Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 334
Filter
Add more filters

Publication year range
1.
Am J Physiol Renal Physiol ; 320(1): F17-F30, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33196322

ABSTRACT

Connexins (Cxs) form gap junctions for intercellular exchange of inorganic ions and messenger molecules. In the kidney, Cxs play essential roles within its compartments, but data on the precise cellular localization and cell type-related function of their isoforms are scarce. We tested whether Cx43 distribution is restricted to vascular and interstitial cells and whether medullary fibroblasts express Cx43 to coordinate profibrotic signaling. Confocal immunofluorescence techniques, ultrastructural labeling, and functional experiments in cell culture were performed. Cx43 was chiefly expressed in the vasculature but was absent from tubular epithelia. All arterial, arteriolar, and lymphatic endothelia showed continuous Cx43 signal along their borders. In the inner medulla, only the interstitium showed Cx43 signals, which were assigned to fibroblasts and their processes. Cultured Cx43-expressing medullary fibroblasts served to study the role of gap junctions in a profibrotic context. In a dye spreading assay, Cx43-sensitive diffusion of Lucifer yellow was dependent on gap junctional passage. The addition of transforming growth factor-ß1 (5 ng/mL for 48 h) activated Cx43 biosynthesis and caused Cx43-sensitive transformation of the fibroblasts into a myofibroblast phenotype. This suggested that Cx43 gap junctional channels enable the coordination of profibrotic signaling between cells of the medullary interstitium. In summary, we demonstrate the presence of Cx43-expressing gap junctions within the two major renal compartments, the vasculature and interstitium. Endothelial Cx43 likely provides functions of an earlier-defined "electrical syncytium" within the vascular wall. Additionally, Cx43 facilitates profibrotic signaling between medullary interstitial fibroblasts.


Subject(s)
Cell Differentiation , Connexin 43/metabolism , Endothelial Cells/metabolism , Fibroblasts/metabolism , Kidney Medulla/blood supply , Kidney Medulla/metabolism , Animals , Cell Communication , Cell Line , Endothelial Cells/ultrastructure , Fibroblasts/ultrastructure , Fibrosis , Gap Junctions/metabolism , Gap Junctions/ultrastructure , Humans , Kidney Medulla/ultrastructure , Male , Mice, Inbred C57BL , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure , Phenotype , Rats, Wistar
2.
Am J Physiol Regul Integr Comp Physiol ; 314(4): R563-R573, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351422

ABSTRACT

In general, the mammalian whole body mass-specific metabolic rate correlates positively with maximal urine concentration (Umax) irrespective of whether or not the species have adapted to arid or mesic habitat. Accordingly, we hypothesized that the thick ascending limb (TAL) of a rodent with markedly higher whole body mass-specific metabolism than rat exhibits a substantially higher TAL metabolic rate as estimated by Na+-K+-ATPase activity and Na+-K+-ATPase α1-gene and protein expression. The kangaroo rat inner stripe of the outer medulla exhibits significantly higher mean Na+-K+-ATPase activity (~70%) compared with two rat strains (Sprague-Dawley and Munich-Wistar), extending prior studies showing rat activity exceeds rabbit. Furthermore, higher expression of Na+-K+-ATPase α1-protein (~4- to 6-fold) and mRNA (~13-fold) and higher TAL mitochondrial volume density (~20%) occur in the kangaroo rat compared with both rat strains. Rat TAL Na+-K+-ATPase α1-protein expression is relatively unaffected by body hydration status or, shown previously, by dietary Na+, arguing against confounding effects from two unavoidably dissimilar diets: grain-based diet without water (kangaroo rat) or grain-based diet with water (rat). We conclude that higher TAL Na+-K+-ATPase activity contributes to relationships between whole body mass-specific metabolic rate and high Umax. More vigorous TAL Na+-K+-ATPase activity in kangaroo rat than rat may contribute to its steeper Na+ and urea axial concentration gradients, adding support to a revised model of the urine concentrating mechanism, which hypothesizes a leading role for vigorous active transport of NaCl, rather than countercurrent multiplication, in generating the outer medullary axial osmotic gradient.


Subject(s)
Body Weight , Energy Metabolism , Kidney Concentrating Ability , Kidney Medulla/enzymology , Loop of Henle/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium/urine , Animals , Dipodomys , Gene Expression Regulation, Enzymologic , Kidney Medulla/ultrastructure , Loop of Henle/ultrastructure , Mitochondria/enzymology , Osmoregulation , Rats, Sprague-Dawley , Rats, Wistar , Renal Elimination , Renal Reabsorption , Species Specificity
3.
Am J Physiol Renal Physiol ; 313(4): F1026-F1037, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28701310

