We found that 30-min exposure of CMECs to hypoxia ranging from 7.5% to 0.5% O2 (vs. 19% normoxic O2) significantly increased cotransporter activity as did 7.5% or 2% O2 for up to 2 h. This was not associated with reduction in Na+-K+-ATPase activity or ATP content. CMEC intracellular volume increased only after 4 to 5 h of hypoxia. Furthermore, glucose and pyruvate deprivation increased cotransporter activity under both normoxic and hypoxic conditions. Finally, we found that hypoxia increased phosphorylation but not abundance of the cotransporter protein. These findings support the hypothesis that hypoxia stimulation of the BBB Na+-K+-Cl- cotransporter contributes to ischemia-induced brain edema formation.


Asunto(s)
Corteza Cerebral/irrigación sanguínea , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Transporte Biológico Activo , Bovinos , Hipoxia de la Célula , Tamaño de la Célula , Células Cultivadas , Células Endoteliales/citología , Endotelio Vascular/citología , Microcirculación/citología , Microcirculación/metabolismo , Microcirculación/patología , Fosforilación , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
20.
Am J Physiol Renal Physiol ; 289(6): F1341-5, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16077079

RESUMEN

Short-term regulation of members of the Na-K-Cl cotransporter family takes place by phosphorylation/dephosphorylation events. Three NH(2)-terminal threonines have been previously identified as phosphoacceptors involved in activation of the ubiquitous/secretory Na-K-Cl cotransporter (NKCC1). In this study, we demonstrate that the corresponding threonines are also involved in the regulation of the renal Na-K-Cl cotransporter (NKCC2). The transport activity of NKCC2, exogenously expressed in Xenopus laevis oocytes, is shown to be stimulated by hypertonicity. Mutagenic analysis demonstrated that threonines T99, T104, and T117 comprise a regulatory domain responsible for the activation of NKCC2 in hypertonic solutions: although none of the threonines was found to be individually necessary or sufficient for regulation, the three residues together are required to obtain the full hypertonic response. Under isotonic and hypotonic conditions, NKCC2 retains 50% of its activity in the absence of phosphorylation of the threonine-regulatory domain. Selective deletions of peptide segments revealed only a minor role for the NH(2)-terminal cytosolic domain of NKCC2 upstream of the threonine regulatory domain, including the recently identified proline alanine-rich Ste-20-related kinase-binding motif. A chimeric NKCC containing the first 104 amino acids of NKCC1 on the NKCC2 backbone behaved essentially the same as NKCC2, further arguing against a major role for this upstream region in NKCC2 regulation.


Asunto(s)
Simportadores de Cloruro de Sodio-Potasio/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Femenino , Regulación de la Expresión Génica , Humanos , Mutagénesis , Oocitos/metabolismo , Concentración Osmolar , Fosforilación , Isoformas de Proteínas , Conejos , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12 , Treonina/genética , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA