Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Am J Physiol Renal Physiol ; 324(2): F152-F167, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36454701

ABSTRACT

Vasopressin (VP)-regulated aquaporin-2 (AQP2) trafficking between cytoplasmic vesicles and the plasma membrane of kidney principal cells is essential for water homeostasis. VP affects AQP2 phosphorylation at several serine residues in the COOH-terminus; among them, serine 256 (S256) appears to be a major regulator of AQP2 trafficking. Mutation of this serine to aspartic acid, which mimics phosphorylation, induces constitutive membrane expression of AQP2. However, the intracellular location(s) at which S256 phosphorylation occurs remains elusive. Here, we used strategies to block AQP2 trafficking at different cellular locations in LLC-PK1 cells and monitored VP-stimulated phosphorylation of S256 at these sites by immunofluorescence and Western blot analysis with phospho-specific antibodies. Using methyl-ß-cyclodextrin, cold block or bafilomycin, and taxol, we blocked AQP2 at the plasma membrane, in the perinuclear trans-Golgi network, and in scattered cytoplasmic vesicles, respectively. Regardless of its cellular location, VP induced a significant increase in S256 phosphorylation, and this effect was not dependent on a functional microtubule cytoskeleton. To further investigate whether protein kinase A (PKA) was responsible for S256 phosphorylation in these cellular compartments, we created PKA-null cells and blocked AQP2 trafficking using the same procedures. We found that S256 phosphorylation was no longer increased compared with baseline, regardless of AQP2 localization. Taken together, our data indicate that AQP2 S256 phosphorylation can occur at the plasma membrane, in the trans-Golgi network, or in cytoplasmic vesicles and that this event is dependent on the expression of PKA in these cells.NEW & NOTEWORTHY Phosphorylation of aquaporin-2 by PKA at serine 256 (S256) occurs in various subcellular locations during its recycling itinerary, suggesting that the protein complex necessary for AQP2 S256 phosphorylation is present in these different recycling stations. Furthermore, we showed, using PKA-null cells, that PKA activity is required for vasopressin-induced AQP2 phosphorylation. Our data reveal a complex spatial pattern of intracellular AQP2 phosphorylation at S256, shedding new light on the role of phosphorylation in AQP2 membrane accumulation.


Subject(s)
Aquaporin 2 , Serine , Animals , Aquaporin 2/genetics , Aquaporin 2/metabolism , LLC-PK1 Cells , Phosphorylation , Serine/metabolism , Swine , Vasopressins/pharmacology , Vasopressins/metabolism , Intracellular Space/metabolism
2.
Annu Rev Pharmacol Toxicol ; 60: 175-194, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31561739

ABSTRACT

The ability to regulate water movement is vital for the survival of cells and organisms. In addition to passively crossing lipid bilayers by diffusion, water transport is also driven across cell membranes by osmotic gradients through aquaporin water channels. There are 13 aquaporins in human tissues, and of these, aquaporin-2 (AQP2) is the most highly regulated water channel in the kidney: The expression and trafficking of AQP2 respond to body volume status and plasma osmolality via the antidiuretic hormone, vasopressin (VP). Dysfunctional VP signaling in renal epithelial cells contributes to disorders of water balance, and research initially focused on regulating the major cAMP/PKA pathway to normalize urine concentrating ability. With the discovery of novel and more complex signaling networks that regulate AQP2 trafficking, promising therapeutic targets have since been identified. Several strategies based on data from preclinical studies may ultimately translate to the care of patients with defective water homeostasis.


Subject(s)
Kidney/metabolism , Water-Electrolyte Imbalance/physiopathology , Water/metabolism , Animals , Aquaporin 2/metabolism , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Neurophysins/metabolism , Protein Precursors/metabolism , Signal Transduction/physiology , Vasopressins/metabolism , Water-Electrolyte Balance/physiology
3.
Am J Physiol Renal Physiol ; 321(2): F179-F194, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34180716

ABSTRACT

The trafficking of proteins such as aquaporin-2 (AQP2) in the exocytotic pathway requires an active actin cytoskeleton network, but the mechanism is incompletely understood. Here, we show that the actin-related protein (Arp)2/3 complex, a key factor in actin filament branching and polymerization, is involved in the shuttling of AQP2 between the trans-Golgi network (TGN) and the plasma membrane. Arp2/3 inhibition (using CK-666) or siRNA knockdown blocks vasopressin-induced AQP2 membrane accumulation and induces the formation of distinct AQP2 perinuclear patches positive for markers of TGN-derived clathrin-coated vesicles. After a 20°C cold block, AQP2 formed perinuclear patches due to continuous endocytosis coupled with inhibition of exit from TGN-associated vesicles. Upon rewarming, AQP2 normally leaves the TGN and redistributes into the cytoplasm, entering the exocytotic pathway. Inhibition of Arp2/3 blocked this process and trapped AQP2 in clathrin-positive vesicles. Taken together, these results suggest that Arp2/3 is essential for AQP2 trafficking, specifically for its delivery into the post-TGN exocytotic pathway to the plasma membrane.NEW & NOTEWORTHY Aquaporin-2 (AQP2) undergoes constitutive recycling between the cytoplasm and plasma membrane, with an intricate balance between endocytosis and exocytosis. By inhibiting the actin-related protein (Arp)2/3 complex, we prevented AQP2 from entering the exocytotic pathway at the post-trans-Golgi network level and blocked AQP2 membrane accumulation. Arp2/3 inhibition, therefore, enables us to separate and target the exocytotic process, while not affecting endocytosis, thus allowing us to envisage strategies to modulate AQP2 trafficking and treat water balance disorders.


Subject(s)
Actin-Related Protein 2/metabolism , Actin-Related Protein 3/metabolism , Aquaporin 2/metabolism , Exocytosis/physiology , Kidney/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Membrane/metabolism , Endocytosis/physiology , LLC-PK1 Cells , Phosphorylation , Protein Transport/physiology , Rats , Swine
4.
Proc Natl Acad Sci U S A ; 114(45): E9559-E9568, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29078380

ABSTRACT

Alterations in the activity/levels of the extralarge G protein α-subunit (XLαs) are implicated in various human disorders, such as perinatal growth retardation. Encoded by GNAS, XLαs is partly identical to the α-subunit of the stimulatory G protein (Gsα), but the cellular actions of XLαs remain poorly defined. Following an initial proteomic screen, we identified sorting nexin-9 (SNX9) and dynamins, key components of clathrin-mediated endocytosis, as binding partners of XLαs. Overexpression of XLαs in HEK293 cells inhibited internalization of transferrin, a process that depends on clathrin-mediated endocytosis, while its ablation by CRISPR/Cas9 in an osteocyte-like cell line (Ocy454) enhanced it. Similarly, primary cardiomyocytes derived from XLαs knockout (XLKO) pups showed enhanced transferrin internalization. Early postnatal XLKO mice showed a significantly higher degree of cardiac iron uptake than wild-type littermates following iron dextran injection. In XLKO neonates, iron and ferritin levels were elevated in heart and skeletal muscle, where XLαs is normally expressed abundantly. XLKO heart and skeletal muscle, as well as XLKO Ocy454 cells, showed elevated SNX9 protein levels, and siRNA-mediated knockdown of SNX9 in XLKO Ocy454 cells prevented enhanced transferrin internalization. In transfected cells, XLαs also inhibited internalization of the parathyroid hormone and type 2 vasopressin receptors. Internalization of transferrin and these G protein-coupled receptors was also inhibited in cells expressing an XLαs mutant missing the Gα portion, but not Gsα or an N-terminally truncated XLαs mutant unable to interact with SNX9 or dynamin. Thus, XLαs restricts clathrin-mediated endocytosis and plays a critical role in iron/transferrin uptake in vivo.


Subject(s)
Clathrin/metabolism , Endocytosis/physiology , GTP-Binding Protein alpha Subunits/metabolism , Iron/metabolism , Animals , CRISPR-Cas Systems/physiology , Cell Line , HEK293 Cells , Heart/physiology , Humans , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Osteocytes/metabolism , Proteomics/methods , Receptors, Vasopressin/metabolism , Sorting Nexins/metabolism , Transferrin/metabolism
5.
J Physiol ; 597(6): 1627-1642, 2019 03.
Article in English | MEDLINE | ID: mdl-30488437

ABSTRACT

KEY POINTS: Aquaporin-2 (AQP2) is crucial for water homeostasis, and vasopressin (VP) induces AQP2 membrane trafficking by increasing intracellular cAMP, activating PKA and causing phosphorylation of AQP2 at serine 256, 264 and 269 residues and dephosphorylation of serine 261 residue on the AQP2 C-terminus. It is thought that serine 256 is the master regulator of AQP2 trafficking, and its phosphorylation has to precede the change of phosphorylation state of other serine residues. We found that Src inhibition causes serine 256-independent AQP2 membrane trafficking and induces phosphorylation of serine 269 independently of serine 256. This targeted phosphorylation of serine 269 is important for Src inhibition-induced AQP2 membrane accumulation; without serine 269, Src inhibition exerts no effect on AQP2 trafficking. This result helps us better understand the independent pathways that can target different AQP2 residues, and design new strategies to induce or sustain AQP2 membrane expression when VP signalling is defective. ABSTRACT: Aquaporin-2 (AQP2) is essential for water homeostasis. Upon stimulation by vasopressin, AQP2 is phosphorylated at serine 256 (S256), S264 and S269, and dephosphorylated at S261. It is thought that S256 is the master regulator of AQP2 trafficking and membrane accumulation, and that its phosphorylation has to precede phosphorylation of other serine residues. In this study, we found that VP reduces Src kinase phosphorylation: by suppressing Src using the inhibitor dasatinib and siRNA, we could increase AQP2 membrane accumulation in cultured AQP2-expressing cells and in kidney collecting duct principal cells. Src inhibition increased exocytosis and inhibited clathrin-mediated endocytosis of AQP2, but exerted its effect in a cAMP, PKA and S256 phosphorylation (pS256)-independent manner. Despite the lack of S256 phosphorylation, dasatinib increased phosphorylation of S269, even in S256A mutant cells in which S256 phosphorylation cannot occur. To confirm the importance of pS269 in AQP2 re-distribution, we expressed an AQP2 S269A mutant in LLC-PK1 cells, and found that dasatinib no longer induced AQP2 membrane accumulation. In conclusion, Src inhibition causes phosphorylation of S269 independently of pS256, and induces AQP2 membrane accumulation by inhibiting clathrin-mediated endocytosis and increasing exocytosis. We conclude that S269 can be phosphorylated without pS256, and pS269 alone is important for AQP2 apical membrane accumulation under some conditions. These data increase our understanding of the independent pathways that can phosphorylate different residues in the AQP2 C-terminus, and suggest new strategies to target distinct AQP2 serine residues to induce membrane expression of this water channel when VP signalling is defective.


Subject(s)
Aquaporin 2/metabolism , Exocytosis , src-Family Kinases/metabolism , Animals , Aquaporin 2/genetics , Cell Line , Dasatinib/pharmacology , Male , Mutation, Missense , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Swine , Vasopressins/metabolism , src-Family Kinases/antagonists & inhibitors
6.
Blood ; 129(4): 405-414, 2017 01 26.
Article in English | MEDLINE | ID: mdl-27864295

ABSTRACT

Bone morphogenetic protein 6 (BMP6) signaling in hepatocytes is a central transcriptional regulator of the iron hormone hepcidin that controls systemic iron balance. How iron levels are sensed to regulate hepcidin production is not known, but local induction of liver BMP6 expression by iron is proposed to have a critical role. To identify the cellular source of BMP6 responsible for hepcidin and iron homeostasis regulation, we generated mice with tissue-specific ablation of Bmp6 in different liver cell populations and evaluated their iron phenotype. Efficiency and specificity of Cre-mediated recombination was assessed by using Cre-reporter mice, polymerase chain reaction of genomic DNA, and quantitation of Bmp6 messenger RNA expression from isolated liver cell populations. Localization of the BMP co-receptor hemojuvelin was visualized by immunofluorescence microscopy. Analysis of the Bmp6 conditional knockout mice revealed that liver endothelial cells (ECs) expressed Bmp6, whereas resident liver macrophages (Kupffer cells) and hepatocytes did not. Loss of Bmp6 in ECs recapitulated the hemochromatosis phenotype of global Bmp6 knockout mice, whereas hepatocyte and macrophage Bmp6 conditional knockout mice exhibited no iron phenotype. Hemojuvelin was localized on the hepatocyte sinusoidal membrane immediately adjacent to Bmp6-producing sinusoidal ECs. Together, these data demonstrate that ECs are the predominant source of BMP6 in the liver and support a model in which EC BMP6 has paracrine actions on hepatocyte hemojuvelin to regulate hepcidin transcription and maintain systemic iron homeostasis.


Subject(s)
Bone Morphogenetic Protein 6/genetics , Endothelial Cells/metabolism , Hemochromatosis/genetics , Hepcidins/genetics , Iron/metabolism , Membrane Proteins/genetics , RNA, Messenger/genetics , Animals , Bone Morphogenetic Protein 6/deficiency , Endothelial Cells/pathology , Female , GPI-Linked Proteins , Gene Expression Regulation , Hemochromatosis Protein , Hepatocytes/metabolism , Hepatocytes/pathology , Hepcidins/metabolism , Homeostasis/genetics , Immunophenotyping , Integrases/genetics , Integrases/metabolism , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver/metabolism , Liver/pathology , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Paracrine Communication , RNA, Messenger/metabolism , Signal Transduction , Transcription, Genetic
7.
Am J Physiol Renal Physiol ; 313(2): F404-F413, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28381458

ABSTRACT

Aquaporin 2 (AQP2) trafficking is regulated by phosphorylation and dephosphorylation of serine residues in the AQP2 COOH terminus. Vasopressin (VP) binding to its receptor (V2R) leads to a cascade of events that result in phosphorylation of serine 256 (S256), S264, and S269, but dephosphorylation of S261. To identify which phosphatase is responsible for VP-induced S261 dephosphorylation, we pretreated cells with different phosphatase inhibitors before VP stimulation. Sanguinarine, a specific protein phosphatase (PP) 2C inhibitor, but not inhibitors of PP1, PP2A (okadaic acid), or PP2B (cyclosporine), abolished VP-induced S261 dephosphorylation. However, sanguinarine and VP significantly increased phosphorylation of ERK, a kinase that can phosphorylate S261; inhibition of ERK by PD98059 partially decreased baseline S261 phosphorylation. These data support a role of ERK in S261 phosphorylation but suggest that, upon VP treatment, increased phosphatase activity overcomes the increase in ERK activity, resulting in overall dephosphorylation of S261. We also found that sanguinarine abolished VP-induced S261 dephosphorylation in cells expressing mutated AQP2 S256A, suggesting that the phosphorylation state of S261 is independent of S256. Sanguinarine alone did not induce AQP2 membrane trafficking, nor did it inhibit VP-induced AQP2 membrane accumulation in cells and kidney tissues, suggesting that S261 does not play an observable role in acute AQP2 membrane accumulation. In conclusion, PP2C activity is required for S261 AQP2 dephosphorylation upon VP stimulation, which occurs independently of S256 phosphorylation. Understanding the pathways involved in modulating PP2C will help elucidate the role of S261 in cellular events involving AQP2.


Subject(s)
Aquaporin 2/metabolism , Kidney/drug effects , Vasopressins/pharmacology , Animals , Aquaporin 2/genetics , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , In Vitro Techniques , Kidney/enzymology , LLC-PK1 Cells , Mutation , Phosphorylation , Protein Phosphatase 2C/antagonists & inhibitors , Protein Phosphatase 2C/metabolism , Protein Transport , Rats, Sprague-Dawley , Receptors, Vasopressin/agonists , Receptors, Vasopressin/metabolism , Serine , Signal Transduction/drug effects , Swine , Transfection , Vasopressins/metabolism
8.
J Am Soc Nephrol ; 27(10): 3105-3116, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27694161

ABSTRACT

Nephrogenic diabetes insipidus (NDI) is caused by impairment of vasopressin (VP) receptor type 2 signaling. Because potential therapies for NDI that target the canonical VP/cAMP/protein kinase A pathway have so far proven ineffective, alternative strategies for modulating aquaporin 2 (AQP2) trafficking have been sought. Successful identification of compounds by our high-throughput chemical screening assay prompted us to determine whether EGF receptor (EGFR) inhibitors stimulate AQP2 trafficking and reduce urine output. Erlotinib, a selective EGFR inhibitor, enhanced AQP2 apical membrane expression in collecting duct principal cells and reduced urine volume by 45% after 5 days of treatment in mice with lithium-induced NDI. Similar to VP, erlotinib increased exocytosis and decreased endocytosis in LLC-PK1 cells, resulting in a significant increase in AQP2 membrane accumulation. Erlotinib increased phosphorylation of AQP2 at Ser-256 and Ser-269 and decreased phosphorylation at Ser-261 in a dose-dependent manner. However, unlike VP, the effect of erlotinib was independent of cAMP, cGMP, and protein kinase A. Conversely, EGF reduced VP-induced AQP2 Ser-256 phosphorylation, suggesting crosstalk between VP and EGF in AQP2 trafficking and a role of EGF in water homeostasis. These results reveal a novel pathway that contributes to the regulation of AQP2-mediated water reabsorption and suggest new potential therapeutic strategies for NDI treatment.


Subject(s)
Aquaporin 2/drug effects , Aquaporin 2/physiology , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/pharmacology , Kidney/metabolism , Water/metabolism , Animals , Cell Membrane/metabolism , Mice
9.
Am J Physiol Cell Physiol ; 307(7): C597-605, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24944200

ABSTRACT

A reduction or loss of plasma membrane aquaporin 2 (AQP2) in kidney principal cells due to defective vasopressin (VP) signaling through the VP receptor causes excessive urine production, i.e., diabetes insipidus. The amount of AQP2 on the plasma membrane is regulated by a balance of exocytosis and endocytosis and is the rate limiting step for water reabsorption in the collecting duct. We describe here a systematic approach using high-throughput screening (HTS) followed by in vitro and in vivo assays to discover novel compounds that enhance vasopressin-independent AQP2 membrane expression. We performed initial chemical library screening with a high-throughput exocytosis fluorescence assay using LLC-PK1 cells expressing soluble secreted yellow fluorescent protein and AQP2. Thirty-six candidate exocytosis enhancers were identified. These compounds were then rescreened in AQP2-expressing cells to determine their ability to increase AQP2 membrane accumulation. Effective drugs were then applied to kidney slices in vitro. Three compounds, AG-490, ß-lapachone, and HA14-1 increased AQP2 membrane accumulation in LLC-PK1 cells, and both AG-490 and ß-lapachone were also effective in MDCK cells and principal cells in rat kidney slices. Finally, one compound, AG-490 (an EGF receptor and JAK-2 kinase inhibitor), decreased urine volume and increased urine osmolality significantly in the first 2-4 h after a single injection into VP-deficient Brattleboro rats. In conclusion, we have developed a systematic procedure for identifying new compounds that modulate AQP2 trafficking using initial HTS followed by in vitro assays in cells and kidney slices, and concluding with in vivo testing in an animal model.


Subject(s)
Aquaporin 2/drug effects , Cell Membrane/drug effects , High-Throughput Screening Assays , Kidney Concentrating Ability/drug effects , Kidney/drug effects , Tyrphostins/pharmacology , Urological Agents/pharmacology , Animals , Aquaporin 2/genetics , Aquaporin 2/metabolism , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Cell Membrane/metabolism , Dogs , Exocytosis/drug effects , In Vitro Techniques , Kidney/metabolism , LLC-PK1 Cells , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Madin Darby Canine Kidney Cells , Microscopy, Fluorescence , Osmolar Concentration , Protein Transport , Rats, Brattleboro , Rats, Sprague-Dawley , Reproducibility of Results , Swine , Time Factors , Transfection , Up-Regulation
10.
J Biol Chem ; 288(39): 27849-60, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23935101

ABSTRACT

The vasopressin type 2 receptor (V2R) is a critical G protein-coupled receptor (GPCR) for vertebrate physiology, including the balance of water and sodium ions. It is unclear how its two native hormones, vasopressin (VP) and oxytocin (OT), both stimulate the same cAMP/PKA pathway yet produce divergent antinatriuretic and antidiuretic effects that are either strong (VP) or weak (OT). Here, we present a new mechanism that differentiates the action of VP and OT on V2R signaling. We found that vasopressin, as opposed to OT, continued to generate cAMP and promote PKA activation for prolonged periods after ligand washout and receptor internalization in endosomes. Contrary to the classical model of arrestin-mediated GPCR desensitization, arrestins bind the VP-V2R complex yet extend rather than shorten the generation of cAMP. Signaling is instead turned off by the endosomal retromer complex. We propose that this mechanism explains how VP sustains water and Na(+) transport in renal collecting duct cells. Together with recent work on the parathyroid hormone receptor, these data support the existence of a novel "noncanonical" regulatory pathway for GPCR activation and response termination, via the sequential action of ß-arrestin and the retromer complex.


Subject(s)
Arrestins/metabolism , Gene Expression Regulation , Receptors, Vasopressin/metabolism , Signal Transduction , Animals , Antidiuretic Agents/pharmacology , Aquaporin 2/metabolism , Cell Membrane/metabolism , Cyclic AMP/metabolism , Dogs , Endosomes/metabolism , HEK293 Cells , Humans , Kidney/metabolism , Ligands , Madin Darby Canine Kidney Cells , Oxytocin/chemistry , Phosphorylation , Receptors, G-Protein-Coupled/metabolism , Sodium/metabolism , beta-Arrestins
11.
Am J Physiol Cell Physiol ; 304(1): C38-48, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23015545

ABSTRACT

The aquaporin-2 (AQP2) water channel relocates mainly to the apical plasma membrane of collecting duct principal cells after vasopressin (VP) stimulation. AQP2 transport to this membrane domain is assumed to be a direct route involving recycling of intracellular vesicles. However, basolateral plasma membrane expression of AQP2 is observed in vivo in principal cells. Here, we asked whether there is a transcytotic pathway of AQP2 trafficking between apical and basolateral membranes. We used MDCK cells in which AQP2 normally accumulates apically after VP exposure. In contrast, both site-specific biotinylation and immunofluorescence showed that AQP2 is strongly accumulated in the basolateral membrane, along with the endocytic protein clathrin, after a brief cold shock (4°C). This suggests that AQP2 may be constitutively targeted to basolateral membranes and then retrieved by clathrin-mediated endocytosis at physiological temperatures. Rab11 does not accumulate in basolateral membranes after cold shock, suggesting that the AQP2 in this location is not associated with Rab11-positive vesicles. After rewarming (37°C), basolateral AQP2 staining is diminished and it subsequently accumulates at the apical membrane in the presence of VP/forskolin, suggesting that transcytosis can be followed by apical insertion of AQP2. This process is inhibited by treatment with colchicine. Our data suggest that the cold shock procedure reveals the presence of microtubule-dependent AQP2 transcytosis, which represents an indirect pathway of apical AQP2 delivery in these cells. Furthermore, our data indicate that protein polarity data obtained from biotinylation assays, which require cells to be cooled to 4°C during the labeling procedure, should be interpreted with caution.


Subject(s)
Aquaporin 2/metabolism , Microtubules/metabolism , Animals , Aquaporin 2/genetics , Cell Polarity/physiology , Dogs , Madin Darby Canine Kidney Cells , Microtubules/drug effects , Protein Transport/drug effects , Protein Transport/physiology , Rats
12.
Am J Physiol Renal Physiol ; 304(5): F553-64, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23235478

ABSTRACT

Vasopressin (VP) binds to the vasopressin type 2 receptor (V2R) to trigger physiological effects including body fluid homeostasis and blood pressure regulation. Signaling is terminated by receptor downregulation involving clathrin-mediated endocytosis and V2R degradation. We report here that both native and epitope-tagged V2R are internalized from the plasma membrane of LLC-PK1 kidney epithelial cells in the presence of another ligand, transferrin (Tf). The presence of iron-saturated Tf (holo-Tf; 4 h) reduced V2R binding sites at the cell surface by up to 33% while iron-free (apo-Tf) had no effect. However, no change in green fluorescent protein-tagged V2R distribution was observed in the presence of bovine serum albumin, atrial natriuretic peptide, or ANG II. Conversely, holo-Tf did not induce the internalization of another G protein-coupled receptor, the parathyroid hormone receptor. In contrast to the effect of VP, Tf did not increase intracellular cAMP or modify aquaporin-2 distribution in these cells, although addition of VP and Tf together augmented VP-induced V2R internalization. Tf receptor coimmunoprecipitated with V2R, suggesting that they interact closely, which may explain the additive effect of VP and Tf on V2R endocytosis. Furthermore, Tf-induced V2R internalization was abolished in cells expressing a dominant negative dynamin (K44A) mutant, indicating the involvement of clathrin-coated pits. We conclude that Tf can induce heterologous downregulation of the V2R and this might desensitize VP target cells without activating downstream V2R signaling events. It also provides new insights into urine-concentrating defects observed in rat models of hemochromatosis.


Subject(s)
Down-Regulation/drug effects , Epithelial Cells/drug effects , Kidney/drug effects , Receptors, Vasopressin/metabolism , Transferrin/pharmacology , Vasopressins/pharmacology , Animals , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Membrane/metabolism , Endocytosis/drug effects , Endocytosis/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Kidney/cytology , Kidney/metabolism , Receptors, Vasopressin/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Swine
13.
Am J Physiol Cell Physiol ; 302(10): C1421-33, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22460710

ABSTRACT

Maintaining tight control over body fluid and acid-base homeostasis is essential for human health and is a major function of the kidney. The collecting duct is a mosaic of two cell populations that are highly specialized to perform these two distinct processes. The antidiuretic hormone vasopressin (VP) and its receptor, the V2R, play a central role in regulating the urinary concentrating mechanism by stimulating accumulation of the aquaporin 2 (AQP2) water channel in the apical membrane of collecting duct principal cells. This increases epithelial water permeability and allows osmotic water reabsorption to occur. An understanding of the basic cell biology/physiology of AQP2 regulation and trafficking has informed the development of new potential treatments for diseases such as nephrogenic diabetes insipidus, in which the VP/V2R/AQP2 signaling axis is defective. Tubule acidification due to the activation of intercalated cells is also critical to organ function, and defects lead to several pathological conditions in humans. Therefore, it is important to understand how these "professional" proton-secreting cells respond to environmental and cellular cues. Using epididymal proton-secreting cells as a model system, we identified the soluble adenylate cyclase (sAC) as a sensor that detects luminal bicarbonate and activates the vacuolar proton-pumping ATPase (V-ATPase) via cAMP to regulate tubular pH. Renal intercalated cells also express sAC and respond to cAMP by increasing proton secretion, supporting the hypothesis that sAC could function as a luminal sensor in renal tubules to regulate acid-base balance. This review summarizes recent advances in our understanding of these fundamental processes.


Subject(s)
Acid-Base Equilibrium/physiology , Epithelial Cells/physiology , Kidney/cytology , Kidney/physiology , Water-Electrolyte Balance/physiology , Animals , Homeostasis/physiology , Humans , Water/physiology
14.
Kidney Int ; 82(1): 7-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22699378

ABSTRACT

The protease corin generates atrial natriuretic peptide and affects blood pressure and salt-water homeostasis. Under dietary salt challenge, corin knockout mice show blood pressure exacerbation and significant weight gain due to water and salt retention. This phenotype involves the epithelial sodium channel but is independent of the renin-angiotensin-aldosterone system. This suggests that corin has an important role in a new adaptive mechanism of the response to variations of salt in the diet.


Subject(s)
Blood Pressure , Hypertension/enzymology , Kidney/enzymology , Natriuresis , Serine Endopeptidases/deficiency , Animals , Male
15.
J Am Soc Nephrol ; 22(1): 59-72, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21071524

ABSTRACT

The most common cause of hereditary nephrogenic diabetes insipidus is a nonfunctional vasopressin (VP) receptor type 2 (V2R). Calcitonin, another ligand of G-protein-coupled receptors, has a VP-like effect on electrolytes and water reabsorption, suggesting that it may affect AQP2 trafficking. Here, calcitonin increased intracellular cAMP and stimulated the membrane accumulation of AQP2 in LLC-PK1 cells. Pharmacologic inhibition of protein kinase A (PKA) and deficiency of a critical PKA phosphorylation site on AQP2 both prevented calcitonin-induced membrane accumulation of AQP2. Fluorescence assays showed that calcitonin led to a 70% increase in exocytosis and a 20% decrease in endocytosis of AQP2. Immunostaining of rat kidney slices demonstrated that calcitonin induced a significant redistribution of AQP2 to the apical membrane of principal cells in cortical collecting ducts and connecting segments but not in the inner stripe or inner medulla. Calcitonin-treated VP-deficient Brattleboro rats had a reduced urine flow and two-fold higher urine osmolality during the first 12 hours of treatment compared with control groups. Although this VP-like effect of calcitonin diminished over the following 72 hours, the tachyphylaxis was reversible. Taken together, these data show that calcitonin induces cAMP-dependent AQP2 trafficking in cortical collecting and connecting tubules in parallel with an increase in urine concentration. This suggests that calcitonin has a potential therapeutic use in nephrogenic diabetes insipidus.


Subject(s)
Aquaporin 2/metabolism , Calcitonin/pharmacology , Kidney/drug effects , Kidney/metabolism , Urine/physiology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclic AMP/metabolism , Endocytosis/drug effects , Endocytosis/physiology , Exocytosis/drug effects , Exocytosis/physiology , Kidney/cytology , LLC-PK1 Cells , Male , Models, Animal , Osmolar Concentration , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Brattleboro , Swine , Vasopressins/pharmacology
16.
Nat Commun ; 13(1): 2422, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35504916

ABSTRACT

Chronic kidney diseases and acute kidney injury are mechanistically distinct kidney diseases. While chronic kidney diseases are associated with podocyte injury, acute kidney injury affects renal tubular epithelial cells. Despite these differences, a cardinal feature of both acute and chronic kidney diseases is dysregulated actin cytoskeleton. We have shown that pharmacological activation of GTPase dynamin ameliorates podocyte injury in murine models of chronic kidney diseases by promoting actin polymerization. Here we establish dynamin's role in modulating stiffness and polarity of renal tubular epithelial cells by crosslinking actin filaments into branched networks. Activation of dynamin's crosslinking capability by a small molecule agonist stabilizes the actomyosin cortex of the apical membrane against injury, which in turn preserves renal function in various murine models of acute kidney injury. Notably, a dynamin agonist simultaneously attenuates podocyte and tubular injury in the genetic murine model of Alport syndrome. Our study provides evidence for the feasibility and highlights the benefits of novel holistic nephron-protective therapies.


Subject(s)
Acute Kidney Injury , Podocytes , Renal Insufficiency, Chronic , Actin Cytoskeleton , Acute Kidney Injury/prevention & control , Animals , Dynamins , Female , Humans , Kidney/physiology , Male , Mice , Renal Insufficiency, Chronic/drug therapy
17.
Am J Physiol Renal Physiol ; 301(2): F309-18, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21511701

ABSTRACT

Statins are 3-hydroxyl-3-methyglutaryl-CoA reductase inhibitors that are commonly used to inhibit cholesterol biosynthesis. Emerging data have suggested that they also have "pleotropic effects," including modulating actin cytoskeleton reorganization. Here, we report an effect of simvastatin on the trafficking of aquaporin-2 (AQP2). Specifically, simvastatin induced the membrane accumulation of AQP2 in cell cultures and kidneys in situ. The effect of simvastatin was independent of protein kinase A activation and phosphorylation at AQP2-Ser(256), a critical event involved in vasopressin (VP)-regulated AQP2 trafficking. Further investigation showed that simvastatin inhibited endocytosis in parallel with downregulation of RhoA activity. Overexpression of active RhoA attenuated simvastatin's effect, suggesting the involvement of this small GTPase in simvastatin-mediated AQP2 trafficking. Finally, the effect of simvastatin on urinary concentration was investigated in VP-deficient Brattleboro rats. Simvastatin acutely (3-6 h) increased urinary concentration and decreased urine output in these animals. In summary, simvastatin regulates AQP2 trafficking in vitro and urinary concentration in vivo via events involving downregulation of Rho GTPase activity and inhibition of endocytosis. Our study provides an alternative mechanism to regulate AQP2 trafficking, bypassing the VP-vasopressin receptor signaling pathway.


Subject(s)
Aquaporin 2/urine , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kidney Tubules, Collecting/drug effects , Simvastatin/pharmacology , rho GTP-Binding Proteins/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation , Endocytosis/drug effects , Exocytosis/drug effects , In Vitro Techniques , Kidney Tubules, Collecting/metabolism , Phosphorylation/drug effects , Rats , Rats, Brattleboro
18.
Nat Chem Biol ; 5(10): 734-42, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19701185

ABSTRACT

Cell signaling mediated by the G protein-coupled parathyroid hormone receptor type 1 (PTHR) is fundamental to bone and kidney physiology. It has been unclear how the two ligand systems--PTH, endocrine and homeostatic, and PTH-related peptide (PTHrP), paracrine--can effectively operate with only one receptor and trigger different durations of the cAMP responses. Here we analyze the ligand response by measuring the kinetics of activation and deactivation for each individual reaction step along the PTHR signaling cascade. We found that during the time frame of G protein coupling and cAMP production, PTHrP(1-36) action was restricted to the cell surface, whereas PTH(1-34) had moved to internalized compartments where it remained associated with the PTHR and Galpha(s), potentially as a persistent and active ternary complex. Such marked differences suggest a mechanism by which PTH and PTHrP induce differential responses, and these results indicate that the central tenet that cAMP production originates exclusively at the cell membrane must be revised.


Subject(s)
Cyclic AMP/biosynthesis , Endocytosis/physiology , Receptor, Parathyroid Hormone, Type 1/physiology , Signal Transduction/physiology , Animals , Bone Resorption/metabolism , Cell Line , Cell Membrane/metabolism , Fluorescence Resonance Energy Transfer , GTP-Binding Proteins/metabolism , Humans , Kinetics , Ligands , Mice , Microscopy, Confocal , Osteoblasts/metabolism , Parathyroid Hormone/metabolism , Protein Conformation , Protein Transport , Receptor, Parathyroid Hormone, Type 1/agonists , Receptor, Parathyroid Hormone, Type 1/metabolism
19.
J Am Soc Nephrol ; 21(4): 666-77, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20167703

ABSTRACT

The neuronal adhesion protein Dragon acts as a bone morphogenetic protein (BMP) coreceptor that enhances BMP signaling. Given the importance of BMP signaling in nephrogenesis and its putative role in the response to injury in the adult kidney, we studied the localization and function of Dragon in the kidney. We observed that Dragon localized predominantly to the apical surfaces of tubular epithelial cells in the thick ascending limbs, distal convoluted tubules, and collecting ducts of mice. Dragon expression was weak in the proximal tubules and glomeruli. In mouse inner medullary collecting duct (mIMCD3) cells, Dragon generated BMP signals in a ligand-dependent manner, and BMP4 is the predominant endogenous ligand for the Dragon coreceptor. In mIMCD3 cells, BMP4 normally signaled through BMPRII, but Dragon enhanced its signaling through the BMP type II receptor ActRIIA. Dragon and BMP4 increased transepithelial resistance (TER) through the Smad1/5/8 pathway. In epithelial cells isolated from the proximal tubule and intercalated cells of collecting ducts, we observed coexpression of ActRIIA, Dragon, and BMP4 but not BMPRII. Taken together, these results suggest that Dragon may enhance BMP signaling in renal tubular epithelial cells and maintain normal renal physiology.


Subject(s)
Bone Morphogenetic Proteins/physiology , Epithelial Cells/physiology , Nerve Tissue Proteins/physiology , Neural Cell Adhesion Molecules/physiology , Animals , Cells, Cultured , Kidney , Mice , Signal Transduction , Urothelium/cytology
20.
Cells ; 9(4)2020 04 23.
Article in English | MEDLINE | ID: mdl-32340337

ABSTRACT

We previously showed that in polarized Madin-Darby canine kidney (MDCK) cells, aquaporin-2 (AQP2) is continuously targeted to the basolateral plasma membrane from which it is rapidly retrieved by clathrin-mediated endocytosis. It then undertakes microtubule-dependent transcytosis toward the apical plasma membrane. In this study, we found that treatment with chlorpromazine (CPZ, an inhibitor of clathrin-mediated endocytosis) results in AQP2 accumulation in the basolateral, but not the apical plasma membrane of epithelial cells. In MDCK cells, both AQP2 and clathrin were concentrated in the basolateral plasma membrane after CPZ treatment (100 µM for 15 min), and endocytosis was reduced. Then, using rhodamine phalloidin staining, we found that basolateral, but not apical, F-actin was selectively reduced by CPZ treatment. After incubation of rat kidney slices in situ with CPZ (200 µM for 15 min), basolateral AQP2 and clathrin were increased in principal cells, which simultaneously showed a significant decrease of basolateral compared to apical F-actin staining. These results indicate that clathrin-dependent transcytosis of AQP2 is an essential part of its trafficking pathway in renal epithelial cells and that this process can be inhibited by selectively depolymerizing the basolateral actin pool using CPZ.


Subject(s)
Actins/metabolism , Aquaporin 2/metabolism , Chlorpromazine/pharmacology , Endocytosis , Epithelial Cells/metabolism , Kidney/cytology , Polymerization , Animals , Clathrin/metabolism , Cold-Shock Response , Colforsin/pharmacology , Dogs , Endocytosis/drug effects , Epithelial Cells/drug effects , Madin Darby Canine Kidney Cells , Phosphorylation/drug effects , Protein Transport/drug effects , Rats , Transferrin/metabolism , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL