Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Clin Invest ; 134(11)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38598837

ABSTRACT

Tissue regeneration is limited in several organs, including the kidney, contributing to the high prevalence of kidney disease globally. However, evolutionary and physiological adaptive responses and the presence of renal progenitor cells suggest an existing remodeling capacity. This study uncovered endogenous tissue remodeling mechanisms in the kidney that were activated by the loss of body fluid and salt and regulated by a unique niche of a minority renal cell type called the macula densa (MD). Here, we identified neuronal differentiation features of MD cells that sense the local and systemic environment and secrete angiogenic, growth, and extracellular matrix remodeling factors, cytokines and chemokines, and control resident progenitor cells. Serial intravital imaging, MD nerve growth factor receptor and Wnt mouse models, and transcriptome analysis revealed cellular and molecular mechanisms of these MD functions. Human and therapeutic translation studies illustrated the clinical potential of MD factors, including CCN1, as a urinary biomarker and therapeutic target in chronic kidney disease. The concept that a neuronally differentiated key sensory and regulatory cell type responding to organ-specific physiological inputs controls local progenitors to remodel or repair tissues may be applicable to other organs and diverse tissue-regenerative therapeutic strategies.


Subject(s)
Cell Differentiation , Regeneration , Animals , Mice , Humans , Kidney/metabolism , Neurons/metabolism , Neurons/pathology , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/genetics , Male
2.
J Am Soc Nephrol ; 23(3): 429-37, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22193389

ABSTRACT

Renal tubular atrophy accompanies many proteinuric renal diseases, suggesting that glomerular proteinuria injures the tubules. However, local or systemic inflammation and filtration of abnormal proteins known to directly injure tubules are also present in many of these diseases and animal models; therefore, whether glomerular proteinuria directly causes tubular injury is unknown. Here, we examined the renal response to proteinuria induced by selective podocyte loss. We generated mice that express the diphtheria toxin receptor exclusively in podocytes, allowing reproducible dose-dependent, specific ablation of podocytes by administering diphtheria toxin. Ablation of <20% of podocytes resulted in profound albuminuria that resolved over 1-2 weeks after the re-establishment of normal podocyte morphology. Immediately after the onset of albuminuria, proximal tubule cells underwent a transient burst of proliferation without evidence of tubular damage or increased apoptosis, resulting in an increase in total tubular cell numbers. The proliferative response coincided with detection of the growth factor Gas6 in the urine and phosphorylation of the Gas6 receptor Axl in the apical membrane of renal tubular cells. In contrast, ablation of >40% of podocytes led to progressive glomerulosclerosis, profound tubular injury, and renal failure. These data suggest that glomerular proteinuria in the absence of severe structural glomerular injury activates tubular proliferation, potentially as an adaptive response to minimize the loss of filtered proteins.


Subject(s)
Albuminuria/physiopathology , Cell Proliferation , Kidney Glomerulus/physiopathology , Kidney Tubules, Proximal/pathology , Podocytes/pathology , Proteinuria/physiopathology , Albuminuria/metabolism , Albuminuria/pathology , Animals , Disease Models, Animal , Female , Heparin-binding EGF-like Growth Factor , Integrases/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Proteinuria/metabolism , Proteinuria/pathology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Axl Receptor Tyrosine Kinase
3.
Dev Cell ; 13(3): 377-90, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17765681

ABSTRACT

Mutations in the inositol 5-phosphatase OCRL are responsible for Lowe syndrome, whose manifestations include mental retardation and renal Fanconi syndrome. OCRL has been implicated in membrane trafficking, but disease mechanisms remain unclear. We show that OCRL visits late-stage, endocytic clathrin-coated pits and binds the Rab5 effector APPL1 on peripheral early endosomes. The interaction with APPL1, which is mediated by the ASH-RhoGAP-like domains of OCRL and is abolished by disease mutations, provides a link to protein networks implicated in the reabsorptive function of the kidney and in the trafficking and signaling of growth factor receptors in the brain. Crystallographic studies reveal a role of the ASH-RhoGAP-like domains in positioning the phosphatase domain at the membrane interface and a clathrin box protruding from the RhoGAP-like domain. Our results support a role of OCRL in the early endocytic pathway, consistent with the predominant localization of its preferred substrates, PI(4,5)P(2) and PI(3,4,5)P(3), at the cell surface.


Subject(s)
Endocytosis/physiology , Endosomes/metabolism , Phosphoric Monoester Hydrolases/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , COS Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Chlorocebus aethiops , Clathrin-Coated Vesicles/metabolism , Crystallography, X-Ray , Endosomes/enzymology , Glutathione Transferase/metabolism , Green Fluorescent Proteins/metabolism , Humans , Kidney/cytology , Models, Biological , Models, Molecular , Molecular Sequence Data , Mutation , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/isolation & purification , Phosphorylation , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Time Factors
4.
Am J Physiol Cell Physiol ; 300(6): C1366-74, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21325636

ABSTRACT

A disintegrin and metalloprotease 10 (ADAM10) is a zinc protease that mediates ectodomain shedding of numerous receptors including Notch and members of the amyloid precursor protein family (APP, APLP1, and APLP2). Ectodomain shedding frequently activates a process called regulated intramembrane proteolysis (RIP) that links cellular events with gene regulation. To characterize ADAM10 in kidney and in opossum kidney proximal tubule (OKP) cells, we performed indirect immunofluorescence microscopy and immunoblotting of renal membrane fractions using specific antibodies. These studies show that ADAM10 and APLP2 are coexpressed in the proximal tubule and in OKP cells. To study the role of ADAM10 activity in the proximal tubule, we stably overexpressed wild-type ADAM10 or an inactive mutant ADAM10 in OKP cells. We found a direct correlation between the amount of active ADAM10 expressed and 1) the amount of APLP2 ectodomain shed into the culture supernatant and 2) the amount of Na(+)/H(+) exchanger 3 (NHE3) and megalin mRNA and protein expressed compared with control proteins. To establish a link between ADAM10-mediated shedding of APLP2 and the effect on NHE3 and megalin mRNA expression we performed RNA interference experiments using APLP2-specific short hairpin RNA (shRNA) in OKP cells. Cells expressing the APLP2 shRNA showed >80% knock down of APLP2 protein and mRNA as well as 60-70% reduction in NHE3 protein and mRNA. Levels of megalin and Na-K-ATPase protein and mRNA were not changed. These studies show 1) ADAM10 and APLP2 are expressed in proximal tubule cells and, 2) ADAM10 activity has a pronounced effect on expression of specific brush-border proteins. We postulate that ADAM10 and APLP2 may represent elements of a here-to-fore unknown signaling pathway in proximal tubule that link events at the brush border with control of gene expression.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Kidney Tubules, Proximal/cytology , ADAM Proteins/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Cattle , Cells, Cultured , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Mice , Mice, Inbred BALB C , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism
5.
Cytoskeleton (Hoboken) ; 67(3): 178-92, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20175219

ABSTRACT

Myosin VI (Myo6) is an actin-based molecular motor involved in clathrin-mediated endocytosis that is highly expressed in the renal proximal tubule brush border. We investigated the renal physiological consequences of loss of Myo6 function by performing renal clearance and physiological measurements on Myo6 functional null Snell's waltzer (sv/sv) and control heterozygous (+/sv) mice. Sv/sv mice showed reduced body weight and elevated blood pressure compared with controls; no differences were observed for glomerular flow rate, urine volume, blood acid-base parameters, and plasma concentrations and urinary excretions of Na(+) and K(+). To assess the integrity of endocytosis-mediated protein absorption by the kidney, urinary albumin excretion was measured, and the proximal tubular uptake of intravenously injected endocytic marker horseradish peroxidase (HRP) was examined. Albumin excretion was increased nearly 4-fold in sv/sv mice relative to controls. Conversely, HRP uptake was reduced and delayed in proximal tubule cells of the sv/sv kidney observed by electron microscopy at 5 and 30 min after injection. Consistent with impaired endocytosis, we also observed defects indicating alterations along the endocytic pathway in sv/sv proximal tubule cells: (1) decreased membrane association of the clathrin adaptor subunit, adaptin beta, and Disabled-2 (Dab2) after sedimentation of renal homogenates and (2) reduced apical vacuole number. In addition, proximal tubular dilation and fibrosis, likely secondary effects of the loss of Myo6, were observed in sv/sv kidneys. These results indicate that Myo6 plays a key role in endocytosis-mediated protein absorption in the mouse kidney proximal tubule.


Subject(s)
Endocytosis/physiology , Kidney Tubules, Proximal/physiology , Myosin Heavy Chains/deficiency , Actins/metabolism , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Transdifferentiation , Horseradish Peroxidase/metabolism , Kidney Tubules, Proximal/pathology , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Mice , Mice, Neurologic Mutants , Microfilament Proteins/metabolism , Microvilli/genetics , Microvilli/metabolism , Vesicular Transport Proteins/metabolism
6.
Am J Physiol Cell Physiol ; 295(2): C529-37, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18495814

ABSTRACT

We recently reported that megalin is subjected to regulated intramembrane proteolysis (RIP) and includes 1) protein kinase C (PKC)-regulated, metalloprotease-mediated ectodomain shedding producing a membrane-bound megalin COOH-terminal fragment (MCTF) and 2) gamma-secretase-mediated cleavage of the MCTF producing a soluble megalin intracellular domain (MICD). Based on studies of RIP of other receptors, the MICD is predicted to target to the nucleus and regulate gene expression. To determine whether RIP of megalin regulates proximal tubule gene expression, we stably expressed the transfected MCTF (tMCTF) or transfected MICD (tMICD) in opossum kidney proximal tubule (OKP) cells and examined the resulting phenotype. Immunoblotting and immunocytochemical analysis of tMCTF cells showed the tMCTF was expressed and constitutively processed by gamma-secretase. Analysis of specific protein expression in tMCTF- and tMICD-transfected cells using Western blot showed endogenous megalin and Na(+)/H(+) exchanger 3 (NHE3) protein expression to be dramatically lower than that of control cells. Expression of other proteins including myosin VI, beta-adaptin, and the Na-K-ATPase appeared unchanged. Analysis of specific mRNA expression using quantitative real-time PCR showed megalin and NHE3 mRNA levels were significantly lower in tMCTF- and tMICD-transfected cells compared with controls. Inhibition of gamma-secretase activity in tMCTF cells resulted in an 8- to 10-fold recovery of megalin mRNA within 4 h. These data show that the COOH-terminal domain of megalin regulates expression of specific proteins in OKP cells and provides the first evidence that RIP of megalin may be part of a signaling pathway linking protein absorption and gene expression in proximal tubule.


Subject(s)
Gene Expression Regulation , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Binding Sites , Cell Line , Cytoplasm/metabolism , Endosomes/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Golgi Apparatus/metabolism , Kidney Tubules, Proximal/cytology , Low Density Lipoprotein Receptor-Related Protein-2/chemistry , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Microvilli/metabolism , Opossums , Peptide Fragments/genetics , Peptide Fragments/metabolism , Plasmids/genetics , Protease Inhibitors/pharmacology , Protein Processing, Post-Translational , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Transfection
7.
Am J Physiol Renal Physiol ; 295(2): F380-7, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18463315

ABSTRACT

MinK-related peptides (MiRPs) are single-span membrane proteins that assemble with specific voltage-gated K+ (Kv) channel alpha-subunits to establish gating kinetics, unitary conductance, expression level, and pharmacology of the mixed complex. MiRP3 (encoded by the KCNE4 gene) has been shown to alter the behavior of some Kv alpha-subunits in vitro but its natural partners and physiologic functions are unknown. Seeking in vivo partners for MiRP3, immunohistochemistry was used to localize its expression to a unique subcellular site, the apical membrane of renal intercalated cells, where one potassium channel type has been recorded, the calcium- and voltage-gated channel BK. Overlapping staining of these two proteins was found in rabbit intercalated cells, and MiRP3 and BK subunits expressed in tissue culture cells were found to form detergent-stable complexes. Electrophysiologic and biochemical evaluation showed MiRP3 to act on BK to reduce current density in two fashions: shifting the current-voltage relationship to more depolarized voltages in a calcium-dependent fashion ( approximately 10 mV at normal intracellular calcium levels) and accelerating degradation of MiRP3-BK complexes. The findings suggest a role for MiRP3 modulation of BK-dependent urinary potassium excretion.


Subject(s)
Kidney/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Potassium Channels, Voltage-Gated/metabolism , Protein Subunits/metabolism , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Humans , Kidney/cytology , Membrane Potentials/physiology , Oocytes/cytology , Oocytes/metabolism , Patch-Clamp Techniques , Potassium/urine , Potassium Channels, Voltage-Gated/genetics , Rabbits , Rats , Transfection
8.
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
9.
Am J Physiol Renal Physiol ; 289(2): F249-58, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15687252

ABSTRACT

Transfection studies using mutant constructs have implicated one or both protein kinase A (PKA) consensus phosphorylation sites [serines 552 and 605 in rat Na(+)/H(+) exchanger type 3 (NHE3)] as critical for mediating inhibition of NHE3 in response to several stimuli including dopamine. However, whether one or both of these sites is actually phosphorylated in endogenous NHE3 in proximal tubule cells is unknown. The purpose of this study was to generate phosphospecific antibodies so that the state of phosphorylation of these serine residues in endogenous NHE3 could be assessed in vitro and in vivo. To this end, polyclonal and monoclonal phosphospecific peptide antibodies were generated against each PKA consensus site. Phosphospecificity was established by ELISA and Western blot assays. We then used these antibodies in vitro to evaluate the effect of dopamine on phosphorylation of the corresponding PKA sites (serines 560 and 613) in NHE3 endogenously expressed in opossum kidney cells. Baseline phosphorylation of both sites was detected that was significantly increased by dopamine. Next, we determined the baseline phosphorylation state of each serine in rat kidney NHE3 in vivo. We found that serine 552 of NHE3 is phosphorylated to a much greater extent than serine 605 at baseline in vivo. Moreover, we detected a distinct subcellular localization for NHE3 phosphorylated at serine 552 compared with total NHE3. Specifically, NHE3 phosphorylated at serine 552 localized to the coated pit region of the brush-border membrane, where NHE3 is inactive, while total NHE3 was found throughout the brush-border membrane. These findings strongly suggest that phosphorylation of NHE3 plays a role in its subcellular trafficking in vivo. In conclusion, we successfully generated phosphospecific antibodies that should be useful to assess the phosphorylation of endogenous NHE3 at its two PKA consensus sites under a variety of physiological conditions in vitro and in vivo.


Subject(s)
Antibodies , Cyclic AMP-Dependent Protein Kinases/metabolism , Phosphorylation , Sodium-Hydrogen Exchangers/metabolism , Animals , Antibodies/isolation & purification , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibody Specificity , COS Cells , Cell Line , Chlorocebus aethiops , Dopamine/pharmacology , Electrophoresis, Polyacrylamide Gel , Immunohistochemistry , Kidney/metabolism , Male , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Microvilli/metabolism , Opossums , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Sodium-Hydrogen Exchanger 3 , Subcellular Fractions/metabolism , Transfection
10.
Curr Opin Nephrol Hypertens ; 12(5): 533-41, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12920402

ABSTRACT

PURPOSE OF REVIEW: The proximal tubule sodium/hydrogen exchanger continuously reabsorbs the bulk of the filtered sodium, controlling salt delivery to the distal nephron which is critical for tubuloglomerular feedback autoregulation and for fine control of salt excretion in the distal nephron. This review focuses on recent studies of the mechanisms of regulation of sodium transport in the proximal tubule, and addresses whether results from studies in proximal tubule cell lines are applicable to the proximal tubule in situ. RECENT FINDINGS: Recent in-vivo studies provided evidence that sodium/hydrogen exchanger isoform 3 can move into and out of the apical microvilli accompanied by parallel changes in renal sodium transport: the exchanger is retracted from the microvilli in response to hypertension, parathyroid hormone or dopamine treatment and moved into the microvilli in response to sympathetic nervous system stimulation, puromycin aminonucleoside induced nephritic syndrome, and insulin treatment. Studies in cultured opossum kidney proximal tubule cells provided evidence for clathrin coated vesicle mediated, dynamin dependent, cytoskeleton dependent internalization of sodium/hydrogen exchanger isoform 3 from the surface to an endosomal pool in response to dopamine or parathyroid hormone. In the intact proximal tubule there is evidence for a two-step internalization process: (1) from villi to the intermicrovillar cleft region and (2) to a higher density membrane pool that may be either below the microvilli or deep in intermicrovillar clefts. Recent studies have described a significant inactive pool of the exchanger in the intermicrovillar region in vivo that may serve as a storage and recruitable pool. SUMMARY: The molecular mechanisms responsible for increasing or decreasing sodium transport in the proximal tubule appear to include redistribution of sodium/hydrogen exchanger isoform 3 to or from the microvillar region. Detailed studies in cultured proximal tubule cell lines provide evidence for endocytosis and exocytosis of the exchanger dependent on cytoskeleton and clathrin coated vesicles. In vivo, the apical membrane is differentiated into discrete villar and intermicrovillar membrane domains and the intermicrovillar domain, not observed in cultured cells, may serve as a recruitable storage pool for sodium/hydrogen exchanger isoform 3.


Subject(s)
Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/ultrastructure , Sodium-Hydrogen Exchangers/metabolism , Animals , Biological Transport, Active/physiology , Cells, Cultured , Humans , Insulin/metabolism , Ion Transport/physiology , Parathyroid Hormone/metabolism
11.
Am J Physiol Renal Physiol ; 282(5): F785-94, 2002 May.
Article in English | MEDLINE | ID: mdl-11934687

ABSTRACT

Myosin VI is a reverse-direction molecular motor implicated in membrane transport events. Because myosin VI is most highly expressed in the kidney, we investigated its renal localization by using high-resolution immunocytochemical and biochemical methods. Indirect immunofluorescence microscopy revealed myosin VI at the base of the brush border in proximal tubule cells. Horseradish peroxidase uptake studies, which labeled endosomes, and double staining for clathrin adapter protein-2 showed that myosin VI was closely associated with the intermicrovillar (IMV) coated-pit region of the brush border. Localization of myosin VI to the IMV region was confirmed at the electron microscopic level by colloidal gold labeling of ultrathin cryosections. In addition, antigen retrieval demonstrated a small but significant pool of myosin VI on the microvilli. To confirm the association of myosin VI with the IMV compartment, these membranes were separated from other membrane compartments by using 15-25% OptiPrep density gradients. Immunoblotting of the gradient fractions confirmed that myosin VI was enriched with markers for the IMV microdomain of the brush border, suggesting that myosin VI associates with proteins in this compartment. Finally, we examined the expression of myosin VI during nephron development. We found myosin VI present in a diffuse cytoplasmic pattern at stage II (S-shaped body phase) and that it was only redistributed fully to the brush border in the stage IV nephron. These studies support a model for myosin VI function in the endocytic process of the proximal tubule.


Subject(s)
Endocytosis , Kidney Tubules, Proximal/growth & development , Kidney Tubules, Proximal/metabolism , Myosin Heavy Chains/metabolism , Animals , Animals, Newborn , Centrifugation, Density Gradient , Endosomes/metabolism , Endosomes/ultrastructure , Fluorescent Antibody Technique , Horseradish Peroxidase , Immunoblotting , Kidney Tubules, Proximal/ultrastructure , Microscopy, Confocal , Microscopy, Immunoelectron , Microvilli/metabolism , Microvilli/ultrastructure , Rats , Rats, Sprague-Dawley
12.
J Biol Chem ; 279(33): 34302-10, 2004 Aug 13.
Article in English | MEDLINE | ID: mdl-15180987

ABSTRACT

Megalin, a member of the low density lipoprotein receptor gene family, is required for efficient protein absorption in the proximal tubule. Recent studies have shown that the low density lipoprotein receptor-related protein, another member of this gene family, is proteolytically processed by gamma-secretase implying a role for low density lipoprotein receptor-related protein in a Notchlike signaling pathway. This pathway has been shown to involve: 1) metalloprotease-mediated ectodomain shedding and gamma-secretase-mediated intramembrane proteolysis of some receptors. Experiments were performed to determine whether megalin undergoes similar processing. By immunocytochemistry, immunoblotting, and a fluorogenic enzyme assay presenilin-1 (required for gamma-secretase activity) and gamma-secretase activity were found in the brush border of proximal kidney tubules where megalin is localized. Using a fluorogenic peptide containing an amyloid precursor protein gamma-secretase cleavage site and Compound E, a specific gamma-secretase inhibitor, we found high levels of gamma-secretase activity in renal brush border membrane vesicles. Immunoblotting analysis of renal microsomes and opossum kidney proximal tubule (OKP) cells using antibodies directed to the cytosolic domain of megalin showed a 35-40-kDa, membrane-associated, carboxyl-terminal fragment of megalin (MCTF). When cells were incubated with 200 nm phorbol 12-myristate 13-acetate, the appearance of the MCTF increased 2.5-fold and was blocked by metalloprotease inhibitors. When the cells were incubated with gamma-secretase inhibitor Compound E, it caused a 2-fold increase in MCTF. Finally, incubating the cells with 1 microm vitamin D-binding protein resulted in a 25% increase in the appearance of the MCTF. In summary, the MCTF is produced by protein kinase C regulated, metalloprotease-mediated ectodomain shedding and is the substrate for gamma-secretase. We postulate that the enzymatic processing of megalin represents part of a novel ligand-dependent signaling pathway in the proximal tubule that links receptor-mediated endocytosis with cell signaling.


Subject(s)
Endocytosis , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Signal Transduction , Amyloid Precursor Protein Secretases , Animals , Antibodies, Monoclonal/chemistry , Aspartic Acid Endopeptidases , Binding Sites , Culture Media, Serum-Free/pharmacology , Cytosol/metabolism , Electrophoresis, Polyacrylamide Gel , Endopeptidases/metabolism , Fluorescent Antibody Technique, Indirect , Glutathione Transferase/metabolism , Immunoblotting , Immunohistochemistry , Kidney/metabolism , Lipoproteins, LDL/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Matrix Metalloproteinases/metabolism , Membrane Proteins/metabolism , Metalloproteases/metabolism , Mice , Mice, Inbred BALB C , Microsomes/metabolism , Mitochondria/metabolism , Molecular Sequence Data , Opossums , Peptides/chemistry , Presenilin-1 , Protein Kinase C/metabolism , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , Tetradecanoylphorbol Acetate , Vitamin D-Binding Protein/metabolism
13.
J Am Soc Nephrol ; 13(12): 2831-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12444201

ABSTRACT

Potassium (K) channels regulate cell membrane potential and modulate a number of important cellular functions. KCNA10 is a cyclic nucleotide-gated, voltage-activated K channel that is detected in kidney, heart, and aorta by Northern blot and postulated to participate in renal K metabolism and to regulate vascular tone. The aim of this study was to establish the cellular and subcellular localization of KCNA10 in kidney and vascular tissues. An anti-KCNA10 polyclonal antibody was generated, and immunocytochemical studies were performed on rat kidney. KCNA10 protein was easily detectable at the apical membrane of rat proximal tubular cells, and a weaker signal was also evident in the glomerulus. In situ hybridization experiments confirmed the immunocytochemical studies and revealed KCNA10 expression in human proximal tubular cells, glomerular and vascular endothelial cells, and also in vascular smooth muscle cells. The data suggest that KCNA10 may facilitate proximal tubular sodium absorption by stabilizing cell membrane voltage. Furthermore, its presence in endothelial and vascular smooth muscle cells supports the notion that it also regulates vascular tone.


Subject(s)
Kidney Glomerulus/metabolism , Kidney Tubules, Proximal/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Animals , Cell Membrane/metabolism , Endothelium/cytology , Endothelium/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Kidney Glomerulus/cytology , Kidney Tubules, Proximal/cytology , Kv1.1 Potassium Channel , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Sprague-Dawley , Renal Circulation , Subcellular Fractions/metabolism , Tissue Distribution
14.
Kidney Int ; 61(4): 1351-64, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11918742

ABSTRACT

BACKGROUND: Depolymerization of microtubules in proximal tubule (PT) cells of colchicine-treated rats causes disruption of vesicle recycling and redistribution of some brush-border membrane (BBM) transporters into cytoplasmic vesicles. NHE3, an isoform of the Na+/H+ exchanger in the PT cell BBM, is acutely regulated by a variety of mechanisms, including protein trafficking and interaction with the PDZ protein, NHERF. The effects of microtubule disruption by colchicine on NHE3 trafficking in PT and the potential role of NHERF in this process have not been studied. METHODS: Immunofluorescence and immunogold cytochemistry were performed on cryosections of kidney tissue, and immunoblotting of BBM isolated from the renal cortex and outer stripe of control and colchicine-treated (3.2 mg/kg, IP, a single dose 12 hours before sacrifice) rats. RESULTS: In cells of the convoluted PT (S1/S2 segments) of control rats, NHE3 was located mainly in the BBM; subapical endosomes were weakly stained. In cells of the straight PT (S3 segment), NHE3 was present in the BBM and in lysosomes. In colchicine-treated rats, there was a marked redistribution of NHE3 from the BBM into intracellular vesicles and the basolateral plasma membrane in the S1/S2 segments. In the S3 segment, the abundance of BBM NHE3 was not visibly changed, but NHE3-positive intracellular organelles largely disappeared, and the antigen was detectable in the basolateral plasma membrane. The PDZ protein NHERF followed a similar pattern: in control animals, it was strong in the BBM and negative in the basolateral membrane in cells along the PT. After colchicine treatment, expression of NHERF in the basolateral membrane strongly increased in all PT segments, where it colocalized with NHE3. CONCLUSIONS: The data indicate that: (a) microtubules are involved in the apical targeting of NHE3 and NHERF in renal PT cells, and (b) the parallel basolateral insertion of NHE3 and NHERF may represent an indirect targeting pathway that involves transient, microtubule-independent basolateral insertion of these proteins, followed by microtubule-dependent, vesicle-mediated transcytosis to the BBM.


Subject(s)
Colchicine/pharmacology , Intracellular Membranes/metabolism , Kidney Tubules, Proximal/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Kidney Tubules, Proximal/drug effects , Male , Microtubules/drug effects , Microtubules/metabolism , Microvilli/metabolism , Rats , Rats, Wistar , Reference Values , Sodium-Hydrogen Exchanger 3
SELECTION OF CITATIONS
SEARCH DETAIL