Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Science ; 366(6464): 505-508, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31649201

ABSTRACT

Cation-chloride cotransporters (CCCs) mediate the coupled, electroneutral symport of cations with chloride across the plasma membrane and are vital for cell volume regulation, salt reabsorption in the kidney, and γ-aminobutyric acid (GABA)-mediated modulation in neurons. Here we present cryo-electron microscopy (cryo-EM) structures of human potassium-chloride cotransporter KCC1 in potassium chloride or sodium chloride at 2.9- to 3.5-angstrom resolution. KCC1 exists as a dimer, with both extracellular and transmembrane domains involved in dimerization. The structural and functional analyses, along with computational studies, reveal one potassium site and two chloride sites in KCC1, which are all required for the ion transport activity. KCC1 adopts an inward-facing conformation, with the extracellular gate occluded. The KCC1 structures allow us to model a potential ion transport mechanism in KCCs and provide a blueprint for drug design.


Subject(s)
Sodium-Potassium-Chloride Symporters/chemistry , Symporters/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cryoelectron Microscopy , HEK293 Cells , Humans , Ion Transport , Mice , Molecular Dynamics Simulation , Oocytes , Protein Domains , Protein Multimerization , Protein Structure, Quaternary , Sequence Alignment , Xenopus laevis , K Cl- Cotransporters
2.
J Physiol ; 597(6): 1451-1465, 2019 03.
Article in English | MEDLINE | ID: mdl-30659612

ABSTRACT

In the early 80s, renal microperfusion studies led to the identification of a basolateral K+ -Cl- cotransport mechanism in the proximal tubule, thick ascending limb of Henle and collecting duct. More than ten years later, this mechanism was found to be accounted for by three different K+ -Cl- cotransporters (KCC1, KCC3 and KCC4) that are differentially distributed along the renal epithelium. Two of these isoforms (KCC1 and KCC3) were also found to be expressed in arterial walls, the myocardium and a variety of neurons. Subsequently, valuable insights have been gained into the molecular and physiological properties of the KCCs in both the mammalian kidney and cardiovascular system. There is now robust evidence indicating that KCC4 sustains distal renal acidification and that KCC3 regulates myogenic tone in resistance vessels. However, progress in understanding the functional significance of these transporters has been slow, probably because each of the KCC isoforms is not identically distributed among species and some of them share common subcellular localizations with other KCC isoforms or sizeable conductive Cl- pathways. In addition, the mechanisms underlying the process of K+ -Cl- cotransport are still ill defined. The present review focuses on the knowledge gained regarding the roles and properties of KCCs in renal and cardiovascular tissues.


Subject(s)
Cardiovascular System/metabolism , Kidney/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Chlorides/metabolism , Humans , Potassium/metabolism , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/genetics
3.
Compr Physiol ; 8(2): 871-901, 2018 03 25.
Article in English | MEDLINE | ID: mdl-29687903

ABSTRACT

Two genes encode the Na+ -K+ -2Cl- cotransporters, NKCC1 and NKCC2, that mediate the tightly coupled movement of 1Na+ , 1K+ , and 2Cl- across the plasma membrane of cells. Na+ -K+ -2Cl- cotransport is driven by the chemical gradient of the three ionic species across the membrane, two of them maintained by the action of the Na+ /K+ pump. In many cells, NKCC1 accumulates Cl- above its electrochemical potential equilibrium, thereby facilitating Cl- channel-mediated membrane depolarization. In smooth muscle cells, this depolarization facilitates the opening of voltage-sensitive Ca2+ channels, leading to Ca2+ influx, and cell contraction. In immature neurons, the depolarization due to a GABA-mediated Cl- conductance produces an excitatory rather than inhibitory response. In many cell types that have lost water, NKCC is activated to help the cells recover their volume. This is specially the case if the cells have also lost Cl- . In combination with the Na+ /K+ pump, the NKCC's move ions across various specialized epithelia. NKCC1 is involved in Cl- -driven fluid secretion in many exocrine glands, such as sweat, lacrimal, salivary, stomach, pancreas, and intestine. NKCC1 is also involved in K+ -driven fluid secretion in inner ear, and possibly in Na+ -driven fluid secretion in choroid plexus. In the thick ascending limb of Henle, NKCC2 activity in combination with the Na+ /K+ pump participates in reabsorbing 30% of the glomerular-filtered Na+ . Overall, many critical physiological functions are maintained by the activity of the two Na+ -K+ -2Cl- cotransporters. In this overview article, we focus on the functional roles of the cotransporters in nonpolarized cells and in epithelia. © 2018 American Physiological Society. Compr Physiol 8:871-901, 2018.


Subject(s)
Sodium-Potassium-Chloride Symporters/physiology , Animals , Biological Transport/physiology , Cell Membrane/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/genetics , Structure-Activity Relationship
4.
Bioorg Med Chem ; 25(14): 3845-3852, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28566208

ABSTRACT

Pseudohypoaldosteronism type II (PHAII) is characterized by hyperkalemia and hypertension despite a normal glomerular filtration rate. Abnormal activation of the signal cascade of with-no-lysine kinase (WNK) with OSR1 (oxidative stress-responsive kinase 1)/SPAK (STE20/SPS1-related proline/alanine-rich kinase) and NCC (NaCl cotransporter) results in characteristic salt-sensitive hypertension. Thus, inhibitors of the WNK-OSR1/SPAK-NCC cascade are candidates for a new class of antihypertensive drugs. In this study, we developed novel inhibitors of this signal cascade from the 9-aminoacridine lead compound 1, one of the hit compounds obtained by screening our chemical library for WNK-SPAK binding inhibitors. Among the synthesized acridine derivatives, several acridine-3-amide and 3-urea derivatives, such as 10 (IC50: 6.9µM), 13 (IC50: 2.6µM), and 20 (IC50: 4.8µM), showed more potent inhibitory activity than the lead compound 1 (IC50: 15.4µM). Compounds 10 and 20 were confirmed to inhibit phosphorylation of NCC in vivo.


Subject(s)
Antihypertensive Agents/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Minor Histocompatibility Antigens/metabolism , Protein Serine-Threonine Kinases/metabolism , Aminacrine/chemistry , Aminacrine/metabolism , Aminacrine/pharmacology , Animals , Antihypertensive Agents/metabolism , Antihypertensive Agents/pharmacology , Cell Survival/drug effects , HEK293 Cells , Humans , Immunoblotting , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Kidney/drug effects , Kidney/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/metabolism , Structure-Activity Relationship , WNK Lysine-Deficient Protein Kinase 1
5.
PLoS One ; 12(6): e0179968, 2017.
Article in English | MEDLINE | ID: mdl-28662098

ABSTRACT

Cation Chloride Cotransporters (CCCs) comprise secondary active membrane proteins mainly mediating the symport of cations (Na+, K+) coupled with chloride (Cl-). They are divided into K+-Cl- outward transporters (KCCs), the Na+-K+-Cl- (NKCCs) and Na+-Cl- (NCCs) inward transporters, the cation chloride cotransporter interacting protein CIP1, and the polyamine transporter CCC9. KCCs and N(K)CCs are established in the genome since eukaryotes and metazoans, respectively. Most of the physiological and functional data were obtained from vertebrate species. To get insights into the basal functional properties of KCCs and N(K)CCs in the metazoan lineage, we cloned and characterized KCC and N(K)CC from the cnidarian Hydra vulgaris. HvKCC is composed of 1,032 amino-acid residues. Functional analyses revealed that hvKCC mediates a Na+-independent, Cl- and K+ (Tl+)-dependent cotransport. The classification of hvKCC as a functional K-Cl cotransporter is furthermore supported by phylogenetic analyses and a similar structural organization. Interestingly, recently obtained physiological analyses indicate a role of cnidarian KCCs in hyposmotic volume regulation of nematocytes. HvN(K)CC is composed of 965 amino-acid residues. Phylogenetic analyses and structural organization suggest that hvN(K)CC is a member of the N(K)CC subfamily. However, no inorganic ion cotransport function could be detected using different buffer conditions. Thus, hvN(K)CC is a N(K)CC subfamily member without a detectable inorganic ion cotransporter function. Taken together, the data identify two non-bilaterian solute carrier 12 (SLC12) gene family members, thereby paving the way for a better understanding of the evolutionary paths of this important cotransporter family.


Subject(s)
Hydra/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Protein Structure, Secondary , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/genetics
6.
Article in English | MEDLINE | ID: mdl-27591837

ABSTRACT

Although iono-regulatory processes are critical for survival of crustaceans during the molt cycle, the mechanisms involved are still not clear. The Na+/K+/2Cl- cotransporter (NKCC), a SLC12A family protein that transports Na+, K+ and 2Cl- into cells, is essential for cell ionic and osmotic regulation. To better understand the role of NKCC in the molt osmoregulation, we cloned and characterized a NKCC gene from the mud crab, Scylla paramamosain (designated as SpNKCC). The predicted SpNKCC protein is well conserved, and phylogenetic analysis revealed that this protein was clustered with crustacean NKCC. Expression of SpNKCC was detected in all the tissues examined but was highest in the posterior gills. Transmission electron microscopy revealed that posterior gills had a thick type of epithelium for ion regulation while the anterior gills possessed a thin phenotype related to gas exchange. During the molting cycle, hemolymph osmolality and ion concentrations (Na+ and Cl-) increased significantly over the postmolt period, remained stable in the intermolt and premolt stages and then decreased at ecdysis. Meanwhile, the expression of SpNKCC mRNA was significantly elevated (26.7 to 338.8-fold) at the ion re-establishing stages (postmolt) as compared with baseline molt level. This pattern was consistent with the coordinated regulation of Na+/K+-ATPase α-subunit (NKA α), carbonic anhydrase cytoplasmic (CAc) isoform and Na+/H+ exchanger (NHE) genes in the posterior gills. These data suggest that SpNKCC may be important in mediating branchial ion uptake during the molt cycle, especially at the postmolt stages.


Subject(s)
Crustacea/metabolism , DNA, Complementary/genetics , Gills/metabolism , Sodium-Potassium-Chloride Symporters/genetics , Amino Acid Sequence , Animals , Base Sequence , Crustacea/physiology , Molting , Osmolar Concentration , Sequence Homology, Amino Acid , Sodium-Potassium-Chloride Symporters/chemistry
7.
J Biol Chem ; 289(11): 7569-79, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24451383

ABSTRACT

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.


Subject(s)
Ion Transport , Sodium-Potassium-Chloride Symporters/chemistry , Amino Acid Sequence , Binding Sites , Cell Line , Chlorides/chemistry , Copper/chemistry , Cross-Linking Reagents/chemistry , Disulfides/chemistry , Homeostasis , Humans , Ions , Kinetics , Microscopy, Confocal , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Mutation , Phenanthrolines/chemistry , Phosphorylation , Protein Structure, Tertiary , Rubidium Radioisotopes/chemistry , Sequence Homology, Amino Acid , Solute Carrier Family 12, Member 2/chemistry , Solute Carrier Family 12, Member 2/genetics
8.
Cell Physiol Biochem ; 31(2-3): 257-76, 2013.
Article in English | MEDLINE | ID: mdl-23466888

ABSTRACT

BACKGROUND/AIMS: Chelerythrine [CET], a protein kinase C [PKC] inhibitor, is a prop-apoptotic BH3-mimetic binding to BH1-like motifs of Bcl-2 proteins. CET action was examined on PKC phosphorylation-dependent membrane transporters (Na+/K+ pump/ATPase [NKP, NKA], Na+-K+-2Cl+ [NKCC] and K+-Cl- [KCC] cotransporters, and channel-supported K+ loss) in human lens epithelial cells [LECs]. METHODS: K+ loss and K+ uptake, using Rb+ as congener, were measured by atomic absorption/emission spectrophotometry with NKP and NKCC inhibitors, and Cl- replacement by NO3ˉ to determine KCC. 3H-Ouabain binding was performed on a pig renal NKA in the presence and absence of CET. Bcl-2 protein and NKA sequences were aligned and motifs identified and mapped using PROSITE in conjunction with BLAST alignments and analysis of conservation and structural similarity based on prediction of secondary and crystal structures. RESULTS: CET inhibited NKP and NKCC by >90% (IC50 values ~35 and ~15 µM, respectively) without significant KCC activity change, and stimulated K+ loss by ~35% at 10-30 µM. Neither ATP levels nor phosphorylation of the NKA α1 subunit changed. 3H-ouabain was displaced from pig renal NKA only at 100 fold higher CET concentrations than the ligand. Sequence alignments of NKA with BH1- and BH3-like motifs containing pro-survival Bcl-2 and BclXl proteins showed more than one BH1-like motif within NKA for interaction with CET or with BH3 motifs. One NKA BH1-like motif (ARAAEILARDGPN) was also found in all P-type ATPases. Also, NKA possessed a second motif similar to that near the BH3 region of Bcl-2. CONCLUSION: Findings support the hypothesis that CET inhibits NKP by binding to BH1-like motifs and disrupting the α1 subunit catalytic activity through conformational changes. By interacting with Bcl-2 proteins through their complementary BH1- or BH3-like-motifs, NKP proteins may be sensors of normal and pathological cell functions, becoming important yet unrecognized signal transducers in the initial phases of apoptosis. CET action on NKCC1 and K+ channels may involve PKC-regulated mechanisms; however, limited sequence homologies to BH1-like motifs cannot exclude direct effects.


Subject(s)
Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Benzophenanthridines/pharmacology , Proto-Oncogene Proteins c-bcl-2/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Humans , Molecular Sequence Data , Ouabain/pharmacology , Phosphorylation , Potassium/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/metabolism , Rubidium/metabolism , Sequence Alignment , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Swine , Tritium/chemistry
9.
Cell Physiol Biochem ; 32(7): 14-31, 2013.
Article in English | MEDLINE | ID: mdl-24429812

ABSTRACT

The homologous genes encoding the electroneutral solute carrier family 12A (SLC12A) were identified more than 20 years ago, however, over the last few years, it has become clear that each of the genes within this family potentially encode for more than one cation-chloride cotransporter (CCC). Even more surprising, despite more than 30 years of functional studies and a wealth of knowledge on the activators, inhibitors, ion affinities, and kinetics of these cotransporters, we still cannot sufficiently explain why some cells express only one CCC isoform, while others express two, three, or more CCC isoforms. In 2009, Drs. Alvarez-Leefmans and Di Fulvio published an extensive in silico molecular analysis of the potential splice variants of the Na(+)-dependent cation-chloride cotransporters. In this review, we will look at the exceptionally large variety of potential splice variants within the Na(+)-independent cation-chloride cotransporter (SLC12A4-SLC12A7) genes, their initial tissue identification, and their physiological relevance.


Subject(s)
Chlorides/metabolism , Ion Transport/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 4/metabolism , Symporters/metabolism , Carrier Proteins/chemistry , Cations/metabolism , Humans , Kinetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 4/chemistry , Solute Carrier Family 12, Member 4/genetics , Symporters/chemistry , Symporters/genetics
10.
Curr Top Membr ; 70: 215-37, 2012.
Article in English | MEDLINE | ID: mdl-23177987

ABSTRACT

The cation chloride cotransporters (CCCs) represent an important family of transporters that plays key roles in vectorial electrolyte movement across epithelia and in intracellular chloride homeostasis of neurons and muscle cells. The CCCs are composed of three broad groups, two of which include multiple isoforms: Na-Cl cotransporter (NCC; SLC12A3), Na-K-2Cl cotransporter (NKCC; SLC12A1-2), and K-Cl cotransporter (KCC; SLC12A4-7). The CCCs are inhibited by clinically relevant drugs, including loop diuretics that inhibit NKCC2 in the renal thick ascending limb and thiazide diuretics that inhibit NCC in the renal distal tubule. For many years, much research on this gene family has centered on understanding ion binding and inhibitor interaction which represent important features of the molecular operation of these transporters. Recently, high resolution structures of bacterial transport proteins related to the CCCs have become available, thus permitting structural context in which to evaluate previous ion and inhibitor studies of the CCCs. In this article, I review past molecular and structure-function studies that have provided key pieces of information about ion binding and inhibitor interaction primarily of NKCC for which we have the most information. I then place these findings into the structural context of recent homology models of NKCC based on the outward-facing open and occluded conformations of the related bacterial transporters. These homology models provide our first glimpse into the fine details of the molecular operation of the CCCs.


Subject(s)
Chlorides/metabolism , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Binding Sites , Cations , Humans , Kidney/metabolism , Models, Molecular , Muscle Cells/metabolism , Neurons/metabolism , Protein Conformation , Sodium Chloride Symporter Inhibitors/metabolism , Structure-Activity Relationship
11.
J Biol Chem ; 287(51): 42642-53, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23105100

ABSTRACT

Mutations in the apical Na-K-2Cl co-transporter, NKCC2, cause type I Bartter syndrome, a life-threatening kidney disease. Yet the mechanisms underlying the regulation of NKCC2 trafficking in renal cells are scarcely known. We previously showed that naturally occurring mutations depriving NKCC2 of its distal COOH-terminal tail and interfering with the (1081)LLV(1083) motif result in defects in the ER exit of the co-transporter. Here we show that this motif is necessary but not sufficient for anterograde trafficking of NKCC2. Indeed, we have identified two additional hydrophobic motifs, (1038)LL(1039) and (1048)LI(1049), that are required for ER exit and surface expression of the co-transporter. Double mutations of (1038)LL(1039) or (1048)LI(1049) to di-alanines disrupted glycosylation and cell surface expression of NKCC2, independently of the expression system. Pulse-chase analysis demonstrated that the absence of the terminally glycosylated form of NKCC2 was not due to reduced synthesis or increased rates of degradation of mutant co-transporters, but was instead caused by defects in maturation. Co-immunolocalization experiments revealed that (1038)AA(1039) and (1048)AA(1049) were trapped mainly in the ER as indicated by extensive co-localization with the ER marker calnexin. Remarkably, among several analyzed motifs present in the NKCC2 COOH terminus, only those required for ER exit and surface expression of NKCC2 are evolutionarily conserved in all members of the SLC12A family, a group of cation-chloride co-transporters that are targets of therapeutic drugs and mutated in several human diseases. Based upon these data, we propose abnormal anterograde trafficking as a common mechanism associated with mutations depriving NKCC2, and also all other members of the SLC12A family, of their COOH terminus.


Subject(s)
Conserved Sequence , Evolution, Molecular , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Cell Line , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/genetics , Opossums , Protein Structure, Tertiary , Protein Transport , Sodium Chloride Symporters/metabolism , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 1 , Structure-Activity Relationship
12.
Am J Physiol Cell Physiol ; 303(11): C1198-205, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23034389

ABSTRACT

X-ray crystallography of the catalytic domain of oxidative stress response 1 (OSR1) has provided evidence for dimerization and domain swapping. However, the functional significance of dimer formation or domain swapping has yet to be addressed. In this study, we used nine glutamine residues to link the carboxyl end of one SPAK (related Ste20 kinase) monomer to the amino end of another SPAK monomer to assess the role of kinase monomers versus dimers in Na-K-2Cl cotransporter 1 (NKCC1) activation. Transport studies in Xenopus laevis oocytes show that forcing dimerization of two wild-type SPAK molecules results in cotransporter activation when calcium-binding protein 39 (Cab39) is coexpressed, indicating that the presence of Cab39 can bypass the upstream phosphorylation requirement of SPAK normally associated with kinase activation. We determined that monomers are the functional units of the kinase as concatamers consisting of an active and various inactive monomers were still functional. Furthermore, we found that two different nonfunctional SPAK mutants could be linked together in a concatamer and activated, presumably by domain swapping, indicating that dimerization and domain swapping are both important components of kinase activation. Finally, we demonstrate rescue of a nonfunctional SPAK mutant by domain swapping with wild-type OSR1, indicating that heterodimers of the two Ste20-related kinases are possible and therefore potentially relevant to the regulation of NKCC1 activity.


Subject(s)
Calcium-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Biological Transport , Calcium-Binding Proteins/chemistry , Crystallography, X-Ray , Female , Glutamine/chemistry , Humans , Molecular Sequence Data , Oocytes , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 2 , Xenopus laevis
13.
J Comp Physiol B ; 182(4): 491-506, 2012 May.
Article in English | MEDLINE | ID: mdl-22179410

ABSTRACT

The freshwater climbing perch, Anabas testudineus, is an obligatory air-breathing teleost which can acclimate to seawater, survive long period of emersion, and actively excrete ammonia against high concentrations of environmental ammonia. This study aimed to clone and sequence the Na⁺:K⁺:2Cl⁻ cotransporter (nkcc) from the gills of A. testudineus, and to determine the effects of seawater acclimation or exposure to 100 mmol l⁻¹ NH4Cl in freshwater on its branchial mRNA expression. The complete coding cDNA sequence of nkcc from the gills of A. testudineus consisted of 3,495 bp, which was translated into a protein with 1,165 amino acid residues and an estimated molecular mass of 127.4 kDa. A phylogenetic analysis revealed that the translated Nkcc of A. testudineus was closer to fish Nkcc1a than to fish Nkcc1b or Nkcc2. After a progressive increase in salinity, there were significant increases in the mRNA expression and protein abundance of nkcc1a in the gills of fish acclimated to seawater as compared with that of the freshwater control. Hence, it can be concluded that similar to marine teleosts, Cl⁻ excretion through basolateral Nkcc1 of mitochondrion-rich cells (MRCs) was essential to seawater acclimation in A. testudineus. Exposure of A. testudineus to 100 mmol l⁻¹ NH4Cl for 1 or 6 days also resulted in significant increases in the mRNA expression of nkcc1a in the gills, indicating a functional role of Nkcc1a in active ammonia excretion. It is probable that NH4⁺ enter MRCs through basolateral Nkcc1a before being actively transported across the apical membrane. Since the operation of Nkcc1a would lead to an increase in the intracellular Na⁺ concentration, it can be deduced that an upregulation of basolateral Na⁺/K⁺-ATPase (Nka) activity would be necessary to compensate for the increased influx of Na⁺ into MRCs during active NH4⁺ excretion. This would imply that the main function of Nka in active NH4⁺ excretion is to maintain intracellular Na⁺ and K⁺ homeostasis instead of transporting NH4⁺ directly into MRCs as proposed previously. In conclusion, active salt secretion during seawater acclimation and active NH4⁺ excretion during exposure to ammonia in freshwater could involve similar transport mechanisms in the gills of A. testudineus.


Subject(s)
Acclimatization , Ammonium Chloride/pharmacology , Fish Proteins/metabolism , Gills/metabolism , Perches/physiology , Seawater , Sodium-Potassium-Chloride Symporters/metabolism , Up-Regulation , Acclimatization/drug effects , Amino Acid Sequence , Animals , Fish Proteins/chemistry , Fish Proteins/genetics , Gills/drug effects , Molecular Sequence Data , Molecular Weight , Phylogeny , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Salinity , Seawater/chemistry , Sequence Alignment , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 2 , Stress, Physiological , Up-Regulation/drug effects , Water Pollutants, Chemical/pharmacology , Water-Electrolyte Balance/drug effects
14.
J Endocrinol Invest ; 35(8): 735-41, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22082806

ABSTRACT

Thyroid disorders are accompanied by major changes in renal sodium handling and blood pressure. Sodium transporters play a crucial role in regulating sodium excretion. We determined the function and expression of type 3 Na/H (NHE3) exchanger, type 2 Na+K+2Cl co-transporter (NKCC2) co-transporter, NaCl co-transporter (NCC) cotransporter, and epithelial sodium channel (ENaC) in hypoand hyperthyroid rats at 6 weeks after each thyroid disorder induction. We measured the renal response to functional blockade of the tubular sodium transporters, using acetazolamide to inhibit the activity of NHE3, furosemide for NKCC2, hydrochlorotiazide for NCC, and amiloride for ENaC. Expression of sodium transporters was analyzed by measuring the protein abundance by Western blot. The responsiveness to NHE3 inhibition and NHE3 protein was lower in hypothyroid rats and higher in hyperthyroid rats vs controls. Hypothyroid rats showed greater diuretic and natriuretic responses to NKCC2 and ENaC blockade and higher protein abundance of NKCC2 vs controls. Hyperthyroid rats showed greater protein expression of NKCC2 and NCC vs controls. Groups did not differ in responsiveness to NCC blockade. The expression and activity of ENaC were lower in hyperthyroid rats. In conclusion, reduced NHE3 activity may participate in the low blood pressure of hypothyroid rats and elevated NHE3 activity in the high blood pressure of hyperthyroid rats. These proximal alterations are counter-balanced by functional upregulation of NKCC2 and ENaC in downstream nephron segments of hypothyroid rats and by downregulation of αENaC activity and expression in hyperthyroid rats.


Subject(s)
Epithelial Sodium Channels/metabolism , Hyperthyroidism/metabolism , Hypothyroidism/metabolism , Kidney/pathology , Sodium Chloride Symporters/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Epithelial Sodium Channels/chemistry , Hyperthyroidism/chemically induced , Hyperthyroidism/pathology , Hypothyroidism/chemically induced , Hypothyroidism/pathology , Kidney/drug effects , Kidney/metabolism , Male , Rats , Rats, Wistar , Sodium Chloride Symporters/chemistry , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Potassium-Chloride Symporters/chemistry , Solute Carrier Family 12, Member 1
15.
J Biol Chem ; 287(3): 2210-20, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22121194

ABSTRACT

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.


Subject(s)
Fish Proteins/chemistry , Models, Molecular , Protein Multimerization , Sodium-Potassium-Chloride Symporters/chemistry , Animals , Fish Proteins/genetics , Fish Proteins/metabolism , Fluorescence Resonance Energy Transfer/methods , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Movement , Sharks , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 2
16.
Biochemistry ; 50(45): 9857-64, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21970294

ABSTRACT

The "secretory" Na(+)-K(+)-2Cl(-) cotransporter, NKCC1, belongs to the SLC12 gene family of electroneutral cation-chloride cotransporters. A number of these proteins, including NKCC1 itself, exist as homodimers in the membrane, suggesting that this may be a common feature of the SLC12 family. We have previously demonstrated that replacing the C-terminus of NKCC1 with that of its close homologue NKCC2 produced a fully functional chimeric protein that formed homodimers but did not dimerize with NKCC1. Here we employ a novel co-immunoprecipitation assay to study the dimerization interaction of NKCC1 using additional NKCC1/NKCC2 C-terminal chimeras and point mutants. Our results indicate that the substitution of a number of regions of the C-terminus of NKCC1 with the corresponding sequence from NKCC2 results in weakened dimerization with wild-type NKCC1, demonstrating that various residues play a role in this interaction. Most interestingly, however, we find that the replacement of a single NKCC1 residue, G812, with cysteine, the corresponding amino acid in NKCC2, results in a point mutant that displays no significant dimerization with the wild-type protein. In addition to this effect on heterodimer formation, we also find that G812 mutants can nevertheless form homodimers but that this interaction can be weaker than that observed for wild-type NKCC1. We demonstrate that our results are consistent with at least one established mechanism of protein dimer formation, that of "domain swapping", as well as with a recently reported crystal structure of the C-terminus of a bacterial SLC12 homologue.


Subject(s)
Sodium-Potassium-Chloride Symporters/chemistry , Amino Acid Substitution , Animals , Dimerization , HEK293 Cells , Humans , Models, Molecular , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 1 , Solute Carrier Family 12, Member 2 , Transfection
17.
Cell Physiol Biochem ; 28(1): 147-56, 2011.
Article in English | MEDLINE | ID: mdl-21865857

ABSTRACT

We have recently reported that Na(+)/K(+)/2Cl(-) cotransporter isoform 1 (NKCC1) plays an essential role in nerve growth factor (NGF)-induced neurite outgrowth in PC12D cells. On the other hand, it has been reported that dietary flavonoids, such as quercetin, apigenin, and luteolin, stimulate various ion transporters. In the present report, we investigated the effect of quercetin, a flavonoid, on NGF-induced neurite outgrowth in PC12 cells (the parental strain of PC12D cells). Quercetin stimulated the NGF-induced neurite outgrowth in a dose-dependent manner. Knockdown of NKCC1 by RNAi methods abolished the stimulatory effect of flavonoid. Quercetin stimulated NKCC1 activity (measured as bumetanide-sensitive (86)Rb influx) without any increase in the expression level of NKCC1 protein. The stimulatory effect of quercetin on neurite outgrowth was dependent upon extracellular Cl(-). These observations indicate that quercetin stimulates the NGF-induced neurite outgrowth via an increase in Cl(-) incorporation into the intracellular space by activating NKCC1 in PC12 cell.


Subject(s)
Antioxidants/pharmacology , Nerve Growth Factors/metabolism , Neurites/drug effects , Quercetin/pharmacology , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Chlorides/pharmacology , Neurites/metabolism , PC12 Cells , RNA Interference , RNA, Small Interfering/metabolism , Rats , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 2
18.
Hum Mol Genet ; 20(5): 855-66, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21131289

ABSTRACT

WNK1 (with-no-lysine[K]-1) is a protein kinase of which mutations cause a familial hypertension and hyperkalemia syndrome known as pseudohypoaldosteronism type 2 (PHA2). Kidney-specific (KS) WNK1 is an alternatively spliced form of WNK1 kinase missing most of the kinase domain. KS-WNK1 downregulates the Na(+)-Cl(-) cotransporter NCC by antagonizing the effect of full-length WNK1 when expressed in Xenopus oocytes. The physiological role of KS-WNK1 in the regulation of NCC and potentially other Na(+) transporters in vivo is unknown. Here, we report that mice overexpressing KS-WNK1 in the kidney exhibited renal Na(+) wasting, elevated plasma levels of angiotensin II and aldosterone yet lower blood pressure relative to wild-type littermates. Immunofluorescent staining revealed reduced surface expression of total and phosphorylated NCC and the Na(+)-K(+)-2Cl(-) cotransporter NKCC2 in the distal convoluted tubule and the thick ascending limb of Henle's loop, respectively. Conversely, mice with targeted deletion of exon 4A (the first exon for KS-WNK1) exhibited Na(+) retention, elevated blood pressure on a high-Na(+) diet and increased surface expression of total and phosphorylated NCC and NKCC2 in respective nephron segments. Thus, KS-WNK1 is a negative regulator of NCC and NKCC2 in vivo and plays an important role in the control of Na(+) homeostasis and blood pressure. These results have important implications to the pathogenesis of PHA2 with WNK1 mutations.


Subject(s)
Down-Regulation , Gene Silencing , Kidney/enzymology , Protein Serine-Threonine Kinases/metabolism , Pseudohypoaldosteronism/metabolism , Receptors, Drug/genetics , Sodium-Potassium-Chloride Symporters/genetics , Symporters/genetics , Amino Acid Sequence , Animals , Blood Pressure , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Minor Histocompatibility Antigens , Molecular Sequence Data , Organ Specificity , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Pseudohypoaldosteronism/enzymology , Pseudohypoaldosteronism/genetics , Pseudohypoaldosteronism/physiopathology , Receptors, Drug/metabolism , Sodium/metabolism , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 1 , Solute Carrier Family 12, Member 3 , Symporters/metabolism , WNK Lysine-Deficient Protein Kinase 1
19.
Dongwuxue Yanjiu ; 31(6): 601-9, 2010 Dec.
Article in Chinese | MEDLINE | ID: mdl-21174349

ABSTRACT

The gills are the major apparatus for osmoregulation in fish to acclimate the changes of salinities. Na+/K+/2Cl(-)cotransporter 1-alpha (NKCC1 alpha) is one of the key ion cotransporter locoalized in gill chloride cells which has been associated with the maintence of osmotic homeostasis. The transport process mediated by NKCC1 alpha is characterized by electroneutrality with a stoichiometry of 1Na:1K:2Cl. Sarotherodon melanotheron is one of the most euryhaline teleosts able to withstand variations in environmental salinity ranging from freshwater to hyper-saline waters. In this study, the reverse transcription-polymerase chain reaction and rapid amplification of 3' and 5'cDNA ends methods were used to identify the full cDNA of the NKCC1 alpha with an Open Reading Frame which contains 1 151aa of S.melanotheron. The amino acid multiple alignment and phylogenetic analysis showed that this isoform is more similar with isoforms in Oreochromis mossambicus, Salmo salar and Anguilla anguilla, and there is the highest homologous of 99% between Sarotherodon and Mossambique. The predicted protein secondly structure of NKCC1 alpha contains 10 transmenbrane domains, which were highly conserved in sequences and locoalization sites relatively to other species. The quantitative real time polymerase chain reaction (qRT-PCR) assay was developed to estimate the mRNA expression levels in gill, liver, intestine and kidney in freshwater, the results showed a tissue-specific model. Furthermore, the sanility significantly affects the relative expression level of NKCC1 alpha mRNA in gill with a 4.9 times higher in 136 salinity water than that in 0 salinity. The results suggest that the NKCC1 alpha is closely related to the salt tolerance in S.melanotheron.


Subject(s)
Cichlids/genetics , RNA, Messenger/analysis , Sodium-Potassium-Chloride Symporters/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Molecular Sequence Data , Protein Isoforms , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Potassium-Chloride Symporters/chemistry , Solute Carrier Family 12, Member 2
20.
Article in English | MEDLINE | ID: mdl-20576485

ABSTRACT

This study used the brackish medaka (Oryzias dancena) to characterize Na+, K+, 2Cl- cotransporter (NKCC) expression from the genetic to cellular level in gills. Using RT-PCR to survey tissue distribution of nkcc1a, 1b, and 2, we report that gills of brackish medaka prominently express Odnkcc1a. The full-length cDNA of Odnkcc1a was cloned from gill tissue. In situ hybridization indicates that Odnkcc1a was localized to mitochondrion-rich (MR) cells. Higher mRNA levels of Odnkcc1a were found in gills from seawater (SW) and brackish water (BW) medaka when compared to freshwater (FW) fish. Furthermore, higher amounts of NKCC1a-like protein were detected by the monoclonal antibody in gills of SW and BW medaka compared to FW medaka. Double immunofluorescence staining revealed that NKCC1a-like protein colocalizes with Na+, K+-ATPase on the basolateral membrane of MR cells in BW and SW fish. In addition, transfer of brackish medaka from SW to FW revealed that expression of NKCC1a-like protein in gills was retained until 7days, which is a likely mechanism for maintaining hyposmoregulatory endurance. The study illustrates salinity-dependent expression of NKCC1a in branchial MR cells from brackish medaka and suggests a critical role for NKCC1a in hyposmoregulatory endurance of this fish.


Subject(s)
Adaptation, Physiological , Gills/metabolism , Oryzias/metabolism , Salinity , Sodium-Potassium-Chloride Symporters/genetics , Animals , Fresh Water/chemistry , Gills/cytology , Gills/drug effects , Oryzias/blood , Oryzias/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Seawater/chemistry , Sodium Chloride/pharmacology , Sodium-Potassium-Chloride Symporters/chemistry , Sodium-Potassium-Chloride Symporters/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...