Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Am Soc Nephrol ; 18(10): 2672-82, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17855646

ABSTRACT

Pendrin is an apical anion exchanger found in type B and nonA-nonB intercalated cells that is involved in bicarbonate secretion. The purpose of this study was to establish the origin and fate of pendrin-positive intercalated cells in the mouse kidney. Using immunohistochemistry, we found that pendrin-positive cells first appeared in the connecting tubule at embryonic day 14 (E14) and subsequently in the medullary collecting duct at E18. Most of the pendrin-positive cells in the connecting tubule were nonA-nonB intercalated cells, wheras those in the medullary collecting duct were type B intercalated cells. In the cortical collecting duct, pendrin-positive cells appeared in the inner part at day 4 after birth and in the outer part at day 7. Pendrin-positive cells gradually disappeared by apoptosis from the inner part of the medullary collecting duct two weeks after birth. Using 5-bromo-2'deoxy-uridine (BrdU) to follow cell proliferation, we determined that selective proliferation of pendrin-positive intercalated cells does not occur; instead, these cells may arise from undifferentiated precursor cells from separate foci, one in the connecting tubule and one in the collecting duct.


Subject(s)
Anion Transport Proteins/metabolism , Cell Differentiation/physiology , Fetal Development/physiology , Fetus/metabolism , Kidney/growth & development , Animals , Animals, Newborn , Anion Exchange Protein 1, Erythrocyte/metabolism , Cell Proliferation , Kidney/cytology , Kidney/metabolism , Mice , Proton-Translocating ATPases/metabolism , Sulfate Transporters
2.
Am J Physiol Renal Physiol ; 291(5): F1052-60, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16735462

ABSTRACT

Recent studies of the distribution of PKC isoenzymes in the mouse kidney demonstrated that PKC-alpha, -beta(I), and -delta are expressed in intercalated cells. The purpose of this study was to identify the intercalated cell subtypes that express the different PKC isoenzymes and determine the location of the PKC isoenzymes within these cells. Adult C57BL/6 mice kidney tissues were processed for multiple-labeling immunohistochemistry. Antibodies against the vacuolar H(+)-ATPase and pendrin were used to identify intercalated cell subtypes, whereas antibodies against calbindin D(28K) and aquaporin-2 (AQP2) were used to identify connecting tubule cells and principal cells of the collecting duct, respectively. Within type A intercalated cells, PKC-delta was highly expressed in the apical part of the cells, whereas immunoreactivity for both PKC-alpha and PKC-beta(I) was weak. Type B intercalated cells exhibited strong expression of PKC-alpha, -beta(I), and -delta. PKC-alpha and -beta(I) were localized throughout the cytoplasm, whereas PKC-delta was restricted to the basal domain. Within non-A-non-B cells, immunoreactivity for both PKC-alpha and PKC-beta(I) was high in intensity and localized diffusely in the cytoplasm, whereas PKC-delta was localized in the apical part of the cells. None of the PKC isoenzymes (PKC-alpha, -beta(I), or -delta) were expressed in the calbindin D(28K)-positive connecting tubule cells. Within AQP2-positive principal cells of the collecting duct, PKC-alpha was expressed on the basolateral plasma membrane, but no significant staining was detected for PKC-beta(I) and -delta. In summary, this study demonstrates distinct and differential expression patterns of PKC-alpha, -beta(I), and -delta in the three subtypes of intercalated cells in the mouse kidney.


Subject(s)
Kidney/enzymology , Protein Kinase C/metabolism , Animals , Cell Polarity , Cytoplasm/enzymology , Immunohistochemistry , Kidney/cytology , Male , Mice , Mice, Inbred C57BL , Protein Kinase C beta , Protein Kinase C-alpha/metabolism , Protein Kinase C-delta/metabolism
3.
Am J Physiol Regul Integr Comp Physiol ; 290(2): R479-92, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16179486

ABSTRACT

Urea transport in the kidney is mediated by a family of transporter proteins, including renal urea transporters (UT-A) and erythrocyte urea transporters (UT-B). We aimed to determine whether hydration status affects the subcellular distribution of urea transporters. Male Sprague-Dawley rats were divided into three groups: dehydrated rats (WD) given minimum water, hydrated rats (WL) given 3% sucrose in water for 3 days before death, and control rats given free access to water. We labeled kidney sections with antibodies against UT-A1 and UT-A2 (L194), UT-A3 (Q2), and UT-B using preembedding immunoperoxidase and immunogold methods. In control animals, UT-A1 and UT-A3 immunoreactivities were observed throughout the cytoplasm in inner medullary collecting duct (IMCD) cells, and weak labeling was observed on the basolateral plasma membrane. UT-A2 immunoreactivity in the descending thin limbs (DTL) was observed mainly on the apical and basolateral membranes of type I epithelium, and very faint labeling was observed in the long-loop DTL at the border between the outer and inner medulla. UT-A1 immunoreactivity intensity was markedly lower, and UT-A3 immunoreactivity was higher in IMCD of WD vs. controls. UT-A2 immunoreactivity intensities in the plasma membrane and cytoplasm of type I, II, and III epithelia of DTL were greater in WD vs. controls. In contrast, UT-A1 expression was greater and UT-A2 and UT-A3 expressions were lower in WL vs. controls. The subcellular distribution of UT-A in DTL or IMCD did not differ between control and experimental animals. UT-B was expressed in the plasma membrane of the descending vasa recta of both control and experimental animals. UT-B intensity was higher in WD and lower in WL vs. controls. These data indicate that changes in hydration status over 3 days affected urea transporter protein expression without changing its subcellular distribution.


Subject(s)
Body Water/metabolism , Kidney/metabolism , Kidney/ultrastructure , Membrane Transport Proteins/metabolism , Animals , Male , Protein Transport , Rats , Rats, Sprague-Dawley , Urea Transporters
4.
Proc Natl Acad Sci U S A ; 102(20): 7251-6, 2005 May 17.
Article in English | MEDLINE | ID: mdl-15878989

ABSTRACT

Interleukin 10 (IL-10) is a pleiotropic cytokine with well known antiinflammatory, immunosuppressive, and immunostimulatory properties. Chronic allograft rejection, characterized by vascular neointimal proliferation, is a major cause of organ transplant loss, particularly in heart and kidney transplant recipients. In a Dark Agouti to Lewis rat model of aortic transplantation, we evaluated the effects of a single intramuscular injection of a recombinant adeno-associated viral vector (serotype 1) encoding IL-10 (rAAV1-IL-10) on neointimal proliferation and inflammation. rAAV1-IL-10 treatment resulted in a significant reduction of neointimal proliferation and graft infiltration with macrophages and T and B lymphocytes. The mechanism underlying the protective effects of IL-10 in aortic allografts involved heme oxygenase 1 (HO-1) because inhibition of HO activity reversed not only neointimal proliferation but also inflammatory cell infiltration. Our results indicate that IL-10 attenuates neointimal proliferation and inflammatory infiltration and strongly imply that HO-1 is an important intermediary through which IL-10 regulates the inflammatory responses associated with chronic vascular rejection.


Subject(s)
Aorta/transplantation , Cell Proliferation , Graft Rejection/prevention & control , Heat-Shock Proteins/metabolism , Inflammation/prevention & control , Interleukin-10/metabolism , Oxygenases/metabolism , Analysis of Variance , Animals , Blotting, Western , Dependovirus , Endothelial Cells/cytology , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , Graft Rejection/metabolism , Heme Oxygenase (Decyclizing) , Inflammation/metabolism , Interleukin-10/blood , Interleukin-10/genetics , Rats , Transgenes/genetics , Transplantation, Homologous
5.
Hum Gene Ther ; 16(2): 235-47, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15761263

ABSTRACT

Recombinant adeno-associated virus (rAAV) has become an attractive tool for gene therapy because of its ability to transduce both dividing and nondividing cells, elicit a limited immune response, and the capacity for imparting long-term transgene expression. Previous studies have utilized rAAV serotype 2 predominantly and found that transduction of vascular cells is relatively inefficient. The purpose of the present study was to evaluate the transduction efficiency of rAAV serotypes 1 through 5 in human and rat aortic endothelial cells (HAEC and RAEC). rAAV vectors with AAV2 inverted terminal repeats containing the human alpha1-antitrypsin (hAAT) gene were transcapsidated using helper plasmids to provide viral capsids for the AAV1 through 5 serotypes. True type rAAV2 and 5 vectors encoding beta-galactosidase or green fluorescence protein were also studied. Infection with rAAV1 resulted in the most efficient transduction in both HAEC and RAEC compared to other serotypes (p < 0.001) at 7 days posttransduction. Interestingly, expression was increased in cells transduced with rAAV5 to levels surpassing rAAV1 by day 14 and 21. Transduction with rAAV1 was completely inhibited by removal of sialic acid with sialidase, while heparin had no effect. These studies are the first demonstration that sialic acid residues are required for rAAV1 transduction in endothelial cells. Transduction of rat aortic segments ex vivo and in vivo demonstrated significant transgene expression in endothelial and smooth muscle cells with rAAV1 and 5 serotype vectors, in comparison to rAAV2. These results suggest the unique potential of rAAV1 and rAAV5-based vectors for vascular-targeted gene-based therapeutic strategies.


Subject(s)
DNA, Recombinant/genetics , Dependovirus/genetics , Endothelium, Vascular/metabolism , Genetic Vectors , Myocytes, Smooth Muscle/metabolism , Transduction, Genetic , Animals , Aorta/metabolism , Capsid/metabolism , Cells, Cultured , Endothelium, Vascular/chemistry , Endothelium, Vascular/virology , Green Fluorescent Proteins/metabolism , Heparin/metabolism , Humans , Male , Myocytes, Smooth Muscle/chemistry , Myocytes, Smooth Muscle/virology , N-Acetylneuraminic Acid/metabolism , Neuraminidase/pharmacology , Rats , Rats, Inbred Lew , alpha 1-Antitrypsin/genetics , beta-Galactosidase/metabolism
6.
Am J Physiol Renal Physiol ; 288(4): F694-702, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15572524

ABSTRACT

Endothelium-derived nitric oxide (NO) is synthesized within the developing kidney and may play a crucial role in the regulation of renal hemodynamics. The purpose of this study was to establish the expression and intrarenal localization of the NO-synthesizing enzyme endothelial NO synthase (eNOS) during kidney development. Rat kidneys from 14 (E14)-, 16 (E16)-, 18 (E18)-, and 20-day-old (E20) fetuses and 1 (P1)-, 3 (P3)-, 7 (P7)-, 14 (P14)-, and 21-day-old (P21) pups were processed for immunocytochemical and immunoblot analysis. In fetal kidneys, expression of eNOS was first observed in the endothelial cells of the undifferentiated intrarenal capillary network at E14. At E16, strong eNOS immunoreactivity was observed in the endothelial cells of renal vesicles, S-shaped bodies (stage II glomeruli), and stage III glomeruli at the corticomedullary junction. At E18-20, early-stage developing glomeruli located in the subcapsular region showed less strong eNOS immunoreactivity than those of E16. The eNOS-positive immature glomeruli were observed in the nephrogenic zone until 7 days after birth. In fetal kidneys, eNOS was also expressed in the medulla in the endothelial cells of the capillaries surrounding medullary collecting ducts. After birth, eNOS immunostaining gradually increased in the developing vascular bundles and peritubular capillaries in the medulla and was highest at P21. Surprisingly, eNOS was also expressed in proximal tubules, in the endocytic vacuolar apparatus, only at P1. The strong expression of eNOS in the early stages of developing glomeruli and vasculature suggests that eNOS may play a role in regulating renal hemodynamics of the immature kidney.


Subject(s)
Kidney/embryology , Kidney/enzymology , Nitric Oxide Synthase/metabolism , Age Factors , Animals , Blotting, Western , Female , Immunohistochemistry , Kidney/growth & development , Male , Nitric Oxide Synthase Type III , Pregnancy , Rats , Rats, Sprague-Dawley , Renal Circulation
7.
Am J Physiol Renal Physiol ; 285(6): F1210-24, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12952854

ABSTRACT

Urea transport in the kidney is mediated by a family of transporter proteins that include the renal urea transporter (UT-A) and the erythrocyte urea transporter (UT-B). The purpose of this study was to determine the location of the urea transporter isoforms in the mouse kidney and to examine the effects of prolonged potassium depletion on the expression and distribution of these transporters by ultrastructural immunocytochemistry. C57BL6 mice were fed a low-potassium diet for 2 wk, and control animals received normal chow. After 2 wk on a low-potassium diet, urinary volume increased and urinary osmolality decreased (833 +/- 30 vs. 1,919 +/- 174 mosmol/kgH2O), as previously demonstrated. Kidneys were processed for immunocytochemistry with antibodies against UT-A1 (L446), UT-A1 and UT-A2 (L194), UT-A3 (Q2), and UT-B. In normal mice, UT-A1 and UT-A3 were expressed mainly in the cytoplasm of the terminal inner medullary collecting duct (IMCD). UT-A2 immunoreactivity was observed mainly on the basolateral membrane of the type 1 epithelium of the descending thin limb (DTL) of short-looped nephrons. The intensity of UT-A1 and UT-A3 immunoreactivity in the IMCD was markedly reduced in potassium-depleted mice. In contrast, there was a significant increase in UT-A2 immunoreactivity in the DTL. The intensity of UT-B immunoreactivity in the descending vasa recta (DVR) was reduced in potassium-depleted animals compared with controls. In control animals, UT-B immunoreactivity was predominantly observed in the plasma membrane, whereas in potassium-depleted mice, it was mainly observed in cytoplasmic granules in endothelial cells of the DVR. In summary, potassium depletion is associated with reduced expression of UT-A1, UT-A3, and UT-B but increased expression of UT-A2. We conclude that reduced expression of urea transporters may play a role in the impaired urine-concentrating ability associated with potassium deprivation.


Subject(s)
Loop of Henle/metabolism , Membrane Transport Proteins/metabolism , Potassium/metabolism , Animals , Antibodies/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Immunohistochemistry , Kidney Concentrating Ability/physiology , Loop of Henle/cytology , Male , Membrane Transport Modulators , Membrane Transport Proteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Urea Transporters
8.
Am J Physiol Renal Physiol ; 285(6): F1244-57, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12944321

ABSTRACT

Prolonged lithium treatment of humans and rodents often results in hyperchloremic metabolic acidosis. This is thought to be caused by diminished net H+ secretion and/or excessive back-diffusion of acid equivalents. To explore whether lithium treatment is associated with changes in the expression of key renal acid-base transporters, semiquantitative immunoblotting and immunocytochemistry were performed using kidneys from lithium-treated (n = 6) and control (n = 6) rats. Rats treated with lithium for 28 days showed decreased urine pH, whereas no significant differences in blood pH and plasma HCO3- levels were observed. Immunoblot analysis revealed that lithium treatment induced a significant increase in the expression of the H+-ATPase (B1-subunit) in cortex (190 +/- 18%) and inner stripe of the outer medulla (190 +/- 9%), and a dramatic increase in inner medulla (900 +/- 104%) in parallel to an increase in the expression of type 1 anion exchanger (400 +/- 40%). This was confirmed by immunocytochemistry and immunoelectron microscopy, which also revealed increased density of intercalated cells. Moreover, immunoblotting and immunocytochemistry revealed a significant increase in the expression of the type 1 electrogenic Na+-HCO3- cotransporter (NBC) in cortex (200 +/- 23%) and of the electroneutral NBCn1 in inner stripe of the outer medulla (250 +/- 54%). In contrast, there were no changes in the expression of Na+/H+ exchanger-3 or of the Cl-/HCO3- exchanger pendrin. These results demonstrate that the expression of specific renal acid-base transporters is markedly altered in response to long-term lithium treatment. This is likely to represent direct or compensatory effects to increase the capacity for HCO3- reabsorption, NH4+ reabsorption, and proton secretion to prevent the development of systemic metabolic acidosis.


Subject(s)
Acidosis/metabolism , Anion Exchange Protein 1, Erythrocyte/metabolism , Diabetes Insipidus, Nephrogenic/metabolism , Membrane Transport Proteins , Sodium-Hydrogen Exchangers/metabolism , Acids/pharmacokinetics , Acids/urine , Animals , Anion Exchange Protein 1, Erythrocyte/immunology , Antibody Specificity , Blotting, Western , Carrier Proteins/metabolism , Diabetes Insipidus, Nephrogenic/chemically induced , Hydrogen-Ion Concentration , Immunohistochemistry , Kidney Cortex/metabolism , Kidney Tubules, Collecting/metabolism , Lithium , Male , Proton-Translocating ATPases/immunology , Proton-Translocating ATPases/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Bicarbonate Symporters/immunology , Sodium-Bicarbonate Symporters/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/immunology , Sulfate Transporters , Urine
9.
Am J Physiol Renal Physiol ; 285(3): F550-7, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12759225

ABSTRACT

Phenotype analysis has demonstrated that aquaporin-1 (AQP1) null mice are polyuric and manifest a urinary concentrating defect because of an inability to create a hypertonic medullary interstitium. We report here that deletion of AQP1 is also associated with a decrease in urinary pH from 6.15 +/- (SE) 0.1 to 5.63 +/- 0.07. To explore the mechanism of the decrease in urinary pH, we examined the expression of H+-ATPase in kidneys of AQP1 null mice. There was strong labeling for H+-ATPase in intercalated cells and proximal tubule cells in both AQP1 null and wild-type mice. Strong H+-ATPase immunostaining was also present in the apical plasma membrane of inner medullary collecting duct (IMCD) cells in AQP1 null mice, whereas no H+-ATPase labeling was observed in IMCD cells in wild-type mice. In addition, there was an increase in the prevalence of type A intercalated cells in the IMCD of AQP1 null mice, suggesting that the deletion of intercalated cells from the IMCD, which normally occurs during postnatal kidney development, was impaired. Western blot analysis of H+-ATPase expression in the different regions of the kidney demonstrated a significant increase in H+-ATPase protein in the inner medulla of AQP1 null mice compared with wild-type mice. There were no changes in H+-ATPase expression in the cortex or outer medulla. These results represent the first demonstration of apical H+-ATPase immunoreactivity in IMCD cells in vivo and suggest that the decrease in urinary pH observed in AQP1 null mice is due to upregulation of H+-ATPase in the IMCD. The induction of H+-ATPase expression in IMCD cells of AQP1 null mice may be related to the chronically low interstitial osmolality in these animals. The challenge will be to identify the molecular signal(s) responsible for the de novo H+-ATPase expression.


Subject(s)
Aquaporins/deficiency , Aquaporins/genetics , Gene Expression Regulation, Enzymologic , Kidney Tubules, Collecting/enzymology , Proton-Translocating ATPases/metabolism , Animals , Aquaporin 1 , Blotting, Western , Hydrogen-Ion Concentration , Immunohistochemistry , Kidney Tubules, Collecting/cytology , Mice , Mice, Knockout , Mice, Transgenic
10.
J Am Soc Nephrol ; 14(4): 947-58, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12660329

ABSTRACT

Gene therapy has the potential to provide a therapeutic strategy for numerous renal diseases such as diabetic nephropathy, chronic rejection, Alport syndrome, polycystic kidney disease, and inherited tubular disorders. In previous studies using cationic liposomes or adenoviral or retroviral vectors to deliver genes into the kidney, transgene expression has been transient and often associated with adverse host immune responses, particularly with the use of adenoviral vectors. The unique properties of recombinant adeno-associated viral (rAAV) vectors permit long-term stable transgene expression with a relatively low host immune response. The purpose of the present study was to evaluate gene expression in the rat kidney after intrarenal arterial infusion of a rAAV (serotype 2) vector encoding green fluorescence protein (GFP) induced by a cytomegalovirus-chicken beta-actin hybrid promoter. The left kidney of experimental animals was treated with either saline or transduced with rAAV2-GFP (0.125 ml/100 g body wt, 1 x 10(10)/ml infectious units) through the renal artery. A time-dependent expression of GFP was observed in all kidneys injected with rAAV2-GFP, with maximal expression observed at 6 wk posttransduction. The expression of GFP was restricted to cells in the S(3) segment of the proximal tubule and intercalated cells in the collecting duct, the latter identified by co-localization with H(+)-ATPase. No transduction was observed in the glomeruli or the intrarenal vasculature. These studies demonstrate successful transgene expression in tubular epithelial cells, specifically in the S(3) segment of the proximal tubule and intercalated cells, after intrarenal administration of a rAAV vector and provide the impetus for further studies to exploit its use as a tool for gene therapy in the kidney.


Subject(s)
Dependovirus/genetics , Epithelial Cells/physiology , Gene Expression/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Kidney Tubules/physiology , Transgenes/genetics , Animals , Genes, Reporter/genetics , Green Fluorescent Proteins , Indicators and Reagents/administration & dosage , Infusions, Intra-Arterial , Kidney/physiology , Luminescent Proteins/administration & dosage , Male , Rats , Rats, Inbred Lew , Renal Artery
11.
Nephron Physiol ; 93(3): p76-86, 2003.
Article in English | MEDLINE | ID: mdl-12660494

ABSTRACT

Osteopontin (OPN) is a secreted phosphoprotein expressed constitutively in the descending thin limb (DTL) and papillary surface epithelium (PSE) of the kidney. Although its function is not fully established, a role for OPN in the regulation of calcium-mediated or calcium-dependent processes has been proposed. The aim of this study was to examine the effects of 1,25-dihydroxyvitamin D(3) (vitD), a hormone involved in the regulation of calcium homeostasis, on renal OPN expression. Four groups of rats were studied: acute vehicle (single intraperitoneal [i.p.] injection of 0.1 ml 10% ethanol-90% propylene glycol, 12 h before being killed); acute vitD (single injection of vitD, 2 ng/g i.p., 12 h before being killed); chronic vehicle (daily subcutaneous [s.c.] injection of 0.1 ml 10% ethanol-90% propylene glycol for 7 days); and chronic vitD (daily s.c. injection of vitD, 0.5 ng/g, for 7 days). Kidneys were processed for light and electron microscope immunocytochemistry, in situ hybridization, and Western blot analysis. In vehicle-treated animals, OPN mRNA and protein were expressed primarily in the DTL and PSE. In the acute vitD group, OPN mRNA and immunoreactivity appeared in the thick ascending limb (TAL) of the inner stripe of the outer medulla, and increased slightly in the DTL and PSE. The proximal tubules exhibited strong OPN immunoreactivity, but no hybridization signal. In the chronic vitD group, there was a marked increase in OPN mRNA and immunoreactivity in the distal tubule, including the TAL, as well as in the DTL and PSE. A weak hybridization signal and immunostaining were also observed in some proximal tubules. Administration of vitD causes a marked increase in OPN mRNA and protein in the rat kidney, mainly in the distal nephron, but also in the DTL, PSE, and proximal tubules. These results indicate that vitD is involved in the regulation of OPN expression in the kidney.


Subject(s)
Calcitriol/pharmacology , Kidney/drug effects , Sialoglycoproteins/biosynthesis , Animals , Blotting, Western/methods , Calcitriol/administration & dosage , Cells, Cultured , Drug Delivery Systems/methods , Ethanol/administration & dosage , Ethanol/pharmacology , Immunohistochemistry/methods , In Situ Hybridization/methods , Injections, Intraperitoneal , Injections, Subcutaneous , Kidney/chemistry , Kidney/ultrastructure , Kidney Glomerulus/chemistry , Kidney Glomerulus/drug effects , Kidney Glomerulus/ultrastructure , Kidney Tubules, Proximal/chemistry , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/ultrastructure , Loop of Henle/chemistry , Loop of Henle/cytology , Loop of Henle/drug effects , Loop of Henle/ultrastructure , Male , Microscopy, Immunoelectron/methods , Osteopontin , Propylene Glycol/administration & dosage , Propylene Glycol/pharmacology , Rats , Rats, Sprague-Dawley , Sialoglycoproteins/immunology , Sialoglycoproteins/metabolism
12.
Am J Physiol Renal Physiol ; 284(3): F584-93, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12556366

ABSTRACT

The anion exchanger pendrin is present in the apical plasma membrane of type B and non-A-non-B intercalated cells of the cortical collecting duct (CCD) and connecting tubule and is involved in HCO(3)(-) secretion. In this study, we investigated whether the abundance and subcellular localization of pendrin are regulated in response to experimental metabolic acidosis and alkalosis with maintained water and sodium intake. NH(4)Cl loading (0.033 mmol NH(4)Cl/g body wt for 7 days) dramatically reduced pendrin abundance to 22 +/- 4% of control values (n = 6, P < 0.005). Immunoperoxidase labeling for pendrin showed reduced intensity in NH(4)Cl-loaded animals compared with control animals. Moreover, double-label laser confocal microscopy revealed a reduction in the fraction of cells in the CCD exhibiting pendrin labeling to 65% of the control value (n = 6, P < 0.005). Conversely, NaHCO(3) loading (0.033 mmol NaHCO(3)/g body wt for 7 days) induced a significant increase in pendrin expression to 153 +/- 11% of control values (n = 6, P < 0.01) with no change in the fraction of cells expressing pendrin. Immunoelectron microscopy revealed no major changes in the subcellular distribution, with abundant labeling in both the apical plasma membrane and the intracellular vesicles in all conditions. These results indicate that changes in pendrin protein expression play a key role in the well-established regulation of HCO(3)(-) secretion in the CCD in response to chronic changes in acid-base balance and suggest that regulation of pendrin expression may be clinically important in the correction of acid-base disturbances.


Subject(s)
Ammonium Chloride/pharmacology , Carrier Proteins/metabolism , Kidney/metabolism , Membrane Transport Proteins , Sodium Bicarbonate/pharmacology , Acidosis/metabolism , Alkalosis/metabolism , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Hydrogen-Ion Concentration/drug effects , Immunohistochemistry , Kidney/cytology , Kidney/drug effects , Kidney Tubules, Collecting/metabolism , Male , Microscopy, Immunoelectron , Osmolar Concentration , Potassium/blood , Rats , Rats, Wistar , Sodium/blood , Sulfate Transporters , Urine/chemistry
13.
Am J Physiol Renal Physiol ; 283(4): F744-54, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12217866

ABSTRACT

Recent studies have demonstrated that a novel anion exchanger, pendrin, is expressed in the apical domain of type B intercalated cells in the mammalian collecting duct. The purpose of this study was 1) to determine the expression and distribution of pendrin along the collecting duct and connecting tubule of mouse and rat kidney and establish whether pendrin is expressed in the non-A-non-B intercalated cells and 2) to determine the intracellular localization of pendrin in the different populations of intercalated cells by immunoelectron microscopy. A peptide-derived affinity-purified antibody was generated that specifically recognized pendrin in immunoblots of rat and mouse kidney. Immunohistochemistry and confocal laser scanning microscopy demonstrated the presence of pendrin in apical domains of all type B intercalated cells in mouse and rat connecting tubule and collecting duct. In addition, strong pendrin immunostaining was observed in non-A-non-B intercalated cells. There was no labeling of type A intercalated cells. Immunoelectron microscopy demonstrated that pendrin was located in the apical plasma membrane and intracellular vesicles of both type B intercalated cells and non-A-non-B cells; the latter was identified by the presence of H(+)-ATPase in the apical plasma membrane. The results of this study demonstrate that both pendrin and H(+)-ATPase are expressed in the apical plasma membrane of non-A-non-B intercalated cells, suggesting that these cells are capable of both HCO and proton secretion. Furthermore, the presence of pendrin in both the apical plasma membrane and the apical intracellular vesicles of type B and non-A-non-B intercalated cells suggests that HCO secretion may be regulated by trafficking of pendrin between the two membrane compartments.


Subject(s)
Carrier Proteins/biosynthesis , Kidney/metabolism , Membrane Transport Proteins , Amino Acid Sequence , Animals , Blotting, Western , Carrier Proteins/genetics , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Immunohistochemistry , Kidney/cytology , Kidney/ultrastructure , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Male , Membrane Proteins/biosynthesis , Membrane Proteins/chemistry , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Immunoelectron , Molecular Sequence Data , Proton-Translocating ATPases/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Subcellular Fractions/metabolism , Sulfate Transporters
14.
Am J Physiol Renal Physiol ; 283(3): F481-91, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12167599

ABSTRACT

Newborn rats are not capable of producing concentrated urine. With development of the concentrating system and a hypertonic medullary interstitium, intracellular osmolytes, such as sorbitol, accumulate in the renal medulla. Sorbitol is produced from glucose in a reaction catalyzed by aldose reductase (AR). The purpose of this study was to establish the time of expression and distribution of AR in the developing rat kidney. Kidneys from 16-, 18-, and 20-day-old fetuses and 1-, 3-, 4-, 5-, 7-, 14-, and 21-day-old pups were processed for immunohistochemistry and immunoblot analysis. In adult animals, AR was expressed only in the inner medulla, in which it was localized in ascending thin limbs (ATLs), inner medullary collecting ducts (IMCDs), and interstitial cells. AR immunoreactivity was not detected in fetal kidneys but was observed in the terminal part of the descending thin limb and IMCD in the renal papilla of 1-day-old pups. At birth, all of the loops of Henle are configured as short loops and there are no ATLs. After birth, papillary thick ascending limbs are gradually transformed into ATLs by a process that involves apoptotic deletion of cells from the thick ascending limb. During this time, AR immunoreactivity appeared in the cells undergoing transformation in the ascending limb, beginning at the papillary tip and ascending to the border between the outer medulla and the inner medulla. However, there was no labeling of apoptotic cells. The expression of AR in both the ATL and the IMCD gradually increased during kidney development. We conclude that AR expression in the inner medulla coincides with the increase in medullary tonicity that is known to occur during the first 3 wk after birth. On the basis of the observation that only AR-negative cells were deleted by apoptosis in the differentiating ATL, we propose that AR may protect ATL cells against apoptosis.


Subject(s)
Aldehyde Reductase/analysis , Kidney/enzymology , Kidney/growth & development , Aging , Aldehyde Reductase/physiology , Animals , Apoptosis , Blotting, Western , Gestational Age , Immunohistochemistry , Kidney/embryology , Kidney Cortex/enzymology , Kidney Medulla/enzymology , Loop of Henle/cytology , Loop of Henle/enzymology , Microscopy, Fluorescence , Rats , Rats, Sprague-Dawley
15.
Am J Physiol Renal Physiol ; 282(3): F530-40, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11832436

ABSTRACT

Urea transport in the kidney is mediated by a family of transporter proteins that includes renal urea transporters (UT-A) and erythrocyte urea transporters (UT-B). Because newborn rats are not capable of producing concentrated urine, we examined the time of expression and the distribution of UT-A and UT-B in the developing rat kidney by light and electron microscopic immunocytochemistry. Kidneys from 16-, 18-, and 20-day-old fetuses, 1-, 4-, 7-, 14-, and 21-day-old pups, and adult animals were studied. In the adult kidney, UT-A was expressed intensely in the inner medullary collecting duct (IMCD) and terminal portion of the short-loop descending thin limb (DTL) and weakly in long-loop DTL in the outer part of the inner medulla. UT-A immunoreactivity was not present in the fetal kidney but was observed in the IMCD and DTL in 1-day-old pups. The intensity of UT-A immunostaining in the IMCD gradually increased during postnatal development. In 4- and 7-day-old pups, UT-A immunoreactivity was present in the DTL at the border between the outer and inner medulla. In 14- and 21-day-old pups, strong UT-A immunostaining was observed in the terminal part of short-loop DTL in the outer medulla, and weak labeling remained in long-loop DTL descending into the outer part of the inner medulla. In the adult kidney, there was intense staining for UT-B in descending vasa recta (DVR) and weak labeling of glomeruli. In the developing kidney, UT-B was first observed in the DVR of a 20-day-old fetus. After birth there was a striking increase in the number of UT-B-positive DVR, in association with the formation of vascular bundles. The intensity of immunostaining remained strong in the outer medulla but gradually decreased in the inner medulla. We conclude that the expression of urea transporters in short-loop DTL and DVR coincides with the development of the ability to produce a concentrated urine.


Subject(s)
Carrier Proteins/analysis , Loop of Henle/chemistry , Loop of Henle/embryology , Membrane Glycoproteins/analysis , Membrane Transport Proteins , Age Factors , Animals , Antibody Specificity , Carrier Proteins/immunology , Immunoenzyme Techniques , Kidney Concentrating Ability/physiology , Loop of Henle/ultrastructure , Membrane Glycoproteins/immunology , Microscopy, Immunoelectron , Rats , Rats, Sprague-Dawley , Urea Transporters
16.
J Am Soc Nephrol ; 12(9): 1795-1804, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11518772

ABSTRACT

Aquaporin-4 (AQP4) is a member of the aquaporin water-channel family. AQP4 is expressed primarily in the brain, but it is also present in the collecting duct of the kidney, where it is located in the basolateral plasma membrane of principal cells and inner medullary collecting duct (IMCD) cells. Recent studies in the mouse also have reported the presence of AQP4 in the basolateral membrane of the proximal tubule. The purpose of this study was to establish the pattern of AQP4 expression during kidney development and in the adult kidney of both the mouse and the rat. Kidneys of adult and 3-, 7-, and 15-d-old mice and rats were preserved for immunohistochemistry and processed using a peroxidase pre-embedding technique. In both the mouse and the rat, strong basolateral immunostaining was observed in IMCD cells and principal cells in the medullary collecting duct at all ages examined. Labeling was weaker in the cortical collecting duct and the connecting tubule, and there was no labeling of connecting tubule cells in the mouse. In adult mouse kidney, strong AQP4 immunoreactivity was observed in the S3 segment of the proximal tubule. However, there was little or no labeling in the cortex or around the corticomedullary junction in 3- and 7-d-old mice. Between 7 and 15 d of age, distinct AQP4 immunoreactivity appeared in the S3 segment of the mouse proximal tubule concomitant with the differentiation of this segment of the nephron. Labeling of proximal tubules was never observed in the rat kidney. These results suggest that there are differences in transepithelial water transport between mouse and rat or that additional, not yet identified water channels exist in the rat proximal tubule.


Subject(s)
Aging/metabolism , Animals, Newborn/metabolism , Aquaporins/metabolism , Kidney/metabolism , Animals , Animals, Newborn/growth & development , Aquaporin 4 , Immunohistochemistry , Mice , Rats , Tissue Distribution
17.
J Am Soc Nephrol ; 12(7): 1410-1421, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11423570

ABSTRACT

In the developing rat kidney, there is no separation of the medulla into an outer and inner zone. At the time of birth, ascending limbs with immature distal tubule epithelium are present throughout the renal medulla, all loops of Henle resemble the short loop of adult animals, and there are no ascending thin limbs. It was demonstrated previously that immature thick ascending limbs in the renal papilla are transformed into ascending thin limbs by apoptotic deletion of cells and transformation of the remaining cells into a thin squamous epithelium. However, it is not known whether this is the only source of ascending thin limb cells or whether cell proliferation occurs in the segment undergoing transformation. This study was designed to address these questions and to identify sites of cell proliferation in the loop of Henle. Rat pups, 1, 3, 5, 7, and 14 d old, received a single injection of 5-bromo-2'-deoxyuridine (BrdU) 18 h before preservation of kidneys for immunohistochemistry. Thick ascending and descending limbs were identified by labeling with antibodies against the serotonin receptor, 5-HT(1A), and aquaporin-1, respectively. Proliferating cells were identified with an antibody against BrdU. BrdU-positive cells in descending and ascending limbs of the loop of Henle were counted and expressed as percentages of the total number of aquaporin-1-positive and 5-HT(1A)-positive cells in the different segments. In the developing kidney, numerous BrdU-positive nuclei were observed in the nephrogenic zone. Outside of this location, BrdU-positive tubule cells were most prevalent in medullary rays in the inner cortex and in the outer medulla. BrdU-labeled cells were rare in the papillary portion of the loop of Henle and were not observed in the lower half of the papilla after 3 d of age. BrdU-labeled nuclei were not observed in segments undergoing transformation or in newly formed ascending thin limb epithelium. It was concluded that the growth zone for the loop of Henle is located around the corticomedullary junction, and the ascending thin limb is mainly, if not exclusively, derived from cells of the thick ascending limb.


Subject(s)
Aging/physiology , Animals, Newborn/growth & development , Loop of Henle/cytology , Aging/metabolism , Animals , Animals, Newborn/metabolism , Bromodeoxyuridine/pharmacokinetics , Cell Division/physiology , Cell Nucleus/metabolism , Loop of Henle/metabolism , Rats , Rats, Sprague-Dawley
18.
J Am Soc Nephrol ; 12(7): 1434-1447, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11423572

ABSTRACT

Few studies have examined the role of the microvasculature in progressive renal disease. It was hypothesized that impaired angiogenesis might occur in the diseased kidney and could contribute to renal scarring. Progressive renal disease was induced in rats by 5/6 renal ablation and those rats were compared with sham-operated control animals at multiple time points, for examination of changes in the microvasculature and the expression of angiogenic factors. An early angiogenic response was documented in remnant kidneys, with increases in the proliferation of peritubular (1 wk) and glomerular (2 wk) endothelial cells. Subsequently, however, there was a decrease in endothelial cell proliferation, which was reduced to levels below those of sham-treated animals, in conjunction with interstitial expression of the antiangiogenic factor thrombospondin-1 (TSP-1) and decreased tubular expression of the proangiogenic factor vascular endothelial growth factor (VEGF). Both the increase in TSP-1 expression and the loss of VEGF expression were correlated with capillary loss and the development of glomerulosclerosis and interstitial fibrosis. Progressive macrophage infiltration was correlated both spatially and quantitatively with the sites of absent or diminished VEGF expression. In addition, macrophage-associated cytokines (interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha) inhibited VEGF mRNA expression and protein secretion by cultured tubular epithelial cells of the medullary thick ascending limb, under both normoxic and hypoxic conditions. Impaired angiogenesis characterizes the remnant kidney model and is correlated with progression. The impaired angiogenesis may be mediated by alterations in the renal expression of TSP-1 and VEGF, with the latter being regulated by macrophage-associated cytokines.


Subject(s)
Endothelial Growth Factors/physiology , Kidney Diseases/physiopathology , Lymphokines/physiology , Neovascularization, Physiologic/physiology , Nephrectomy , Thrombospondin 1/physiology , Animals , Blood Vessels/pathology , Cytokines/physiology , Endothelial Growth Factors/antagonists & inhibitors , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Loop of Henle/cytology , Loop of Henle/metabolism , Lymphokines/antagonists & inhibitors , Macrophages/metabolism , Macrophages/pathology , Male , Microcirculation , Proteinuria/pathology , Proteinuria/physiopathology , Rats , Rats, Sprague-Dawley , Renal Circulation , Renal Insufficiency/pathology , Renal Insufficiency/physiopathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...