Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Histochem Cell Biol ; 133(5): 541-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20369363

ABSTRACT

The SLC26 family represents a group of integral membrane anion transport proteins. Mutations in one member of this protein family, SLC26A2 (DTDST or diastrophic dysplasia sulfate transporter), result in various chondrodysplasias due to undersulfation of proteoglycans in chondrocytes, a major site of DTDST protein expression. DTDST mRNA has been detected in the kidney, but protein expression has not been characterized. Our objective for this study was to determine the protein localization of this sulfate transporter in the kidney. We used immunofluorescence (IMF) techniques with an anti-DTDST monoclonal antibody to examine kidneys harvested from adult rats. Double labeling was performed with antibodies directed against megalin, which is found in the microvillus membrane and coated pits of the proximal tubule. IMF analysis indicated that DTDST protein expression was limited to the microvillus membrane of proximal tubule cells in the renal cortex but absent in glomeruli and other nephron segments. DTDST was also detected in isolated rat kidney proximal tubule microvillus membranes by Western blot analysis, confirming the immunofluorescent localization of the DTDST transporter to this nephron segment. The functional role of the DTDST protein in the kidney is unknown, but it may play a role in proximal tubule sulfate transport.


Subject(s)
Anion Transport Proteins/metabolism , Kidney/metabolism , Animals , Anion Transport Proteins/immunology , Antibodies/immunology , Antibody Specificity/immunology , Glycosylation , Kidney/cytology , Kidney Cortex/cytology , Kidney Cortex/metabolism , Kidney Glomerulus/metabolism , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Microvilli/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/immunology , Sulfate Transporters
2.
Am J Physiol Cell Physiol ; 298(6): C1363-75, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20219950

ABSTRACT

Nephrolithiasis in the Slc26a6(-/-) mouse is accompanied by 50-75% reduction in intestinal oxalate secretion with unchanged intestinal oxalate absorption. The molecular identities of enterocyte pathways for oxalate absorption and for Slc26a6-independent oxalate secretion remain undefined. The reported intestinal expression of SO(4)(2-) transporter SLC26A2 prompted us to characterize transport of oxalate and other anions by human SLC26A2 and mouse Slc26a2 expressed in Xenopus oocytes. We found that hSLC26A2-mediated [(14)C]oxalate uptake (K(1/2) of 0.65 +/- 0.08 mM) was cis-inhibited by external SO(4)(2-) (K(1/2) of 3.1 mM). hSLC26A2-mediated bidirectional oxalate/SO(4)(2-) exchange exhibited extracellular SO(4)(2-) K(1/2) of 1.58 +/- 0.44 mM for exchange with intracellular [(14)C]oxalate, and extracellular oxalate K(1/2) of 0.14 +/- 0.11 mM for exchange with intracellular (35)SO(4)(2-). Influx rates and K(1/2) values for mSlc26a2 were similar. hSLC26A2-mediated oxalate/Cl(-) exchange and bidirectional SO(4)(2-)/Cl(-) exchange were not detectably electrogenic. Both SLC26A2 orthologs exhibited nonsaturable extracellular Cl(-) dependence for efflux of intracellular [(14)C]oxalate, (35)SO(4)(2-), or (36)Cl(-). Rate constants for (36)Cl(-) efflux into extracellular Cl(-), SO(4)(2-), and oxalate were uniformly 10-fold lower than for oppositely directed exchange. Acidic extracellular pH (pH(o)) inhibited all modes of hSLC26A2-mediated anion exchange. In contrast, acidic intracellular pH (pH(i)) selectively activated exchange of extracellular Cl(-) for intracellular (35)SO(4)(2-) but not for intracellular (36)Cl(-) or [(14)C]oxalate. Protein kinase C inhibited hSLC26A2 by reducing its surface abundance. Diastrophic dysplasia mutants R279W and A386V of hSLC26A2 exhibited similar reductions in uptake of both (35)SO(4)(2-) and [(14)C]oxalate. A386V surface abundance was reduced, but R279W surface abundance was at wild-type levels.


Subject(s)
Anion Transport Proteins/metabolism , Cell Membrane/metabolism , Oxalic Acid/metabolism , Sulfates/metabolism , Animals , Anion Transport Proteins/genetics , Biological Transport , Cell Membrane/drug effects , Humans , Hydrogen-Ion Concentration , Kinetics , Mice , Mutation , Oocytes , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Sodium Chloride/metabolism , Sulfate Transporters , Xenopus
3.
Hum Mutat ; 30(4): 599-608, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19204907

ABSTRACT

Hearing loss with enlargement of the vestibular aqueduct (EVA) can be associated with mutations of the SLC26A4 gene encoding pendrin, a transmembrane Cl(-)/I(-)/HCO(3)(-) exchanger. Pendrin's critical transport substrates are thought to be I(-) in the thyroid gland and HCO(3)(-) in the inner ear. We previously reported that bi-allelic SLC26A4 mutations are associated with Pendred syndromic EVA whereas one or zero mutant alleles are associated with nonsyndromic EVA. One study proposed a correlation of nonsyndromic EVA with SLC26A4 alleles encoding pendrin with residual transport activity. Here we describe the phenotypes and SLC26A4 genotypes of 47 EVA patients ascertained since our first report of 39 patients. We sought to determine the pathogenic potential of each variant in our full cohort of 86 patients. We evaluated the trafficking of 11 missense pendrin products expressed in COS-7 cells. Products that targeted to the plasma membrane were expressed in Xenopus oocytes for measurement of anion exchange activity. p.F335L, p.C565Y, p.L597S, p.M775T, and p.R776C had Cl(-)/I(-) and Cl(-)/HCO(3)(-) exchange rate constants that ranged from 13 to 93% of wild type values. p.F335L, p.L597S, p.M775T and p.R776C are typically found as mono-allelic variants in nonsyndromic EVA. The high normal control carrier rate for p.L597S indicates it is a coincidentally detected nonpathogenic variant in this context. We observed moderate differential effects of hypo-functional variants upon exchange of HCO(3)(-) versus I(-) but their magnitude does not support a causal association with nonsyndromic EVA. However, these alleles could be pathogenic in trans configuration with a mutant allele in Pendred syndrome.


Subject(s)
Hearing Loss/genetics , Membrane Transport Proteins/genetics , Mutation , Vestibular Aqueduct/metabolism , Adolescent , Adult , Animals , COS Cells , Cell Membrane/metabolism , Child , Child, Preschool , Chlorocebus aethiops , Female , Genetic Variation , Genotype , Hearing Loss/metabolism , Hearing Loss/pathology , Humans , Infant , Male , Membrane Transport Proteins/metabolism , Oocytes/cytology , Oocytes/metabolism , Phenotype , Polymorphism, Genetic , Protein Transport , Sulfate Transporters , Syndrome , Transfection , Vestibular Aqueduct/abnormalities , Xenopus
4.
J Physiol ; 586(16): 3813-24, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18565999

ABSTRACT

Transcellular Cl(-) and HCO(3)(-) transport is a vital function of secretory epithelia and exit across the luminal membrane is mediated by members of the SLC26 transporters in conjunction with cystic fibrosis transmembrane conductance regulator (CFTR) channel. Typically, secretory epithelia express several SLC26 transporters in the same tissue; however, how their specific function is determined in vivo is not known. In the present work we used the parotid gland duct which expressed Slc26a4 and Slc26a6 and the model systems of Slc26a4(-/-) and Slc26a6(-/-) mice to study the role and regulation of these SLC26 transporters. We examined the transport modes of SLC26A4 expressed in Xenopus oocytes and report that SLC26A4 functions as a coupled, electroneutral I(-)/Cl(-), I(-)/HCO(3)(-) and Cl(-)/HCO(3)(-) exchanger with 1: 1 stoichiometry, with I(-) as the preferred anion. In the duct, Slc26a4 is expressed in the luminal membrane and mainly mediates I(-) secretion with minimal role in luminal HCO(3)(-) transport. By contrast, Slc26a6 mediates luminal Cl(-)/HCO(3)(-) exchange activity with minimal role in I(-) secretion. Furthermore, silencing of CFTR altered Cl(-)/HCO(3)(-) exchange by Slc26a6, but had no effect on I(-) secretion by Slc26a4. Accordingly, deletion of Slc26a6, but not deletion of Slc26a4, results in dysregulation of CFTR. These findings provide the first evidence for a selective role of the SLC26 transporters expressed in the same tissue in epithelial anion transport and suggest that transport specificity is achieved by both the properties of the transporters and the composition of the complexes they form.


Subject(s)
Antiporters/metabolism , Bicarbonates/metabolism , Chloride-Bicarbonate Antiporters/metabolism , Chlorine/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Membrane Transport Proteins/metabolism , Parotid Gland/metabolism , Animals , Cells, Cultured , Gene Expression Regulation/physiology , Hydrogen-Ion Concentration , Iodine/metabolism , Oocytes/metabolism , Signal Transduction/physiology , Sulfate Transporters , Xenopus laevis
5.
Am J Physiol Cell Physiol ; 292(4): C1485-92, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17151144

ABSTRACT

SLC26A6 (CFEX, PAT1) is an anion exchanger expressed in several tissues including renal proximal tubule, pancreatic duct, small intestine, liver, stomach, and heart. It has recently been reported that PKC activation inhibits A6-mediated Cl/HCO(3) exchange by disrupting binding of carbonic anhydrase to A6. However, A6 can operate in HCO(3)-independent exchange modes of physiological importance, as A6-mediated Cl/oxalate exchange plays important roles in proximal tubule NaCl reabsorption and intestinal oxalate secretion. We therefore examined whether PKC activation affects HCO(3)-independent exchange modes of Slc26a6 functionally expressed in Xenopus oocytes. We found that PKC activation inhibited Cl/formate exchange mediated by Slc26a6 but failed to inhibit the related anion exchanger pendrin (SLC26A4) under identical conditions. PKC activation inhibited Slc26a6-mediated Cl/formate exchange, Cl/oxalate exchange, and Cl/Cl exchange to a similar extent. The inhibitor sensitivity profile and the finding that PMA-induced inhibition was calcium independent suggested a potential role for PKC-delta. Indeed, the PKC-delta-selective inhibitor rottlerin significantly blocked PMA-induced inhibition of Slc26a6 activity. Localization of Slc26a6 by immunofluorescence microscopy demonstrated that exposure to PKC activation led to redistribution of Slc26a6 from the oocyte plasma membrane to the intracellular compartment immediately below it. We also observed that PMA decreased the pool of Slc26a6 available to surface biotinylation but had no effect on total Slc26a6 expression. The physiological significance of these findings was supported by the observation that PKC activation inhibited mouse duodenal oxalate secretion, an effect blocked by rottlerin. We conclude that multiple modes of anion exchange mediated by Slc26a6 are negatively regulated by PKC-delta activation.


Subject(s)
Antiporters/metabolism , Protein Kinase C-delta/physiology , Acetophenones/pharmacology , Animals , Anion Transport Proteins/metabolism , Benzopyrans/pharmacology , Biological Transport, Active , Carbazoles/pharmacology , Cell Membrane/metabolism , Chlorides/metabolism , Cytoplasm/metabolism , Enzyme Activation , Female , Formates/metabolism , In Vitro Techniques , Indoles/pharmacology , Maleimides/pharmacology , Mice , Oocytes/metabolism , Oxalates/metabolism , Protein Kinase C-delta/antagonists & inhibitors , Protein Transport , Sulfate Transporters , Tetradecanoylphorbol Acetate/pharmacology , Xenopus
6.
Am J Med Genet A ; 140(11): 1143-7, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16642506

ABSTRACT

Diastrophic dysplasia sulfate transporter (DTDST) is a sulfate transporter required for the synthesis of sulfated proteoglycans in the cartilage. Over 30 mutations have been described in the DTDST gene, which result in a continuous clinical spectrum of recessively inherited chondrodysplasias, including, in order of increasing severity, a recessive form of multiple epiphyseal dysplasia (rMED), diastrophic dysplasia (DTD), atelosteogenesis type II (AO-II) and achondrogenesis 1B (ACG-1B). Correlation between disease severity and residual sulfate transport activity has been reported. Here we report a patient with DTDST mutations, whose manifestations fell in a range between AO-II and DTD. The patient was a compound heterozygote for the recurrent c.835C>T (p.R279W) and novel c.1987G>A (p.G663R) mutations. Immunocytochemical analysis in HEK293 cells showed that the p.G663R mutation was localized within the cytoplasm, and not to the cell membrane, suggesting p.G663R is a loss-of-function mutation. Our case supports the previously described correlation between the severity of the phenotype and the putative level of residual transport function.


Subject(s)
Abnormalities, Multiple/genetics , Fetal Diseases/genetics , Membrane Transport Proteins/genetics , Mutation , Osteochondrodysplasias/pathology , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Abortion, Eugenic , Amino Acid Sequence , Anion Transport Proteins , Base Sequence , Cell Line , DNA Mutational Analysis , Female , Fetal Diseases/metabolism , Fetal Diseases/pathology , Gestational Age , Heterozygote , Humans , Immunohistochemistry , Membrane Transport Proteins/analysis , Molecular Sequence Data , Phenotype , Pregnancy , Sequence Homology, Amino Acid , Sulfate Transporters , Ultrasonography, Prenatal
7.
Hum Mol Genet ; 13(19): 2165-71, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15294877

ABSTRACT

Defects in sulfate transport in chondrocytes lead to undersulfation of the cartilage extracellular matrix proteoglycans. Mutations in the diastrophic dysplasia sulfate transporter (DTDST) gene have been linked to four chondrodysplasias of varying severity. To characterize disease-causing mutations of DTDST, we expressed DTDST-mediated sulfate transport in mammalian HEK-293 cells and determined that the wild-type protein is glycosylated and localized to the cell plasma membrane. Four mutations, A715V, C653S, Q454P and R279W, stimulated sulfate transport at rates only 39-62% of wild-type DTDST. These four mutations were expressed on the plasma membrane of the cell, but the amount of expressed protein was reduced when compared with wild-type DTDST. The Q454P mutant is unique in that it is not properly glycosylated in HEK cells. There was no difference in sulfate transport activity between cells transfected with either the DeltaV340 or the G678V mutations and control HEK cells. Furthermore, the G678V mutation is not expressed along the plasma membrane, but is trapped within the cytoplasm. When comparing the sulfate transport capacity of each DTDST mutation with the chondrodysplasia in which it has been identified, we find that individuals with severe achondrogenesis 1B phenotype have null mutations on both DTDST alleles. Heterozygotes for both a null mutation and a partial-function mutation result in either atelosteogenesis type 2 or DTD, whereas the milder, recessive multiple epiphyseal dysplasia phenotype is homozygous for partial-function mutations. In contrast to previous studies in Xenopus laevis oocytes, we find a strong correlation between the severity of the phenotype and the level of residual transport function in mammalian cells.


Subject(s)
Carrier Proteins/genetics , Mutation/genetics , Anion Transport Proteins , Biological Transport , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cells, Cultured , Cytoplasm , Genotype , Glycosylation , Heterozygote , Humans , Kidney/metabolism , Membrane Transport Proteins , Phenotype , Sulfate Transporters , Sulfates/metabolism
8.
Am J Physiol Renal Physiol ; 283(5): F952-6, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12372770

ABSTRACT

A significant fraction of active chloride reabsorption across the apical membrane of the proximal tubule is mediated by a chloride/formate exchange process, whereby intracellular formate drives the transport of chloride into the cell. When chloride/formate exchange operates in parallel with Na(+)/H(+) exchange and H(+)-coupled recycling of formate, the net result is electroneutral NaCl reabsorption. Pendrin is the protein product of the PDS gene (SLC26A4) and functions in several different anion exchange modes, including chloride/formate exchange. Pendrin is expressed in the kidney and may serve as the transporter responsible for formate-dependent NaCl reabsorption. In the present study, Pds-knockout mice were used to determine the role of pendrin in proximal tubule chloride reabsorption. We show that formate-dependent NaCl absorption in microperfused proximal tubules is similar between wild-type and pendrin-deficient mice. In addition, there is no difference in the rate of formate-mediated chloride transport in brush-border membrane vesicles isolated from wild-type and pendrin-deficient mice. These studies demonstrate that pendrin is not responsible for formate-dependent NaCl reabsorption in the proximal tubule.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Kidney Tubules, Proximal/metabolism , Membrane Transport Proteins , Sodium Chloride/metabolism , Animals , Biological Transport/physiology , Female , Formates/pharmacokinetics , Hemostatics/pharmacokinetics , Male , Mice , Mice, Knockout , Microvilli/metabolism , Sulfate Transporters
SELECTION OF CITATIONS
SEARCH DETAIL
...