Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
J Biol Chem ; 291(9): 4487-502, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26721884

ABSTRACT

Mutations in the renal specific Na-K-2Cl co-transporter (NKCC2) lead to type I Bartter syndrome, a life-threatening kidney disease featuring arterial hypotension along with electrolyte abnormalities. We have previously shown that NKCC2 and its disease-causing mutants are subject to regulation by endoplasmic reticulum-associated degradation (ERAD). The aim of the present study was to identify the protein partners specifically involved in ERAD of NKCC2. To this end, we screened a kidney cDNA library through a yeast two-hybrid assay using NKCC2 C terminus as bait. We identified OS9 (amplified in osteosarcomas) as a novel and specific binding partner of NKCC2. Co-immunoprecipitation assays in renal cells revealed that OS9 association involves mainly the immature form of NKCC2. Accordingly, immunocytochemistry analysis showed that NKCC2 and OS9 co-localize at the endoplasmic reticulum. In cells overexpressing OS9, total cellular NKCC2 protein levels were markedly decreased, an effect blocked by the proteasome inhibitor MG132. Pulse-chase and cycloheximide-chase assays demonstrated that the marked reduction in the co-transporter protein levels was essentially due to increased protein degradation of the immature form of NKCC2. Conversely, knockdown of OS9 by small interfering RNA increased NKCC2 expression by increasing the co-transporter stability. Inactivation of the mannose 6-phosphate receptor homology domain of OS9 had no effect on its action on NKCC2. In contrast, mutations of NKCC2 N-glycosylation sites abolished the effects of OS9, indicating that OS9-induced protein degradation is N-glycan-dependent. In summary, our results demonstrate the presence of an OS9-mediated ERAD pathway in renal cells that degrades immature NKCC2 proteins. The identification and selective modulation of ERAD components specific to NKCC2 and its disease-causing mutants might provide novel therapeutic strategies for the treatment of type I Bartter syndrome.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Kidney/metabolism , Lectins/metabolism , Neoplasm Proteins/metabolism , Solute Carrier Family 12, Member 1/metabolism , Amino Acid Substitution , Animals , Bartter Syndrome/genetics , Bartter Syndrome/metabolism , Cell Line , Endoplasmic Reticulum-Associated Degradation/drug effects , Gene Library , Glycosylation/drug effects , HEK293 Cells , Humans , Immunoprecipitation , Kidney/drug effects , Lectins/antagonists & inhibitors , Lectins/chemistry , Lectins/genetics , Mutation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Opossums , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proteasome Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Stability/drug effects , Protein Structure, Tertiary , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Solute Carrier Family 12, Member 1/antagonists & inhibitors , Solute Carrier Family 12, Member 1/chemistry , Solute Carrier Family 12, Member 1/genetics
2.
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
3.
Hum Mutat ; 32(4): 476-83, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21305656

ABSTRACT

Mutations in the electrogenic Cl(-)/H(+) exchanger ClC-5 gene CLCN5 are frequently associated with Dent disease, an X-linked recessive disorder affecting the proximal tubules. Here, we investigate the consequences in Xenopus laevis oocytes and in HEK293 cells of nine previously reported, pathogenic, missense mutations of ClC-5, most of them which are located in regions forming the subunit interface. Two mutants trafficked normally to the cell surface and to early endosomes, and displayed complex glycosylation at the cell surface like wild-type ClC-5, but exhibited reduced currents. Three mutants displayed improper N-glycosylation, and were nonfunctional due to being retained and degraded at the endoplasmic reticulum. Functional characterization of four mutants allowed us to identify a novel mechanism leading to ClC-5 dysfunction in Dent disease. We report that these mutant proteins were delayed in their processing, and that the stability of their complex glycosylated form was reduced, causing lower cell surface expression. The early endosome distribution of these mutants was normal. Half of these mutants displayed reduced currents, whereas the other half showed abolished currents. Our study revealed distinct cellular mechanisms accounting for ClC-5 loss of function in Dent disease.


Subject(s)
Chloride Channels/genetics , Dent Disease/genetics , Mutation , Amino Acid Sequence , Animals , Cells, Cultured , Chloride Channels/metabolism , Dent Disease/metabolism , HEK293 Cells , Humans , Kidney Tubules, Proximal/metabolism , Molecular Sequence Data , Oocytes/metabolism , Sequence Alignment , Xenopus laevis
4.
J Biol Chem ; 286(11): 9489-502, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21205824

ABSTRACT

The renal-specific Na-K-2Cl co-transporter, NKCC2, plays a pivotal role in regulating body salt levels and blood pressure. NKCC2 mutations lead to type I Bartter syndrome, a life-threatening kidney disease. Regulation of NKCC2 trafficking behavior serves as a major mechanism in controlling NKCC2 activity across the plasma membrane. However, the identities of the protein partners involved in cell surface targeting of NKCC2 are largely unknown. To gain insight into these processes, we used a yeast two-hybrid system to screen a kidney cDNA library for proteins that interact with the NKCC2 C terminus. One binding partner we identified was SCAMP2 (secretory carrier membrane protein 2). Microscopic confocal imaging and co-immunoprecipitation assays confirmed NKCC2-SCAMP2 interaction in renal cells. SCAMP2 associated also with the structurally related co-transporter NCC, suggesting that the interaction with SCAMP2 is a common feature of sodium-dependent chloride co-transporters. Heterologous expression of SCAMP2 specifically decreased cell surface abundance as well as transport activity of NKCC2 across the plasma membrane. Co-immunolocalization experiments revealed that intracellularly retained NKCC2 co-localizes with SCAMP2 in recycling endosomes. The rate of NKCC2 endocytic retrieval, assessed by the sodium 2-mercaptoethane sulfonate cleavage assay, was not affected by SCAMP2. The surface-biotinylatable fraction of newly inserted NKCC2 in the plasma membrane was reduced by SCAMP2, demonstrating that SCAMP2-induced decrease in surface NKCC2 is due to decreased exocytotic trafficking. Finally, a single amino acid mutation, cysteine 201 to alanine, within the conserved cytoplasmic E peptide of SCAMP2, which is believed to regulate exocytosis, abolished SCAMP2-mediated down-regulation of the co-transporter. Taken together, these data are consistent with a model whereby SCAMP2 regulates NKCC2 transit through recycling endosomes and limits the cell surface targeting of the co-transporter by interfering with its exocytotic trafficking.


Subject(s)
Carrier Proteins/metabolism , Cell Membrane/metabolism , Exocytosis/physiology , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Carrier Proteins/genetics , Cell Membrane/genetics , HEK293 Cells , Humans , Male , Mice , Opossums , Protein Transport/physiology , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 1
5.
J Biol Chem ; 284(32): 21752-64, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19535327

ABSTRACT

Mutations in the apically located Na(+)-K(+)-2Cl(-) co-transporter, NKCC2, lead to type I Bartter syndrome, a life-threatening kidney disorder, yet the mechanisms underlying the regulation of mutated NKCC2 proteins in renal cells have not been investigated. Here, we identified a trihydrophobic motif in the distal COOH terminus of NKCC2 that was required for endoplasmic reticulum (ER) exit and surface expression of the co-transporter. Indeed, microscopic confocal imaging showed that a naturally occurring mutation depriving NKCC2 of its distal COOH-terminal region results in the absence of cell surface expression. Biotinylation assays revealed that lack of cell surface expression was associated with abolition of mature complex-glycosylated NKCC2. Pulse-chase analysis demonstrated that the absence of mature protein was not caused by reduced synthesis or increased rates of degradation of mutant co-transporters. Co-immunolocalization experiments revealed that these mutants co-localized with the ER marker protein-disulfide isomerase, demonstrating that they are retained in the ER. Cell treatment with proteasome or lysosome inhibitors failed to restore the loss of complex-glycosylated NKCC2, further eliminating the possibility that mutant co-transporters were processed by the Golgi apparatus. Serial truncation of the NKCC2 COOH terminus, followed by site-directed mutagenesis, identified hydrophobic residues (1081)LLV(1083) as an ER exit signal necessary for maturation of NKCC2. Mutation of (1081)LLV(1083) to AAA within the context of the full-length protein prevented NKCC2 ER exit independently of the expression system. This trihydrophobic motif is highly conserved in the COOH-terminal tails of all members of the cation-chloride co-transporter family, and thus may function as a common motif mediating their transport from the ER to the cell surface. Taken together, these data are consistent with a model whereby naturally occurring premature terminations that interfere with the LLV motif compromise co-transporter surface delivery through defective trafficking.


Subject(s)
Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Sodium-Potassium-Chloride Symporters/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , Chlorides/chemistry , Glycosylation , Mice , Models, Biological , Molecular Sequence Data , Mutation , Opossums , Protein Structure, Tertiary , Protein Transport , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 1
6.
J Biol Chem ; 282(46): 33817-33830, 2007 Nov 16.
Article in English | MEDLINE | ID: mdl-17848580

ABSTRACT

Apical bumetanide-sensitive Na(+)-K(+)-2Cl(-) co-transporter, termed NKCC2, is the major salt transport pathway in kidney thick ascending limb. NKCC2 surface expression is subject to regulation by intracellular protein trafficking. However, the protein partners involved in the intracellular trafficking of NKCC2 remain unknown. Moreover, studies aimed at under-standing the post-translational regulation of NKCC2 have been hampered by the difficulty to express NKCC2 protein in mammalian cells. Here we were able to express NKCC2 protein in renal epithelial cells by tagging its N-terminal domain. To gain insights into the regulation of NKCC2 trafficking, we screened for interaction partners of NKCC2 with the yeast two-hybrid system, using the C-terminal tail of NKCC2 as bait. Aldolase B was identified as a dominant and novel interacting protein. Real time PCR on renal microdissected tubules demonstrated the expression of aldolase B in the thick ascending limb. Co-immunoprecipitation and co-immunolocalization experiments confirmed NKCC2-aldolase interaction in renal cells. Biotinylation assays showed that aldolase co-expression reduces NKCC2 surface expression. In the presence of aldolase substrate, fructose 1,6-bisphosphate, aldolase binding was disrupted, and aldolase co-expression had no further effect on the cell surface level of NKCC2. Finally, functional studies demonstrated that aldolase-induced down-regulation of NKCC2 at the plasma membrane was associated with a decrease in its transport activity. In summary, we identified aldolase B as a novel NKCC2 binding partner that plays a key role in the modulation of NKCC2 surface expression, thereby revealing a new regulatory mechanism governing the co-transporter intracellular trafficking. Furthermore, NKCC2 protein expression in mammalian cells and its regulation by protein-protein interactions, described here, may open new and important avenues in studying the cell biology and post-transcriptional regulation of the co-transporter.


Subject(s)
Epithelial Cells/metabolism , Fructose-Bisphosphate Aldolase/chemistry , Gene Expression Regulation , Kidney/metabolism , Sodium-Potassium-Chloride Symporters/physiology , Animals , Biotinylation , Cell Membrane/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Biological , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Protein Transport , Solute Carrier Family 12, Member 1 , Two-Hybrid System Techniques
7.
Kidney Int ; 64(3): 939-49, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12911544

ABSTRACT

BACKGROUND: Low-concentration angiotensin II (Ang II) stimulates Na+/H+ exchanger 3 (NHE3) activity in renal proximal tubule mainly via angiotensin II type 1 (AT1) receptors. The mechanisms that mediate the increase in NHE3 activity elicited by Ang II remain incompletely settled. METHODS: To assess a potential role of NHE3 trafficking in the Ang II effect, NHE3 activity was measured by H+-driven initial rate of 22Na uptake resistant to 50 micromol/L of the Na+/H+ exchange inhibitor cariporide (HOE642), and sensitive to 300 micromol/L ethyl isopropyl amiloride (EIPA), in a model of cultured proximal tubular cells (MKCC), in which functional apical NHE3 and AT receptors are normally present. Apical expression of NHE3 protein was determined by cell surface biotinylation and immunoblotting. RESULTS: Ang II (10-10 mol/L, 43 minutes) increased NHE3 activity and biotinylated NHE3 protein without any change in total amount of NHE3 protein. Both effects were suppressed by specific AT1 receptor antagonists. When 2-mercaptoethanesulphonic acid (MESNA) was used to cleave biotin from all apical proteins, intracellular biotinylated NHE3 protein remained unchanged after Ang II incubation compared to control. When sulfo-N-hydrosuccinimide (NHS)-acetate was used first to block all apical reactive sites, an increase in biotinylated NHE3 protein was observed following Ang II incubation. To evaluate the role of phosphatidylinositol 3-kinase (PI 3-kinase), the specific inhibitor wortmannin was used. It suppressed Ang II-induced increase in NHE3 activity and trafficking. Furthermore, latrunculin B, inhibitor of actin filament polymerization, prevented both Ang II stimulatory effects. CONCLUSION: Ang II stimulates NHE3 activity, at least in part, by exocytic insertion of the protein into the apical membrane. This effect is mediated by PI 3-kinase and required integrity of actin cytoskeleton.


Subject(s)
Angiotensin II/physiology , Exocytosis/physiology , Kidney Cortex/metabolism , Phosphatidylinositol 3-Kinases/physiology , Sodium-Hydrogen Exchangers/metabolism , Actins/metabolism , Angiotensin II/administration & dosage , Animals , Cell Membrane/metabolism , Cells, Cultured , Cytoskeleton/physiology , Dose-Response Relationship, Drug , Endocytosis/physiology , Kidney Cortex/cytology , Mice , Sodium-Hydrogen Exchanger 3
SELECTION OF CITATIONS
SEARCH DETAIL
...