ABSTRACT

The renal collecting duct (CD) contains two major cell types, intercalated (ICs) and principal cells (PCs). A previous report showed that deletion of ß1-integrin in the entire renal CD causes defective CD morphogenesis resulting in kidney dysfunction. However, subsequent deletion of ß1-integrin specifically in ICs and PCs, respectively, did not cause any morphological defects in the CDs. The discrepancy between these studies prompts us to reinvestigate the role of ß1-integrin in CD cells, specifically in the PCs. We conditionally deleted ß1-integrin in mouse CD PCs using a specific aquaporin-2 (AQP2) promoter Cre-LoxP system. The resulting mutant mice, ß-1f/fAQP2-Cre+, had lower body weight, failed to thrive, and died around 8-12 wk. Their CD tubules were dilated, and some of them contained cellular debris. Increased apoptosis and proliferation of PCs were observed in the dilated CDs. Trichrome staining and electron microscopy revealed the presence of peritubular and interstitial fibrosis that is associated with increased production of extracellular matrix proteins including collagen type IV and fibronectin, as detected by immunoblotting. Further analysis revealed a significantly increased expression of transforming growth factor-ß (TGF-ß)-induced protein, fibronectin, and TGF-ß receptor-1 mRNAs and concomitantly increased phosphorylation of SMAD-2 that indicates the activation of the TGF-ß signaling pathway. Therefore, our data reveal that normal expression of ß1-integrin in PCs is a critical determinant of CD structural and functional integrity and further support the previously reported critical role of ß1-integrin in the development and/or maintenance of the CD structure and function.


Subject(s)
Extracellular Matrix/metabolism , Gene Deletion , Integrin beta1/metabolism , Kidney Medulla/metabolism , Kidney Tubules, Collecting/metabolism , Polyuria/metabolism , Renal Insufficiency/metabolism , Age Factors , Animals , Apoptosis , Aquaporin 2/genetics , Cell Proliferation , Extracellular Matrix/ultrastructure , Failure to Thrive/genetics , Failure to Thrive/metabolism , Failure to Thrive/pathology , Fibrosis , Genetic Predisposition to Disease , Integrases/genetics , Integrin beta1/genetics , Kidney Medulla/ultrastructure , Kidney Tubules, Collecting/ultrastructure , Mice, Knockout , Phenotype , Phosphorylation , Polyuria/genetics , Polyuria/pathology , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Renal Insufficiency/genetics , Renal Insufficiency/pathology , Signal Transduction , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism
4.
Am J Physiol Renal Physiol ; 313(1): F20-F29, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28298361

ABSTRACT

The natriuretic hormone CCK exhibits its gene transcripts in total kidney extracts. To test the possibility of CCK acting as an intrarenal mediator of sodium excretion, we examined mouse kidneys by 1) an in situ hybridization technique for CCK mRNA in animals fed a normal- or a high-sodium diet; 2) immuno-electron microscopy for the CCK peptide, 3) an in situ hybridization method and immunohistochemistry for the CCK-specific receptor CCKAR; 4) confocal image analysis of receptor-mediated Ca2+ responses in isolated renal tubules; and 5) metabolic cage experiments for the measurement of urinary sodium excretion in high-salt-fed mice either treated or untreated with the CCKAR antagonist lorglumide. Results showed the CCK gene to be expressed intensely in the inner medulla and moderately in the inner stripe of the outer medulla, with the expression in the latter being enhanced by high sodium intake. Immunoreactivity for the CCK peptide was localized to the rough endoplasmic reticulum of the medullary interstitial cells in corresponding renal regions, confirming it to be a secretory protein. Gene transcripts, protein products, and the functional activity for CCKAR were consistently localized to the late proximal tubule segments (S2 and S3) in the medullary rays, and the outer stripe of the outer medulla. Lorglumide significantly diminished natriuretic responses of mice to a dietary sodium load without altering the glomerular filtration rate. These findings suggest that the medullary interstitial cells respond to body fluid expansion by CCK release for feedback regulation of the late proximal tubular reabsorption.


Subject(s)
Cholecystokinin/metabolism , Kidney Medulla/metabolism , Kidney Tubules, Proximal/metabolism , Natriuresis , Signal Transduction , Sodium, Dietary/administration & dosage , Water-Electrolyte Balance , Animals , Calcium/metabolism , Cholecystokinin/antagonists & inhibitors , Cholecystokinin/genetics , Feedback, Physiological , Hormone Antagonists/pharmacology , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kidney Medulla/drug effects , Kidney Medulla/ultrastructure , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/ultrastructure , Male , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Immunoelectron , Natriuresis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin A/metabolism , Signal Transduction/drug effects , Time Factors , Water-Electrolyte Balance/drug effects
5.
Am J Physiol Renal Physiol ; 305(5): F745-52, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23825077

ABSTRACT

Every collecting duct (CD) of the rat inner medulla is uniformly surrounded by about four abutting ascending vasa recta (AVR) running parallel to it. One or two ascending thin limbs (ATLs) lie between and parallel to each abutting AVR pair, opposite the CD. These structures form boundaries of axially running interstitial compartments. Viewed in transverse sections, these compartments appear as four interstitial nodal spaces (INSs) positioned symmetrically around each CD. The axially running compartments are segmented by interstitial cells spaced at regular intervals. The pairing of ATLs and CDs bounded by an abundant supply of AVR carrying reabsorbed water, NaCl, and urea make a strong argument that the mixing of NaCl and urea within the INSs and countercurrent flows play a critical role in generating the inner medullary osmotic gradient. The results of this study fully support that hypothesis. We quantified interactions of all structures comprising INSs along the corticopapillary axis for two rodent species, the Munich-Wistar rat and the kangaroo rat. The results showed remarkable similarities in the configurations of INSs, suggesting that the structural arrangement of INSs is a highly conserved architecture that plays a fundamental role in renal function. The number density of INSs along the corticopapillary axis directly correlated with a loop population that declines exponentially with distance below the outer medullary-inner medullary boundary. The axial configurations were consistent with discrete association between near-bend loop segments and INSs and with upper loop segments lying distant from INSs.


Subject(s)
Kidney Medulla/ultrastructure , Kidney Tubules, Collecting/ultrastructure , Animals , Dipodomys , Female , Kidney Medulla/metabolism , Male , Rats , Rats, Wistar
6.
J Electron Microsc (Tokyo) ; 59(6): 481-94, 2010.
Article in English | MEDLINE | ID: mdl-20670932

ABSTRACT

This work defines the ultrastructural responses of immortalized cells from the inner medullary collecting duct cells (IMCD3 cells) to hypertonic challenges. The cultured cells were either acutely exposed to hypertonic medium (550 mOsm/kgH2O) for 24-72 h or gradually adapted to 600 or 900 mOsm/kgH2O media with sodium chloride. After short (24 h) hypertonic challenges, there was an expansion of the Golgi apparatus with distinct expression of the γ subunit of Na,K-ATPase. The frequency of active caspase-3-positive cells was unchanged as was also the measured activity of caspase-3. Immunoelectron microscopy showed that active caspase-3 in the positive cells was localized in cytoplasmic bodies 0.5-1 µm in diameter but not in other structures. Apoptotic bodies with the nuclei were only rarely observed following acute hypertonicity for 24 to 72 h. Following prolonged hypertonic challenges, some cells showed condensation of the chromatin but still few apoptotic bodies. Gradual hypertonicity to 900 mOsm/kgH2O led to a decrease of microvilli, dilated cisternae of the endoplasmic reticulum (ER), increased abundance of free ribosomes and longitudinal mitochondrial cristae. Virus particles were present inside and outside the cells in all experimental conditions and appeared unrelated to the apoptotic process. The results suggest that cultured IMCD3 cells are resistant to short hypertonic challenge or gradual adaptation to moderate hypertonicity and only rarely exhibit more ultrastructural apoptotic changes than control cells. The presence of caspase-3-containing bodies is a novel finding, and we suggest that they arise from the ER and are involved in the apoptotic signaling system.


Subject(s)
Kidney Medulla/ultrastructure , Kidney Tubules, Collecting/ultrastructure , Saline Solution, Hypertonic/pharmacology , Adaptation, Physiological , Animals , Apoptosis , Caspase 3/metabolism , Cell Line , Kidney Medulla/cytology , Kidney Medulla/metabolism , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Mice , Microscopy, Immunoelectron , Osmolar Concentration , Sodium Chloride/pharmacology
7.
J Cell Biol ; 91(3 Pt 1): 803-13, 1981 Dec.
Article in English | MEDLINE | ID: mdl-6276410

ABSTRACT

Na+,K+-ATPase was localized at the ultrastructural level in rat and rabbit kidney medulla. The cytochemical method for the K+-dependent phosphatase component of the enzyme, using p-nitrophenylphosphate (NPP) as substrate, was employed to demonstrate the distribution of Na+, K+-ATPase in tissue-chopped sections from kidneys perfusion-fixed with 1% paraformaldehyde-0.25% glutaraldehyde. In other outer medulla of rat kidney, ascending thick limbs (MATL) were sites of intense K+-dependent NPPase (K+-NPPase) activity, whereas descending thick limbs and collecting tubules were barely reactive. Although descending thin limbs (DTL) of short loop nephrons were unstained, DTL from long loop nephrons in outer medulla were sites of moderate K+-NPPase activity. In rat inner medulla, DTL and ascending thin limbs (ATL) were unreactive for K+-NPPase. In rabbit medulla, only MATL were sites of significant K+-NPPase activity. The specificity of the cytochemical localization of Na+,K+-ATPase at reactive sites in rat and rabbit kidney medulla was demonstrated by K+-dependence of reaction product deposition, localization of reaction product (precipitated phosphate hydrolyzed from NPP) to the cytoplasmic side of basolateral plasma membranes, insensitivity of the reaction to inhibitors of nonspecific alkaline phosphatase, and, in the glycoside-sensitive rabbit kidney, substantial inhibition of staining by ouabain. The observed pattern of distribution of the sodium transport enzyme in kidney medulla is particularly relevant to current models for urine concentration. The presence of substantial Na+,K+-ATPase in MATL is consistent with the putative role of this segment as the driving force for the countercurrent multiplication system in the outer medulla. The absence of significant activity in inner medullary ATL and DTL, however, implies that interstitial solute accumulation in this region probably occurs by passive processes. The localization of significant Na+,K+-ATPase in outer medullary DTL of long loop nephrons in the rat suggests that solute addition in this segment may occur in part by an active salt secretory mechanism that could ultimately contribute to the generation of inner medullary interstitial hypertonicity and urine concentration.


Subject(s)
Kidney Medulla/enzymology , Kidney Tubules/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Histocytochemistry , Kidney Medulla/ultrastructure , Microscopy, Electron/methods , Rabbits , Rats
8.
J Cell Biol ; 88(2): 274-80, 1981 Feb.
Article in English | MEDLINE | ID: mdl-7204493

ABSTRACT

The electrolyte and water content of cellular and interstitial compartments in the renal papilla of the rat was determined by x-ray microanalysis of frozen-hydrated tissue sections. Papillae from rats on ad libitum water were rapidly frozen in a slush of Freon 12, and sectioned in a cryomicrotome at -30 to -40 degrees C. Frozen 0.5-micrometer sections were mounted on carbon-coated nylon film over a Be grid, transferred cold to the scanning microscope, and maintained at -175 degrees C during analysis. The scanning transmission mode was used for imaging. Structural preservation was of good quality and allowed identification of tissue compartments. Tissue mass (solutes + water) was determined by continuum radiation from regions of interest. After drying in the SEM, elemental composition of morphologically defined compartments (solutes) was determined by analysis of specific x-rays, and total dry mass by continuum. Na, K, Cl, and H2O contents in collecting-duct cells (CDC), papillary epithelial cells (PEC), and interstitial cells (IC) and space were measured. Cells had lower water content (mean 58.7%) than interstitium (77.5%). Intracellular K concentrations (millimoles per kilogram wet weight) were unremarkable (79-156 mm/kg wet weight); P was markedly higher in cells than in interstitium. S was the same in all compartments. Intracellular Na levels were extremely high (CDC, 344 +/- 127 SD mm/kg wet weight; PEC, 287 +/- 105; IC, 898 +/- 194). Mean interstitial Na was 590 +/- 119 mm/kg wet weight. CI values paralleled those for Na. If this Na is unbound, then these data suggest that renal papillary interstitial cells adapt to their hyperosmotic environment by a Na-uptake process.


Subject(s)
Electrolytes/analysis , Electron Probe Microanalysis , Kidney Medulla/analysis , Animals , Cytoplasm/analysis , Frozen Sections , Kidney Medulla/ultrastructure , Kidney Tubules, Collecting/analysis , Male , Microscopy, Electron, Scanning , Rats
9.
J Cell Biol ; 86(3): 746-54, 1980 Sep.
Article in English | MEDLINE | ID: mdl-6251096

ABSTRACT

To study the size and structure of the Na,K-pump molecule, the ultrastructure of phospholipid vesicles was examined after incorporation of purified Na,K-ATPase which catalyzes active coupled transport of Na+ and K+ in a ratio close to 3Na/2K. The vesicles were analyzed by thin sectioning and freeze-fracture electron microscopy after reconstitution with different ratios of Na,K-ATPase protein to lipid, and the ultrastructural observations were correlated to the cation transport capacity. The purified Na,K-ATPase reconstituted with phospholipids to form a very uniform population of vesicles. Thin sections of preparations fixed with glutaraldehyde and osmium tetroxide showed vesicles limited by a single membrane which in samples stained with tannic acid appeared triple-layered with a thickness of 70 A. Also, freeze-fracture electron microscopy demonstrated uniform vesicles with diameters in the range of 700-1,100 A and an average value close to 900 A. The vesicle diameter was independent of the amount of protein used for reconstitution. Intramembrane particles appeared only in the vesicle membrane after introduction of Na,K-ATPase and the frequency of intramembrane particles was proportional to the amount of Na,K-ATPase protein used in the reconstitution. The particles were evenly distributed on the inner and the outer leaflet of the vesicle membrane. The diameter of the particles was 90 A and similar to our previous values for the diameter of intramembrane particles in the purified Na,K-ATPase. The capacity for active cation transport in the reconstituted vesicles was proportional to the frequency of intramembrane particles over a range of 0.2-16 particles per vesicle. The data therefore show that active coupled Na,K transport can be carried out by units of Na,K-ATPase which appear as single intramembrane particles with diameters close fo 90 A in the freeze-fracture micrographs.


Subject(s)
Ion Channels/metabolism , Kidney Medulla/enzymology , Membrane Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Biological Transport, Active , Cell-Free System , Freeze Fracturing , Kidney Medulla/ultrastructure , Macromolecular Substances , Swine
10.
J Cell Biol ; 111(2): 379-89, 1990 Aug.
Article in English | MEDLINE | ID: mdl-1696262

ABSTRACT

The water permeability of the kidney collecting duct epithelium is regulated by vasopressin (VP)-induced recycling of water channels between an intracellular vesicular compartment and the plasma membrane of principal cells. To test whether the water channels pass through an acidic endosomal compartment during the endocytic portion of this pathway, we measured ATP-dependent acidification of FITC-dextran-labeled endosomes in isolated microsomal fractions from different regions of Brattleboro rat kidneys. Both VP-deficient controls and rat treated with exogenous VP were examined. ATP-dependent acidification was not detectable in endosomes containing water channels from distal papilla (osmotic water permeability Pf = 0.038 +/- 0.004 cm/s). In contrast, the addition of ATP resulted in a strong acidification of renal cortical endosomes (pHmin = 5.8, initial rate = 0.18-0.25 pH U/s). Acidification of cortical endosomes was reversed with nigericin and strongly inhibited by N-ethyl-maleimide. Passive proton permeability was similar and low in both cortical and papillary endosomes from rats treated or not treated with VP. The fraction of labeled endosomes present in microsomal preparations was determined by fluorescence imaging microscopy of microsomes nonspecifically bound to poly-l-lysine-coated coverslips and was 25% in cortical preparations compared to 14% (+VP) and 9% (-VP) in papillary preparations. The fraction of cortical endosomes was enriched 1.5-fold by immunoabsorption to coverslips coated with mAbs against the bovine vacuolar proton pump. In contrast, the fraction of papillary endosomes was depleted more than twofold by immunoabsorption to identical coverslips. Finally, sections of distal papilla stained with antibodies against the lysosomal glycoprotein LGP120 showed that most of the entrapped FITC-dextran did not colocalize with this lysosomal protein. These results demonstrate that vesicles which internalize water channels in kidney collecting duct principal cells lack functional proton pumps, and do not deliver the bulk of their FITC-dextran content to lysosomes. The data suggest that the principal cell contains a specialized nonacidic apical endocytic compartment which functions primarily to recycle membrane components, including water channels, to the plasma membrane.


Subject(s)
Body Water/metabolism , Endocytosis , Fluorescein-5-isothiocyanate/analogs & derivatives , Kidney Medulla/metabolism , Kidney Tubules/metabolism , Proton-Translocating ATPases/analysis , Vasopressins/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Membrane Permeability , Coated Pits, Cell-Membrane/metabolism , Coated Pits, Cell-Membrane/ultrastructure , Dextrans , Endocytosis/drug effects , Fluoresceins , Fluorescent Dyes , Hydrogen-Ion Concentration , Kidney Medulla/drug effects , Kidney Medulla/ultrastructure , Kidney Tubules/ultrastructure , Kinetics , Microscopy, Fluorescence , Rats , Rats, Brattleboro
11.
Ren Fail ; 31(7): 573-81, 2009.
Article in English | MEDLINE | ID: mdl-19839853

ABSTRACT

Haloperidol, a typical antipsychotic, is the most commonly prescribed medication for the treatment of mental health problems such as agitation and psychosis. We attempted to determine the effects of haloperidol treatment on the kidneys of female rats. In addition, we aimed to estimate the numerical density, total number, and height of renal glomeruli and the volume and volumetric fractions of the cortex, medulla, and whole kidneys, and tried to determine whether there was a change in these stereological parameters depending on haloperidol treatment. Both the qualitative and quantitative histological features of the kidney samples were analyzed with conventional histopathological and modern stereological methods at the light microscopic level. The total number of glomeruli and numerical density of glomerulus in the haloperidol-treated groups was not changed by increasing the dose in comparison to the control group. The mean height of the glomerulus significantly increased, especially in low-dose groups. In the haloperidol-treated groups, the volumetric fractions of the cortex to the whole kidney of the rats were significantly decreased by increasing the dose. The volumetric fractions of the medulla to the whole kidney of the rats were increased significantly in parallel by the given dose. In addition, we present quantitative findings showing that haloperidol is associated with many alterations in rat kidneys. It was shown that haloperidol may lead to undesirable changes in the kidney after chronic treatment with especially high doses.


Subject(s)
Haloperidol/adverse effects , Kidney Diseases/pathology , Kidney/drug effects , Kidney/pathology , Animals , Antipsychotic Agents/adverse effects , Antipsychotic Agents/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Haloperidol/pharmacology , Immunohistochemistry , Injections, Intraperitoneal , Kidney/ultrastructure , Kidney Cortex/drug effects , Kidney Cortex/pathology , Kidney Cortex/ultrastructure , Kidney Diseases/chemically induced , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Kidney Medulla/drug effects , Kidney Medulla/pathology , Kidney Medulla/ultrastructure , Microscopy/methods , Organ Size , Probability , Random Allocation , Rats , Rats, Sprague-Dawley , Reference Values
12.
J Magn Reson ; 303: 121-127, 2019 06.
Article in English | MEDLINE | ID: mdl-31051387

ABSTRACT

To map the hemodynamic responses of kidney microstructures at 7.05 T with improved sensitivity, a Wireless Amplified NMR Detector (WAND) with cylindrical symmetry was fabricated as an endoluminal detector that can convert externally provided wireless signal at 600.71 MHz into amplified MR signals at 300.33 MHz. When this detector was inserted inside colonic lumens to sensitively observe adjacent kidneys, it could clearly identify kidney microstructures in the renal cortex and renal medullary. Owing to the higher achievable spatial resolution, differential hemodynamic responses of kidney microstructures under different breathing conditions could be individually quantified to estimate the underlying correlation between oxygen bearing capability and local levels of oxygen unsaturation. The WAND's ability to map Blood Oxygen Level Dependent (BOLD) signal responses in heterogeneous microstructures will pave way for early-stage diagnosis of kidney diseases, without the use of contrast agents for reduced tissue retention and toxicity.


Subject(s)
Kidney/diagnostic imaging , Magnetic Resonance Imaging/instrumentation , Magnetic Resonance Imaging/methods , Animals , Image Processing, Computer-Assisted , Kidney/ultrastructure , Kidney Cortex/diagnostic imaging , Kidney Cortex/ultrastructure , Kidney Medulla/diagnostic imaging , Kidney Medulla/ultrastructure , Male , Oxygen Consumption , Phantoms, Imaging , Rats , Rats, Sprague-Dawley , Renal Circulation , Wireless Technology
13.
Semin Nephrol ; 28(2): 111-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18359392

ABSTRACT

By using intraoperative papillary biopsy material from kidneys of idiopathic calcium oxalate, intestinal bypass for obesity, brushite, cystine, and distal renal tubular acidosis stone formers during percutaneous nephrolithotomy, we have determined that idiopathic calcium oxalate stone formers appear to be the special case, although the most commonly encountered one, in which stones form external to the kidney and by processes that do not involve the epithelial compartments. It is in this one group of patients that we find not only abundant interstitial plaque, but also strong evidence that the plaque is essential to stone formation. The initial site of plaque formation is always in the papillary tip, and must be in the basement membrane of the thin loop of Henle. With time, plaque spreads throughout the papilla tip to the urothelium, which under conditions we do not understand is denuded and thereby exposes the apatite deposits to the urine. It is on this exposed apatite that a stone forms as an overgrowth, first of amorphous apatite and then layers of calcium oxalate. This process generates an attached stone fixed to the side of a papilla, allowing the ever-changing urine to dictate stone growth and composition.


Subject(s)
Apatites/metabolism , Calcium Oxalate/metabolism , Kidney Calculi/physiopathology , Humans , Kidney Calculi/chemistry , Kidney Calculi/metabolism , Kidney Calculi/pathology , Kidney Medulla/metabolism , Kidney Medulla/pathology , Kidney Medulla/ultrastructure , Microscopy, Electron, Transmission
14.
Nephron Physiol ; 109(1): p1-10, 2008.
Article in English | MEDLINE | ID: mdl-18460876

ABSTRACT

BACKGROUND: The genomic response to adaptation of IMCD3 cells to hypertonicity results in both upregulation and downregulation of a variety of genes. METHOD: The present study was undertaken to assess the metabonomic and proteomic response of IMCD3 cells that have been chronically adapted to hypertonicity (600 and 900 mosm/kg H(2)O) as compared to cells under isotonic conditions. RESULTS: Adaptation of IMCD3 cells to hypertonic conditions resulted in a change of a wide range of organic osmolytes, including sorbitol (+8,291%), betaine (+1,099%), myo-inositol (+669%), taurine (+113%) and glycerophosphorylcholine (+61%). Evaluation of the polyol pathway for sorbitol production revealed a reduction in sorbitol dehydrogenase and an increase in aldose reductase mRNA in adapted cells. Proteome analysis revealed increased expression of six glycolytic proteins, including malic enzyme and pyruvate carboxylase, indicating the activation of the pyruvate shunt and changes in glucose metabolism. This study showed that the observed reduction in cell replication could possibly reflect a redirection of cellular energy from cell growth and replication to maintenance of intracellular ion levels in chronically adapted cells. CONCLUSION: The combined metabonomic and proteomic analysis was shown to be a very helpful tool for the analysis of the effects caused by chronic adaptation to hypertonicity. It made it possible to better evaluate the importance of certain changes that occur in the process of adaptation.


Subject(s)
Energy Metabolism , Enzymes/metabolism , Kidney Medulla/metabolism , Kidney Tubules, Collecting/metabolism , Proteomics , Adaptation, Physiological , Amino Acids/metabolism , Animals , Blotting, Western , Cell Line , Cell Proliferation , Electrophoresis, Gel, Two-Dimensional , Enzymes/genetics , Glucose/metabolism , Kidney Medulla/enzymology , Kidney Medulla/ultrastructure , Kidney Tubules, Collecting/enzymology , Kidney Tubules, Collecting/ultrastructure , Mice , Microscopy, Electron, Transmission , Mitochondria/metabolism , Nuclear Magnetic Resonance, Biomolecular , Osmotic Pressure , Phenotype , Phosphates/metabolism , Polymers/metabolism , Proteomics/methods , Reverse Transcriptase Polymerase Chain Reaction , Saline Solution, Hypertonic
15.
Bull Exp Biol Med ; 146(6): 682-6, 2008 Dec.
Article in English, Russian | MEDLINE | ID: mdl-19513354

ABSTRACT

Administration of desmopressin for 4 days to homozygous Brattleboro rats lacking endogenous vasopressin induced an increase in osmotic concentration and was accompanied by typical changes in the morphofunctional state of interstitial cells of the renal papilla. These changes increase the permeability of extracellular matrix, which attested to the involvement of interstitial cells into hydrosmotic reaction to vasopressin.


Subject(s)
Antidiuretic Agents/pharmacology , Deamino Arginine Vasopressin/pharmacology , Kidney/drug effects , Kidney/ultrastructure , Animals , Kidney Medulla/drug effects , Kidney Medulla/ultrastructure , Microscopy, Electron, Transmission , Rats , Rats, Brattleboro , Vasopressins/metabolism
16.
J Clin Invest ; 103(4): 491-6, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10021457

ABSTRACT

It has been controversial whether high water permeability in the thin descending limb of Henle (TDLH) is required for formation of a concentrated urine by the kidney. Freeze-fracture electron microscopy (FFEM) of rat TDLH has shown an exceptionally high density of intramembrane particles (IMPs), which were proposed to consist of tetramers of aquaporin-1 (AQP1) water channels. In this study, transepithelial osmotic water permeability (Pf) was measured in isolated perfused segments (0.5-1 mm) of TDLH in wild-type (+/+), AQP1 heterozygous (+/-), and AQP1 null (-/-) mice. Pf was measured at 37 degrees C using a 100 mM bath-to-lumen osmotic gradient of raffinose, and fluorescein isothiocyanate (FITC)-dextran as the luminal volume marker. Pf was (in cm/s): 0.26 +/- 0.02 ([+/+]; SE, n = 9 tubules), 0.21 +/- 0.01 ([+/-]; n = 12), and 0.031 +/- 0.007 ([-/-]; n = 6) (P < 0.02, [+/+] vs. [+/-]; P < 0.0001, [+/+] vs. [-/-]). FFEM of kidney medulla showed remarkably fewer IMPs in TDLH from (-/-) vs. (+/+) and (+/-) mice. IMP densities were (in microm-2, SD, 5-12 micrographs): 5,880 +/- 238 (+/+); 5,780 +/- 450 (+/-); and 877 +/- 420 (-/-). IMP size distribution analysis revealed mean IMP diameters of 8.4 nm ([+/+] and [+/-]) and 5.2 nm ([-/-]). These results demonstrate that AQP1 is the principal water channel in TDLH and support the view that osmotic equilibration along TDLH by water transport plays a key role in the renal countercurrent concentrating mechanism. The similar Pf and AQP1 expression in TDLH of (+/+) and (+/-) mice was an unexpected finding that probably accounts for the unimpaired urinary concentrating ability in (+/-) mice.


Subject(s)
Aquaporins/physiology , Loop of Henle/physiology , Loop of Henle/ultrastructure , Animals , Aquaporin 1 , Aquaporins/genetics , Female , Kidney Cortex/ultrastructure , Kidney Medulla/ultrastructure , Male , Mice , Mice, Knockout , Osmolar Concentration , Permeability , Rats , Water/metabolism
17.
J Clin Invest ; 92(5): 2448-57, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8227360

ABSTRACT

Low protein diets reverse the urea concentration gradient in the renal inner medulla. To investigate the mechanism(s) for this change, we studied urea transport and cell ultrastructure in initial and terminal inner medullary collecting ducts (IMCD) from rats fed 18% protein or an isocaloric, 8% protein diet for 4 wk. Serum urea, aldosterone, and albumin were significantly lower in rats fed 8% protein, but total protein and potassium were unchanged. Vasopressin stimulated passive urea permeability (Purea) threefold (P < 0.05) in initial IMCDs from rats fed 8% protein, but not from rats fed 18% protein. Luminal phloretin reversibly inhibited vasopressin-stimulated Purea. However, in terminal IMCDs from rats fed either diet, vasopressin stimulated Purea. Net transepithelial urea flux (measured with identical perfusate and bath solutions) was found only in initial IMCDs from rats fed 8% protein. Reducing the temperature reversibly inhibited it, but phloretin did not. Electron microscopy of initial IMCD principal cells from rats fed 8% protein showed expanded Golgi bodies and prominent autophagic vacuoles, and morphometric analysis demonstrated a marked increase in the surface density and boundary length of the basolateral plasma membrane. These ultrastructural changes were not observed in the terminal IMCD. Thus, 8% dietary protein causes two new urea transport processes to appear in initial but not terminal IMCDs. This is the first demonstration that "active" urea transport can be induced in a mammalian collecting duct segment.


Subject(s)
Kidney Medulla/ultrastructure , Kidney Tubules, Collecting/ultrastructure , Protein Deficiency/metabolism , Urea/metabolism , Animals , Biological Transport , Dietary Proteins , Golgi Apparatus/ultrastructure , In Vitro Techniques , Male , Perfusion , Permeability/drug effects , Phloretin/pharmacology , Rats , Rats, Sprague-Dawley , Specific Pathogen-Free Organisms , Vacuoles/ultrastructure , Vasopressins/pharmacology
18.
J Clin Invest ; 82(1): 26-36, 1988 Jul.
Article in English | MEDLINE | ID: mdl-3260605

ABSTRACT

Hepatocytes are considered to be the predominant source of alpha 1-antitrypsin (AAT), the major antiprotease in human plasma. The development of emphysema in the hereditary PiZ AAT deficiency state suggests that inhibition of leukocyte elastase in the lung is a major function of this protein. In addition, patients with AAT deficiency are at increased risk for developing cholestasis in infancy and chronic liver disease as adults. The mechanism for hepatic cell injury, however, is not understood. Transgenic mice that express the normal human AAT gene demonstrate abundant AAT in hepatocytes and specific cell types of numerous nonhepatic tissues. Immunoperoxidase techniques have previously disclosed AAT in many of the cell types seen in transgenic mice; however, the issue of local synthesis vs. endocytosis in these cell types has remained unresolved. In this study, AAT mRNA was seen in a variety of tissues in the transgenic mouse. Immunoelectron microscopy of renal tubular and small intestinal epithelial cells in the transgenic mice demonstrated AAT within the cisternae of the rough endoplasmic reticulum, as in hepatocytes. These findings support the possibility of local synthesis in the various cell types. The results suggest that in addition to maintaining tissue integrity in the lung, the protease/antiprotease balance may have physiological functions in other organs as well.


Subject(s)
Mice, Transgenic/metabolism , alpha 1-Antitrypsin/analysis , Animals , Digestive System/enzymology , Digestive System/ultrastructure , Humans , Kidney Medulla/enzymology , Kidney Medulla/ultrastructure , Liver/enzymology , Liver/ultrastructure , Mice , Organ Specificity , Pancreas/enzymology , Pancreas/ultrastructure , Species Specificity , Tissue Distribution , alpha 1-Antitrypsin/genetics
19.
J Clin Invest ; 97(8): 1960-8, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8621781

ABSTRACT

Prolonged hypokalemia causes vasopressin-resistant polyuria. We have recently shown that another cause of severe polyuria, chronic lithium therapy, is associated with decreased aquaporin-2 (AQP2) water channel expression (Marples, D., S. Christensen, E.I. Christensen, P.D. Ottosen, and S. Nielsen, 1995. J. Clin. Invest., 95: 1838-1845). Consequently, we studied the effect in rats of 11 days' potassium deprivation on urine production and AQP2 expression and distribution. Membrane fractions were prepared from one kidney, while the contralateral kidney was perfusion-fixed for immunocytochemistry. Immunoblotting and densitometry revealed a decrease in AQP2 levels to 27+/-3.4% of control levels (n=11, P<0.001) in inner medulla, and 34+/-15% of controls (n=5, P<0.05) in cortex. Urine production increased in parallel, from 11+/-1.4 to 30+/-4.4 ml/day (n=11, P<0.01). After return to a potassium-containing diet both urine output and AQP2 labels normalized within 7 d. Immunocytochemistry confirmed decreased AQP2 labeling in principal cells of both inner medullary and cortical collecting ducts. AQP2 labeling was predominantly associated with the apical plasma membrane and intracellular vesicles. Lithium treatment for 24 d caused a more extensive reduction of AQP2 levels, to 4+/-1% of control levels in the inner medulla and 4+/-2% in cortex, in association with severe polyuria. The similar degree of downregulation in medulla and cortex suggests that interstitial tonicity is not the major factor in the regulation of AQP2 expression. Consistent with this furosemide treatment did not alter AQP2 levels. In summary,hypokalemia, like lithium treatment, results in a decrease in AQP2 expression in rat collecting ducts, in parallel with the development of polyuria, and the degree of downregulation is consistent with the level of polyuria induced, supporting the view that there is a causative link.


Subject(s)
Aquaporins , Gene Expression Regulation , Hypokalemia/metabolism , Ion Channels/biosynthesis , Kidney Cortex/metabolism , Kidney Medulla/metabolism , Animals , Aquaporin 2 , Aquaporin 6 , Cell Membrane/metabolism , Cell Membrane/pathology , Cell Membrane/ultrastructure , Electrophoresis, Polyacrylamide Gel , Hypokalemia/pathology , Immunoblotting , Ion Channels/analysis , Kidney Cortex/pathology , Kidney Cortex/ultrastructure , Kidney Medulla/pathology , Kidney Medulla/ultrastructure , Male , Microscopy, Electron , Polyuria , Potassium Deficiency/metabolism , Potassium Deficiency/urine , Rats , Rats, Wistar , Thirst
20.
J Clin Invest ; 80(6): 1660-9, 1987 Dec.
Article in English | MEDLINE | ID: mdl-3680519

ABSTRACT

To investigate the mechanisms responsible for urinary acidification in the terminal nephron, primary cultures of cells isolated from the renal papilla were grown as monolayers in a defined medium. Morphologically, cultured cells were epithelial in type, and similar to collecting duct principal cells. Cell pH measured fluorometrically in monolayers grown on glass slides showed recovery from acid loads in Na+-free media. Recovery was inhibited by cyanide, oligomycin A, and N-ethylmaleimide. Cyanide and oligomycin inhibited recovery less in the presence than in the absence of glucose. When cells were first acid loaded in a Na+-free medium and then exposed to external Na+, pH recovery also took place. This recovery exhibited first-order dependence on Na+ concentration and was inhibited by 5-(N-ethyl-N-isopropyl)amiloride. These studies demonstrate that in culture, collecting duct principal cells possess at least two mechanisms for acid extrusion: a proton ATP-ase and an Na+-H+ exchanger. The former may be responsible for some component of the urinary acidification observed in the papillary collecting duct in vivo; the role of the latter in acid-base transport remains uncertain.


Subject(s)
Acid-Base Equilibrium , Kidney Medulla/physiology , Kidney Tubules, Collecting/physiology , Kidney Tubules/physiology , Acid-Base Equilibrium/drug effects , Animals , Cyanides/pharmacology , Ethylmaleimide/pharmacology , Hydrogen-Ion Concentration , Kidney Medulla/drug effects , Kidney Medulla/ultrastructure , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/ultrastructure , Microscopy, Electron , Oligomycins/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL