Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Methods Mol Biol ; 1432: 223-42, 2016.
Article in English | MEDLINE | ID: mdl-27485339

ABSTRACT

This protocol describes the isolation of recombinant human and mammalian membrane proteins expressed in Xenopus laevis frog oocytes for structural studies. The cDNA-derived cRNA of the desired genes is injected into several hundreds of oocytes, which are incubated for several days to allow protein expression. Recombinant proteins are then purified via affinity chromatography. The novelty of this method comes from the design of a plasmid that produces multi-tagged proteins and, most importantly, the development of a protocol for efficiently discarding lipids, phospholipids, and lipoproteins from the oocyte egg yolk, which represent the major contaminants in protein purifications. Thus, the high protein purity and good yield obtained from this method allows protein structure determination by transmission electron microscopy of single detergent-solubilized protein particles and of 2D crystals of membrane protein embedded in lipid bilayers. Additionally, a radiotracer assay for functional analysis of the expressed target proteins in oocytes is described. Overall, this method is a valuable option for structural studies of mammalian and particularly human proteins, for which other expression systems often fail.


Subject(s)
Mammals/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Xenopus laevis/genetics , Animals , Chromatography, Affinity , Cloning, Molecular , DNA, Complementary/genetics , Female , Genetic Vectors/administration & dosage , Humans , Mammals/genetics , Membrane Proteins/genetics , Oocytes/metabolism , Plasmids/genetics , Protein Conformation , RNA, Complementary/genetics , Recombinant Proteins/metabolism , Xenopus laevis/metabolism
4.
Nat Med ; 19(11): 1524-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24097188

ABSTRACT

The K(+)-Cl(-) cotransporter KCC2 is responsible for maintaining low Cl(-) concentration in neurons of the central nervous system (CNS), which is essential for postsynaptic inhibition through GABA(A) and glycine receptors. Although no CNS disorders have been associated with KCC2 mutations, loss of activity of this transporter has emerged as a key mechanism underlying several neurological and psychiatric disorders, including epilepsy, motor spasticity, stress, anxiety, schizophrenia, morphine-induced hyperalgesia and chronic pain. Recent reports indicate that enhancing KCC2 activity may be the favored therapeutic strategy to restore inhibition and normal function in pathological conditions involving impaired Cl(-) transport. We designed an assay for high-throughput screening that led to the identification of KCC2 activators that reduce intracellular chloride concentration ([Cl(-)]i). Optimization of a first-in-class arylmethylidine family of compounds resulted in a KCC2-selective analog (CLP257) that lowers [Cl(-)]i. CLP257 restored impaired Cl(-) transport in neurons with diminished KCC2 activity. The compound rescued KCC2 plasma membrane expression, renormalized stimulus-evoked responses in spinal nociceptive pathways sensitized after nerve injury and alleviated hypersensitivity in a rat model of neuropathic pain. Oral efficacy for analgesia equivalent to that of pregabalin but without motor impairment was achievable with a CLP257 prodrug. These results validate KCC2 as a druggable target for CNS diseases.


Subject(s)
Analgesics/therapeutic use , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Symporters/agonists , Thiazolidines/therapeutic use , Analgesics/chemistry , Animals , CHO Cells , Chlorides/metabolism , Cricetinae , Cricetulus , Disease Models, Animal , HEK293 Cells , High-Throughput Screening Assays , Humans , Intracellular Fluid/metabolism , Ion Transport/drug effects , Male , Neuralgia/drug therapy , Neuralgia/metabolism , Rats , Rats, Sprague-Dawley , Thiazolidines/chemistry , K Cl- Cotransporters
5.
PLoS One ; 6(7): e21901, 2011.
Article in English | MEDLINE | ID: mdl-21760919

ABSTRACT

Structural analyses of heterologously expressed mammalian membrane proteins remain a great challenge given that microgram to milligram amounts of correctly folded and highly purified proteins are required. Here, we present a novel method for the expression and affinity purification of recombinant mammalian and in particular human transport proteins in Xenopus laevis frog oocytes. The method was validated for four human and one murine transporter. Negative stain transmission electron microscopy (TEM) and single particle analysis (SPA) of two of these transporters, i.e., the potassium-chloride cotransporter 4 (KCC4) and the aquaporin-1 (AQP1) water channel, revealed the expected quaternary structures within homogeneous preparations, and thus correct protein folding and assembly. This is the first time a cation-chloride cotransporter (SLC12) family member is isolated, and its shape, dimensions, low-resolution structure and oligomeric state determined by TEM, i.e., by a direct method. Finally, we were able to grow 2D crystals of human AQP1. The ability of AQP1 to crystallize was a strong indicator for the structural integrity of the purified recombinant protein. This approach will open the way for the structure determination of many human membrane transporters taking full advantage of the Xenopus laevis oocyte expression system that generally yields robust functional expression.


Subject(s)
Aquaporin 1/chemistry , Aquaporin 1/metabolism , Oocytes/metabolism , Symporters/chemistry , Symporters/metabolism , Xenopus laevis/metabolism , Animals , Aquaporin 1/isolation & purification , Aquaporin 1/ultrastructure , Blotting, Western , Cell Membrane/metabolism , Crystallization , Egg Yolk/metabolism , Electrophoresis, Polyacrylamide Gel , Genetic Vectors/genetics , Humans , Negative Staining , Oocytes/cytology , Protein Transport , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Silver Staining , Symporters/isolation & purification , Symporters/ultrastructure
6.
J Biol Chem ; 286(13): 11242-53, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21257749

ABSTRACT

Renal excretion of citrate, an inhibitor of calcium stone formation, is controlled mainly by reabsorption via the apical Na(+)-dicarboxylate cotransporter NaDC1 (SLC13A2) in the proximal tubule. Recently, it has been shown that the protein phosphatase calcineurin inhibitors cyclosporin A (CsA) and FK-506 induce hypocitraturia, a risk factor for nephrolithiasis in kidney transplant patients, but apparently through urine acidification. This suggests that these agents up-regulate NaDC1 activity. Using the Xenopus lævis oocyte and HEK293 cell expression systems, we examined first the effect of both anti-calcineurins on NaDC1 activity and expression. While FK-506 had no effect, CsA reduced NaDC1-mediated citrate transport by lowering heterologous carrier expression (as well as endogenous carrier expression in HEK293 cells), indicating that calcineurin is not involved. Given that CsA also binds specifically to cyclophilins, we determined next whether such proteins could account for the observed changes by examining the effect of selected cyclophilin wild types and mutants on NaDC1 activity and cyclophilin-specific siRNA. Interestingly, our data show that the cyclophilin isoform B is likely responsible for down-regulation of carrier expression by CsA and that it does so via its chaperone activity on NaDC1 (by direct interaction) rather than its rotamase activity. We have thus identified for the first time a regulatory partner for NaDC1, and have gained novel mechanistic insight into the effect of CsA on renal citrate transport and kidney stone disease, as well as into the regulation of membrane transporters in general.


Subject(s)
Cyclophilins/metabolism , Dicarboxylic Acid Transporters/metabolism , Molecular Chaperones/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Animals , Calcineurin/genetics , Calcineurin/metabolism , Citric Acid/metabolism , Cyclophilins/genetics , Cyclosporine/pharmacology , Dicarboxylic Acid Transporters/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression/drug effects , HEK293 Cells , Humans , Immunosuppressive Agents/pharmacology , Kidney/metabolism , Kidney Calculi/genetics , Kidney Calculi/metabolism , Molecular Chaperones/genetics , Mutation , Oocytes , Organic Anion Transporters, Sodium-Dependent/genetics , Protein Transport/drug effects , Protein Transport/genetics , Symporters/genetics , Tacrolimus/pharmacology , Xenopus laevis
7.
Cell Calcium ; 49(1): 43-55, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21146870

ABSTRACT

TRPV6 belongs to the vanilloid family of the transient receptor potential channel (TRP) superfamily. This calcium-selective channel is highly expressed in the duodenum and the placenta, being responsible for calcium absorption in the body and fetus. Previous observations have suggested that TRPV6 is not only permeable to calcium but also to other divalent cations in epithelial tissues. In this study, we tested whether TRPV6 is indeed also permeable to cations such as zinc and cadmium. We found that the basal intracellular calcium concentration was higher in HEK293 cells transfected with hTRPV6 than in non-transfected cells, and that this difference almost disappeared in nominally calcium-free solution. Live cell imaging experiments with Fura-2 and NewPort Green DCF showed that overexpression of human TRPV6 increased the permeability for Ca(2+), Ba(2+), Sr(2+), Mn(2+), Zn(2+), Cd(2+), and interestingly also for La(3+) and Gd(3+). These results were confirmed using the patch clamp technique. (45)Ca uptake experiments showed that cadmium, lanthanum and gadolinium were also highly efficient inhibitors of TRPV6-mediated calcium influx at higher micromolar concentrations. Our results suggest that TRPV6 is not only involved in calcium transport but also in the transport of other divalent cations, including heavy metal ions, which may have toxicological implications.


Subject(s)
Cell Membrane Permeability/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Metals, Heavy/pharmacology , TRPV Cation Channels/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadmium/pharmacology , Calcium/metabolism , Cations , Cell Line, Tumor , Female , Fluorescent Dyes/metabolism , Fura-2/metabolism , HEK293 Cells , Humans , Imaging, Three-Dimensional , Intracellular Space/drug effects , Intracellular Space/metabolism , Ion Channel Gating/drug effects , Patch-Clamp Techniques , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Time Factors , Zinc/pharmacology
8.
J Cell Physiol ; 219(3): 787-96, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19206159

ABSTRACT

It has long been stated that the K(+)-Cl(-) cotransporters (KCCs) are activated during cell swelling through dephosphorylation of their cytoplasmic domains by a protein phosphatase (PP) but that other enzymes are involved by targeting this PP or the KCCs directly. To date, however, the role of signaling intermediates in KCC regulation has been deduced from indirect evidence rather than in vitro phosphorylation studies, and examined after simulation of ion transport through cell swelling or N-ethylmaleimide treatment. In this study, the oocyte expression system was used to examine the effects of changes in cell volume (C(VOL)) and intracellular [Cl(-)] ([Cl(-)](i)) on the activity and phosphorylation levels (P(LEV)) of KCC4, and determine whether these effects are mediated by PP1 or phorbol myristate acetate (PMA)-sensitive effectors. We found that (1) low [Cl(-)](i) or low C(VOL) leads to decreased activity but increased P(LEV), (2) high C(VOL) leads to increased activity but no decrease in P(LEV) and (3) calyculin A (Cal A) or PMA treatment leads to decreased activity but no increase in P(LEV). Thus, we have shown for the first time that one of the KCCs can be regulated through direct phosphorylation, that changes in [Cl(-)](i) or C(VOL) modify the activity of signaling enzymes at carrier sites, and that the effectors directly involved do not include a Cal A-sensitive PP in contrast to the widely held view. J. Cell. Physiol. 219: 787-796, 2009. (c) 2009 Wiley-Liss, Inc.


Subject(s)
Chlorides/metabolism , Symporters/metabolism , Animals , Cell Size , Female , In Vitro Techniques , Intracellular Fluid/metabolism , Marine Toxins , Mice , Mutagenesis, Site-Directed , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Oxazoles/pharmacology , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Symporters/chemistry , Symporters/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Xenopus laevis
9.
J Biol Chem ; 282(25): 18083-18093, 2007 Jun 22.
Article in English | MEDLINE | ID: mdl-17462999

ABSTRACT

Little is known regarding the quaternary structure of cation-Cl- cotransporters (CCCs) except that the Na+-dependent CCCs can exist as homooligomeric units. Given that each of the CCCs exhibits unique functional properties and that several of these carriers coexist in various cell types, it would be of interest to determine whether the four K+-Cl- cotransporter (KCC) isoforms and their splice variants can also assemble into such units and, more importantly, whether they can form heterooligomers by interacting with each other or with the secretory Na+-K+-Cl- cotransporter (NKCC1). In the present work, we have addressed these questions by conducting two groups of analyses: 1) yeast two-hybrid and pull-down assays in which CCC-derived protein segments were used as both bait and prey and 2) coimmunoprecipitation and functional studies of intact CCCs coexpressed in Xenopus laevis oocytes. Through a combination of such analyses, we have found that KCC2 and KCC4 could adopt various oligomeric states (in the form of KCC2-KCC2, KCC4-KCC4, KCC2-KCC4, and even KCC4-NKCC1 complexes), that their carboxyl termini were probably involved in carrier assembly, and that the KCC4-NKCC1 oligomers, more specifically, could deploy unique functional features. Through additional coimmunoprecipitation studies, we have also found that KCC1 and KCC3 had the potential of assembling into various types of CCC-CCC oligomers as well, although the interactions uncovered were not characterized as extensively, and the protein segments involved were not identified in yeast two-hybrid assays. Taken together, these findings could change our views on how CCCs operate or are regulated in animal cells by suggesting, in particular, that cation-Cl- cotransport achieves higher levels of functional diversity than foreseen.


Subject(s)
Sodium-Potassium-Chloride Symporters/metabolism , Symporters/physiology , Animals , Biological Transport , Carrier Proteins/chemistry , Epitopes/chemistry , Glutathione Transferase/metabolism , Immunoprecipitation , Ion Transport , Oocytes/metabolism , Protein Isoforms , Sodium-Potassium-Chloride Symporters/chemistry , Solute Carrier Family 12, Member 2 , Symporters/metabolism , Two-Hybrid System Techniques , Xenopus laevis , K Cl- Cotransporters
10.
J Biol Chem ; 281(23): 15959-69, 2006 Jun 09.
Article in English | MEDLINE | ID: mdl-16595678

ABSTRACT

The K+-Cl- cotransporter (KCC) isoforms constitute a functionally heterogeneous group of ion carriers. Emerging evidence suggests that the C terminus (Ct) of these proteins is important in conveying isoform-specific traits and that it may harbor interacting sites for 4beta-phorbol 12-myristate 13-acetate (PMA)-induced effectors. In this study, we have generated KCC2-KCC4 chimeras to identify key functional domains in the Ct of these carriers and single point mutations to determine whether canonical protein kinase C sites underlie KCC2-specific behaviors. Functional characterization of wild-type (wt) and mutant carriers in Xenopus laevis oocytes showed for the first time that the KCCs do not exhibit similar sensitivities to changes in osmolality and that this distinguishing feature as well as differences in transport activity under both hypotonic and isotonic conditions are in part determined by the residue composition of the distal Ct. At the same time, several mutations in this domain and in the proximal Ct of the KCCs were found to generate allosteric-like effects, suggesting that the regions analyzed are important in defining conformational ensembles and that isoform-specific structural configurations could thus account for variant functional traits as well. Characterization of the other mutants in this work showed that KCC2 is not inhibited by PMA through phosphorylation of its canonical protein kinase C sites. Intriguingly, however, the substitutions N728S and S940A were seen to alter the PMA effect paradoxically, suggesting again that allosteric changes in the Ct are important determinants of transport activity and, furthermore, that the structural configuration of this domain can convey specific functional traits by defining the accessibility of cotransporter sites to regulatory intermediates such as PMA-induced effectors.


Subject(s)
Symporters/physiology , Animals , Base Sequence , Blotting, Western , DNA Primers , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Symporters/chemistry , Xenopus laevis , K Cl- Cotransporters
11.
J Gen Physiol ; 126(4): 325-37, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16157691

ABSTRACT

The absorptive Na(+)-K(+)-Cl(-) cotransporter (NKCC2) is a polytopic protein that forms homooligomeric complexes in the apical membrane of the thick ascending loop of Henle (TAL). It occurs in at least four splice variants (called B, A, F, and AF) that are identical to one another except for a short region in the membrane-associated domain. Although each of these variants exhibits unique functional properties and distributions along the TAL, their teleological purpose and structural organization remain poorly defined. In the current work, we provide additional insight in these regards by showing in mouse that the administration of either furosemide or an H(2)O-rich diet, which are predicted to alter NKCC2 expression in the TAL, exerts differential effects on mRNA levels for the variants, increasing those of A (furosemide) but decreasing those of F and AF (furosemide or H(2)O). Based on a yeast two-hybrid mapping analysis, we also show that the formation of homooligomeric complexes is mediated by two self-interacting domains in the COOH terminus (residues 671 to 816 and 910 to 1098), and that these complexes could probably include more than one type of variant. Taken together, the data reported here suggest that A, F, and AF each play unique roles that are adapted to specific physiological needs, and that the accomplishment of such roles is coordinated through the splicing machinery as well as complex NKCC2-NKCC2 interactions.


Subject(s)
Alternative Splicing , Kidney/metabolism , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Fluorescent Antibody Technique , Furosemide/administration & dosage , Furosemide/pharmacology , Humans , Loop of Henle/metabolism , Male , Mice , Mice, Inbred C3H , Oocytes , Protein Isoforms/genetics , RNA, Messenger/metabolism , Sharks , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Sodium-Potassium-Chloride Symporters/chemistry , Solute Carrier Family 12, Member 1 , Solute Carrier Family 12, Member 2 , Transfection , Two-Hybrid System Techniques , Water/administration & dosage , Xenopus laevis
12.
J Biol Chem ; 280(37): 32555-63, 2005 Sep 16.
Article in English | MEDLINE | ID: mdl-16027154

ABSTRACT

Two variants of the renal Na(+)-K(+)-Cl(-) cotransporter (NKCC2), called NKCC2A and NKCC2F, display marked differences in Na(+), Rb(+), and Cl(-) affinities, yet are identical to one another except for a 23-residue membrane-associated domain that is derived from alternatively spliced exons. The proximal portion of these exons is predicted to encode the second transmembrane domain (tm2) in the form of an alpha-helix, and the distal portion, part of the following connecting segment (cs1a). In recent studies, we have taken advantage of the A-F differences in kinetic behavior to determine which regions in tm2-cs1a are involved in ion transport. Functional characterizations of chimeras in which tm2 or cs1a were interchanged between the variants showed that both regions are important in specifying ion affinities, but did not allow delineating the contribution of individual residues. Here, we have extended these structure-function analyses by studying additional mutants in which variant residues between A and F were interchanged individually in the tm2-cs1a region (amino acid number 216, 220, 223, 229, or 233 in NKCC2). None of the substitutions were found to affect K(m (C1-)), suggesting that the affinity difference for anion transport is conveyed by a combination of variant residues in this domain. However, 2 substitutions in the tm2 of F were found to affect cation constants specifically; interestingly, one of these mutations (residue 216) only affected K(m (Rb+)) while the other (residue 220) only affected K(m (Na+)). We have thus identified two novel residues in NKCC2 that play a key role in cation transport. Because such residues should be adjacent to one another on the vertical axis of the tm2 alpha-helix, our results imply, furthermore, that the ion transport sites in NKCC2 could be physically linked.


Subject(s)
Cations , Ions , Sodium-Potassium-Chloride Symporters/chemistry , Animals , Binding Sites , Biological Transport , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Exons , Kinetics , Mutation , Oligonucleotides/chemistry , Oocytes/metabolism , Point Mutation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Rubidium/chemistry , Sodium-Potassium-Chloride Symporters/metabolism , Sodium-Potassium-Chloride Symporters/physiology , Solute Carrier Family 12, Member 1 , Squalus , Structure-Activity Relationship , Xenopus laevis/metabolism
13.
J Biol Chem ; 279(46): 48449-56, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15347682

ABSTRACT

The first isoform of the Na(+)-K(+)-Cl(-) cotransporter (NKCC1) is of central importance for the control of cellular ion concentration and epithelium-mediated salt secretion. Several studies have established that a change in intracellular [Cl(-)] (Cl(-)(i)) represents a key signaling mechanism by which NKCC1-induced Cl(-) movement is autoregulated and by which Cl(-) entry and exit on opposite sides of polarized cells are coordinated. Although this signaling mechanism is coupled to a pathway that leads to post-translational modification of the carrier, no unifying model currently accounts for the ion dependence of NKCC1 regulation. In this paper, evidence is presented for the first time that hsp90 associates with the cytosolic C terminus of NKCC1, probably when the carrier is predominantly in its unfolded form during early biogenesis. Evidence is also presented that the Cl(-)(i)-dependent regulatory pathway can be activated by a thermal stress but that it is no longer operational if NKCC1-expressing cells are pretreated with geldanamycin, an antibiotic that inhibits hsp90, albeit nonspecifically. Taken together, our data indicate that binding of hsp90 to NKCC1 may be required for Na(+)-K(+)-Cl(-) cotransport to occur at the cell surface and that it could play an important role in ion-dependent signaling mechanisms, insofar as the maneuvers that were used to alter the expression or activity of the chaperone do not exert their main effect by inducing other cellular events such as the unfolded protein response. Further studies will be required to elucidate the functional relevance of this novel interaction.


Subject(s)
Chlorides/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Benzoquinones , Cell Line , Enzyme Inhibitors/metabolism , HSP90 Heat-Shock Proteins/genetics , Humans , Lactams, Macrocyclic , Molecular Chaperones/genetics , Protein Binding , Protein Conformation , Quinones/metabolism , Rubidium Radioisotopes/metabolism , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 2 , Squalus acanthias , Temperature , Two-Hybrid System Techniques
14.
J Biol Chem ; 279(7): 5648-54, 2004 Feb 13.
Article in English | MEDLINE | ID: mdl-14645215

ABSTRACT

The 2nd transmembrane domain (tm) of the secretory Na(+)-K(+)-Cl(-) cotransporter (NKCC1) and of the kidney-specific isoform (NKCC2) has been shown to play an important role in cation transport. For NKCC2, by way of illustration, alternative splicing of exon 4, a 96-bp sequence from which tm2 is derived, leads to the formation of the NKCC2A and F variants that both exhibit unique affinities for cations. Of interest, the NKCC2 variants also exhibit substantial differences in Cl- affinity as well as in the residue composition of the first intracellular connecting segment (cs1a), which immediately follows tm2 and which too is derived from exon 4. In this study, we have prepared chimeras of the shark NKCC2A and F (saA and saF) to determine whether cs1a could play a role in Cl- transport; here, tm2 or cs1a in saF was replaced by the corresponding domain from saA (generating saA/F or saF/A, respectively). Functional analyses of these chimeras have shown that cs1a-specific residues account for most of the A-F difference in Cl- affinity. For example, Km(Cl-)s were approximately 8 mm for saF/A and saA, and approximately 70 mm for saA/F and saF. Intriguingly, variant residues in cs1a also affected cation transport; here, Km(Na+)s for the chimeras and for saA were all approximately 20 mM, and Km(Rb+) all approximately 2 mM. Regarding tm2, our studies have confirmed its importance in cation transport and have also identified novel properties for this domain. Taken together, our results demonstrate for the first time that an intracellular loop in NKCC contributes to the transport process perhaps by forming a flexible structure that positions itself between membrane spanning domains.


Subject(s)
Chlorine/chemistry , Sodium-Potassium-Chloride Symporters/chemistry , Alternative Splicing , Amino Acid Sequence , Animals , Binding Sites , Bumetanide/pharmacology , Cations , Cell Membrane/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Exons , Genetic Vectors , Ions , Kinetics , Models, Biological , Molecular Sequence Data , Mutation , Oocytes/metabolism , Protein Isoforms , Protein Structure, Tertiary , Rubidium/pharmacology , Sequence Homology, Amino Acid , Sharks , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 1 , Xenopus , Xenopus laevis
16.
Am J Physiol Renal Physiol ; 285(1): F68-78, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12657561

ABSTRACT

The Na(+)-K(+)-Cl(-) cotransporters (NKCCs), which belong to the cation-Cl(-) cotransporter (CCC) family, are able to translocate NH4(+) across cell membranes. In this study, we have used the oocyte expression system to determine whether the K(+)-Cl(-) cotransporters (KCCs) can also transport NH4(+) and whether they play a role in pH regulation. Our results demonstrate that all of the CCCs examined (NKCC1, NKCC2, KCC1, KCC3, and KCC4) can promote NH4(+) translocation, presumably through binding of the ion at the K(+) site. Moreover, kinetic studies for both NKCCs and KCCs suggest that NH4(+) is an excellent surrogate of Rb(+) or K(+) and that NH4(+) transport and cellular acidification resulting from CCC activity are relevant physiologically. In this study, we have also found that CCCs are strongly and differentially affected by changes in intracellular pH (independently of intracellular [NH4(+)]). Indeed, NKCC2, KCC1, KCC2, and KCC3 are inhibited at intracellular pH <7.5, whereas KCC4 is activated. These results indicate that certain CCC isoforms may be specialized to operate in acidic environments. CCC-mediated NH4(+) transport could bear great physiological implication given the ubiquitous distribution of these carriers.


Subject(s)
Quaternary Ammonium Compounds/metabolism , Symporters/metabolism , Amino Acid Sequence , Animals , Biological Transport, Active , Gene Expression , Humans , Hydrogen-Ion Concentration , Mice , Molecular Sequence Data , Oocytes , Rabbits , Rats , Rubidium/metabolism , Sequence Alignment , Symporters/chemistry , Xenopus laevis , K Cl- Cotransporters
SELECTION OF CITATIONS
SEARCH DETAIL
...