Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
J Biol Chem ; 299(3): 102925, 2023 03.
Article in English | MEDLINE | ID: mdl-36682497

ABSTRACT

Polymorphism of the gene encoding mucin 1 (MUC1) is associated with skeletal and dental phenotypes in human genomic studies. Animals lacking MUC1 exhibit mild reduction in bone density. These phenotypes could be a consequence of modulation of bodily Ca homeostasis by MUC1, as suggested by the previous observation that MUC1 enhances cell surface expression of the Ca2+-selective channel, TRPV5, in cultured unpolarized cells. Using biotinylation of cell surface proteins, we asked whether MUC1 influences endocytosis of TRPV5 and another Ca2+-selective TRP channel, TRPV6, in cultured polarized epithelial cells. Our results indicate that MUC1 reduces endocytosis of both channels, enhancing cell surface expression. Further, we found that mice lacking MUC1 lose apical localization of TRPV5 and TRPV6 in the renal tubular and duodenal epithelium. Females, but not males, lacking MUC1 exhibit reduced blood Ca2+. However, mice lacking MUC1 exhibited no differences in basal urinary Ca excretion or Ca retention in response to PTH receptor signaling, suggesting compensation by transport mechanisms independent of TRPV5 and TRPV6. Finally, humans with autosomal dominant tubulointerstitial kidney disease due to frame-shift mutation of MUC1 (ADTKD-MUC1) exhibit reduced plasma Ca concentrations compared to control individuals with mutations in the gene encoding uromodulin (ADTKD-UMOD), consistent with MUC1 haploinsufficiency causing reduced bodily Ca2+. In summary, our results provide further insight into the role of MUC1 in Ca2+-selective TRP channel endocytosis and the overall effects on Ca concentrations.


Subject(s)
Calcium , Mucin-1 , TRPV Cation Channels , Animals , Female , Humans , Mice , Calcium/blood , Calcium/metabolism , Calcium/urine , Cell Membrane/metabolism , Cells, Cultured , Mucin-1/genetics , Mucin-1/metabolism , TRPV Cation Channels/metabolism , Epithelial Cells/metabolism , Sex Factors , Mutation , Protein Transport/genetics
2.
Antioxidants (Basel) ; 11(9)2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36139884

ABSTRACT

Acute kidney injury (AKI) is a rapid decline in renal function and can occur after ischemia/reperfusion injury (IRI) to the tubular epithelia. The nuclear factor erythroid-2-related factor 2 (NRF2) pathway protects against AKI and AKI-to-chronic kidney disease (CKD) progression, but we previously demonstrated that severe IRI maladaptively reduced NRF2 activity in mice. To understand the mechanism of this response, we subjected C57BL/6J mice to unilateral kidney IRI with ischemia times that were titrated to induce mild to severe injury. Mild IRI increased NRF2 activity and was associated with renal recovery, whereas severe IRI decreased NRF2 activity and led to progressive CKD. Due to these effects of ischemia, we tested the hypothesis that hypoxia-inducible factor-1α (HIF-1α) mediates NRF2 activity. To mimic mild and severe ischemia, we activated HIF-1α in HK-2 cells in nutrient-replete or nutrient-deficient conditions. HIF-1α activation in nutrient-replete conditions enhanced NRF2 nuclear localization and activity. However, in nutrient-deficient conditions, HIF-1α activation suppressed NRF2 nuclear localization and activity. Nuclear localization was rescued with HIF-1α siRNA knockdown. Our results suggest that severe ischemic AKI leads to HIF-1α-mediated suppression of NRF2, leading to AKI-to-CKD progression.

3.
Am J Physiol Renal Physiol ; 321(2): F135-F148, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34151589

ABSTRACT

Cell-associated kidney injury molecule-1 (KIM-1) exerts an anti-inflammatory role following kidney injury by mediating efferocytosis and downregulating the NF-κB pathway. KIM-1 cleavage blunts its anti-inflammatory activities. We reported that mucin 1 (MUC1) is protective in a mouse model of ischemia-reperfusion injury (IRI). As both KIM-1 and MUC1 are induced in the proximal tubule (PT) during IRI and are a disintegrin and metalloprotease 17 (ADAM17) substrates, we tested the hypothesis that MUC1 protects KIM-1 activity. Muc1 knockout (KO) mice and wild-type (WT) littermates were subjected to IRI. KIM-1, MUC1, and ADAM17 levels (and signaling pathways) were assessed by immunoblot analysis. PT localization was assessed by confocal microscopy and an in situ proximity ligation assay. Findings were extended using human kidneys and urine as well as KIM-1-mediated efferocytosis assays in mouse PT cultures. In response to tubular injury in mouse and human kidneys, we observed induction and coexpression of KIM-1 and MUC1 in the PT. Compared with WT mice, Muc1 KO mice had higher urinary KIM-1 and lower kidney KIM-1. KIM-1 was apical in the PT of WT kidneys but predominately with luminal debris in Muc1 KO mice. Efferocytosis was reduced in Muc1 KO PT cultures compared with WT cultures, whereas inflammation was increased in Muc1 KO kidneys compared with WT kidneys. MUC1 was cleaved by ADAM17 in PT cultures and blocked KIM-1 shedding in Madin-Darby canine kidney cells. We conclude that KIM-1-mediated efferocytosis and thus anti-inflammatory activity during IRI is preserved in the injured kidney by MUC1 inhibition of KIM-1 shedding.NEW & NOTEWORTHY KIM-1 plays a key role in the recovery of the tubule epithelium during renal IRI by mediating efferocytosis and associated signaling that suppresses inflammation. Excessive cleavage of KIM-1 by ADAM17 provides a decoy receptor that aggravates efferocytosis and subsequent signaling. Our data from experiments in mice, patients, and cultured cells show that MUC1 is also induced during IRI and competes with KIM-1 for cleavage by ADAM17. Consequently, MUC1 protects KIM-1 anti-inflammatory activity in the damaged kidney.


Subject(s)
Hepatitis A Virus Cellular Receptor 1/metabolism , Inflammation/metabolism , Kidney Tubules, Proximal/metabolism , Kidney/blood supply , Mucin-1/metabolism , Reperfusion Injury/metabolism , ADAM17 Protein/metabolism , Animals , Cell Line , Dogs , Humans , Kidney/metabolism , Mice, Knockout , Mice, Transgenic , Mucin-1/genetics , Phagocytosis/physiology
4.
Am J Physiol Renal Physiol ; 317(2): F303-F321, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31166705

ABSTRACT

The proper function of the organs that make up the urinary tract (kidneys, ureters, bladder, and urethra) depends on their ability to sense and respond to mechanical forces, including shear stress and wall tension. However, we have limited understanding of the mechanosensors that function in these organs and the tissue sites in which these molecules are expressed. Possible candidates include stretch-activated PIEZO channels (PIEZO1 and PIEZO2), which have been implicated in mechanically regulated body functions including touch sensation, proprioception, lung inflation, and blood pressure regulation. Using reporter mice expressing a COOH-terminal fusion of Piezo1 with the sequence for the tandem-dimer Tomato gene, we found that PIEZO1 is expressed in the kidneys, ureters, bladder, and urethra as well as organs in close proximity, including the prostate, seminal vesicles and ducts, ejaculatory ducts, and the vagina. We further found that PIEZO1 expression is not limited to one cell type; it is observed in the endothelial and parietal cells of the renal corpuscle, the basolateral surfaces of many of the epithelial cells that line the urinary tract, the interstitial cells of the bladder and ureters, and populations of smooth and striated muscle cells. We propose that in the urinary tract, PIEZO1 likely functions as a mechanosensor that triggers responses to wall tension.


Subject(s)
Ion Channels/metabolism , Urinary Tract/metabolism , Animals , Female , Gene Expression Regulation , Genes, Reporter , Ion Channels/genetics , Male , Mechanotransduction, Cellular , Mice, Inbred C57BL , Mice, Transgenic , Pressure , Stress, Mechanical , Tissue Distribution , Urinary Tract/cytology
5.
Curr Opin Nephrol Hypertens ; 26(5): 384-391, 2017 09.
Article in English | MEDLINE | ID: mdl-28622163

ABSTRACT

PURPOSE OF REVIEW: Recent studies in the kidney have revealed that the well characterized tumor antigen mucin 1 (MUC1/Muc1) also has numerous functions in the normal and injured kidney. RECENT FINDINGS: Mucin 1 is a transmembrane mucin with a robust glycan-dependent apical targeting signal and efficient recycling from endosomes. It was recently reported that the TRPV5 calcium channel is stabilized on the cell surface by galectin-dependent cross-linking to mucin 1, providing a novel mechanism for regulation of ion channels and normal electrolyte balance.Our recent studies in mice show that Muc 1 is induced after ischemia, stabilizing hypoxia-inducible factor 1 (HIF-1)α and ß-catenin levels, and transactivating the HIF-1 and ß-catenin protective pathways. However, prolonged induction of either pathway in the injured kidney can proceed from apparent full recovery to chronic kidney disease. A very recent report indicates that aberrant activation of mucin 1 signaling after ischemic injury in mice and humans is associated with development of chronic kidney disease and fibrosis. A frameshift mutation in MUC1 was recently identified as the genetic lesion causing medullary cystic kidney disease type 1, now appropriately renamed MUC1 Kidney Disease. SUMMARY: Studies of mucin 1 in the kidney now reveal significant functions for the extracellular mucin-like domain and signaling through the cytoplasmic tail.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/metabolism , Mucin-1/metabolism , Renal Insufficiency, Chronic/metabolism , TRPV Cation Channels/metabolism , Animals , Cell Membrane/metabolism , Galectins/metabolism , Humans , Ischemia/metabolism , Mucin-1/genetics , Polycystic Kidney, Autosomal Dominant/genetics , Signal Transduction , Water-Electrolyte Balance , beta Catenin/metabolism
6.
Am J Physiol Renal Physiol ; 311(5): F890-F900, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27534994

ABSTRACT

Aquaporin-2 (AQP2) is essential to maintain body water homeostasis. AQP2 traffics from intracellular vesicles to the apical membrane of kidney collecting duct principal cells in response to vasopressin [arginine vasopressin (AVP)], a hormone released with low intravascular volume, which causes decreased kidney perfusion. Decreased kidney perfusion activates AMP-activated kinase (AMPK), a metabolic sensor that inhibits the activity of several transport proteins. We hypothesized that AMPK activation also inhibits AQP2 function. These putative AMPK effects could protect interstitial ionic gradients required for urinary concentration during metabolic stress when low intravascular volume induces AVP release. Here we found that short-term AMPK activation by treatment with 5-aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside (AICAR; 75 min) in kidney tissue prevented baseline AQP2 apical accumulation in principal cells, but did not prevent AQP2 apical accumulation in response to the AVP analog desmopressin (dDAVP). Prolonged AMPK activation prevented AQP2 cell membrane accumulation in response to forskolin in mouse collecting duct mpkCCDc14 cells. Moreover, AMPK inhibition accelerated hypotonic lysis of Xenopus oocytes expressing AQP2. We performed phosphorylation assays to elucidate the mechanism by which AMPK regulates AQP2. Although AMPK weakly phosphorylated immunoprecipitated AQP2 in vitro, no direct AMPK phosphorylation of the AQP2 COOH-terminus was detected by mass spectrometry. AMPK promoted Ser-261 phosphorylation and antagonized dDAVP-dependent phosphorylation of other AQP2 COOH-terminal sites in cells. Our findings suggest an increasing, time-dependent antagonism of AMPK on AQP2 regulation with AICAR-dependent inhibition of cAMP-dependent apical accumulation and AVP-dependent phosphorylation of AQP2. This inhibition likely occurs via a mechanism that does not involve direct AQP2 phosphorylation by AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aquaporin 2/metabolism , Kidney Tubules, Collecting/metabolism , Kidney/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cell Line , Kidney/cytology , Kidney/drug effects , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/drug effects , Male , Mice , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology , Xenopus
7.
Am J Physiol Renal Physiol ; 310(11): F1216-28, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26911844

ABSTRACT

Extracellular proton-secreting transport systems that contribute to extracellular pH include the vacuolar H(+)-ATPase (V-ATPase). This pump, which mediates ATP-driven transport of H(+) across membranes, is involved in metastasis. We previously showed (Alzamora R, Thali RF, Gong F, Smolak C, Li H, Baty CJ, Bertrand CA, Auchli Y, Brunisholz RA, Neumann D, Hallows KR, Pastor-Soler NM. J Biol Chem 285: 24676-24685, 2010) that V-ATPase A subunit phosphorylation at Ser-175 is important for PKA-induced V-ATPase activity at the membrane of kidney intercalated cells. However, Ser-175 is also located within a larger phosphorylation consensus sequence for Aurora kinases, which are known to phosphorylate proteins that contribute to the pathogenesis of metastatic carcinomas. We thus hypothesized that Aurora kinase A (AURKA), overexpressed in aggressive carcinomas, regulates the V-ATPase in human kidney carcinoma cells (Caki-2) via Ser-175 phosphorylation. We found that AURKA is abnormally expressed in Caki-2 cells, where it binds the V-ATPase A subunit in an AURKA phosphorylation-dependent manner. Treatment with the AURKA activator anacardic acid increased V-ATPase expression and activity at the plasma membrane of Caki-2 cells. In addition, AURKA phosphorylates the V-ATPase A subunit at Ser-175 in vitro and in Caki-2 cells. Immunolabeling revealed that anacardic acid induced marked membrane accumulation of the V-ATPase A subunit in transfected Caki-2 cells. However, anacardic acid failed to induce membrane accumulation of a phosphorylation-deficient Ser-175-to-Ala (S175A) A subunit mutant. Finally, S175A-expressing cells had decreased migration in a wound-healing assay compared with cells expressing wild-type or a phospho-mimetic Ser-175-to-Asp (S175D) mutant A subunit. We conclude that AURKA activates the V-ATPase in kidney carcinoma cells via phosphorylation of Ser-175 in the V-ATPase A subunit. This regulation contributes to kidney carcinoma V-ATPase-mediated extracellular acidification and cell migration.


Subject(s)
Aurora Kinase A/metabolism , Carcinoma/metabolism , Kidney Neoplasms/metabolism , Kidney/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Anacardic Acids/pharmacology , Carcinoma/pathology , Cell Line, Tumor , Humans , Kidney/drug effects , Kidney/pathology , Kidney Neoplasms/pathology , Phosphorylation/drug effects
8.
Am J Physiol Renal Physiol ; 310(6): F569-79, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26739894

ABSTRACT

The hypoxia-inducible factor (HIF)-1 and ß-catenin protective pathways represent the two most significant cellular responses that are activated in response to acute kidney injury. We previously reported that murine mucin (Muc)1 protects kidney function and morphology in a mouse model of ischemia-reperfusion injury (IRI) by stabilizing HIF-1α, enhancing HIF-1 downstream signaling, and thereby preventing metabolic stress (Pastor-Soler et al. Muc1 is protective during kidney ischemia-reperfusion injury. Am J Physiol Renal Physiol 308: F1452-F1462, 2015). We asked if Muc1 regulates the ß-catenin protective pathway during IRI as 1) ß-catenin nuclear targeting is MUC1 dependent in cultured human cells, 2) ß-catenin is found in coimmunoprecipitates with human MUC1 in extracts of both cultured cells and tissues, and 3) MUC1 prevents ß-catenin phosphorylation by glycogen synthase kinase (GSK)3ß and thereby ß-catenin degradation. Using the same mouse model of IRI, we found that levels of active GSK3ß were significantly lower in kidneys of control mice compared with Muc1 knockout (KO) mice. Consequently, ß-catenin was significantly upregulated at 24 and 72 h of recovery and appeared in the nuclear fraction at 72 h in control mouse kidneys. Both ß-catenin induction and nuclear targeting were absent in Muc1 KO mice. We also found downstream induction of ß-catenin prosurvival factors (activated Akt, survivin, transcription factor T cell factor 4 (TCF4), and its downstream target cyclin D1) and repression of proapoptotic factors (p53, active Bax, and cleaved caspase-3) in control mouse kidneys that were absent or aberrant in kidneys of Muc1 KO mice. Altogether, the data clearly indicate that Muc1 protection during acute kidney injury proceeds by enhancing both the HIF-1 and ß-catenin protective pathways.


Subject(s)
Mucin-1/metabolism , Reperfusion Injury/metabolism , beta Catenin/metabolism , Animals , Apoptosis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cyclin D1/metabolism , Hypoxia-Inducible Factor 1/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/metabolism , Survivin , Transcription Factor 4 , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
9.
Am J Physiol Renal Physiol ; 308(12): F1452-62, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25925251

ABSTRACT

Ischemia-reperfusion injury (IRI) due to hypotension is a common cause of human acute kidney injury (AKI). Hypoxia-inducible transcription factors (HIFs) orchestrate a protective response in renal endothelial and epithelial cells in AKI models. As human mucin 1 (MUC1) is induced by hypoxia and enhances HIF-1 activity in cultured epithelial cells, we asked whether mouse mucin 1 (Muc1) regulates HIF-1 activity in kidney tissue during IRI. Whereas Muc1 was localized on the apical surface of the thick ascending limb, distal convoluted tubule, and collecting duct in the kidneys of sham-treated mice, Muc1 appeared in the cytoplasm and nucleus of all tubular epithelia during IRI. Muc1 was induced during IRI, and Muc1 transcripts and protein were also present in recovering proximal tubule cells. Kidney damage was worse and recovery was blocked during IRI in Muc1 knockout mice compared with congenic control mice. Muc1 knockout mice had reduced levels of HIF-1α, reduced or aberrant induction of HIF-1 target genes involved in the shift of glucose metabolism to glycolysis, and prolonged activation of AMP-activated protein kinase, indicating metabolic stress. Muc1 clearly plays a significant role in enhancing the HIF protective pathway during ischemic insult and recovery in kidney epithelia, providing a new target for developing therapies to treat AKI. Moreover, our data support a role specifically for HIF-1 in epithelial protection of the kidney during IRI as Muc1 is present only in tubule epithelial cells.


Subject(s)
Mucin-1/metabolism , Reperfusion Injury/metabolism , Animals , Cell Line , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/physiopathology
10.
Clin J Am Soc Nephrol ; 10(2): 305-24, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25632105

ABSTRACT

Intercalated cells are kidney tubule epithelial cells with important roles in the regulation of acid-base homeostasis. However, in recent years the understanding of the function of the intercalated cell has become greatly enhanced and has shaped a new model for how the distal segments of the kidney tubule integrate salt and water reabsorption, potassium homeostasis, and acid-base status. These cells appear in the late distal convoluted tubule or in the connecting segment, depending on the species. They are most abundant in the collecting duct, where they can be detected all the way from the cortex to the initial part of the inner medulla. Intercalated cells are interspersed among the more numerous segment-specific principal cells. There are three types of intercalated cells, each having distinct structures and expressing different ensembles of transport proteins that translate into very different functions in the processing of the urine. This review includes recent findings on how intercalated cells regulate their intracellular milieu and contribute to acid-base regulation and sodium, chloride, and potassium homeostasis, thus highlighting their potential role as targets for the treatment of hypertension. Their novel regulation by paracrine signals in the collecting duct is also discussed. Finally, this article addresses their role as part of the innate immune system of the kidney tubule.


Subject(s)
Acid-Base Equilibrium , Epithelial Cells/physiology , Kidney Tubules, Collecting/physiology , Acidosis, Renal Tubular/metabolism , Acidosis, Renal Tubular/physiopathology , Animals , Cell Differentiation , Cell Lineage , Epithelial Cells/immunology , Epithelial Cells/metabolism , Humans , Immunity, Innate , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/immunology , Kidney Tubules, Collecting/metabolism , Phenotype , Renin-Angiotensin System
11.
Nat Chem Biol ; 10(9): 707-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25064832

ABSTRACT

The PTH receptor is to our knowledge one of the first G protein-coupled receptor (GPCR) found to sustain cAMP signaling after internalization of the ligand-receptor complex in endosomes. This unexpected model is adding a new dimension on how we think about GPCR signaling, but its mechanism is incompletely understood. We report here that endosomal acidification mediated by the PKA action on the v-ATPase provides a negative feedback mechanism by which endosomal receptor signaling is turned off.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Endosomes/metabolism , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Vacuolar Proton-Translocating ATPases/physiology , Arrestins/chemistry , Arrestins/metabolism , Cholera Toxin/pharmacology , Cyclic AMP/physiology , Feedback, Physiological , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Phosphorylation , Protein Binding , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptor, Parathyroid Hormone, Type 1/physiology , beta-Arrestins
12.
Am J Physiol Renal Physiol ; 306(9): F981-95, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24553431

ABSTRACT

The vacuolar H(+)-ATPase (V-ATPase) mediates ATP-driven H(+) transport across membranes. This pump is present at the apical membrane of kidney proximal tubule cells and intercalated cells. Defects in the V-ATPase and in proximal tubule function can cause renal tubular acidosis. We examined the role of protein kinase A (PKA) and AMP-activated protein kinase (AMPK) in the regulation of the V-ATPase in the proximal tubule as these two kinases coregulate the V-ATPase in the collecting duct. As the proximal tubule V-ATPases have different subunit compositions from other nephron segments, we postulated that V-ATPase regulation in the proximal tubule could differ from other kidney tubule segments. Immunofluorescence labeling of rat ex vivo kidney slices revealed that the V-ATPase was present in the proximal tubule both at the apical pole, colocalizing with the brush-border marker wheat germ agglutinin, and in the cytosol when slices were incubated in buffer alone. When slices were incubated with a cAMP analog and a phosphodiesterase inhibitor, the V-ATPase accumulated at the apical pole of S3 segment cells. These PKA activators also increased V-ATPase apical membrane expression as well as the rate of V-ATPase-dependent extracellular acidification in S3 cell monolayers relative to untreated cells. However, the AMPK activator AICAR decreased PKA-induced V-ATPase apical accumulation in proximal tubules of kidney slices and decreased V-ATPase activity in S3 cell monolayers. Our results suggest that in proximal tubule the V-ATPase subcellular localization and activity are acutely coregulated via PKA downstream of hormonal signals and via AMPK downstream of metabolic stress.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Kidney Tubules, Proximal/enzymology , Vacuolar Proton-Translocating ATPases/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cytosol/enzymology , Enzyme Activation , Enzyme Activators/pharmacology , Hydrogen-Ion Concentration , Isoenzymes , Kidney Tubules, Proximal/drug effects , Mice , Microvilli/enzymology , Phosphodiesterase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Transport , Rats , Rats, Sprague-Dawley , Signal Transduction
13.
Am J Physiol Renal Physiol ; 305(7): F943-56, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23863464

ABSTRACT

The vacuolar H(+)-ATPase (V-ATPase) in intercalated cells contributes to luminal acidification in the kidney collecting duct and nonvolatile acid excretion. We previously showed that the A subunit in the cytoplasmic V1 sector of the V-ATPase (ATP6V1A) is phosphorylated by the metabolic sensor AMP-activated protein kinase (AMPK) in vitro and in kidney cells. Here, we demonstrate that treatment of rabbit isolated, perfused collecting ducts with the AMPK activator 5-aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside (AICAR) inhibited V-ATPase-dependent H(+) secretion from intercalated cells after an acid load. We have identified by mass spectrometry that Ser-384 is a major AMPK phosphorylation site in the V-ATPase A subunit, a result confirmed by comparing AMPK-dependent phosphate labeling of wild-type A-subunit (WT-A) with that of a Ser-384-to-Ala A subunit mutant (S384A-A) in vitro and in intact HEK-293 cells. Compared with WT-A-expressing HEK-293 cells, S384A-A-expressing cells exhibited greater steady-state acidification of HCO3(-)-containing media. Moreover, AICAR treatment of clone C rabbit intercalated cells expressing the WT-A subunit reduced V-ATPase-dependent extracellular acidification, an effect that was blocked in cells expressing the phosphorylation-deficient S384A-A mutant. Finally, expression of the S384A-A mutant prevented cytoplasmic redistribution of the V-ATPase by AICAR in clone C cells. In summary, direct phosphorylation of the A subunit at Ser-384 by AMPK represents a novel regulatory mechanism of the V-ATPase in kidney intercalated cells. Regulation of the V-ATPase by AMPK may couple V-ATPase activity to cellular metabolic status with potential relevance to ischemic injury in the kidney and other tissues.


Subject(s)
AMP-Activated Protein Kinases/physiology , Kidney Tubules, Collecting/enzymology , Vacuolar Proton-Translocating ATPases/metabolism , Acid-Base Equilibrium , Animals , Cytosol/enzymology , Female , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Mass Spectrometry , Phosphorylation , Rabbits
14.
Am J Physiol Renal Physiol ; 301(3): F622-33, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21677144

ABSTRACT

Galectins (Gal) are ß-galactoside-binding proteins that function in epithelial development and homeostasis. An overlapping role for Gal-3 and Gal-7 in wound repair was reported in stratified epithelia. Although Gal-7 was thought absent in simple epithelia, it was reported in a proteomic analysis of cilia isolated from cultured human airway, and we recently identified Gal-7 transcripts in Madin-Darby canine kidney (MDCK) cells (Poland PA, Rondanino C, Kinlough CL, Heimburg-Molinaro J, Arthur CM, Stowell SR, Smith DF, Hughey RP. J Biol Chem 286: 6780-6790, 2011). We now report that Gal-7 is localized exclusively on the primary cilium of MDCK, LLC-PK(1) (pig kidney), and mpkCCD(c14) (mouse kidney) cells as well as on cilia in the rat renal proximal tubule. Gal-7 is also present on most cilia of multiciliated cells in human airway epithelia primary cultures. Interestingly, exogenous glutathione S-transferase (GST)-Gal-7 bound the MDCK apical plasma membrane as well as the cilium, while the lectin Ulex europeaus agglutinin, with glycan preferences similar to Gal-7, bound the basolateral plasma membrane as well as the cilium. In pull-down assays, ß1-integrin isolated from either the basolateral or apical/cilia membranes of MDCK cells was similarly bound by GST-Gal-7. Selective localization of Gal-7 to cilia despite the presence of binding sites on all cell surfaces suggests that intracellular Gal-7 is specifically delivered to cilia rather than simply binding to surface glycoconjugates after generalized secretion. Moreover, depletion of Gal-7 using tetracycline-induced short-hairpin RNA in mpkCCD(c14) cells significantly reduced cilia length and slowed wound healing in a scratch assay. We conclude that Gal-7 is selectively targeted to cilia and plays a key role in surface stabilization of glycoconjugates responsible for integrating cilia function with epithelial repair.


Subject(s)
Cilia/physiology , Cilia/ultrastructure , Epithelial Cells/physiology , Galectins/physiology , Kidney/physiology , Wound Healing/physiology , Animals , Cell Membrane/physiology , Cells, Cultured , Dogs , Epithelial Cells/cytology , Epithelial Cells/ultrastructure , Galectins/genetics , Humans , Integrin beta1/physiology , Kidney/cytology , Kidney/ultrastructure , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/physiology , Kidney Tubules, Proximal/ultrastructure , Mice , Mice, Knockout , Protein Binding/physiology , Rats , Swine
15.
Biopharm Drug Dispos ; 31(8-9): 506-15, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21104926

ABSTRACT

P-glycoprotein is an efflux pump belonging to the ATP-binding cassette super-family that influences the bioavailability and disposition of many drugs. Mammary epithelial cells express various drug transporters including P-glycoprotein, albeit at low level during lactation. During inflammatory reactions, which can be associated with changes in epithelial barrier functions, pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α) are elevated in milk and serum. In this study, the role of TNF-α in the regulation of P-glycoprotein was determined in cultured BME-UV cells, an immortalized bovine mammary epithelial cell line. The protein production of P-glycoprotein and mRNA expression of bABCB1, the gene encoding P-glycoprotein, were increased after 24 h of TNF-α exposure. The highest observed effects for TNF-α on the regulation of P-glycoprotein was after 72 h of exposure. Protein and mRNA expression also increased significantly after 120 h of TNF-α exposure, but was lower than the level observed in the cells exposed to TNF-α for 72 h. The apical to basolateral flux of digoxin, a P-glycoprotein substrate, was decreased in the TNF-α-exposed epithelium. This effect was reversed when verapamil or ketoconazole, compounds known to interact with P-glycoprotein, were added together with digoxin into the donor compartment. Probenecid, a compound known to interact with organic anion transporters, but not P-glycoprotein, did not increase the flux of digoxin. This model has important implications for understanding the barrier function of the mammary epithelium and provides insight into the role of P-glycoprotein in the accumulation and/or removal of xenobiotics from milk and/or plasma.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Cytokines/metabolism , Mammary Glands, Animal/metabolism , Tumor Necrosis Factor-alpha/metabolism , 14-alpha Demethylase Inhibitors/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Animals , Biological Transport/drug effects , Calcium Channel Blockers/pharmacology , Cattle , Cell Line, Transformed , Cell Membrane Permeability/drug effects , Digoxin/metabolism , Enzyme Inhibitors/metabolism , Epithelial Cells/metabolism , Female , Inflammation/metabolism , Ketoconazole/pharmacology , RNA, Messenger/metabolism , Verapamil/pharmacology
16.
J Proteome Res ; 6(5): 1822-32, 2007 May.
Article in English | MEDLINE | ID: mdl-17432893

ABSTRACT

Glycomic profiles derived from human blood sera of 10 healthy males were compared to those from 24 prostate cancer patients. The profiles were acquired using MALDI-MS of permethylated N-glycans released from 10-microL sample aliquots. Quantitative permethylation was attained using solid-phase permethylation. Principal component analysis of the glycomic profiles revealed significant differences among the two sets, allowing their distinct clustering. The first principal component distinguished the 24 prostate cancer patients from the healthy individuals. It was determined that fucosylation of glycan structures is generally higher in cancer samples (ANOVA test p-value of 0.0006). Although more than 50 N-glycan structures were determined, 12 glycan structures, of which six were fucosylated, were significantly different between the two sample sets. Significant differences were confirmed through two independent statistical tests (ANOVA and ROC analyses). Ten of these structures had significantly higher relative intensities in the case of the cancer samples, while the other two were less abundant in the cancer samples. All 12 structures were statistically significant, as suggested by their very low ANOVA scores (<0.001) and ROC analysis, with area under the curve values close to 1 or 0. Accordingly, these structures can be considered as cancer-specific glycans and potential prostate cancer biomarkers. Therefore, serum glycomic profiling appears worthy of further investigation to define its role in cancer early detection and prognostication.


Subject(s)
Blood Proteins/chemistry , Glycoproteins/chemistry , Polysaccharides/analysis , Prostatic Neoplasms , Area Under Curve , Blood Proteins/metabolism , Carbohydrate Conformation , Carbohydrate Sequence , Glycoproteins/metabolism , Humans , Male , Molecular Sequence Data , Neoplasm Metastasis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Array Analysis , ROC Curve , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL