Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 207
Filter
Add more filters










Publication year range
1.
Kidney Int ; 72(9): 1043-5, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17943150

ABSTRACT

Normal urine contains thousands of proteins, largely due to the presence of 'exosomes,' tiny vesicles secreted into the urine by renal epithelial cells. These exosomes, demonstrated by Keller and colleagues to be also retrievable from amniotic fluid, offer great promise for future disease biomarker discovery studies.


Subject(s)
Kidney/metabolism , Proteinuria/metabolism , Secretory Vesicles/metabolism , Amniotic Fluid/chemistry , Amniotic Fluid/metabolism , Biomarkers/urine , CD24 Antigen/urine , Calcium-Binding Proteins/urine , Carrier Proteins/urine , Cell Cycle Proteins/urine , Endosomal Sorting Complexes Required for Transport , Epithelium/metabolism , Epithelium/pathology , Humans , Kidney/pathology , Proteinuria/pathology , RNA, Messenger/urine , Ubiquitin/urine
2.
Kidney Int ; 70(10): 1847-57, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17021608

ABSTRACT

Urinary exosomes containing apical membrane and intracellular fluid are normally secreted into the urine from all nephron segments, and may carry protein markers of renal dysfunction and structural injury. We aimed to discover biomarkers in urinary exosomes to detect acute kidney injury (AKI), which has a high mortality and morbidity. Animals were injected with cisplatin. Urinary exosomes were isolated by differential centrifugation. Protein changes were evaluated by two-dimensional difference in gel electrophoresis and changed proteins were identified by mass spectrometry. The identified candidate biomarkers were validated by Western blotting in individual urine samples from rats subjected to cisplatin injection; bilateral ischemia and reperfusion (I/R); volume depletion; and intensive care unit (ICU) patients with and without AKI. We identified 18 proteins that were increased and nine proteins that were decreased 8 h after cisplatin injection. Most of the candidates could not be validated by Western blotting. However, exosomal Fetuin-A increased 52.5-fold at day 2 (1 day before serum creatinine increase and tubule damage) and remained elevated 51.5-fold at day 5 (peak renal injury) after cisplatin injection. By immunoelectron microscopy and elution studies, Fetuin-A was located inside urinary exosomes. Urinary Fetuin-A was increased 31.6-fold in the early phase (2-8 h) of I/R, but not in prerenal azotemia. Urinary exosomal Fetuin-A also increased in three ICU patients with AKI compared to the patients without AKI. We conclude that (1) proteomic analysis of urinary exosomes can provide biomarker candidates for the diagnosis of AKI and (2) urinary Fetuin-A might be a predictive biomarker of structural renal injury.


Subject(s)
Acute Kidney Injury/urine , Blood Proteins/urine , Proteomics/methods , Reperfusion Injury/urine , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Biomarkers/urine , Cell Membrane/metabolism , Cisplatin/adverse effects , Cisplatin/pharmacology , Female , Humans , Kidney/drug effects , Kidney/injuries , Kidney/pathology , Male , Middle Aged , Models, Animal , Rats , Reperfusion Injury/etiology , Reperfusion Injury/pathology , alpha-2-HS-Glycoprotein , alpha-Fetoproteins/urine
3.
Kidney Int ; 70(6): 1026-37, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16850027

ABSTRACT

We aimed to investigate the molecular mechanisms underlying the renal wasting of Na(+), K(+), Ca(2+), and Mg(2+) in gentamicin (GM)-treated rats. Male Wistar rats were injected with GM (40 or 80 mg/kg/day for 7 days, respectively; GM-40 or GM-80). The expression of NHE3, Na-K-ATPase, NKCC2, ROMK, NCC, alpha-, beta- and gamma-ENaC, and CaSR was examined in the kidney by immunoblotting and immunohistochemistry. Urinary fractional excretion of Na(+), K(+), Ca(2+), and Mg(2+) was increased and urinary concentration was decreased in both GM-40 and GM-80 rats. In cortex and outer stripe of outer medulla (cortex) in GM-80 rats, the expression of NHE3, Na-K-ATPase, and NKCC2 was decreased; NCC expression was unchanged; and CaSR was upregulated compared to controls. In the inner stripe of outer medulla (ISOM) in GM-80 rats, NKCC2 and Na-K-ATPase expression was decreased, whereas CaSR was upregulated, and NHE3 and ROMK expression remained unchanged. In GM-40 rats, NKCC2 expression was decreased in the cortex and ISOM, whereas NHE3, Na-K-ATPase, CaSR, ROMK, and NCC abundance was unchanged in both cortex and ISOM. Immunoperoxidase labeling confirmed decreased expression of NKCC2 in the thick ascending limb (TAL) in both GM-80- and GM-40-treated rats. Immunoblotting and immunohistochemical analysis revealed increased expression of alpha-, beta-, and gamma-ENaC in cortex in GM-80 rats, but not in GM-40 rats. These findings suggest that the decrease in NKCC2 in TAL seen in response to low-dose (40 mg/kg/day) gentamicin treatment may play an essential role for the increased urinary excretion of Mg(2+) and Ca(2+), and play a significant role for the development of the urinary concentrating defect, and increased urinary excretion of Na(+) and K(+). At high-dose gentamicin, both proximal and TAL sodium transporter downregulation is likely to contribute to this.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gentamicins/pharmacology , Kidney/metabolism , Sodium Channels/drug effects , Sodium/metabolism , Animals , Anti-Bacterial Agents/pharmacokinetics , Calcium/urine , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gentamicins/pharmacokinetics , Immunohistochemistry , Kidney/drug effects , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Kidney Medulla/drug effects , Kidney Medulla/metabolism , Magnesium/urine , Male , Potassium/urine , Rats , Rats, Wistar , Receptors, Calcium-Sensing/metabolism , Sodium/urine , Sodium Chloride Symporters/metabolism , Sodium-Bicarbonate Symporters/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
4.
Kidney Int ; 70(4): 660-8, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16807546

ABSTRACT

In the proximal tubule, angiotensin II (Ang-II) regulates HCO(-)(3) reabsorption and H+ secretion by binding the type 1 Ang-II (AT1) receptor, stimulating Na(+)/HCO(-)(3) cotransport and Na(+)/H(+) exchange. Studies were carried out to determine if long-term changes in Ang-II receptor occupation alter the abundance of the basolateral Na(+)/HCO(-)(3) cotransporter (NBC1) or the apical membrane type 3 Na(+)/H(+) exchanger (NHE3). In the first set of experiments, rats eating a low-sodium diet were infused with the AT1 blocker, candesartan, or vehicle. In the second, lisinopril-infused rats were infused with either Ang II or vehicle. Transporter abundances were determined in whole kidney homogenates (WKH) and in brush border membrane (BBM) preparations by semiquantitative immunoblotting. Tissue distribution of transporters was assessed by immunocytochemistry. Blockade of the AT1 receptor by candesartan caused decreased abundance of NBC1 in WKH (59 +/- 9% of control; P<0.05) and Ang-II infusion increased abundance (130 +/- 7% of control; P<0.05). Changes in NBC1 in response to candesartan were confirmed immunohistochemically. Neither candesartan nor Ang II infusion affected the abundance of NHE3 in WKH or cortical homogenates. Candesartan decreased type 2 sodium-phosphate cotransporter abundance in both WKH (52 +/- 7% of control; P<0.05) and BBM (32 +/- 7% of control; P<0.05). Serum bicarbonate was decreased by candesartan and increased by Ang-II. Candesartan also decreased urinary ammonium excretion (P<0.05). The long-term effects of Ang-II in the proximal tubule may be mediated in part by regulation of NBC1 abundance, modifying bicarbonate reabsorption.


Subject(s)
Angiotensin II/physiology , Kidney Tubules, Proximal/metabolism , Sodium-Bicarbonate Symporters/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium-Phosphate Cotransporter Proteins, Type II/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin Receptor Antagonists , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Kidney Tubules, Proximal/drug effects , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Sodium-Bicarbonate Symporters/genetics , Sodium-Hydrogen Exchangers/genetics , Sodium-Phosphate Cotransporter Proteins, Type II/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Tetrazoles/pharmacology
5.
Kidney Int ; 69(8): 1471-6, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16501490

ABSTRACT

Urinary exosomes containing apical membrane and intracellular fluid are normally secreted into the urine from all nephron segments, and may carry protein markers of renal dysfunction and structural injury. We studied methods for collection, storage, and preservation of urinary exosomal proteins. We collected urine from healthy volunteers, added protease inhibitors, and stored urine samples at 4, -20, and -80 degrees C for 1 week or 7 months. Samples were thawed with and without extensive vortexing, and three fractions were isolated: urinary sediment, supernatant, and exosome fraction. Protein concentration, electrophoresis patterns, and abundance of seven exosome-associated proteins were measured. Exosome-associated proteins were not detected in sediment or supernatant fractions. Protease inhibitors prevented degradation of exosome-associated proteins. Freezing at -20 degrees C caused a major loss in exosomes compared to fresh urine. In contrast, recovery after freezing at -80 degrees C was almost complete. Extensive vortexing after thawing markedly increased exosome recovery in urine frozen at -20 or -80 degrees C, even if frozen for 7 months. The recovery from first and second morning urine was similar. The abundance of cytosolic exosome-associated proteins did not decrease during long-term storage. We concluded: (1) protease inhibitors are essential for preservation; (2) storage at -80 degrees C with extensive vortexing after thawing maximizes the recovery of urinary exosomes; (3) the difference between first and second morning urine exosome-associated protein was small, suggesting minimal protein degradation in the urinary tract/bladder; (4) urinary exosomes remain intact during long-term storage. These urine collection, storage, and processing conditions may be useful for future biomarker discovery efforts.


Subject(s)
Biomarkers/urine , Cryopreservation , Membrane Proteins/urine , Peptide Fragments/urine , Symporters/urine , Blotting, Western , Cryopreservation/instrumentation , Cryopreservation/methods , Cytosol/chemistry , Electrophoresis, Polyacrylamide Gel , Humans , Membrane Proteins/isolation & purification , Peptide Fragments/analysis , Peptide Fragments/isolation & purification , Protease Inhibitors/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Time Factors
6.
Kidney Int ; 69(1): 89-98, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16374428

ABSTRACT

We hypothesize that dysregulation of the epithelial sodium channel (ENaC) may be responsible for the increased sodium retention in liver cirrhosis. Liver cirrhosis was induced by common bile duct ligation (CBDL). We examined the abundance of ENaC subunits and type 2 isoform of 11beta-hydroxysteroid dehydrogenase (11betaHSD2) in the kidney by immunoblotting and immunohistochemistry at 6 or 8 weeks after operation. At 6 weeks, cirrhotic rats had developed ascites and displayed a positive sodium balance. The urinary sodium excretion and fractional excretion of sodium were decreased, while plasma aldosterone was unchanged. The abundance of ENaC subunits was not changed in the cortex and outer stripe of the outer medulla (OSOM). In contrast, immunoperoxidase microscopy revealed an increased apical targeting of alpha-, beta- and gammaENaC in late distal convoluted tubule, connecting tubule and collecting duct. Moreover, 11betaHSD2 abundance was decreased in the cortex/OSOM and inner stripe of the outer medulla. At 8 weeks, urinary sodium excretion and fractional excretion of sodium were not changed, while the plasma aldosterone level was decreased. The expression of ENaC subunits was decreased in the cortex/OSOM. Immunoperoxidase microscopy confirmed decreased expression of ENaC subunits, whereas subcellular localization was not changed. These results suggest that increased apical targeting of ENaC subunits and diminished abundance of 11betaHSD2 may contribute to promote sodium retention in the sodium-retaining stage of liver cirrhosis (at 6 weeks). The subsequent decreased expression and reduced targeting of ENaC subunits may play a role in promoting sodium excretion in the later stage of liver cirrhosis (at 8 weeks).


Subject(s)
Liver Cirrhosis, Experimental/metabolism , Sodium Channels/physiology , Sodium/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/analysis , Aldosterone/blood , Aldosterone/physiology , Animals , Cell Membrane/metabolism , Common Bile Duct , Epithelial Sodium Channels , Kidney/chemistry , Kidney/metabolism , Ligation , Male , Protein Subunits , Protein Transport , Rats , Rats, Wistar , Sodium Channels/analysis , Sodium-Potassium-Chloride Symporters/analysis
7.
J Membr Biol ; 212(2): 119-31, 2006.
Article in English | MEDLINE | ID: mdl-17264985

ABSTRACT

Urea movement across plasma membranes is modulated by specialized urea transporter proteins. These proteins are proposed to play key roles in the urinary concentrating mechanism and fluid homeostasis. To date, two urea-transporter genes have been cloned; UT-A (Slc14a2), encoding at least five proteins and UT-B (Slc14a1) encoding a single protein isoform. Recently we engineered mice that lack the inner medullary collecting duct (IMCD) urea transporters, UT-A1 and UT-A3 (UT-A1/3 -/- mice). This article includes 1) a historical review of the role of renal urea transporters in renal function; 2) a review of our studies utilizing the UT-A1/3 -/- mice; 3) description of an additional line of transgenic mice in which beta-galactosidase expression is driven by the alpha-promoter of the UT-A gene, which is allowing better physiological definition of control mechanisms for UT-A expression; and 4) a discussion of the implications of the studies in transgenic mice for the teaching of kidney physiology.


Subject(s)
Kidney Tubules, Collecting/metabolism , Kidney/physiology , Membrane Transport Proteins/metabolism , Mice/physiology , Urea/metabolism , Animals , Mice, Transgenic , Models, Animal , Models, Biological , Urea Transporters
8.
Acta Physiol Scand ; 173(1): 11-21, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11678722

ABSTRACT

Building on extensive physiological characterization of sodium transport mechanisms along the renal tubule over the past 30 years, complementary DNAs for almost all of the major transporters and channels responsible for renal tubular sodium reabsorption have been cloned over the past 10 years. The consequence is the generation of a broad range of cDNA and antibody probes which can be used to investigate physiological mechanisms on a molecular level. An ensemble of such probes can be exploited for comprehensive analysis of integrative physiological processes, approaches which are referred to as 'physiological genomics' or 'physiological proteomics'. In this review, we describe a targeted proteomic approach to comprehensive analysis of sodium transporter and water channel protein abundance along the renal tubule using an ensemble of rabbit polyclonal antibodies directed to the major sodium transporters and water channels expressed in each renal tubule segment. We discuss preparation and characterization of the antibodies, strategies for quantification of transporter protein abundance, and provide examples of the application of antibody-based targeted proteomics analysis of kidney tissue, revealing the effects of elevations of circulating aldosterone levels and circulating vasopressin levels on sodium transporter, sodium channel, and water channel abundance in kidney.


Subject(s)
Kidney/chemistry , Kidney/physiology , Proteome/analysis , Proteome/immunology , Animals , Antibodies , Aquaporins/analysis , Aquaporins/immunology , Epithelial Sodium Channels , Immunoblotting , Sodium Channels/analysis , Sodium Channels/immunology
9.
Exp Neurol ; 171(2): 227-34, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11573975

ABSTRACT

Vasopressin plays a role in both salt and water balance in the kidney. Classic studies, utilizing isolated perfused tubules, have revealed that vasopressin increases sodium reabsorption in the kidney thick ascending limb and the collecting duct. Furthermore, the activity of several sodium transport proteins expressed in these segments, such as the bumetanide-sensitive Na-K-2Cl cotransporter (NKCC2) and the epithelial sodium channel (ENaC), have been shown to be directly increased by vasopressin. Increased protein abundance might be one means through which sodium transporter and channel activity is enhanced. We have used immunoblotting and immunohistochemistry in order to investigate the regulation of abundance of the major sodium transporters and channels expressed along the renal tubule in response to vasopressin. Chronic (7-day) studies were performed in which vasopressin levels were elevated either endogenously by water restriction of Sprague-Dawley rats or exogenously through infusion of the vasopressin V2-receptor-selective agonist, dDAVP (1-deamino-8d-arginine-vasopressin), to Brattleboro rats. We found a significant increase in protein abundance for NKCC2 and the beta- and gamma-subunits of ENaC with either water restriction or dDAVP infusion. The alpha-subunit of Na-K-ATPase was increased by water restriction, but not by dDAVP infusion, and alpha-ENaC and the thiazide-sensitive cotransporter (NCC) were increased by dDAVP infusion but not by water restriction. Acute (60-min) in vivo exposure to dDAVP led to an increase in both beta- and gamma-ENaC abundance in kidney cortex homogenates, displaying the rapid nature of some of these changes. Overall these increases in sodium transporter and channel abundances likely contribute to both the antidiuretic and antinatriuretic actions of vasopressin.


Subject(s)
Kidney/physiology , Sodium Channels/physiology , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Deamino Arginine Vasopressin/pharmacology , Epithelial Sodium Channels , Homeostasis , Humans , Protein Subunits , Sodium Channels/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Solute Carrier Family 12, Member 1 , Water-Electrolyte Balance
10.
Am J Physiol Renal Physiol ; 281(4): F639-48, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11553510

ABSTRACT

Renal sodium retention, as a result of increased abundance of sodium transporters, may play a role in the development and/or maintenance of the increased blood pressure in obesity. To address this hypothesis, we evaluated the relative abundances of renal sodium transporters in lean and obese Zucker rats at 2 and 4 mo of age by semiquantitative immunoblotting. Mean systolic blood pressure was higher in obese rats relative to lean at 3 mo, P < 0.02. Furthermore, circulating insulin levels were 6- or 13-fold higher in obese rats compared with lean at 2 or 4 mo of age, respectively. The abundances of the alpha(1)-subunit of Na-K-ATPase, the thiazide-sensitive Na-Cl cotransporter (NCC or TSC), and the beta-subunit of the epithelial sodium channel (ENaC) were all significantly increased in the obese rats' kidneys. There were no differences for the sodium hydrogen exchanger (NHE3), the bumetanide-sensitive Na-K-2Cl cotransporter (NKCC2 or BSC1), the type II sodium-phosphate cotransporter (NaPi-2), or the alpha-subunit of ENaC. These selective increases could possibly increase sodium retention by the kidney and therefore could play a role in obesity-related hypertension.


Subject(s)
Carrier Proteins/metabolism , Kidney Cortex/metabolism , Obesity/metabolism , Sodium Channels/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Symporters , Animals , Blood Pressure , Epithelial Sodium Channels , Hyperinsulinism/metabolism , Loop of Henle/metabolism , Male , Rats , Rats, Zucker , Sodium/metabolism , Sodium Chloride/pharmacology , Sodium Chloride Symporters , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type II , Sodium-Potassium-Chloride Symporters
11.
Curr Opin Nephrol Hypertens ; 10(5): 655-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11496061

ABSTRACT

The regulation of sodium transport in the kidney is important for maintenance of extracellular fluid volume and arterial blood-pressure regulation. The major sodium transporters and channels in individual renal tubule segments have been identified via physiological techniques, and complementary DNAs for all of the key sodium transporters and channels expressed along the renal tubule have been cloned. Complementary DNA probes and antibodies are now being used to investigate the molecular basis of renal tubule sodium-transport regulation. This review summarizes some of the major observations made in the past year that are relevant to the regulation of the major sodium transporters in the proximal tubule (the type 3 sodium-hydrogen exchanger, NHE3), the thick ascending limb of Henle (the bumetanide-sensitive sodium-potassium-chloride cotransporter, NKCC2), and the distal convoluted tubule (the thiazide-sensitive sodium-chloride cotransporter, NCC).


Subject(s)
Carrier Proteins/metabolism , Kidney/metabolism , Receptors, Drug/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Symporters , Animals , Humans , Proteome/metabolism , Sodium Chloride Symporters , Sodium-Hydrogen Exchanger 3 , Solute Carrier Family 12, Member 1 , Solute Carrier Family 12, Member 3
12.
J Clin Invest ; 108(2): 215-22, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11457874

ABSTRACT

The kidneys "escape" from the Na-retaining effects of aldosterone when circulating levels of aldosterone are inappropriately elevated in the setting of normal or expanded extracellular fluid volume, e.g., in primary aldosteronism. Using a targeted proteomics approach, we screened renal protein extracts with rabbit polyclonal antibodies directed to each of the major Na transporters expressed along the nephron to determine whether escape from aldosterone-mediated Na retention is associated with decreased abundance of one or more of renal Na transporters. The analysis revealed that the renal abundance of the thiazide-sensitive Na-Cl cotransporter (NCC) was profoundly and selectively decreased. None of the other apical solute-coupled Na transporters displayed decreases in abundance, nor were the total abundances of the three ENaC subunits significantly altered. Immunocytochemistry showed a strong decrease in NCC labeling in distal convoluted tubules of aldosterone-escape rats with no change in the cellular distribution of NCC. Ribonuclease protection assays (RPAs) revealed that the decrease in NCC protein abundance was not associated with altered NCC mRNA abundance. Thus, the thiazide-sensitive Na-Cl cotransporter of the distal convoluted tubule appears to be the chief molecular target for regulatory processes responsible for mineralocorticoid escape, decreasing in abundance via a posttranscriptional mechanism.


Subject(s)
Aldosterone/metabolism , Carrier Proteins/metabolism , Kidney Cortex/metabolism , Kidney Medulla/metabolism , Kidney Tubules, Distal/metabolism , Symporters , Aldosterone/administration & dosage , Aldosterone/blood , Animals , Body Weight , Carrier Proteins/analysis , Carrier Proteins/immunology , Creatinine/blood , Male , Models, Animal , Natriuresis , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Sodium/urine , Sodium Channels/analysis , Sodium Chloride Symporters , Sodium Chloride, Dietary/administration & dosage , Sodium Chloride, Dietary/metabolism , Time Factors
13.
Am J Physiol Renal Physiol ; 281(1): F133-43, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11399654

ABSTRACT

UT-A1 is an extremely hydrophobic 929-amino acid integral membrane protein, expressed in the renal inner medullary collecting duct, with a central role in the urinary concentrating mechanism. Previous immunoblotting studies in rats have revealed that UT-A1 is present in kidney in 97- and 117-kDa monomeric forms and that the relative abundance of the two forms is altered by vasopressin treatment and other treatments that altered urinary inner medullary urea concentration. The present studies were carried out using protein chemistry techniques to determine the origin of the two forms. Peptide-directed polyclonal antibodies targeted to five sites along the polypeptide sequence from the NH2 to the COOH terminus labeled both forms, thus failing to demonstrate a significant deletion in the primary amino acid chain. The 97- and 117-kDa monomeric forms were both reduced to 88 kDa by deglycosylation with N-glycosidase F, indicating that a single polypeptide chain is glycosylated to two different extents. Studies using nonionic detergents for membrane solubilization or using homobifunctional cross-linkers demonstrated that UT-A1 exists as a 206-kDa protein complex in native kidney membranes. The mobility of this complex was also increased by deglycosylation. Both the 97- and 117-kDa proteins, as well as the 206-kDa complex, were immunoprecipitated with UT-A1 antibodies. We conclude that UT-A1 is a glycoprotein and that the two monomeric forms (97 and 117 kDa) in inner medullary collecting duct are the consequence of different states of glycosylation.


Subject(s)
Carrier Proteins/analysis , Kidney Tubules, Collecting/metabolism , Membrane Glycoproteins/analysis , Membrane Transport Proteins , Animals , Antibodies/immunology , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/immunology , Cell Membrane/metabolism , Cross-Linking Reagents , Electrophoresis , Epitopes/immunology , Glycosylation , Hexosaminidases/pharmacology , Immunoblotting , Male , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/immunology , Precipitin Tests , Protein Isoforms/chemistry , Rats , Rats, Sprague-Dawley , Vasopressins/pharmacology , Urea Transporters
14.
Am J Physiol Renal Physiol ; 281(1): F163-71, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11399657

ABSTRACT

Previously, we demonstrated that 24 h of bilateral ureteral obstruction (BUO) and short-term release of BUO was associated with a decrease in the expression of aquaporin-2 (AQP2), polyuria, and a reduced urinary concentrating capacity (10). The purposes of the present study were to examine whether BUO and the long-term release of BUO (BUO-R) for 3, 14, and 30 days were associated with changes in the expression of renal AQP1, AQP2, and AQP3 and whether such changes were associated with parallel changes in urinary output and urinary concentrating capacity. Rats (n = 4-7 in each group) were kept in metabolic cages for measurements of urinary output. Kidneys were removed to determine the expression levels of AQP1, AQP2, and AQP3 by semiquantitative immunoblotting. AQP2 was downregulated after 24 h of BUO (42 +/- 3%). Downregulation of AQP2 persisted 3 (43 +/- 14%; P < 0.01) and 15 days after BUO-R (48 +/- 11%; P < 0.01) but was normalized 30 days after BUO-R. AQP3 showed a similar pattern. Moreover, AQP1 was downregulated in response to BUO (65 +/- 7%) and remained downregulated 3 days after BUO-R (41 +/- 5%), 14 days after BUO-R (57 +/- 8%), and 30 days after BUO-R (59 +/- 5%). BUO-R resulted in a significant polyuria that gradually decreased, although it remained significant at day 30. Urinary concentrating capacity remained significantly impaired when determined 3, 14, and 30 days after BUO-R in response to a 24-h period of thirst (1,712 +/- 270 vs. 2,880 +/- 91 mosmol/kgH2O at day 30, P < 0.05). In conclusion, the expression of AQP1, AQP2, and AQP3 were long-term downregulated after BUO-R, suggesting that dysregulation of aquaporins located at the proximal tubule, thin descending limb of the loop of Henle, and the collecting duct may contribute to the long-term polyuria and impairment of urinary concentrating capacity associated with obstructive nephropathy.


Subject(s)
Aquaporins/metabolism , Nephrons/physiopathology , Ureteral Obstruction/physiopathology , Animals , Aquaporin 1 , Aquaporin 2 , Aquaporin 3 , Aquaporin 6 , Aquaporins/analysis , Cell Fractionation , Down-Regulation , Immunoblotting , Kidney Concentrating Ability , Kidney Tubules, Collecting/physiopathology , Loop of Henle/metabolism , Male , Osmolar Concentration , Polyuria/physiopathology , Rats , Rats, Wistar , Time Factors , Ureteral Obstruction/urine
15.
Am J Physiol Renal Physiol ; 280(6): F1093-106, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352848

ABSTRACT

Epithelial sodium channel (ENaC) subunit (alpha, beta, and gamma) mRNA and protein have been localized to the principal cells of the connecting tubule (CNT), cortical collecting duct (CCD), and outer medullary collecting duct (OMCD) in rat kidney. However, the subcellular localization of ENaC subunits in the principal cells of these cells is undefined. The cellular and subcellular localization of ENaC subunits in rat kidney was therefore examined. Immunocytochemistry demonstrated the presence of all three subunits in principal cells of the CNT, CCD, OMCD, and IMCD. In cortex and outer medulla, confocal microscopy demonstrated a difference in the subcellular localization of subunits. alpha-ENaC was localized mainly in a zone in the apical domains, whereas beta- and gamma-ENaC were found throughout the cytoplasm. Immunoelectron microscopy confirmed the presence of ENaC subunits in both the apical plasma membrane and intracellular vesicles. In contrast to the labeling pattern seen in cortex, alpha-ENaC labeling in IMCD cells was distributed throughout the cytoplasm. In the urothelium covering pelvis, ureters, and bladder, immunoperoxidase and confocal microscopy revealed differences the presence of all ENaC subunits. As seen in CCD, alpha-ENaC was present in a narrow zone near the apical plasma membrane, whereas beta- and gamma-ENaC were dispersed throughout the cytoplasm. In conclusion, all three subunits of ENaC are expressed throughout the collecting duct (CD), including the IMCD as well as in the urothelium. The intracellular vesicular pool in CD principal cells suggests ENaC trafficking as a potential mechanism for the regulation of Na(+) reabsorption.


Subject(s)
Kidney Tubules, Collecting/chemistry , Sodium Channels/analysis , Aldosterone/physiology , Animals , Antibody Specificity , Aquaporin 2 , Aquaporin 6 , Aquaporins/analysis , Aquaporins/immunology , Epithelial Sodium Channels , Immunoblotting , Immunohistochemistry , Kidney Cortex/chemistry , Kidney Cortex/metabolism , Kidney Cortex/ultrastructure , Kidney Medulla/chemistry , Kidney Medulla/metabolism , Kidney Medulla/ultrastructure , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/ultrastructure , Microscopy, Confocal , Microscopy, Immunoelectron , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Sodium Channels/immunology , Sodium Channels/metabolism , Urothelium/chemistry , Urothelium/ultrastructure
16.
Am J Physiol Regul Integr Comp Physiol ; 280(6): R1632-41, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11353665

ABSTRACT

Nifedipine, a calcium antagonist, has diuretic and natriuretic properties. However, the molecular mechanisms by which these effects are produced are poorly understood. We examined kidney abundance of aquaporins (AQP1, AQP2, and AQP3) and major sodium transporters [type 3 Na/H exchanger (NHE-3); type 2 Na-Pi cotransporter (NaPi-2); Na-K-ATPase; type 1 bumetanide-sensitive cotransporter (BSC-1); and thiazide-sensitive Na-Cl cotransporter (TSC)] as well as inner medullary abundance of AQP2, phosphorylated-AQP2 (p-AQP2), AQP3, and calcium-sensing receptor (CaR). Rats treated with nifedipine orally (700 mg/kg) for 19 days had a significant increase in urine output, whereas urinary osmolality and solute-free water reabsorption were markedly reduced. Consistent with this, immunoblotting revealed a significant decrease in the abundance of whole kidney AQP2 (47 +/- 7% of control rats, P < 0.05) and in inner medullary AQP2 (60 +/- 7%) as well as in p-AQP2 abundance (17 +/- 6%) in nifedipine-treated rats. In contrast, whole kidney AQP3 abundance was significantly increased (219 +/- 28%). Of potential importance in modulating AQP2 levels, the abundance of CaR in the inner medulla was significantly increased (295 +/- 25%) in nifedipine-treated rats. Nifedipine treatment was also associated with increased urinary sodium excretion. Consistent with this, semiquantitative immunoblotting revealed significant reductions in the abundance of proximal tubule Na(+) transporters: NHE-3 (3 +/- 1%), NaPi-2 (53 +/- 12%), and Na-K-ATPase (74 +/- 5%). In contrast, the abundance of the distal tubule Na-Cl cotransporter (TSC) was markedly increased (240 +/- 29%), whereas BSC-1 in the thick ascending limb was not altered. In conclusion, 1) increased urine output and reduced urinary concentration in nifedipine-treated-rats may, in part, be due to downregulation of AQP2 and p-AQP2 levels; 2) CaR might be involved in the regulation of water reabsorption in the inner medulla collecting duct; 3) reduced expression of proximal tubule Na(+) transporters (NHE-3, NaPi-2, and Na, K-ATPase) may be involved in the increased urinary sodium excretion; and 4) increase in TSC expression may occur as a compensatory mechanism.


Subject(s)
Aquaporins/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Carrier Proteins/metabolism , Kidney/metabolism , Nifedipine/pharmacology , Sodium/metabolism , Symporters , Animals , Carrier Proteins/antagonists & inhibitors , Diuresis/drug effects , Female , Kidney/drug effects , Kidney Medulla/drug effects , Kidney Medulla/metabolism , Natriuresis/drug effects , Osmolar Concentration , Rats , Rats, Wistar , Receptors, Calcium-Sensing , Receptors, Cell Surface/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type I , Sodium-Phosphate Cotransporter Proteins, Type II , Urine/chemistry
17.
Biochem Biophys Res Commun ; 282(1): 194-9, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11263991

ABSTRACT

The SNARE hypothesis, describing a protein assembly-disassembly pathway, was recently proposed for the sequential steps of synaptic vesicle docking, activation, and fusion. To determine if SNARE proteins are involved in regulated exocytosis in eosinophils, the presence and functional role of SNAREs was examined in human blood eosinophils. Immunoblotting, subcellular fractionation, and immunocytochemistry documented that vesicle-associated membrane protein-2 (VAMP-2), a vesicle-SNARE, was expressed in human eosinophils. Syntaxin 4 and SNAP-25 were also detected. Sequencing of cloned RT-PCR products amplified from a domain conserved among VAMP isoforms revealed identity only to VAMP-2 but not to VAMP-1 or cellubrevin. Functional experiments revealed that tetanus toxin pretreatment, which cleaved VAMP-2 in eosinophils, significantly inhibited both IgE receptor- and phorbol ester-mediated exocytosis of eosinophil cationic protein (ECP) from streptolysin-O-permeabilized eosinophils. Thus, these results strongly suggest a critical role of SNAREs in regulated exocytosis in eosinophils.


Subject(s)
Blood Proteins/metabolism , Eosinophils/metabolism , Exocytosis/physiology , Membrane Proteins/physiology , Ribonucleases , Animals , Base Sequence , DNA, Complementary , Eosinophil Granule Proteins , Humans , Immunohistochemistry , Membrane Proteins/genetics , Molecular Sequence Data , R-SNARE Proteins , Rats , Rats, Wistar
18.
Am J Physiol Renal Physiol ; 280(4): F715-26, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11249863

ABSTRACT

Diabetes mellitus (DM) is associated with osmotic diuresis and natriuresis. At day 15, rats with DM induced by streptozotocin (n = 13) had severe hyperglycemia (27.1 +/- 0.4 vs. 4.7 +/- 0.1 mM in controls) and had a fivefold increase in water intake (123 +/- 5 vs. 25 +/- 2 ml/day) and urine output. Semiquantitative immunoblotting revealed a significant increase in inner medullary AQP2 (201 +/- 12% of control rats, P < 0.05) and phosphorylated (Ser(256)) AQP2 (p-AQP2) abundance (299 +/- 32%) in DM rats. Also, the abundance of inner medullary AQP3 was markedly increased to 171 +/- 19% of control levels (100 +/- 4%, n = 7, P < 0.05). In contrast, the abundance of whole kidney AQP1 (90 +/- 3%) and inner medullary AQP4 (121 +/- 16%) was unchanged in rats with DM. Immunoelectron microscopy further revealed an increased labeling of AQP2 in the apical plasma membrane of collecting duct principal cells (with less labeling in the intracellular vesicles) of DM rats, indicating enhanced trafficking of AQP2 to the apical plasma membrane. There was a marked increase in urinary sodium excretion in DM. Only Na(+)/H(+) exchanger NHE3 was downregulated (67 +/- 10 vs. 100 +/- 11%) whereas there were no significant changes in abundance of type 2 Na-phosphate cotransporter (128 +/- 6 vs. 100 +/- 10%); the Na-K-2Cl cotransporter (125 +/- 19 vs. 100 +/- 10%); the thiazide-sensitive Na-Cl cotransporter (121 +/- 9 vs. 100 +/- 10%); the alpha(1)-subunit of the Na-K-ATPase (106 +/- 7 vs. 100 +/- 5%); and the proximal tubule Na-HCO(3) cotransporter (98 +/- 16 vs. 100 +/- 7%). In conclusion, DM rats had an increased AQP2, p-AQP2, and AQP3 abundance as well as high AQP2 labeling of the apical plasma membrane, which is likely to represent a vasopressin-mediated compensatory increase in response to the severe polyuria. In contrast, there were no major changes in the abundance of AQP1, AQP4, and several major proximal and distal tubule Na(+) transporters except NHE3 downregulation, which may participate in the increased sodium excretion.


Subject(s)
Aquaporins/genetics , Aquaporins/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Kidney Concentrating Ability/physiology , Animals , Aquaporin 1 , Aquaporin 2 , Aquaporin 3 , Aquaporin 4 , Aquaporin 6 , Aquaporins/analysis , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Female , Fluorescent Antibody Technique , Kidney Tubules, Collecting/chemistry , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/ultrastructure , Kidney Tubules, Proximal/chemistry , Kidney Tubules, Proximal/metabolism , Microscopy, Electron , Natriuresis/physiology , Phosphorylation , Rats , Rats, Wistar , Serine/metabolism , Sodium/metabolism , Sodium-Bicarbonate Symporters , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Water/metabolism
19.
Proc Natl Acad Sci U S A ; 98(7): 4221-6, 2001 Mar 27.
Article in English | MEDLINE | ID: mdl-11274445

ABSTRACT

Pendrin is an anion transporter encoded by the PDS/Pds gene. In humans, mutations in PDS cause the genetic disorder Pendred syndrome, which is associated with deafness and goiter. Previous studies have shown that this gene has a relatively restricted pattern of expression, with PDS/Pds mRNA detected only in the thyroid, inner ear, and kidney. The present study examined the distribution and function of pendrin in the mammalian kidney. Immunolocalization studies were performed using anti-pendrin polyclonal and monoclonal antibodies. Labeling was detected on the apical surface of a subpopulation of cells within the cortical collecting ducts (CCDs) that also express the H(+)-ATPase but not aquaporin-2, indicating that pendrin is present in intercalated cells of the CCD. Furthermore, pendrin was detected exclusively within the subpopulation of intercalated cells that express the H(+)-ATPase but not the anion exchanger 1 (AE1) and that are thought to mediate bicarbonate secretion. The same distribution of pendrin was observed in mouse, rat, and human kidney. However, pendrin was not detected in kidneys from a Pds-knockout mouse. Perfused CCD tubules isolated from alkali-loaded wild-type mice secreted bicarbonate, whereas tubules from alkali-loaded Pds-knockout mice failed to secrete bicarbonate. Together, these studies indicate that pendrin is an apical anion transporter in intercalated cells of CCDs and has an essential role in renal bicarbonate secretion.


Subject(s)
Bicarbonates/metabolism , Carrier Proteins/metabolism , Kidney/metabolism , Membrane Transport Proteins , Animals , Biological Transport , Carrier Proteins/genetics , Fluorescent Antibody Technique , Humans , In Vitro Techniques , Kidney Tubules, Collecting/metabolism , Mice , Molecular Sequence Data , RNA, Messenger/metabolism , Rats , Sulfate Transporters , Tissue Distribution
20.
Am J Physiol Renal Physiol ; 280(2): F325-32, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11208608

ABSTRACT

UT-A3 has recently been identified as a splicing variant transcript of the UT-A gene present in the kidney. To study the cellular and subcellular localization of UT-A3, we raised a new polyclonal antibody to its COOH-terminal end. Immunoblots identified bands at 44 and 67 kDa predominately in the inner medulla and showed that the antibody does not recognize UT-A1. Deglycosylation with PNGase decreased the molecular mass of both forms to 40 kDa. UT-A3 is most abundant in the inner third of the inner medulla and is present in membrane fractions. Cell fractionation studies showed that UT-A3 is only detectable in inner medullary collecting duct (IMCD) cells. These observations were confirmed with immunolocalization studies demonstrating an exclusive labeling of IMCD cells. Double-labeling studies with anti-Na-K-ATPase demonstrated UT-A3 in intracellular membranes and in the apical region but were incompatible with a basolateral site for UT-A3. Although the function of this isoform in the inner medulla is unknown, the large abundance suggests that it may be important in the renal handling of urea.


Subject(s)
Carrier Proteins/metabolism , Kidney Tubules, Collecting/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Animals , Male , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Urea Transporters
SELECTION OF CITATIONS
SEARCH DETAIL
...