Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters











Publication year range
1.
Am J Surg Pathol ; 42(12): 1571-1584, 2018 12.
Article in English | MEDLINE | ID: mdl-30285995

ABSTRACT

Our recent study revealed recurrent chromosomal losses and somatic mutations of genes in the Hippo pathway in mucinous tubular and spindle cell carcinoma (MTSCC). Here, we performed an integrative analysis of 907 renal cell carcinoma (RCC) samples (combined from The Cancer Genome Atlas and in-house studies) and the Knepper data set of microdissected rat nephrons. We identified VSTM2A and IRX5 as novel cancer-specific and lineage-specific biomarkers in MTSCC. We then assessed their expression by RNA in situ hybridization (ISH) in 113 tumors, including 33 MTSCC, 40 type 1 papillary RCC, 8 type 2 papillary RCC, 2 unclassified RCC, 15 clear cell RCC, and 15 chromophobe RCC. Sensitivity and specificity were calculated as the area under the receiver operating characteristics curve (AUC). All MTSCC tumors demonstrated moderate to high expression of VSTM2A (mean ISH score=255). VSTM2A gene expression assessed by RNA sequencing strongly correlated with VSTM2A ISH score (r(2)=0.81, P=0.00016). The majority of non-MTSCC tumors demonstrated negative or low expression of VSTM2A. IRX5, nominated as a lineage-specific biomarker, showed moderate to high expression in MTSCC tumors (mean ISH score=140). IRX5 gene expression assessed by RNA sequencing strongly correlated with IRX5 ISH score (r(2)=0.69, P=0.00291). VSTM2A (AUC: 99.2%) demonstrated better diagnostic efficacy than IRX5 (AUC: 87.5%), and may thus serve as a potential diagnostic marker to distinguish tumors with overlapping histology. Furthermore, our results suggest MTSCC displays an overlapping phenotypic expression pattern with the loop of Henle region of normal nephrons.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Biomarkers, Tumor/genetics , Carcinoma, Papillary/genetics , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Membrane Proteins/genetics , Adenocarcinoma, Mucinous/pathology , Adult , Aged , Aged, 80 and over , Animals , Canada , Carcinoma, Papillary/pathology , Carcinoma, Renal Cell/pathology , Diagnosis, Differential , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , In Situ Hybridization , Kidney Neoplasms/pathology , Loop of Henle/chemistry , Male , Middle Aged , Neoplasm Grading , Predictive Value of Tests , Rats , Reproducibility of Results , Transcription Factors/genetics , Tumor Burden , United States , Up-Regulation , Young Adult
2.
Urolithiasis ; 46(4): 333-341, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29234857

ABSTRACT

Most of kidney stones are supposed to originate from Randall's plaque at the tip of the papilla or from papillary tubular plugs. Nevertheless, the frequency and the composition of crystalline plugs remain only partly described. The objective was to assess the frequency, the composition and the topography of papillary plugs in human kidneys. A total of 76 papillae from 25 kidneys removed for cancer and without stones were analysed by immunohistochemistry combined with Yasue staining, field emission-scanning electron microscopy and Fourier transformed infrared micro-spectroscopy. Papillary tubular plugs have been observed by Yasue staining in 23/25 patients (92%) and 52/76 papillae (68%). Most of these plugs were made of calcium phosphate, mainly carbonated apatite and amorphous calcium phosphate, and rarely octacalcium phosphate pentahydrate. Calcium and magnesium phosphate (whitlockite) have also been observed. Based upon immunostaining coupled to Yasue coloration, most of calcium phosphate plugs were located in the deepest part of the loop of Henle. Calcium oxalate monohydrate and dihydrate tubular plugs were less frequent and stood in collecting ducts. At last, we observed calcium phosphate plugs deforming and sometimes breaking adjacent collecting ducts. Papillary tubular plugging, which may be considered as a potential first step toward kidney stone formation, is a very frequent setting, even in kidneys of non-stone formers. The variety in their composition and the distal precipitation of calcium oxalate suggest that plugs may occur in various conditions of urine supersaturation. Plugs were sometimes associated with collecting duct deformation.


Subject(s)
Kidney Calculi/etiology , Kidney Tubules, Collecting/pathology , Loop of Henle/pathology , Aged , Calcium Phosphates/analysis , Humans , Kidney Calculi/chemistry , Kidney Calculi/epidemiology , Kidney Calculi/ultrastructure , Kidney Tubules, Collecting/chemistry , Kidney Tubules, Collecting/diagnostic imaging , Loop of Henle/chemistry , Loop of Henle/diagnostic imaging , Microscopy, Electron, Scanning , Middle Aged , Spectroscopy, Fourier Transform Infrared
3.
Histochem Cell Biol ; 146(1): 1-12, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27091563

ABSTRACT

A new intermediate type of Henle's loop has been reported that it extends into the inner medulla and turns within the first millimeter beyond the outer medulla. This study aimed to identify the descending thin limb (DTL) of the intermediate loop in the adult C57Bl/6 mouse kidney using aquaporin 1 (AQP1) and urea transporter A2 (UT-A2) antibodies. In the upper part of the inner stripe of the outer medulla (ISOM), AQP1 was expressed strongly in the DTL with type II epithelium of the long loop, but not in type I epithelium of the short loop. The DTL of the intermediate loop exhibited weak AQP1 immunoreactivity. UT-A2 immunoreactivity was not observed in the upper part of any DTL type. AQP1 expression was similar in the upper and middle parts of the ISOM. UT-A2 expression was variable, being expressed strongly in the DTL with type I epithelium of the short loop, but not in type II epithelium of the long loop. In the innermost part of the ISOM, AQP1 was expressed only in type III epithelium of the long loop. UT-A2-positive and UT-A2-negative cells were intermingled in type I epithelium of the intermediate loop, but were not observed in type III epithelium of the long loop. UT-A2-positive DTLs of the intermediate loop extended into the UT-A2/AQP1-negative type I epithelium in the initial part of the inner medulla. These results demonstrate that the DTL of the intermediate loop is composed of type I epithelium and expresses both AQP1 and UT-A2. The functional role of the DTL of the intermediate loop may be distinct from the short or long loops.


Subject(s)
Aquaporin 1/metabolism , Kidney Medulla/metabolism , Kidney/metabolism , Loop of Henle/metabolism , Membrane Transport Proteins/metabolism , Animals , Aquaporin 1/analysis , Kidney/chemistry , Kidney Medulla/chemistry , Loop of Henle/chemistry , Male , Membrane Transport Proteins/analysis , Mice , Mice, Inbred C57BL , Urea Transporters
4.
Urologe A ; 47(5): 538, 540-44, 2008 May.
Article in German | MEDLINE | ID: mdl-18392604

ABSTRACT

Calcium oxalate (CaOx) urolithiasis is a very common disorder. Surprisingly, the pathogenetic mechanisms leading to CaOx stone formation have been largely unknown so far. The long-accepted simple explanation by an exceeding of the solubility product of lithogenic substances in the urine cannot sufficiently describe the complex processes. Deviating from the hypothesis that proclaims that the initial crystal deposition takes place in the lumens of renal tubules, new insights suggest a primary plaque formation in the interstitial space of the renal papilla. Initially, calcium phosphate (CaPh) crystals and organic matrix are deposited along the basement membranes of the thin loops of Henle and extend further in the interstitial space to the urothelium, constituting the so-called Randall's plaques that can be regularly found during endoscopy of CaOx-stone-forming patients. These CaPh crystals seem to be the origin for the development of future CaOx stones, which form by the attachment of further matrix molecules and CaOx from the urine to the plaque. The driving forces, the exact pathogenetic mechanisms, and the involved matrix molecules remain largely unknown. Possibly, completely different pathomechanisms lead to the common clinical diagnosis of"CaOx stone former."


Subject(s)
Calcium Oxalate/analysis , Kidney Calculi/chemistry , Kidney Calculi/pathology , Acid-Base Equilibrium/physiology , Apatites/analysis , Calcium Phosphates/analysis , Crystallization , Extracellular Matrix/chemistry , Extracellular Matrix/pathology , Extracellular Space/chemistry , Humans , Kidney Medulla/chemistry , Kidney Medulla/pathology , Kidney Tubules/chemistry , Kidney Tubules/pathology , Loop of Henle/chemistry , Loop of Henle/pathology , Urothelium/chemistry , Urothelium/pathology
5.
J Ren Nutr ; 16(3): 199-203, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16825019

ABSTRACT

Indoxyl sulfate shows nephrotoxicity and is a stimulating factor for progression of chronic renal failure (CRF). Indoxyl sulfate is taken up by renal proximal tubular cells through organic anion transporters 1 and 3 (OAT1/3), and is accumulated in the renal proximal tubular cells of uremic rats. To determine whether indoxyl sulfate is accumulated in human OAT1/3 (hOAT1/3)-positive renal proximal tubular cells, localization of indoxyl sulfate and hOAT1/3 in the kidneys of CRF patients was determined by immunohistochemistry. Kidney samples were obtained by autopsy from 9 CRF patients (mean serum creatinine 4.7 mg/dL, ranging from 2.0 to 14.5 mg/dL) and 9 patients with non-kidney disease (mean serum creatinine 0.6 mg/dL, ranging from 0.4 to 0.9 mg/dL). Immunohistochemistry was performed using antibodies against indoxyl sulfate, hOAT1, and hOAT3. Indoxyl sulfate was localized in the hOAT1- and hOAT3-positive renal tubular cells in the kidneys of CRF patients. The indoxyl sulfate-positive area in the kidneys was markedly increased in the kidneys of CRF patients compared with patients with non-kidney disease. The indoxyl sulfate-positive area was positively correlated with serum creatinine. In conclusion, in CRF patients, indoxyl sulfate is accumulated in the tubular cells with hOAT1 and/or hOAT3 localized at the basolateral membrane. The extent of indoxyl sulfate accumulation in the kidneys is more prominent in those patients with more severe CRF.


Subject(s)
Indican/metabolism , Kidney Failure, Chronic/metabolism , Kidney Tubules/metabolism , Organic Anion Transport Protein 1/analysis , Organic Anion Transporters, Sodium-Independent/analysis , Aged , Aged, 80 and over , Female , Humans , Immunohistochemistry , Indican/analysis , Kidney/chemistry , Kidney Tubules/chemistry , Kidney Tubules, Distal/chemistry , Kidney Tubules, Proximal/chemistry , Loop of Henle/chemistry , Male , Middle Aged
6.
J Vet Med Sci ; 68(5): 453-63, 2006 May.
Article in English | MEDLINE | ID: mdl-16757888

ABSTRACT

Claudin-16 is one of the tight junction protein claudins and has been shown to contribute to reabsorption of divalent cations in the human kidney. In cattle, total deficiency of claudin-16 causes severe renal tubular dysplasia without aberrant metabolic changes of divalent cations, suggesting that bovine claudin-16 has some roles in renal tubule formation and paracellular transport that are somewhat different from those expected from the pathology of human disease. As the first step to clarify these roles, we examined the expression and distribution of claudin-16 and several other major claudin subtypes, claudins 1-4 and 10, in bovine renal tubular segments by immunofluorescence microscopy. Claudin-16 was exclusively distributed to the tight junction in the tubular segment positive for Tamm-Horsfall glycoprotein, the thick ascending limb (TAL) of Henle's loop, and was found colocalized with claudins 3, 4, and 10. This study also demonstrates that bovine kidneys possess segment-specific expression patterns for claudins 2-4 and 10 that are different from those reported for mice. Particularly, distribution of claudin-4 in the TAL and distal convoluted tubules was characteristic of bovine nephrons as were differences in the expression patterns of claudins 2 and 3. These findings demonstrate that the total lack of claudin-16 in the TAL segment is the sole cause of renal tubular dysplasia in cattle and suggest that the tight junctions in distinct tubular segments including the TAL have barrier functions in paracellular permeability that are different among animal species.


Subject(s)
Cattle/metabolism , Loop of Henle/metabolism , Membrane Proteins/metabolism , Tight Junctions/metabolism , Amino Acid Sequence , Animals , Base Sequence , Claudin-1 , Claudin-3 , Claudin-4 , Claudins , Gene Expression , Loop of Henle/chemistry , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Microscopy, Fluorescence/veterinary , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis/veterinary , Species Specificity , Tight Junctions/chemistry , Tissue Distribution
7.
Kidney Int ; 69(12): 2227-35, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16710357

ABSTRACT

We have biopsied the papillae of patients who have cystine stones asking if this stone type is associated with specific tissue changes. We studied seven cystine stone formers (SF) treated with percutaneous nephrolithotomy using digital video imaging of renal papillae for mapping and obtained papillary biopsies. Biopsies were analyzed by routine light and electron microscopy, infrared spectroscopy, electron diffraction, and micro-CT. Many ducts of Bellini (BD) had an enlarged ostium, and all such were plugged with cystine crystals, and had injured or absent lining cells with a surrounding interstitium that was inflamed to fibrotic. Crystal plugs often projected into the urinary space. Many inner medullary collecting ducts (IMCD) were dilated with or without crystal plugging. Apatite crystals were identified in the lumens of loops of Henle and IMCD. Abundance of interstitial Randall's plaque was equivalent in amount to that of non-SF. In the cortex, glomerular obsolescence and interstitial fibrosis exceeded normal. Cystine crystallizes in BD with the probable result of cell injury, interstitial reaction, nephron obstruction, and with the potential of inducing cortical change and loss of IMCD tubular fluid pH regulation, resulting in apatite formation. The pattern of IMCD dilation, and loss of medullary structures is most compatible with such obstruction, either from BD lumen plugs or urinary tract obstruction from stones themselves.


Subject(s)
Cystine/analysis , Kidney Calculi/chemistry , Kidney Calculi/pathology , Kidney Tubules, Collecting/pathology , Loop of Henle/pathology , Adolescent , Adult , Apatites/analysis , Biopsy , Crystallization , Cystinuria/pathology , Female , Humans , Kidney Tubules, Collecting/chemistry , Loop of Henle/chemistry , Male , Microscopy, Electron , Microscopy, Electron, Transmission , Middle Aged , Nephrostomy, Percutaneous , Spectroscopy, Near-Infrared
8.
Am J Physiol Renal Physiol ; 291(2): F350-7, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16597609

ABSTRACT

Hypertension is associated with increased levels of oxidative stress and medullary renal injury. Previous studies have shown that elevations in renal perfusion pressure increase Na(+) delivery to the medullary thick ascending limb (mTAL), and enhancement of NaCl transport in the outer medulla has been reported in many experimental forms of hypertension. This study examined the effects of increased Na(+) and fluid delivery in mTAL perfused in vitro on the generation of superoxide. Osmolality was maintained constant between low- and high-Na(+) perfusates by adjusting with choline Cl(-). Real-time fluorescent microscopic techniques were used to determine the generation of superoxide and nitric oxide in individual mTAL cells using dihydroethidium and DAF-FM dyes, respectively. Increasing the Na(+) concentration of the perfusate from 60 to 149 mM or luminal flow rate from 5 to 20 nl/min (with fixed Na(+) concentration of 60 mM) significantly increased superoxide generation and decreased nitric oxide in mTAL. These effects were inhibited when active transport of Na(+) was inhibited by ouabain. We conclude that increases in luminal Na(+) concentration and/or flow rate can increase the generation of superoxide in mTAL and reduce nitric oxide bioavailability. This may lead to reduction in medullary blood flow and promote hypoxia and tubular necrosis within the renal medulla during in hypertension.


Subject(s)
Loop of Henle/metabolism , Nitric Oxide/metabolism , Sodium/pharmacology , Superoxides/metabolism , Animals , Biological Availability , Biological Transport/drug effects , Biological Transport/physiology , Enzyme Inhibitors/pharmacology , Hydrogen Peroxide/pharmacology , Hypertension/physiopathology , Kidney Medulla/blood supply , Kidney Medulla/physiopathology , Loop of Henle/chemistry , Male , Nitric Oxide/analysis , Ouabain/pharmacology , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Regional Blood Flow/physiology , Sodium/analysis , Sodium/pharmacokinetics , Sodium Chloride/metabolism , Superoxides/analysis
9.
J Gen Physiol ; 127(2): 205-15, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16446508

ABSTRACT

This study investigates the presence and properties of Na+-activated K+ (K(Na)) channels in epithelial renal cells. Using real-time PCR on mouse microdissected nephron segments, we show that Slo2.2 mRNA, which encodes for the K(Na) channels of excitable cells, is expressed in the medullary and cortical thick ascending limbs of Henle's loop, but not in the other parts of the nephron. Patch-clamp analysis revealed the presence of a high conductance K+ channel in the basolateral membrane of both the medullary and cortical thick ascending limbs. This channel was highly K+ selective (P(K)/P(Na) approximately 20), its conductance ranged from 140 to 180 pS with subconductance levels, and its current/voltage relationship displayed intermediate, Na+-dependent, inward rectification. Internal Na+ and Cl- activated the channel with 50% effective concentrations (EC50) and Hill coefficients (nH) of 30 +/- 1 mM and 3.9 +/- 0.5 for internal Na+, and 35 +/- 10 mM and 1.3 +/- 0.25 for internal Cl-. Channel activity was unaltered by internal ATP (2 mM) and by internal pH, but clearly decreased when internal free Ca2+ concentration increased. This is the first demonstration of the presence in the epithelial cell membrane of a functional, Na+-activated, large-conductance K+ channel that closely resembles native K(Na) channels of excitable cells. This Slo2.2 type, Na+- and Cl--activated K+ channel is primarily located in the thick ascending limb, a major renal site of transcellular NaCl reabsorption.


Subject(s)
Loop of Henle/chemistry , Potassium Channels/analysis , Potassium Channels/physiology , Urothelium/chemistry , Adenosine Triphosphate/physiology , Animals , Cell Membrane/chemistry , Cell Membrane/physiology , Chlorides/physiology , Epithelial Cells/chemistry , Epithelial Cells/physiology , Hydrogen-Ion Concentration , Large-Conductance Calcium-Activated Potassium Channels/analysis , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Large-Conductance Calcium-Activated Potassium Channels/physiology , Loop of Henle/cytology , Loop of Henle/physiology , Male , Mice , Mice, Inbred Strains , Nerve Tissue Proteins , Patch-Clamp Techniques , Potassium Channels/genetics , Potassium Channels, Sodium-Activated , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sodium/physiology , Urothelium/physiology
10.
Am J Physiol Renal Physiol ; 290(6): F1443-52, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16434569

ABSTRACT

The primary mechanism by which the kidneys mediate net acid excretion is through ammonia metabolism. In the current study, we examined whether chronic metabolic acidosis, which increases ammonia metabolism, alters the cell-specific and/or the subcellular expression of the ammonia transporter family member, Rhcg, in the outer medullary collecting duct in the inner stripe (OMCDi). Chronic metabolic acidosis was induced in normal SD rats by HCl ingestion for 7 days; controls were pair-fed. The subcellular distribution of Rhcg was determined using immunogold electron microscopy and morphometric analyses. In intercalated cells, acidosis increased total Rhcg, apical plasma membrane Rhcg, and the proportion of total cellular Rhcg in the apical plasma membrane. Intracellular Rhcg decreased significantly, and basolateral Rhcg was unchanged. Because apical plasma membrane length increased in parallel with apical Rhcg immunolabel, apical plasma membrane Rhcg density was unchanged. In principal cells, acidosis increased total Rhcg, apical plasma membrane Rhcg, and the proportion of total cellular Rhcg in the apical plasma membrane while decreasing the intracellular proportion. In contrast to the intercalated cell, chronic metabolic acidosis did not significantly alter apical boundary length; accordingly, apical plasma membrane Rhcg density increased. In addition, basolateral Rhcg immunolabel increased in response to chronic metabolic acidosis. These results indicate that in the rat OMCDi 1) chronic metabolic acidosis increases apical plasma membrane Rhcg in both the intercalated cell and principal cell where it may contribute to enhanced apical ammonia secretion; 2) increased apical plasma membrane Rhcg results from both increased total protein and changes in the subcellular distribution of Rhcg; 3) the mechanism of Rhcg subcellular redistribution differs in intercalated and principal cells; and 4) Rhcg may contribute to regulated basolateral ammonia transport in the principal cell.


Subject(s)
Acidosis/metabolism , Cation Transport Proteins/analysis , Kidney/ultrastructure , Membrane Glycoproteins/analysis , Subcellular Fractions/chemistry , Acidosis/chemically induced , Ammonia/metabolism , Animals , Cell Membrane/chemistry , Chronic Disease , Cytoplasm/chemistry , Hydrochloric Acid , Immunohistochemistry , Kidney Medulla/chemistry , Kidney Medulla/ultrastructure , Loop of Henle/chemistry , Loop of Henle/ultrastructure , Male , Microscopy, Electron , Rats , Rats, Sprague-Dawley
11.
Am J Physiol Renal Physiol ; 290(4): F937-45, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16263805

ABSTRACT

Previous studies have indicated that a major fraction of the filtered Cl(-) is reabsorbed via apical membrane Cl(-)/base exchange in the proximal tubule. Recent studies in Slc26a6 null mice have suggested that this transporter mediates only a portion of proximal tubule Cl(-)/base exchange, raising the possibility that one or more unidentified apical membrane transporters may additionally contribute. Recent studies have identified Slc26a7 as another Cl(-)/base exchanger expressed in the kidney. We therefore generated Slc26a7-specific polyclonal and monoclonal antibodies to examine cellular and subcellular sites of expression in mouse kidney. The specificity of each antibody was verified by immunoblotting and immunofluorescence of COS-7 cells transiently transfected with mouse Slc26a7. Immunofluorescence microscopy of mouse kidney detected the expression of Slc26a7 subapically in proximal tubule cells, and on the basolateral surface of thick ascending limb cells. Similar staining patterns were demonstrated with two antibodies shown to react with different epitopes on Slc26a7. Immunolocalization of Slc26a7 to proximal tubule and thick ascending limb was also observed in rat kidney. We conclude that Slc26a7 is expressed in the proximal tubule and thick ascending limb of the loop of Henle, and it may therefore contribute to anion transport in these nephron segments.


Subject(s)
Chloride-Bicarbonate Antiporters/biosynthesis , Ion Transport/physiology , Kidney Tubules, Proximal/physiology , Loop of Henle/physiology , Animals , Anions , Chloride-Bicarbonate Antiporters/analysis , Fluorescent Antibody Technique , Kidney Tubules, Proximal/chemistry , Loop of Henle/chemistry , Mice , Sulfate Transporters
12.
Am J Physiol Renal Physiol ; 287(4): F767-74, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15187004

ABSTRACT

Functional reconstruction of inner medullary thin limbs of Henle and collecting ducts (CDs) has enabled us to characterize distinctive three-dimensional vertical and lateral relationships between these segments. We previously reported that inner medullary descending thin limbs (DTLs) that form a bend at a distance greater than approximately 1 mm below the inner medullary base express detectable aquaporin (AQP) 1 only along the initial 40% of the segment before the bend, whereas ClC-K1 is expressed continuously along all ascending thin limbs (ATLs), beginning with the prebend segment. We have now reconstructed individual CDs that are grouped together in single clusters at the base of the inner medulla; CDs belonging to each separate cluster coalesce into a single CD in the deep papilla. DTLs are positioned predominantly at the periphery of each individual CD cluster at all levels of the inner medulla and are absent from within the cluster. In contrast, ATLs are distributed near uniformly among the CDs and DTLs at all levels of the inner medulla. A second population of inner medullary DTLs averages approximately 700 microm in length from base to bend and, as previously reported, expresses no detectable AQP1 and expresses ClC-K1 continuously beginning with the prebend segment. ATLs located within the interior of the CD clusters arise predominantly from these short AQP1-null inner medullary DTLs, suggesting there may be functional interdependence between IMCD1 segments and short-length inner medullary thin limbs exhibiting minimal water permeability along their descending segments. AQP1-expressing DTLs and CDs are apparently separated into two structurally distinct lateral compartments. A similar lateral compartmentation between the ATLs and CDs is not apparent. This architectural arrangement indicates that fluid and solutes may be preferentially transported transversely between multiple inner medullary compartments.


Subject(s)
Imaging, Three-Dimensional , Kidney Tubules, Collecting/anatomy & histology , Loop of Henle/anatomy & histology , Animals , Antibodies , Aquaporin 1 , Aquaporins/analysis , Aquaporins/immunology , Kidney Concentrating Ability/physiology , Kidney Medulla/anatomy & histology , Kidney Medulla/chemistry , Kidney Medulla/physiology , Kidney Tubules, Collecting/chemistry , Kidney Tubules, Collecting/physiology , Loop of Henle/chemistry , Loop of Henle/physiology , Male , Rats , Rats, Wistar
13.
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
14.
Am J Physiol Renal Physiol ; 284(1): F133-43, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12388392

ABSTRACT

Furosemide and hydrochlorothiazide (HCTZ) exert their diuretic actions by binding to apical Na(+) transporters, viz., the Na(+)-K(+)-2Cl(-) cotransporter in the thick ascending limb and the Na(+)-Cl(-) cotransporter in the distal convoluted tubule, respectively. We carried out semiquantitative immunoblotting and immunohistochemistry of rat kidneys to investigate whether chronic administration of furosemide or HCTZ is associated with compensatory changes in the abundance of Na(+) transporters downstream from the primary site of action. Osmotic minipumps were implanted into Sprague-Dawley rats to deliver furosemide (12 mg/day) or HCTZ (3.75 mg/day) for 7 days. To prevent volume depletion, all animals were offered tap water and a solution containing 0.8% NaCl and 0.1% KCl as drinking fluid. The diuretic/natriuretic response was quantified in response to both agents by using quantitative urine collections. Semiquantitative immunoblotting revealed that the abundances of thick ascending limb Na(+)-K(+)-2Cl(-) cotransporter and all three subunits of the epithelial Na(+) channel (ENaC) were increased by furosemide infusion. HCTZ infusion increased the abundances of thiazide-sensitive Na(+)-Cl(-) cotransporter and beta-ENaC in the cortex and beta- and gamma-ENaC in the outer medulla. Consistent with these results, beta-ENaC immunohistochemistry showed a remarkable increase in immunoreactivity in the principal cells of collecting ducts with either diuretic treatment. These increases in the abundance of Na(+) transporters in response to chronic diuretic treatment may account for the generation of diuretic tolerance associated with long-term diuretic use.


Subject(s)
Hydrochlorothiazide/pharmacology , Sodium Channels/metabolism , Sodium Chloride Symporter Inhibitors/pharmacology , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Blotting, Western , Diuretics/pharmacology , Epithelial Sodium Channels , Furosemide/pharmacology , Immunohistochemistry , Kidney Tubules, Collecting/chemistry , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/metabolism , Loop of Henle/chemistry , Loop of Henle/drug effects , Loop of Henle/metabolism , Male , Rats , Rats, Sprague-Dawley , Sodium Channels/analysis , Sodium-Potassium-Chloride Symporters/analysis , Up-Regulation/drug effects
15.
Am J Physiol Renal Physiol ; 282(3): F393-407, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11832419

ABSTRACT

Renal Na(+)-K(+)-ATPase is associated with the gamma-subunit (FXYD2), a single-span membrane protein that modifies ATPase properties. There are two splice variants with different amino termini, gamma(a) and gamma(b). Both were found in the inner stripe of the outer medulla in the thick ascending limb. Coimmunoprecipitation with each other and the alpha-subunit indicated that they were associated in macromolecular complexes. Association was controlled by ligands that affect Na(+)-K(+)-ATPase conformation. In the cortex, the proportion of the gamma(b)-subunit was markedly lower, and the gamma(a)-subunit predominated in isolated proximal tubule cells. By immunofluorescence, the gamma(b)-subunit was detected in the superficial cortex only in the distal convoluted tubule and connecting tubule, which are rich in Na(+)-K(+)-ATPase but comprise a minor fraction of cortex mass. In the outer stripe of the outer medulla and for a short distance in the deep cortex, the thick ascending limb predominantly expressed the gamma(b)-subunit. Because different mechanisms maintain and regulate Na(+) homeostasis in different nephron segments, the splice forms of the gamma-subunit may have evolved to control the renal Na(+) pump through pump properties, gene expression, or both.


Subject(s)
Alternative Splicing/physiology , Kidney Medulla/chemistry , Kidney Medulla/physiology , Sodium-Potassium-Exchanging ATPase/analysis , Sodium-Potassium-Exchanging ATPase/genetics , Amino Acid Sequence , Animals , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic , Kidney Tubules, Proximal/chemistry , Kidney Tubules, Proximal/physiology , Loop of Henle/chemistry , Loop of Henle/physiology , Microscopy, Confocal , Nephrons/chemistry , Nephrons/physiology , Organ Specificity , Peptide Fragments/immunology , Precipitin Tests , Rats , Sodium-Potassium-Exchanging ATPase/immunology
16.
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
17.
Am J Physiol Renal Physiol ; 282(3): F553-7, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11832438

ABSTRACT

We utilized immunofluorescent immunolabeling of renal tissue sections to identify and count tubules at specified depths of the rat renal inner medulla. We used primary antibodies to aquaporin-1 (AQP1; labeling thin descending limbs), aquaporin-2 (AQP2; labeling inner medullary collecting ducts), the rat kidney-specific chloride channel (ClC-K1; labeling thin ascending limbs), and von Willebrand factor (labeling descending vasa recta). Secondary antibodies conjugated to different fluorophores were used, giving up to a three-color display. Labeled structures were then identified and counted. At each level sampled in the inner medulla, many more thin limbs were labeled by ClC-K1 than AQP1. In addition, thin limbs were found to label with antibodies to ClC-K1 on both sides of their hairpin turns. We conclude that the descending thin limbs shift from expressing AQP1 to expressing ClC-K1 some distance before the point where they turn and begin to ascend. Mathematical models can use our quantitative data to explore implications for the urine-concentrating mechanism.


Subject(s)
Aquaporins/analysis , Chloride Channels/analysis , Kidney Medulla/chemistry , Animals , Antibodies , Aquaporin 1 , Aquaporin 2 , Aquaporin 6 , Aquaporins/immunology , Chickens , Chloride Channels/immunology , Fluorescent Antibody Technique , Kidney Concentrating Ability/physiology , Kidney Medulla/cytology , Loop of Henle/chemistry , Loop of Henle/cytology , Male , Rabbits , Rats , Rats, Brattleboro , von Willebrand Factor/analysis , von Willebrand Factor/immunology
18.
Am J Physiol Renal Physiol ; 282(1): F34-44, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11739110

ABSTRACT

Chronic hypercalcemia (HC) is accompanied by urinary concentration defects, and functional studies indicate defects in the thick ascending limb (TAL). We hypothesize that dysregulation of renal sodium transporters may play an important role in this. Vitamin D-induced HC in rats resulted in polyuria, natriuresis, and phosphaturia. Immunoblotting revealed a marked reduction in the abundance of rat type 1 bumetanide-sensitive Na-K-2Cl cotransporter (BSC-1) in inner stripe of the outer medullary (ISOM; 36 +/- 5%) and whole kidney (51 +/- 11%) in HC. Consistent with this finding, immunocytochemistry and immunoelectron microscopy demonstrated reduced BSC-1 labeling of the apical plasma membrane. Immunoblotting and immunohistochemical labeling of the K channel Kir 1.1 (ROMK) was also reduced in HC. In contrast, there were no reductions in the expression of Na/H exchanger (NHE)3 and Na,K-ATPase in ISOM. The abundance of the proximal tubule type II Na-P(i) cotransporter (NaPi-2) (but not Na,K-ATPase and NHE3) was significantly reduced (25 +/- 4%), consistent with a dramatic increase in urinary phosphate excretion. In conclusion, 1) the reduced abundance of BSC-1 and ROMK in TAL is likely to play a major role in the urinary concentration defects associated with HC and 2) the reduced abundance of NaPi-2 is likely to play a role in the increased urinary phosphate excretion.


Subject(s)
Hypercalcemia/physiopathology , Kidney Concentrating Ability/physiology , Kidney Medulla/physiology , Natriuresis/physiology , Potassium Channels, Inwardly Rectifying , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Hypercalcemia/chemically induced , Hypercalcemia/metabolism , Immunoblotting , Immunohistochemistry , Kidney Medulla/chemistry , Kidney Medulla/ultrastructure , Kidney Tubules, Proximal/chemistry , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/ultrastructure , Loop of Henle/chemistry , Loop of Henle/metabolism , Loop of Henle/ultrastructure , Male , Microscopy, Immunoelectron , Phosphates/metabolism , Polyuria/chemically induced , Polyuria/metabolism , Polyuria/physiopathology , Potassium Channels/analysis , Potassium Channels/biosynthesis , Rats , Rats, Wistar , Sodium/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/analysis , Sodium-Potassium-Chloride Symporters/analysis , Sodium-Potassium-Chloride Symporters/biosynthesis , Sodium-Potassium-Exchanging ATPase/analysis , Solute Carrier Family 12, Member 1 , Vitamin D , Water-Electrolyte Balance/physiology
19.
J Acquir Immune Defic Syndr ; 28(1): 9-13, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11579271

ABSTRACT

OBJECTIVE: To determine the probable site of the nephron and the plasma indinavir (IDV) concentration at which intrarenal IDV crystallization occurs. DESIGN: We performed in vitro crystallization experiments in IDV solutions simulating conditions found in the nephron. METHODS: To determine intrarenal IDV concentrations at which conditions in the nephron allow crystallization, several concentrations of IDV basic solutions (0-800 mM) were titrated from pH 4.0 to higher pH values until crystals formed within 1 minute. Based on the combination of pH and ionic strength at which crystals formed, we determined the site of the nephron at which this combination was first attained. Based on the capacity for concentration at that site, we were able to measure the corresponding plasma IDV concentration. RESULTS: Under conditions normally found at the proximal tubule (i.e., pH 6.7 and ionic strength of 200 mM), IDV crystallized at 200 mg/L. Under conditions applying to the loop of Henle, pH 7.4 and ionic strength of 200 mM, IDV crystallized at 125 mg/L, which would correspond to a plasma IDV concentration of 8 mg/L. CONCLUSIONS: IDV crystallization is most likely in the loop of Henle and may already start at plasma IDV concentrations as low as 8 mg/L. Increasing hydration does not reduce the risk of IDV crystallization in the loop of Henle but instead prevents IDV crystallization and aggregation in the lower urinary tract. It remains to be confirmed whether prevention of high IDV plasma concentrations will reduce the risk of IDV crystallization in the loop of Henle.


Subject(s)
HIV Protease Inhibitors/chemistry , Indinavir/chemistry , Loop of Henle/chemistry , Crystallization , Humans , Hydrogen-Ion Concentration , Solubility
20.
Am J Physiol Renal Physiol ; 281(5): F966-74, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11592954

ABSTRACT

The proximal nephron possesses a leaky epithelium with unique paracellular permeability properties that underlie its high rate of passive NaCl and water reabsorption, but the molecular basis is unknown. The claudins are a large family of transmembrane proteins that are part of the tight junction complex and likely form structural components of a paracellular pore. To localize claudin-2 in the mouse kidney, we performed in situ hybridization using an isoform-specific riboprobe and immunohistochemistry using a polyclonal antibody directed against a COOH-terminal peptide. Claudin-2 mRNA and protein were found throughout the proximal tubule and in the contiguous early segment of the thin descending limb of long-looped nephrons. The level of expression demonstrated an axial increase from proximal to distal segments. In confocal images, the subcellular localization of claudin-2 protein coincided with that of the tight junction protein ZO-1. Our findings suggest that claudin-2 is a component of the paracellular pathway of the most proximal segments of the nephron and that it may be responsible for their uniquely leaky permeability properties.


Subject(s)
Kidney Tubules, Proximal/chemistry , Membrane Proteins/analysis , Nephrons/chemistry , Animals , Claudins , Fluorescent Antibody Technique , Glutathione Transferase/genetics , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Loop of Henle/chemistry , Membrane Proteins/genetics , Mice , RNA Probes , RNA, Antisense , RNA, Messenger/analysis , Recombinant Fusion Proteins , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL