1
|
D'Agostino C, Zindy E, Conrard L, Takkal A, Gregoire F, Bolaky N, Törnroth-Horsefield S, Perret J, Delporte C. AQP5 trafficking is regulated by its C-terminal tail and interaction with prolactin-inducible protein. Biol Direct 2025; 20:53. [PMID: 40241160 PMCID: PMC12001509 DOI: 10.1186/s13062-025-00647-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Aquaporin-5 (AQP5) is a crucial membrane protein involved in water transport across cellular membranes, particularly within exocrine glands such as salivary glands. Dysregulation of AQP5, including its mislocalization, has been associated with various diseases, emphasizing the need to understand the molecular mechanisms governing its trafficking. This study investigates the multifaceted regulatory mechanisms of AQP5 trafficking, with specific emphasis on the role of the carboxyl-terminal (C-terminal) tail and the functional involvement of prolactin-inducible protein (PIP) as an interacting protein partner. METHODS An innovative 2D-custom model employing SNAP-tag human AQP5 constructs together with a novel automated algorithm-based methodology was used following immunofluorescence and confocal microscopy to assess hAQP5 localization to the plasma membrane of stably transfected normal salivary gland-SV40 transformed-acinar cells (NS-SV-AC). The expression of the constructs was verified by Western blot analysis. RESULTS The expression of SNAP-hAQP5 constructs expressed in stably transfected NS-SV-AC cells allowed to explore the involvement of hAQP5 C-terminal tail and the hAQP5-hPIP interaction in hAQP5 trafficking upon stimulation. The use of C-terminal truncation constructs revealed distinct responses to intracellular 3',5'-cyclic adenosine monophosphate (cAMP) and calcium increase, shedding light on the importance of specific regions within the highly flexible distal part of the C-terminal tail for AQP5 trafficking. Furthermore, our investigation of the interplay between hAQP5 and hPIP revealed that PIP promotes AQP5 translocation to the plasma membrane, blunting the effects of calcium- and cAMP-dependent pathways on AQP5 sub-cellular localization. CONCLUSION In summary, this study advances our understanding of AQP5 trafficking dynamics and provides critical insights into the regulatory roles of the C-terminal tail and its interaction with PIP. The innovative methodology to assess AQP5 translocation to the plasma membrane sets the stage for future investigations to identify the role of individual amino acids and phosphorylation sites within the distal AQP5 C-terminus in the trafficking mechanism and protein-protein interaction, and to explore the dynamic of the process by high resolution live cell imaging. Further research in this area is warranted to uncover critical insights into the regulation of AQP5, offering opportunities for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Claudia D'Agostino
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| | - Egor Zindy
- Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles, Gosselies, Belgium
| | - Louise Conrard
- Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles, Gosselies, Belgium
| | - Amel Takkal
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| | - Françoise Gregoire
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| | - Nargis Bolaky
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Jason Perret
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| | - Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
2
|
Babicz RSE, Baylor N, Terlouw A, Faber DA, Fukushima K, Biondi RM, Bouley R, Brown D. Ribosomal s6 kinase is a mediator of aquaporin-2 S256 phosphorylation and membrane accumulation after EGFR inhibition with erlotinib. Am J Physiol Renal Physiol 2025; 328:F344-F359. [PMID: 39823198 DOI: 10.1152/ajprenal.00353.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025] Open
Abstract
Vasopressin (VP) activates protein kinase A (PKA), resulting in phosphorylation events and membrane accumulation of aquaporin-2 (AQP2). Epidermal growth factor receptor (EGFR) inhibition with erlotinib also induces AQP2 membrane trafficking with a phosphorylation pattern similar to VP, but without increasing PKA activity. Here, we identify the ribosomal s6 kinase (RSK) as a major mediator phosphorylating AQP2 in this novel, erlotinib-induced pathway. We found that RSK was expressed in collecting duct principal cells in rat kidneys. RSK inhibition with BI-D1870 blocked erlotinib-induced AQP2 serine 256 (S256) phosphorylation and membrane accumulation. CRISPR-generated RSK knockout (KO) cells failed to show increased S256 phosphorylation in response to erlotinib. Like PKA, RSK was able to phosphorylate AQP2 S256 in vitro. Inhibition of phosphoinositide-dependent kinase-1 (PDK1), a known activator of RSK, blocked erlotinib-induced AQP2 S256 phosphorylation and membrane accumulation. We conclude that RSK is a crucial terminal kinase phosphorylating AQP2 at S256 upon EGFR inhibition by erlotinib.NEW & NOTEWORTHY Epidermal growth factor receptor (EGFR) inhibition with erlotinib induces aquaporin-2 (AQP2) membrane accumulation with a phosphorylation pattern similar to vasopressin (VP). Here, we identify the ribosomal s6 kinase (RSK) as a major mediator phosphorylating AQP2 in this novel, erlotinib-induced pathway. In addition, we show that phosphoinositide-dependent kinase-1 (PDK1), a known activator of RSK, is implicated in this pathway: PDK1 inhibition blocks erlotinib-induced AQP2 S256 phosphorylation and membrane accumulation.
Collapse
Affiliation(s)
- Richard S E Babicz
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Noah Baylor
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Abby Terlouw
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Daphne A Faber
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Kazuhiko Fukushima
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Ricardo M Biondi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck SocietyBuenos AiresArgentina
| | - Richard Bouley
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Dennis Brown
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
3
|
Levtchenko E, Ariceta G, Arguedas Flores O, Bichet DG, Bockenhauer D, Emma F, Hoorn EJ, Koster-Kamphuis L, Nijenhuis T, Trepiccione F, Vargas-Poussou R, Walsh SB, Knoers NVAM. International expert consensus statement on the diagnosis and management of congenital nephrogenic diabetes insipidus (arginine vasopressin resistance). Nat Rev Nephrol 2025; 21:83-96. [PMID: 39438674 DOI: 10.1038/s41581-024-00897-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 10/25/2024]
Abstract
Congenital nephrogenic diabetes insipidus (NDI; also known as arginine vasopressin resistance) is a rare inherited disorder of water homeostasis, caused by insensitivity of the distal nephron to arginine vasopressin. Consequently, the kidney loses its ability to concentrate urine, which leads to polyuria, polydipsia and the risk of hypertonic dehydration. The diagnosis and management of NDI are very challenging and require an integrated, multidisciplinary approach. Here, we present 36 recommendations for diagnosis, treatment and follow-up in both children and adults, as well as emergency management, genetic counselling and family planning, for patients with NDI. These recommendations were formulated and graded by an international group of experts in NDI from paediatric and adult nephrology, urology and clinical genetics from the European Rare Kidney Disease Reference Network and the European Society of Paediatric Nephrology, as well as patient advocates, and were validated by a voting panel in a Delphi process. The goal of these recommendations is to provide guidance to health care professionals who care for patients with NDI and to patients and their families. In addition, we emphasize the need for further research on different aspects of this potentially life-threatening disorder to support the development of evidence-based guidelines in the future.
Collapse
Affiliation(s)
- Elena Levtchenko
- Department of Paediatric Nephrology, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam, The Netherlands.
| | - Gema Ariceta
- Department of Paediatric Nephrology, Hospital Vall d' Hebron, Autonomous University of Barcelona, Barcelona, Spain
| | - Olga Arguedas Flores
- Department of Paediatric Urology, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Daniel G Bichet
- Nephrology Service, Hôpital du Sacré-Coeur de Montréal, Departments of Medicine, Pharmacology and Physiology, University of Montreal, Québec, Canada
| | - Detlef Bockenhauer
- Paediatric Nephrology, University Hospital and Catholic University Leuven, Leuven, Belgium
- Great Ormond Street Hospital for Children NHS Foundation Trust and Department of Renal Medicine, University College London, London, UK
| | - Francesco Emma
- Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Ewout J Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Linda Koster-Kamphuis
- Department of Paediatric Nephrology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tom Nijenhuis
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Francesco Trepiccione
- Department of Medical Translational Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- Biogem Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
| | - Rosa Vargas-Poussou
- Department of Genomic Medicine for Rare Diseases, Reference Centre for Hereditary Kidney Diseases of Children and Adults MARHEA, Assistance Publique Hôpitaux de Paris - Hôpital Européen Georges Pompidou, Paris, France
| | - Stephen B Walsh
- London Tubular Centre, Department of Renal Medicine, Department of Renal Medicine, University College London, London, UK
| | - Nine V A M Knoers
- Department of Genetics, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
4
|
Tchakal-Mesbahi A, He J, Zhu S, Huang M, Fukushima K, Bouley R, Brown D, Lu HAJ. Focal Adhesion Kinase (FAK) inhibition induces membrane accumulation of aquaporin2 (AQP2) through endocytosis inhibition and actin depolymerization in renal epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617300. [PMID: 39416213 PMCID: PMC11482834 DOI: 10.1101/2024.10.08.617300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Cellular trafficking of the water channel aquaporin 2 (AQP2) is regulated by the actin cytoskeleton in collecting duct principal cells (PC) to maintain proper water balance in animals. Critical actin depolymerization/polymerization events are involved in both constitutive AQP2 recycling, and the pathway stimulated by vasopressin receptor signaling. Focal adhesion kinase (FAK) plays an important role in modulating the actin cytoskeleton through inhibiting small GTPases, and multiple studies have shown the involvement of FAK in insulin and cholesterol trafficking through actin regulation. To understand whether FAK contributes to water reabsorption by the kidney, we performed a series of in vitro experiments to examine the involvement of FAK and its signaling in mediating AQP2 trafficking in cultured renal epithelial cells. Our data showed that FAK inhibition by specific inhibitors caused membrane accumulation of AQP2 in AQP2expressing LLCPK1 cells by immunofluorescence staining. AQP2 membrane accumulation induced by FAK inhibition is associated with significantly reduced endocytosis of AQP2 via the clathrin-mediated endocytosis pathway. Moreover, AQP2 membrane accumulation induced by FAK inhibition also occurred in cells expressing the constitutive dephosphorylation mutant of AQP2, S256A. This was confirmed by immunoblotting using a specific antibody against phospho-serine 256 AQP2, supporting a phosphorylation independent mechanism. Finally, we demonstrated that inhibition of FAK caused reduced RhoA signaling and promoted F-actin depolymerization. In conclusion, our study identifies FAK signaling as a pathway that could provide a novel therapeutical avenue for AQP2 trafficking regulation in water balance disorders.
Collapse
|
5
|
Ando F, Hara Y, Uchida S. Identification of protein kinase A signalling molecules in renal collecting ducts. J Physiol 2024; 602:3057-3067. [PMID: 37013848 DOI: 10.1113/jp284178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Body water homeostasis is maintained by the correct balance between water intake and water loss through urine, faeces, sweat and breath. It is known that elevated circulating levels of the antidiuretic hormone vasopressin decrease urine volume to prevent excessive water loss from the body. Vasopressin/cAMP/protein kinase A (PKA) signalling is the canonical pathway in renal collecting ducts for phosphorylating aquaporin-2 (AQP2) water channels, which leads to the reabsorption of water from urine via AQP2. Although recent omics data have verified various downstream targets of PKA, crucial regulators that mediate PKA-induced AQP2 phosphorylation remain unknown, mainly because vasopressin is usually used to activate PKA as a positive control. Vasopressin is extremely potent and phosphorylates various PKA substrates non-specifically, making it difficult to narrow down the candidate mediators responsible for AQP2 phosphorylation. The intracellular localization of PKA is tightly regulated by its scaffold proteins, also known as A-kinase anchoring proteins (AKAPs). Furthermore, each AKAP has a target domain that determines its intracellular localization, enabling the creation of a local PKA signalling network. Although vasopressin activates most PKAs independently of their intracellular localization, some chemical compounds preferentially act on PKAs localized on AQP2-containing vesicles while simultaneously phosphorylating AQP2 and its surrounding PKA substrates. Immunoprecipitation with antibodies against phosphorylated PKA substrates followed by mass spectrometry analysis revealed that the PKA substrate in proximity to AQP2 was lipopolysaccharide-responsive and beige-like anchor (LRBA). Furthermore, Lrba knockout studies revealed that LRBA was required for vasopressin-induced AQP2 phosphorylation.
Collapse
Affiliation(s)
- Fumiaki Ando
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yu Hara
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shinichi Uchida
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
6
|
Kharin A, Klussmann E. Many kinases for controlling the water channel aquaporin-2. J Physiol 2024; 602:3025-3039. [PMID: 37440212 DOI: 10.1113/jp284100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/26/2023] [Indexed: 07/14/2023] Open
Abstract
Aquaporin-2 (AQP2) is a member of the aquaporin water channel family. In the kidney, AQP2 is expressed in collecting duct principal cells where it facilitates water reabsorption in response to antidiuretic hormone (arginine vasopressin, AVP). AVP induces the redistribution of AQP2 from intracellular vesicles and its incorporation into the plasma membrane. The plasma membrane insertion of AQP2 represents the crucial step in AVP-mediated water reabsorption. Dysregulation of the system preventing the AQP2 plasma membrane insertion causes diabetes insipidus (DI), a disease characterised by an impaired urine concentrating ability and polydipsia. There is no satisfactory treatment of DI available. This review discusses kinases that control the localisation of AQP2 and points out potential kinase-directed targets for the treatment of DI.
Collapse
Affiliation(s)
- Andrii Kharin
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Enno Klussmann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
7
|
Kamegawa A, Suzuki S, Suzuki H, Nishikawa K, Numoto N, Fujiyoshi Y. Structural analysis of the water channel AQP2 by single-particle cryo-EM. J Struct Biol 2023; 215:107984. [PMID: 37315821 DOI: 10.1016/j.jsb.2023.107984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/19/2023] [Accepted: 06/10/2023] [Indexed: 06/16/2023]
Abstract
Water channels, which are small membrane proteins almost entirely buried in lipid membranes, are challenging research targets for single-particle cryo-electron microscopy (cryo-EM), a powerful technique routinely used to determine the structures of membrane proteins. Because the single-particle method enables structural analysis of a whole protein with flexible parts that interfere with crystallization, we have focused our efforts on analyzing water channel structures. Here, utilizing this system, we analyzed the structure of full-length aquaporin-2 (AQP2), a primary regulator of vasopressin-dependent reabsorption of water at the renal collecting ducts. The 2.9 Å resolution map revealed a cytoplasmic extension of the cryo-EM density that was presumed to be the highly flexible C-terminus at which the localization of AQP2 is regulated in the renal collecting duct cells. We also observed a continuous density along the common water pathway inside the channel pore and lipid-like molecules at the membrane interface. Observations of these constructions in the AQP2 structure analyzed without any fiducial markers (e.g., a rigidly bound antibody) indicate that single-particle cryo-EM will be useful for investigating water channels in native states as well as in complexes with chemical compounds.
Collapse
Affiliation(s)
- Akiko Kamegawa
- Cellular and Structural Physiology Laboratory (CeSPL), Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Shota Suzuki
- Cellular and Structural Physiology Laboratory (CeSPL), Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Hiroshi Suzuki
- Cellular and Structural Physiology Laboratory (CeSPL), Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Kouki Nishikawa
- Joint Research Course for Advanced Biomolecular Characterization, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Nobutaka Numoto
- Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8501, Japan
| | - Yoshinori Fujiyoshi
- Cellular and Structural Physiology Laboratory (CeSPL), Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
8
|
Cheung PW, Boukenna M, Babicz RSE, Mitra S, Kay A, Paunescu TC, Baylor N, Liu CCS, Nair AV, Bouley R, Brown D. Intracellular sites of AQP2 S256 phosphorylation identified using inhibitors of the AQP2 recycling itinerary. Am J Physiol Renal Physiol 2023; 324:F152-F167. [PMID: 36454701 PMCID: PMC9844975 DOI: 10.1152/ajprenal.00123.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
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-β-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.
Collapse
Affiliation(s)
- Pui W Cheung
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mey Boukenna
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Richard S E Babicz
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Shimontini Mitra
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Anna Kay
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Theodor C Paunescu
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Noah Baylor
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Chen-Chung Steven Liu
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Anil V Nair
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Richard Bouley
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dennis Brown
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
9
|
Genome-Engineered mpkCCDc14 Cells as a New Resource for Studying AQP2. Int J Mol Sci 2023; 24:ijms24021684. [PMID: 36675199 PMCID: PMC9866188 DOI: 10.3390/ijms24021684] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/02/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
mpkCCDc14 cells, a polarized epithelial cell line derived from mouse kidney cortical collecting ducts, are known to express the vasopressin V2 receptor (V2R) and aquaporin-2 (AQP2) that are responsive to vasopressin. However, a low abundance of the endogenous AQP2 protein in the absence of vasopressin and heterogeneity of AQP2 protein abundance among the cultured cells may limit the further application of the cell line in AQP2 studies. To overcome the limitation, we aimed to establish mpkCCDc14 cells constitutively expressing V2R and AQP2 via CRISPR/Cas9-mediated genome engineering technology (i.e., V2R-AQP2 cells). 3'- and 5'-Junction PCR revealed that the V2R-AQP2 expression cassette with a long insert size (~2.2 kb) was correctly integrated. Immunoblotting revealed the expression of products of integrated Aqp2 genes. Cell proliferation rate and dDAVP-induced cAMP production were not affected by the knock-in of Avpr2 and Aqp2 genes. The AQP2 protein abundance was significantly higher in V2R-AQP2 cells compared with control mpkCCDc14 cells in the absence of dDAVP and the integrated AQP2 was detected. Immunocytochemistry demonstrated that V2R-AQP2 cells exhibited more homogenous and prominent AQP2 labeling intensity in the absence of dDAVP stimulation. Moreover, prominent AQP2 immunolabeling (both AQP2 and pS256-AQP2) in the apical domain of the genome-edited cells was observed in response to dDAVP stimulation, similar to that in the unedited control mpkCCDc14 cells. Taken together, mpkCCDc14 cells constitutively expressing V2R and AQP2 via genome engineering could be exploited for AQP2 studies.
Collapse
|
10
|
Xiong M, Li C, Wang W, Yang B. Protein Structure and Modification of Aquaporins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1398:15-38. [PMID: 36717484 DOI: 10.1007/978-981-19-7415-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Aquaporins (AQPs) allow water molecules and other small, neutral solutes to quickly pass through membrane. The protein structures of AQPs solved by crystallographic methods or cryo-electron microscopy technology show that AQP monomer consists of six membrane-spanning alpha-helices that form the central water-transporting pore. AQP monomers assemble to form tetramers, forming the functional units in the membrane, to transport water or other small molecules. The biological functions of AQPs are regulated by posttranslational modifications, e.g., phosphorylation, ubiquitination, glycosylation, subcellular distribution, degradation and protein interactions. Modifications of AQP combined with structural properties contribute to a better functional mechanism of AQPs. Insight into the molecular mechanisms responsible for AQP modifications as well as gating and transport properties proved to be fundamental to the development of new therapeutic targets or reliable diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Mengyao Xiong
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chunling Li
- Institute of Hypertension and Kidney Research, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Weidong Wang
- Institute of Hypertension and Kidney Research, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Baoxue Yang
- School of Basic Medical Sciences, Peking University, Beijing, China.
| |
Collapse
|
11
|
Sun Z, Shao X, Wu H, Zhao Y, Cao Y, Li D, Sun Y, Wang Q. Loss of Pten in Renal Tubular Cells Leads to Water Retention by Upregulating AQP2. KIDNEY DISEASES (BASEL, SWITZERLAND) 2022; 9:58-71. [PMID: 36756085 PMCID: PMC9900467 DOI: 10.1159/000528010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 10/26/2022] [Indexed: 12/02/2022]
Abstract
Introduction Phosphatase and tensin (PTEN) is a multifunctional gene associated with the normal development and physiological function of various tissues including the kidney. However, its role in renal tubular reabsorption function has not been well elucidated. Methods We generated a renal tubule-specific Pten knockout mouse model by crossing Ptenfl/fl mice with Ksp-Cre transgenic mice, evaluated the effect of Pten loss on renal tubular function, and investigated the underlying mechanisms. Results Pten loss resulted in abnormal renal structure and function and water retention in multiple organs. Our results also demonstrated that aquaporin-2 (AQP2), an important water channel protein, was upregulated and concentrated on the apical plasma membrane of collecting duct cells, which could be responsible for the impaired water balance in Pten loss mice. The regulation of Pten loss on AQP2 was mediated by protein kinase B (AKT) activation. Conclusions Our results reveal a connection between PTEN gene inactivation and water retention, suggesting the importance of PTEN in normal kidney development and function.
Collapse
Affiliation(s)
- Zhuo Sun
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China
| | - Xiaotong Shao
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China
| | - Haotian Wu
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China
| | - Yaxian Zhao
- Department of Pathology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, China
| | - Yidan Cao
- Department of Pathology, Wuxi No.2 People's Hospital, Wuxi, China
| | - Danhua Li
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China
| | - Ying Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Qingling Wang
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China,*Qingling Wang,
| |
Collapse
|
12
|
Abstract
Protein kinase A (PKA) directly phosphorylates aquaporin-2 (AQP2) water channels in renal collecting ducts to reabsorb water from urine for the maintenance of systemic water homeostasis. More than 50 functionally distinct PKA-anchoring proteins (AKAPs) respectively create compartmentalized PKA signaling to determine the substrate specificity of PKA. Identification of an AKAP responsible for AQP2 phosphorylation is an essential step toward elucidating the molecular mechanisms of urinary concentration. PKA activation by several compounds is a novel screening strategy to uncover PKA substrates whose phosphorylation levels were nearly perfectly correlated with that of AQP2. The leading candidate in this assay proved to be an AKAP termed lipopolysaccharide-responsive and beige-like anchor protein (LRBA). We found that LRBA colocalized with AQP2 in vivo, and Lrba knockout mice displayed a polyuric phenotype with severely impaired AQP2 phosphorylation. Most of the PKA substrates other than AQP2 were adequately phosphorylated by PKA in the absence of LRBA, demonstrating that LRBA-anchored PKA preferentially phosphorylated AQP2 in renal collecting ducts. Furthermore, the LRBA-PKA interaction, rather than other AKAP-PKA interactions, was robustly dissociated by PKA activation. AKAP-PKA interaction inhibitors have attracted attention for their ability to directly phosphorylate AQP2. Therefore, the LRBA-PKA interaction is a promising drug target for the development of anti-aquaretics.
Collapse
|
13
|
AQP2 trafficking in health and diseases: an updated overview. Int J Biochem Cell Biol 2022; 149:106261. [DOI: 10.1016/j.biocel.2022.106261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 05/25/2022] [Accepted: 06/30/2022] [Indexed: 11/23/2022]
|
14
|
Mukherjee K, Gu C, Collins A, Mettlen M, Samelko B, Altintas MM, Sudhini YR, Wang X, Bouley R, Brown D, Pedro BP, Bane SL, Gupta V, Brinkkoetter PT, Hagmann H, Reiser J, Sever S. Simultaneous stabilization of actin cytoskeleton in multiple nephron-specific cells protects the kidney from diverse injury. Nat Commun 2022; 13:2422. [PMID: 35504916 PMCID: PMC9065033 DOI: 10.1038/s41467-022-30101-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/04/2022] [Indexed: 02/07/2023] Open
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.
Collapse
Affiliation(s)
- Kamalika Mukherjee
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Changkyu Gu
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Agnieszka Collins
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Marcel Mettlen
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Beata Samelko
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | | | - Xuexiang Wang
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Richard Bouley
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Dennis Brown
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Bradley P Pedro
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Susan L Bane
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, NY, USA
| | - Vineet Gupta
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Paul T Brinkkoetter
- Department of Internal Medicine-Center for Molecular Medicine Cologne, University of Cologne and Faculty of Medicine-University Hospital Cologne, Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne (Sybacol), Cologne, Germany
| | - Henning Hagmann
- Department of Internal Medicine-Center for Molecular Medicine Cologne, University of Cologne and Faculty of Medicine-University Hospital Cologne, Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne (Sybacol), Cologne, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA.
| | - Sanja Sever
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
15
|
Dutta A, Das M. Deciphering the Role of Aquaporins in Metabolic Diseases: A Mini Review. Am J Med Sci 2022; 364:148-162. [DOI: 10.1016/j.amjms.2021.10.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 06/16/2021] [Accepted: 10/21/2021] [Indexed: 12/23/2022]
|
16
|
Noda Y, Sasaki S. Updates and Perspectives on Aquaporin-2 and Water Balance Disorders. Int J Mol Sci 2021; 22:ijms222312950. [PMID: 34884753 PMCID: PMC8657825 DOI: 10.3390/ijms222312950] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 12/31/2022] Open
Abstract
Ensuring the proper amount of water inside the body is essential for survival. One of the key factors in the maintenance of body water balance is water reabsorption in the collecting ducts of the kidney, a process that is regulated by aquaporin-2 (AQP2). AQP2 is a channel that is exclusively selective for water molecules and impermeable to ions or other small molecules. Impairments of AQP2 result in various water balance disorders, including nephrogenic diabetes insipidus (NDI), which is a disease characterized by a massive loss of water through the kidney and consequent severe dehydration. Dysregulation of AQP2 is also a cause of water retention with hyponatremia in heart failure, hepatic cirrhosis, and syndrome of inappropriate antidiuretic hormone secretion (SIADH). Antidiuretic hormone vasopressin is an upstream regulator of AQP2. Its binding to the vasopressin V2 receptor promotes AQP2 targeting to the apical membrane and thus enables water reabsorption. Tolvaptan, a vasopressin V2 receptor antagonist, is effective and widely used for water retention with hyponatremia. However, there are no studies showing improvement in hard outcomes or long-term prognosis. A possible reason is that vasopressin receptors have many downstream effects other than AQP2 function. It is expected that the development of drugs that directly target AQP2 may result in increased treatment specificity and effectiveness for water balance disorders. This review summarizes recent progress in studies of AQP2 and drug development challenges for water balance disorders.
Collapse
Affiliation(s)
- Yumi Noda
- Department of Nephrology, Nitobe Memorial Nakano General Hospital, Tokyo 164-8607, Japan
- Department of Nephrology, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
- Correspondence: ; Tel.: +81-3-3382-1231; Fax: +81-3-3382-1588
| | - Sei Sasaki
- Department of Nephrology, Cellular and Structural Physiology Laboratory, Tokyo Medical and Dental University, Tokyo 113-8519, Japan;
| |
Collapse
|
17
|
Liu CCS, Cheung PW, Dinesh A, Baylor N, Paunescu TC, Nair AV, Bouley R, Brown D. Actin-related protein 2/3 complex plays a critical role in the aquaporin-2 exocytotic pathway. Am J Physiol Renal Physiol 2021; 321:F179-F194. [PMID: 34180716 PMCID: PMC8424666 DOI: 10.1152/ajprenal.00015.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
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.
Collapse
Affiliation(s)
- Chen-Chung Steven Liu
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Pui Wen Cheung
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anupama Dinesh
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Noah Baylor
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Theodor C. Paunescu
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anil V. Nair
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Richard Bouley
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dennis Brown
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Mukherjee K, Chio TI, Gu H, Sackett DL, Bane SL, Sever S. A Novel Fluorogenic Assay for the Detection of Nephrotoxin-Induced Oxidative Stress in Live Cells and Renal Tissue. ACS Sens 2021; 6:2523-2528. [PMID: 34214393 PMCID: PMC8314269 DOI: 10.1021/acssensors.1c00422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Drug-induced kidney
injury frequently leads to aborted clinical
trials and drug withdrawals. Sufficiently sensitive sensors capable
of detecting mild signs of chemical insult in cell-based screening
assays are critical to identifying and eliminating potential toxins
in the preclinical stage. Oxidative stress is a common early manifestation
of chemical toxicity, and biomolecule carbonylation is an irreversible
repercussion of oxidative stress. Here, we present a novel fluorogenic
assay using a sensor, TFCH, that responds to biomolecule carbonylation
and efficiently detects modest forms of renal injury with much greater
sensitivity than standard assays for nephrotoxins. We demonstrate
that this sensor can be deployed in live kidney cells and in renal
tissue. Our robust assay may help inform preclinical decisions to
recall unsafe drug candidates. The application of this sensor in identifying
and analyzing diverse pathologies is envisioned.
Collapse
Affiliation(s)
- Kamalika Mukherjee
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Tak Ian Chio
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Han Gu
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Dan L. Sackett
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Susan L. Bane
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Sanja Sever
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
19
|
Wang R, Wu ST, Yang X, Qian Y, Choi JP, Gao R, Song S, Wang Y, Zhuang T, Wong JJ, Zhang Y, Han Z, Lu HA, Alexander SI, Liu R, Xia Y, Zheng X. Pdcd10-Stk24/25 complex controls kidney water reabsorption by regulating Aqp2 membrane targeting. JCI Insight 2021; 6:e142838. [PMID: 34156031 PMCID: PMC8262504 DOI: 10.1172/jci.insight.142838] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 05/12/2021] [Indexed: 11/17/2022] Open
Abstract
PDCD10, also known as CCM3, is a gene found to be associated with the human disease cerebral cavernous malformations (CCMs). PDCD10 forms a complex with GCKIII kinases including STK24, STK25, and MST4. Studies in C. elegans and Drosophila have shown a pivotal role of the PDCD10-GCKIII complex in maintaining epithelial integrity. Here, we found that mice deficient of Pdcd10 or Stk24/25 in the kidney tubules developed polyuria and displayed increased water consumption. Although the expression levels of aquaporin genes were not decreased, the levels of total and phosphorylated aquaporin 2 (Aqp2) protein in the apical membrane of tubular epithelial cells were decreased in Pdcd10- and Stk24/25-deficient mice. This loss of Aqp2 was associated with increased expression and membrane targeting of Ezrin and phosphorylated Ezrin, Radixin, Moesin (p-ERM) proteins and impaired intracellular vesicle trafficking. Treatment with Erlotinib, a tyrosine kinase inhibitor promoting exocytosis and inhibiting endocytosis, normalized the expression level and membrane abundance of Aqp2 protein, and partially rescued the water reabsorption defect observed in the Pdcd10-deficient mice. Our current study identified the PDCD10-STK-ERM signaling pathway as a potentially novel pathway required for water balance control by regulating vesicle trafficking and protein abundance of AQP2 in the kidneys.
Collapse
Affiliation(s)
- Rui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, China
| | - Shi-Ting Wu
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, China
| | - Xi Yang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, China
| | - Yude Qian
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, China
| | - Jaesung P Choi
- Lab of Cardiovascular Signaling, Centenary Institute, and Sydney Medical School, University of Sydney, Sydney, Australia
| | - Rui Gao
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, China
| | - Siliang Song
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, China
| | - Yixuan Wang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, China
| | - Tao Zhuang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Justin Jl Wong
- Epigenetics and RNA Biology Program Centenary Institute and Sydney Medical School, University of Sydney, Sydney, Australia
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiming Han
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hua A Lu
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen I Alexander
- Department of Pediatric Nephrology, The Children's Hospital at Westmead and Centre for Kidney Research, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Renjing Liu
- Vascular Epigenetics Laboratory, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Yin Xia
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiangjian Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, China.,Lab of Cardiovascular Signaling, Centenary Institute, and Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
20
|
Hinrichs GR, Mortensen LA, Bistrup C, Dieperink HH, Jensen BL. Treatment of Nephrogenic Diabetes Insipidus Patients With cGMP-Stimulating Drugs Does Not Mitigate Polyuria or Increase Urinary Concentrating Ability. Kidney Int Rep 2020; 5:1319-1325. [PMID: 32775834 PMCID: PMC7403538 DOI: 10.1016/j.ekir.2020.05.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/28/2020] [Accepted: 05/04/2020] [Indexed: 11/30/2022] Open
Affiliation(s)
- Gitte R Hinrichs
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - Line A Mortensen
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - Claus Bistrup
- Department of Nephrology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Hans H Dieperink
- Department of Nephrology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Boye L Jensen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
21
|
Fenton RA, Murali SK, Moeller HB. Advances in aquaporin-2 trafficking mechanisms and their implications for treatment of water balance disorders. Am J Physiol Cell Physiol 2020; 319:C1-C10. [PMID: 32432927 DOI: 10.1152/ajpcell.00150.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In mammals, conservation of body water is critical for survival and is dependent on the kidneys' ability to minimize water loss in the urine during periods of water deprivation. The collecting duct water channel aquaporin-2 (AQP2) plays an essential role in this homeostatic response by facilitating water reabsorption along osmotic gradients. The ability to increase the levels of AQP2 in the apical plasma membrane following an increase in plasma osmolality is a rate-limiting step in water reabsorption, a process that is tightly regulated by the antidiuretic hormone arginine vasopressin (AVP). In this review, the focus is on the role of the carboxyl-terminus of AQP2 as a key regulatory point for AQP2 trafficking. We provide an overview of AQP2 structure, disease-causing mutations in the AQP2 carboxyl-terminus, the role of posttranslational modifications such as phosphorylation and ubiquitylation in the tail domain, and their implications for balanced trafficking of AQP2. Finally, we discuss how various modifications of the AQP2 tail facilitate selective protein-protein interactions that modulate the AQP2 trafficking mechanism.
Collapse
Affiliation(s)
- Robert A Fenton
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Hanne B Moeller
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
22
|
Datta A, Yang CR, Limbutara K, Chou CL, Rinschen MM, Raghuram V, Knepper MA. PKA-independent vasopressin signaling in renal collecting duct. FASEB J 2020; 34:6129-6146. [PMID: 32219907 PMCID: PMC9200475 DOI: 10.1096/fj.201902982r] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 11/11/2022]
Abstract
Vasopressin regulates renal water excretion by binding to a Gα s-coupled receptor (V2R) in collecting duct cells, resulting in increased water permeability through regulation of the aquaporin-2 (AQP2) water channel. This action is widely accepted to be associated with cAMP-mediated activation of protein kinase A (PKA). Here, we use phosphoproteomics in collecting duct cells in which PKA has been deleted (CRISPR-Cas9) to identify PKA-independent responses to vasopressin. The results show that V2R-mediated vasopressin signaling is predominantly, but not entirely, PKA-dependent. Upregulated sites in PKA-null cells include Ser256 of AQP2, which is critical to regulation of AQP2 trafficking. In addition, phosphorylation changes in the protein kinases Stk39 (SPAK) and Prkci (an atypical PKC) are consistent with PKA-independent regulation of these protein kinases. Target motif analysis of the phosphopeptides increased in PKA-null cells indicates that vasopressin activates one or more members of the AMPK/SNF1-subfamily of basophilic protein kinases. In vitro phosphorylation assays using recombinant, purified SNF1-subfamily kinases confirmed postulated target specificities. Of interest, measured IBMX-dependent cAMP levels were an order of magnitude higher in PKA-null than in PKA-intact cells, indicative of a PKA-dependent feedback mechanism. Overall, the findings support the conclusion that V2-receptor mediated signaling in collecting duct cells is in part PKA-independent.
Collapse
Affiliation(s)
- Arnab Datta
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore 575018, Karnataka, India
| | - Chin-Rang Yang
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Kavee Limbutara
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Markus M. Rinschen
- Department of Chemistry, Center for Metabolomics and Mass Spectrometry, The Scripps Research Institute, La Jolla, CA
| | - Viswanathan Raghuram
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark A. Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
23
|
Chlorpromazine Induces Basolateral Aquaporin-2 Accumulation via F-Actin Depolymerization and Blockade of Endocytosis in Renal Epithelial Cells. Cells 2020; 9:cells9041057. [PMID: 32340337 PMCID: PMC7226349 DOI: 10.3390/cells9041057] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/13/2020] [Accepted: 04/19/2020] [Indexed: 12/11/2022] Open
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.
Collapse
|
24
|
Cyclin-Dependent Kinase 18 Controls Trafficking of Aquaporin-2 and Its Abundance through Ubiquitin Ligase STUB1, Which Functions as an AKAP. Cells 2020; 9:cells9030673. [PMID: 32164329 PMCID: PMC7140648 DOI: 10.3390/cells9030673] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/27/2020] [Accepted: 03/09/2020] [Indexed: 12/24/2022] Open
Abstract
Arginine-vasopressin (AVP) facilitates water reabsorption in renal collecting duct principal cells through regulation of the water channel aquaporin-2 (AQP2). The hormone binds to vasopressin V2 receptors (V2R) on the surface of the cells and stimulates cAMP synthesis. The cAMP activates protein kinase A (PKA), which initiates signaling that causes an accumulation of AQP2 in the plasma membrane of the cells facilitating water reabsorption from primary urine and fine-tuning of body water homeostasis. AVP-mediated PKA activation also causes an increase in the AQP2 protein abundance through a mechanism that involves dephosphorylation of AQP2 at serine 261 and a decrease in its poly-ubiquitination. However, the signaling downstream of PKA that controls the localization and abundance of AQP2 is incompletely understood. We carried out an siRNA screen targeting 719 kinase-related genes, representing the majority of the kinases of the human genome and analyzed the effect of the knockdown on AQP2 by high-content imaging and biochemical approaches. The screening identified 13 hits whose knockdown inhibited the AQP2 accumulation in the plasma membrane. Amongst the candidates was the so far hardly characterized cyclin-dependent kinase 18 (CDK18). Our further analysis revealed a hitherto unrecognized signalosome comprising CDK18, an E3 ubiquitin ligase, STUB1 (CHIP), PKA and AQP2 that controls the localization and abundance of AQP2. CDK18 controls AQP2 through phosphorylation at serine 261 and STUB1-mediated ubiquitination. STUB1 functions as an A-kinase anchoring protein (AKAP) tethering PKA to the protein complex and bridging AQP2 and CDK18. The modulation of the protein complex may lead to novel concepts for the treatment of disorders which are caused or are associated with dysregulated AQP2 and for which a satisfactory treatment is not available, e.g., hyponatremia, liver cirrhosis, diabetes insipidus, ADPKD or heart failure.
Collapse
|
25
|
Leipziger J, Praetorius H. Renal Autocrine and Paracrine Signaling: A Story of Self-protection. Physiol Rev 2020; 100:1229-1289. [PMID: 31999508 DOI: 10.1152/physrev.00014.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Autocrine and paracrine signaling in the kidney adds an extra level of diversity and complexity to renal physiology. The extensive scientific production on the topic precludes easy understanding of the fundamental purpose of the vast number of molecules and systems that influence the renal function. This systematic review provides the broader pen strokes for a collected image of renal paracrine signaling. First, we recapitulate the essence of each paracrine system one by one. Thereafter the single components are merged into an overarching physiological concept. The presented survey shows that despite the diversity in the web of paracrine factors, the collected effect on renal function may not be complicated after all. In essence, paracrine activation provides an intelligent system that perceives minor perturbations and reacts with a coordinated and integrated tissue response that relieves the work load from the renal epithelia and favors diuresis and natriuresis. We suggest that the overall function of paracrine signaling is reno-protection and argue that renal paracrine signaling and self-regulation are two sides of the same coin. Thus local paracrine signaling is an intrinsic function of the kidney, and the overall renal effect of changes in blood pressure, volume load, and systemic hormones will always be tinted by its paracrine status.
Collapse
Affiliation(s)
- Jens Leipziger
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; and Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark
| | - Helle Praetorius
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; and Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark
| |
Collapse
|
26
|
Cheung PW, Bouley R, Brown D. Targeting the Trafficking of Kidney Water Channels for Therapeutic Benefit. Annu Rev Pharmacol Toxicol 2020; 60:175-194. [PMID: 31561739 PMCID: PMC7334826 DOI: 10.1146/annurev-pharmtox-010919-023654] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
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.
Collapse
Affiliation(s)
- Pui W. Cheung
- Center for Systems Biology, Program in Membrane Biology, and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Richard Bouley
- Center for Systems Biology, Program in Membrane Biology, and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Dennis Brown
- Center for Systems Biology, Program in Membrane Biology, and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
27
|
|
28
|
Wang Q, Wu ZL, Yuan X, Dong HY, Xu X, Xin H, Wang YH, Zhang JB, Chen L, Li HL, Zhang XM, Zhang WD. Bilobetin induces kidney injury by influencing cGMP-mediated AQP-2 trafficking and podocyte cell cycle arrest. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 64:153073. [PMID: 31542661 DOI: 10.1016/j.phymed.2019.153073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/26/2019] [Accepted: 08/22/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Ginkgo biloba (Gb) extracts have been used as a traditional Chinese medicine. Gb contains flavonoids, which are considered to be its active ingredients and have been used in the treatment of a variety of diseases. However, few scientific research studies on the side effects of flavonoid in Gb have been reported. PURPOSE The present study aimed to investigate the effect of bilobetin on the kidney of Sprague-Dawley (SD) rats. STUDY DESIGN AND RESULT In this study, rats were injected with 50 mg/kg of bilobetin, a biflavone isolated from Gb, for 7 days and aristolochic acid was used as positive controls. The results showed that the body weight and urine output of the rats were dramatically decreased, and urinary protein increased after the intraperitoneal injection of bilobetin compared with the control group. Bilobetin treatment showed vacuolar degeneration in the renal tubular epithelium, glomerular atrophy by histostaining, and podocyte fusion by electron microscopy. This study further showed that bilobetin promoted the trafficking of aquaporin 2 (AQP-2) onto the plasma membrane to achieve the function of urine concentration by in vivo study in rats and in vitro study in IMCD-3 cells. The redistribution of AQP-2 is due to increased expression of cGMP in IMCD-3 cells, which in turn promoted the phosphorylation of AQP-2 at site Ser-256. The proteinuria caused by bilobetin may be attributed to podocyte cell cycle arrest at G2/M transition, which is may associated with AKT and MAPK signaling. CONCLUSIONS The current study showed that bilobetin has some side effects on kidneys at a dose of 50 mg/kg in SD rats and provides insight into the potential detrimental effects of monomeric ingredients in Gb.
Collapse
Affiliation(s)
- Qun Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China; Institute of Interdisciplinary Research Complex, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Zhi Li Wu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Xing Yuan
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Hong Yuan Dong
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Xin Xu
- Institute of Interdisciplinary Research Complex, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yin Hang Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jian Bing Zhang
- WanBangDe Pharmaceutical Group Co.,Ltd. Wenlin, Zhejiang 317500, China
| | - Li Chen
- WanBangDe Pharmaceutical Group Co.,Ltd. Wenlin, Zhejiang 317500, China
| | - Hui Liang Li
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Xue Mei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Wei Dong Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China; School of Pharmacy, Second Military Medical University, Shanghai 200433, China; Institute of Interdisciplinary Research Complex, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
29
|
Törnroth-Horsefield S. Phosphorylation of human AQP2 and its role in trafficking. VITAMINS AND HORMONES 2019; 112:95-117. [PMID: 32061351 DOI: 10.1016/bs.vh.2019.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human Aquaporin 2 (AQP2) is a membrane-bound water channel found in the kidney collecting duct whose regulation by trafficking plays a key role in regulating urine volume. AQP2 trafficking is tightly controlled by the pituitary hormone arginine vasopressin (AVP), which stimulates translocation of AQP2 residing in storage vesicles to the apical membrane. The AVP-dependent translocation of AQP2 to and from the apical membrane is controlled by multiple phosphorylation sites in the AQP2 C-terminus, the phosphorylation of which alters its affinity to proteins within the cellular membrane protein trafficking machinery. The aim of this chapter is to provide a summary of what is currently known about AVP-mediated AQP2 trafficking, dissecting the roles of individual phosphorylation sites, kinases and phosphatases and interacting proteins. From this, the picture of an immensely complex process emerges, of which many structural and molecular details remains to be elucidated.
Collapse
|
30
|
Baltzer S, Klussmann E. Small molecules for modulating the localisation of the water channel aquaporin-2-disease relevance and perspectives for targeting local cAMP signalling. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1049-1064. [PMID: 31300862 DOI: 10.1007/s00210-019-01686-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/26/2019] [Indexed: 12/23/2022]
Abstract
The tight spatial and temporal organisation of cyclic adenosine monophosphate (cAMP) signalling plays a key role in arginine-vasopressin (AVP)-mediated water reabsorption in renal collecting duct principal cells and in a plethora of other processes such as in the control of cardiac myocyte contractility. This review critically discusses in vitro- and cell-based screening strategies for the identification of small molecules that interfere with AVP/cAMP signalling in renal principal cells; it features phenotypic screening and approaches for targeting protein-protein interactions of A-kinase anchoring proteins (AKAPs), which organise local cAMP signalling hubs. The discovery of novel chemical entities for the modulation of local cAMP will not only provide tools for elucidating molecular mechanisms underlying cAMP signalling. Novel chemical entities can also serve as starting points for the development of novel drugs for the treatment of human diseases. Examples illustrate how screening for small molecules can pave the way to novel approaches for the treatment of certain forms of diabetes insipidus, a disease caused by defects in AVP-mediated water reabsorption.
Collapse
Affiliation(s)
- Sandrine Baltzer
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Helmholtz Association, Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| | - Enno Klussmann
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Helmholtz Association, Robert-Rössle-Strasse 10, 13125, Berlin, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany. .,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Vegetative Physiology, Berlin, Germany.
| |
Collapse
|
31
|
Mendoza LD, Hyndman KA. The contribution of collecting duct NOS1 to the concentrating mechanisms in male and female mice. Am J Physiol Renal Physiol 2019; 317:F547-F559. [PMID: 31241990 DOI: 10.1152/ajprenal.00180.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The collecting duct (CD) concentrates the urine, thereby maintaining body water volume and plasma osmolality within a normal range. The endocrine hormone arginine vasopressin acts in the CD to increase water permeability via the vasopressin 2 receptor (V2R)-aquaporin (AQP) axis. Recent studies have suggested that autocrine factors may also contribute to the regulation of CD water permeability. Nitric oxide is produced predominantly by nitric oxide synthase 1 (NOS1) in the CD and acts as a diuretic during salt loading. The present study sought to determine whether CD NOS1 regulates diuresis during changes in hydration status. Male and female control and CD NOS1 knockout (CDNOS1KO) mice were hydrated (5% sucrose water), water deprived, or acutely challenged with the V2R agonist desmopressin. In male mice, water deprivation resulted in decreased urine flow and increased plasma osmolality, copeptin concentration, and kidney AQP2 abundance independent of CD NOS1. In female control mice, water deprivation reduced urine flow, increased plasma osmolality and copeptin, but did not significantly change total AQP2; however, there was increased basolateral AQP3 localization. Surprisingly, female CDNOS1KO mice while on the sucrose water presented with symptoms of dehydration. Fibroblast growth factor 21, an endocrine regulator of sweetness preference, was significantly higher in female CDNOS1KO mice, suggesting that this was reducing their drive to drink the sucrose water. With acute desmopressin challenge, female CDNOS1KO mice failed to appropriately concentrate their urine, resulting in higher plasma osmolality than controls. In conclusion, CD NOS1 plays only a minor role in urine-concentrating mechanisms.
Collapse
Affiliation(s)
- Luciano D Mendoza
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kelly A Hyndman
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
32
|
Cheung PW, Terlouw A, Janssen SA, Brown D, Bouley R. Inhibition of non-receptor tyrosine kinase Src induces phosphoserine 256-independent aquaporin-2 membrane accumulation. J Physiol 2019; 597:1627-1642. [PMID: 30488437 PMCID: PMC6418769 DOI: 10.1113/jp277024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/23/2018] [Indexed: 12/22/2022] Open
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.
Collapse
Affiliation(s)
- Pui W. Cheung
- Center for Systems BiologyProgram in Membrane Biology and Division of NephrologyMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Abby Terlouw
- Center for Systems BiologyProgram in Membrane Biology and Division of NephrologyMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Sam Antoon Janssen
- Center for Systems BiologyProgram in Membrane Biology and Division of NephrologyMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Dennis Brown
- Center for Systems BiologyProgram in Membrane Biology and Division of NephrologyMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Richard Bouley
- Center for Systems BiologyProgram in Membrane Biology and Division of NephrologyMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| |
Collapse
|
33
|
Nesverova V, Törnroth-Horsefield S. Phosphorylation-Dependent Regulation of Mammalian Aquaporins. Cells 2019; 8:cells8020082. [PMID: 30678081 PMCID: PMC6406877 DOI: 10.3390/cells8020082] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/17/2019] [Accepted: 01/21/2019] [Indexed: 12/26/2022] Open
Abstract
Water homeostasis is fundamental for cell survival. Transport of water across cellular membranes is governed by aquaporins—tetrameric integral membrane channels that are highly conserved throughout the prokaryotic and eukaryotic kingdoms. In eukaryotes, specific regulation of these channels is required and is most commonly carried out by shuttling the protein between cellular compartments (trafficking) or by opening and closing the channel (gating). Structural and functional studies have revealed phosphorylation as a ubiquitous mechanism in aquaporin regulation by both regulatory processes. In this review we summarize what is currently known about the phosphorylation-dependent regulation of mammalian aquaporins. Focusing on the water-specific aquaporins (AQP0–AQP5), we discuss how gating and trafficking are controlled by phosphorylation and how phosphorylation affects the binding of aquaporins to regulatory proteins, thereby highlighting structural details and dissecting the contribution of individual phosphorylated residues when possible. Our aim is to provide an overview of the mechanisms behind how aquaporin phosphorylation controls cellular water balance and to identify key areas where further studies are needed.
Collapse
Affiliation(s)
- Veronika Nesverova
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, Box 124, 221 00 Lund, Sweden.
| | - Susanna Törnroth-Horsefield
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, Box 124, 221 00 Lund, Sweden.
| |
Collapse
|
34
|
The Impact of the Nitric Oxide (NO)/Soluble Guanylyl Cyclase (sGC) Signaling Cascade on Kidney Health and Disease: A Preclinical Perspective. Int J Mol Sci 2018; 19:ijms19061712. [PMID: 29890734 PMCID: PMC6032334 DOI: 10.3390/ijms19061712] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/07/2018] [Accepted: 06/08/2018] [Indexed: 12/20/2022] Open
Abstract
Chronic Kidney Disease (CKD) is a highly prevalent disease with a substantial medical need for new and more efficacious treatments. The Nitric Oxide (NO), soluble guanylyl cyclase (sGC), cyclic guanosine monophosphate (cGMP) signaling cascade regulates various kidney functions. cGMP directly influences renal blood flow, renin secretion, glomerular function, and tubular exchange processes. Downregulation of NO/sGC/cGMP signaling results in severe kidney pathologies such as CKD. Therefore, treatment strategies aiming to maintain or increase cGMP might have beneficial effects for the treatment of progressive kidney diseases. Within this article, we review the NO/sGC/cGMP signaling cascade and its major pharmacological intervention sites. We specifically focus on the currently known effects of cGMP on kidney function parameters. Finally, we summarize the preclinical evidence for kidney protective effects of NO-donors, PDE inhibitors, sGC stimulators, and sGC activators.
Collapse
|
35
|
Basolateral cholesterol depletion alters Aquaporin-2 post-translational modifications and disrupts apical plasma membrane targeting. Biochem Biophys Res Commun 2018; 495:157-162. [DOI: 10.1016/j.bbrc.2017.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 11/01/2017] [Indexed: 11/22/2022]
|
36
|
Umejiego EN, Wang Y, Knepper MA, Chou CL. Roflumilast and aquaporin-2 regulation in rat renal inner medullary collecting duct. Physiol Rep 2017; 5:5/2/e13121. [PMID: 28108651 PMCID: PMC5269416 DOI: 10.14814/phy2.13121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/29/2016] [Accepted: 12/12/2016] [Indexed: 12/23/2022] Open
Abstract
Roflumilast is a cyclic nucleotide phosphodiesterase inhibitor that is FDA‐approved for treatment of chronic obstructive pulmonary disease. With a view toward possible use for treatment of patients with X‐linked nephrogenic diabetes insipidus (NDI) due to hemizygous mutations in the V2 vasopressin receptor, this study sought to determine the effect of roflumilast on aquaporin‐2 (AQP2) phosphorylation, AQP2 trafficking, and water permeability in the rat inner medullary collecting duct (IMCD). In the presence of the vasopressin analog dDAVP (0.1 nmol/L), both roflumilast and its active metabolite roflumilast N‐oxide (RNO) significantly increased phosphorylation at S256, S264, and S269, and decreased phosphorylation at S261 (immunoblotting) in IMCD suspensions in a dose‐dependent manner (3–3000 nmol/L). Another commonly used phosphodiesterase inhibitor, IBMX, affected phosphorylation only at the highest concentration in this range. However, neither roflumilast nor RNO had an effect on AQP2 phosphorylation in the absence of vasopressin. Furthermore, roflumilast alone did not increase AQP2 trafficking to the plasma membrane (immunofluorescence) or increase water permeability in freshly microdissected perfused IMCD segments. We conclude that roflumilast can be used to enhance vasopressin's action on AQP2 activity in the renal collecting duct, but has no detectable effect in the absence of vasopressin. These findings suggest that roflumilast may not have a beneficial effect in X‐linked NDI, but could find useful application in acquired NDI.
Collapse
Affiliation(s)
- Ezigbobiara N Umejiego
- Epithelial Systems Biology Laboratory, Systems Biology Center NHLBI National Institutes of Health, Bethesda, Maryland, 20892-1603
| | - Yanhua Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, 30322
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center NHLBI National Institutes of Health, Bethesda, Maryland, 20892-1603
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center NHLBI National Institutes of Health, Bethesda, Maryland, 20892-1603
| |
Collapse
|
37
|
Milano S, Carmosino M, Gerbino A, Svelto M, Procino G. Hereditary Nephrogenic Diabetes Insipidus: Pathophysiology and Possible Treatment. An Update. Int J Mol Sci 2017; 18:ijms18112385. [PMID: 29125546 PMCID: PMC5713354 DOI: 10.3390/ijms18112385] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/03/2017] [Accepted: 11/07/2017] [Indexed: 12/17/2022] Open
Abstract
Under physiological conditions, excessive loss of water through the urine is prevented by the release of the antidiuretic hormone arginine-vasopressin (AVP) from the posterior pituitary. In the kidney, AVP elicits a number of cellular responses, which converge on increasing the osmotic reabsorption of water in the collecting duct. One of the key events triggered by the binding of AVP to its type-2 receptor (AVPR2) is the exocytosis of the water channel aquaporin 2 (AQP2) at the apical membrane the principal cells of the collecting duct. Mutations of either AVPR2 or AQP2 result in a genetic disease known as nephrogenic diabetes insipidus, which is characterized by the lack of responsiveness of the collecting duct to the antidiuretic action of AVP. The affected subject, being incapable of concentrating the urine, presents marked polyuria and compensatory polydipsia and is constantly at risk of severe dehydration. The molecular bases of the disease are fully uncovered, as well as the genetic or clinical tests for a prompt diagnosis of the disease in newborns. A real cure for nephrogenic diabetes insipidus (NDI) is still missing, and the main symptoms of the disease are handled with s continuous supply of water, a restrictive diet, and nonspecific drugs. Unfortunately, the current therapeutic options are limited and only partially beneficial. Further investigation in vitro or using the available animal models of the disease, combined with clinical trials, will eventually lead to the identification of one or more targeted strategies that will improve or replace the current conventional therapy and grant NDI patients a better quality of life. Here we provide an updated overview of the genetic defects causing NDI, the most recent strategies under investigation for rescuing the activity of mutated AVPR2 or AQP2, or for bypassing defective AVPR2 signaling and restoring AQP2 plasma membrane expression.
Collapse
Affiliation(s)
- Serena Milano
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy.
| | - Monica Carmosino
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy.
| | - Andrea Gerbino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy.
| | - Maria Svelto
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy.
| | - Giuseppe Procino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy.
| |
Collapse
|
38
|
Lei L, Huang M, Su L, Xie D, Mamuya FA, Ham O, Tsuji K, Păunescu TG, Yang B, Lu HAJ. Manganese promotes intracellular accumulation of AQP2 via modulating F-actin polymerization and reduces urinary concentration in mice. Am J Physiol Renal Physiol 2017; 314:F306-F316. [PMID: 29046300 DOI: 10.1152/ajprenal.00391.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aquaporin-2 (AQP2) is a water channel protein expressed in principal cells (PCs) of the kidney collecting ducts (CDs) and plays a critical role in mediating water reabsorption and urine concentration. AQP2 undergoes both regulated trafficking mediated by vasopressin (VP) and constitutive recycling, which is independent of VP. For both pathways, actin cytoskeletal dynamics is a key determinant of AQP2 trafficking. We report here that manganese chloride (MnCl2) is a novel and potent regulator of AQP2 trafficking in cultured cells and in the kidney. MnCl2 treatment promoted internalization and intracellular accumulation of AQP2. The effect of MnCl2 on the intracellular accumulation of AQP2 was associated with activation of RhoA and actin polymerization without modification of AQP2 phosphorylation. Although the level of total and phosphorylated AQP2 did not change, MnCl2 treatment impeded VP-induced phosphorylation of AQP2 at its serine-256, -264, and -269 residues and dephosphorylation at serine 261. In addition, MnCl2 significantly promoted F-actin polymerization along with downregulation of RhoA activity and prevented VP-induced membrane accumulation of AQP2. Finally, MnCl2 treatment in mice resulted in significant polyuria and reduced urinary concentration, likely due to intracellular relocation of AQP2 in the PCs of kidney CDs. More importantly, the reduced urinary concentration caused by MnCl2 treatment in animals was not corrected by VP. In summary, our study identified a novel effect of MnCl2 on AQP2 trafficking through modifying RhoA activity and actin polymerization and uncovered its potent impact on water diuresis in vivo.
Collapse
Affiliation(s)
- Lei Lei
- Department of Pharmacology, School of Basic Medical Sciences, Peking University , Beijing , People's Republic of China.,Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts
| | - Ming Huang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University , Beijing , People's Republic of China.,Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts
| | - Limin Su
- Department of Pharmacology, School of Basic Medical Sciences, Peking University , Beijing , People's Republic of China.,Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts
| | - Dongping Xie
- Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts
| | - Fahmy A Mamuya
- Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts.,Harvard Medical School , Boston, Massachusetts
| | - Onju Ham
- Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts.,Harvard Medical School , Boston, Massachusetts
| | - Kenji Tsuji
- Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts.,Harvard Medical School , Boston, Massachusetts
| | - Teodor G Păunescu
- Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts.,Harvard Medical School , Boston, Massachusetts
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University , Beijing , People's Republic of China
| | - Hua A Jenny Lu
- Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts.,Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
39
|
Al Therwani S, Malmberg MES, Rosenbaek JB, Bech JN, Pedersen EB. Effect of tolvaptan on renal handling of water and sodium, GFR and central hemodynamics in autosomal dominant polycystic kidney disease during inhibition of the nitric oxide system: a randomized, placebo-controlled, double blind, crossover study. BMC Nephrol 2017; 18:268. [PMID: 28810844 PMCID: PMC5558668 DOI: 10.1186/s12882-017-0686-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/07/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Tolvaptan slows progression of autosomal dominant polycystic kidney disease (ADPKD) by antagonizing the vasopressin-cAMP axis. Nitric oxide (NO) stimulates natriuresis and diuresis, but its role is unknown during tolvaptan treatment in ADPKD. METHODS Eighteen patients with ADPKD received tolvaptan 60 mg or placebo in a randomized, placebo-controlled, double blind, crossover study. L-NMMA (L-NG-monomethyl-arginine) was given as a bolus followed by continuous infusion during 60 min. We measured: GFR, urine output (UO), free water clearance (CH2O), fractional excretion of sodium (FENa), urinary excretion of aquaporin-2 channels (u-AQP2) and epithelial sodium channels (u-ENaCγ), plasma concentrations of vasopressin (p-AVP), renin (PRC), angiotensinII (p-AngII), aldosterone (p-Aldo), and central blood pressure (cBP). RESULTS During tolvaptan with NO-inhibition, a more pronounced decrease was measured in UO, CH2O (61% vs 43%) and FENa (46% vs 41%) after placebo than after tolvaptan; GFR and u-AQP2 decreased to the same extent; p-AVP increased three fold, whereas u-ENaCγ, PRC, p-AngII, and p-Aldo remained unchanged. After NO-inhibition, GFR increased after placebo and remained unchanged after tolvaptan (5% vs -6%). Central diastolic BP (CDBP) increased to a higher level after placebo than tolvaptan. Body weight fell during tolvaptan treatment. CONCLUSIONS During NO inhibition, tolvaptan antagonized both the antidiuretic and the antinatriuretic effect of L-NMMA, partly via an AVP-dependent mechanism. U-AQP2 was not changed by tolvaptan, presumeably due to a counteracting effect of elevated p-AVP. The reduced GFR during tolvaptan most likely is caused by the reduction in extracellular fluid volume and blood pressure. TRIAL REGISTRATION Clinical Trial no: NCT02527863 . Registered 18 February 2015.
Collapse
Affiliation(s)
- Safa Al Therwani
- University Clinic in Nephrology and Hypertension, Department of Medical Research, Holstebro Hospital and Aarhus University, Hospital Unit Jutland West, Laegaardvej 12, 7500 Holstebro, Denmark
| | - My Emma Sofie Malmberg
- University Clinic in Nephrology and Hypertension, Department of Medical Research, Holstebro Hospital and Aarhus University, Hospital Unit Jutland West, Laegaardvej 12, 7500 Holstebro, Denmark
| | - Jeppe Bakkestroem Rosenbaek
- University Clinic in Nephrology and Hypertension, Department of Medical Research, Holstebro Hospital and Aarhus University, Hospital Unit Jutland West, Laegaardvej 12, 7500 Holstebro, Denmark
| | - Jesper Noergaard Bech
- University Clinic in Nephrology and Hypertension, Department of Medical Research, Holstebro Hospital and Aarhus University, Hospital Unit Jutland West, Laegaardvej 12, 7500 Holstebro, Denmark
| | - Erling Bjerregaard Pedersen
- University Clinic in Nephrology and Hypertension, Department of Medical Research, Holstebro Hospital and Aarhus University, Hospital Unit Jutland West, Laegaardvej 12, 7500 Holstebro, Denmark
| |
Collapse
|
40
|
Cheung PW, Ueberdiek L, Day J, Bouley R, Brown D. Protein phosphatase 2C is responsible for VP-induced dephosphorylation of AQP2 serine 261. Am J Physiol Renal Physiol 2017; 313:F404-F413. [PMID: 28381458 PMCID: PMC5582913 DOI: 10.1152/ajprenal.00004.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/28/2017] [Accepted: 04/03/2017] [Indexed: 12/11/2022] Open
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.
Collapse
Affiliation(s)
- Pui W Cheung
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lars Ueberdiek
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jack Day
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Richard Bouley
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dennis Brown
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
41
|
Gonzalez-Vicente A, Garvin JL. Effects of Reactive Oxygen Species on Tubular Transport along the Nephron. Antioxidants (Basel) 2017; 6:antiox6020023. [PMID: 28333068 PMCID: PMC5488003 DOI: 10.3390/antiox6020023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/15/2017] [Accepted: 03/18/2017] [Indexed: 12/17/2022] Open
Abstract
Reactive oxygen species (ROS) are oxygen-containing molecules naturally occurring in both inorganic and biological chemical systems. Due to their high reactivity and potentially damaging effects to biomolecules, cells express a battery of enzymes to rapidly metabolize them to innocuous intermediaries. Initially, ROS were considered by biologists as dangerous byproducts of respiration capable of causing oxidative stress, a condition in which overproduction of ROS leads to a reduction in protective molecules and enzymes and consequent damage to lipids, proteins, and DNA. In fact, ROS are used by immune systems to kill virus and bacteria, causing inflammation and local tissue damage. Today, we know that the functions of ROS are not so limited, and that they also act as signaling molecules mediating processes as diverse as gene expression, mechanosensation, and epithelial transport. In the kidney, ROS such as nitric oxide (NO), superoxide (O₂-), and their derivative molecules hydrogen peroxide (H₂O₂) and peroxynitrite (ONO₂-) regulate solute and water reabsorption, which is vital to maintain electrolyte homeostasis and extracellular fluid volume. This article reviews the effects of NO, O₂-, ONO₂-, and H₂O₂ on water and electrolyte reabsorption in proximal tubules, thick ascending limbs, and collecting ducts, and the effects of NO and O₂- in the macula densa on tubuloglomerular feedback.
Collapse
Affiliation(s)
- Agustin Gonzalez-Vicente
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires C1113AAD, Argentina.
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
42
|
Al Therwani S, Rosenbæk JB, Mose FH, Bech JN, Pedersen EB. Effect of tolvaptan on renal water and sodium excretion and blood pressure during nitric oxide inhibition: a dose-response study in healthy subjects. BMC Nephrol 2017; 18:86. [PMID: 28288570 PMCID: PMC5347830 DOI: 10.1186/s12882-017-0501-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/07/2017] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Tolvaptan is a selective vasopressin receptor antagonist. Nitric Oxide (NO) promotes renal water and sodium excretion, but the effect is unknown in the nephron's principal cells. In a dose-response study, we measured the effect of tolvaptan on renal handling of water and sodium and systemic hemodynamics, during baseline and NO-inhibition with L-NMMA (L-NG-monomethyl-arginine). METHODS In a randomized, placebo-controlled, double blind, cross over study, 15 healthy subjects received tolvaptan 15, 30 and 45 mg or placebo. L-NMMA was given as a bolus followed by continuous infusion during 60 min. We measured urine output (UO), free water clearance (CH2O), fractional excretion of sodium (FENa), urinary aquaporin-2 channels (u-AQP2) and epithelial sodium channels (u-ENaCγ), plasma vasopressin (p-AVP) and central blood pressure (cBP). RESULTS During baseline, FENa was unchanged. Tolvaptan decreased u-ENaCγ dose-dependently and increased p-AVP threefold, whereas u-AQP2 was unchanged. During tolvaptan with NO-inhibition, UO and CH2O decreased dose-dependently. FENa decreased dose-independently and u-ENaCγ remained unchanged. Central BP increased equally after all treatments. CONCLUSIONS During baseline, fractional excretion of sodium was unchanged. During tolvaptan with NO-inhibition, renal water excretion was reduced dose dependently, and renal sodium excretion was reduced unrelated to the dose, partly via an AVP dependent mechanism. Thus, tolvaptan antagonized the reduction in renal water and sodium excretion during NO-inhibition. Most likely, the lack of decrease in AQP2 excretion by tolvaptan could be attributed to a counteracting effect of the high level of p-AVP. TRIAL REGISTRATION Clinical Trial no: NCT02078973 . Registered 1 March 2014.
Collapse
Affiliation(s)
- Safa Al Therwani
- University Clinic in Nephrology and Hypertension, Department of Medical Research, Holstebro Hospital, Aarhus University, Hospital Unit Jutland West, Laegaardvej 12, 7500 Holstebro, Denmark
| | - Jeppe Bakkestrøm Rosenbæk
- University Clinic in Nephrology and Hypertension, Department of Medical Research, Holstebro Hospital, Aarhus University, Hospital Unit Jutland West, Laegaardvej 12, 7500 Holstebro, Denmark
| | - Frank Holden Mose
- University Clinic in Nephrology and Hypertension, Department of Medical Research, Holstebro Hospital, Aarhus University, Hospital Unit Jutland West, Laegaardvej 12, 7500 Holstebro, Denmark
| | - Jesper Nørgaard Bech
- University Clinic in Nephrology and Hypertension, Department of Medical Research, Holstebro Hospital, Aarhus University, Hospital Unit Jutland West, Laegaardvej 12, 7500 Holstebro, Denmark
| | - Erling Bjerregaard Pedersen
- University Clinic in Nephrology and Hypertension, Department of Medical Research, Holstebro Hospital, Aarhus University, Hospital Unit Jutland West, Laegaardvej 12, 7500 Holstebro, Denmark
| |
Collapse
|
43
|
Abstract
Aquaporins (AQPs ) are a family of membrane water channels that basically function as regulators of intracellular and intercellular water flow. To date, thirteen AQPs , which are distributed widely in specific cell types in various organs and tissues, have been characterized in humans. Four AQP monomers, each of which consists of six membrane-spanning alpha-helices that have a central water-transporting pore, assemble to form tetramers, forming the functional units in the membrane. AQP facilitates osmotic water transport across plasma membranes and thus transcellular fluid movement. The cellular functions of aquaporins are regulated by posttranslational modifications , e.g. phosphorylation, ubiquitination, glycosylation, subcellular distribution, degradation, and protein interactions. Insight into the molecular mechanisms responsible for regulated aquaporin trafficking and synthesis is proving to be fundamental for development of novel therapeutic targets or reliable diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Chunling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, 74# Zhongshan Er Road, Guangzhou, 510080, China
| | - Weidong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, 74# Zhongshan Er Road, Guangzhou, 510080, China.
| |
Collapse
|
44
|
Wang TD, Tan RS, Lee HY, Ihm SH, Rhee MY, Tomlinson B, Pal P, Yang F, Hirschhorn E, Prescott MF, Hinder M, Langenickel TH. Effects of Sacubitril/Valsartan (LCZ696) on Natriuresis, Diuresis, Blood Pressures, and NT-proBNP in Salt-Sensitive Hypertension. Hypertension 2017; 69:32-41. [DOI: 10.1161/hypertensionaha.116.08484] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 09/26/2016] [Accepted: 10/20/2016] [Indexed: 11/16/2022]
Abstract
Salt-sensitive hypertension (SSH) is characterized by impaired sodium excretion and subnormal vasodilatory response to salt loading. Sacubitril/valsartan (LCZ696) was hypothesized to increase natriuresis and diuresis and result in superior blood pressure control compared with valsartan in Asian patients with SSH. In this randomized, double-blind, crossover study, 72 patients with SSH received sacubitril/valsartan 400 mg and valsartan 320 mg once daily for 4 weeks each. SSH was diagnosed if the mean arterial pressure increased by ≥10% when patients switched from low (50 mmol/d) to high (320 mmol/d) sodium diet. The primary outcome was cumulative 6- and 24-hour sodium excretion after first dose administration. Compared with valsartan, sacubitril/valsartan was associated with a significant increase in natriuresis (adjusted treatment difference: 24.5 mmol/6 hours, 50.3 mmol/24 hours, both
P
<0.001) and diuresis (adjusted treatment difference: 291.2 mL/6 hours,
P
<0.001; 356.4 mL/24 hours,
P
=0.002) on day 1, but not on day 28, and greater reductions in office and ambulatory blood pressure on day 28. Despite morning dosing of both drugs, ambulatory blood pressure reductions were more pronounced at nighttime than at daytime or the 24-hour average. Compared with valsartan, sacubitril/valsartan significantly reduced N-terminal pro B-type natriuretic peptide levels on day 28 (adjusted treatment difference: −20%;
P
=0.001). Sacubitril/valsartan and valsartan were safe and well tolerated with no significant changes in body weight or serum sodium and potassium levels with either treatments. In conclusion, sacubitril/valsartan compared with valsartan was associated with short-term increases in natriuresis and diuresis, superior office and ambulatory blood pressure control, and significantly reduced N-terminal pro B-type natriuretic peptide levels in Asian patients with SSH.
Clinical Trial Registration—
URL:
http://www.clinicaltrials.gov
. Unique identifier: NCT01681576.
Collapse
Affiliation(s)
- Tzung-Dau Wang
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Ru-San Tan
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Hae-Young Lee
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Sang-Hyun Ihm
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Moo-Yong Rhee
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Brian Tomlinson
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Parasar Pal
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Fan Yang
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Elizabeth Hirschhorn
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Margaret F. Prescott
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Markus Hinder
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Thomas H. Langenickel
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| |
Collapse
|
45
|
Wnt5a induces renal AQP2 expression by activating calcineurin signalling pathway. Nat Commun 2016; 7:13636. [PMID: 27892464 PMCID: PMC5133730 DOI: 10.1038/ncomms13636] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/20/2016] [Indexed: 12/27/2022] Open
Abstract
Heritable nephrogenic diabetes insipidus (NDI) is characterized by defective urine concentration mechanisms in the kidney, which are mainly caused by loss-of-function mutations in the vasopressin type 2 receptor. For the treatment of heritable NDI, novel strategies that bypass the defective vasopressin type 2 receptor are required to activate the aquaporin-2 (AQP2) water channel. Here we show that Wnt5a regulates AQP2 protein expression, phosphorylation and trafficking, suggesting that Wnt5a is an endogenous ligand that can regulate AQP2 without the activation of the classic vasopressin/cAMP signalling pathway. Wnt5a successfully increases the apical membrane localization of AQP2 and urine osmolality in an NDI mouse model. We also demonstrate that calcineurin is a key regulator of Wnt5a-induced AQP2 activation without affecting intracellular cAMP level and PKA activity. The importance of calcineurin is further confirmed with its activator, arachidonic acid, which shows vasopressin-like effects underlining that calcineurin activators may be potential therapeutic targets for heritable NDI.
Collapse
|
46
|
Mamuya FA, Cano-Peñalver JL, Li W, Rodriguez Puyol D, Rodriguez Puyol M, Brown D, de Frutos S, Lu HAJ. ILK and cytoskeletal architecture: an important determinant of AQP2 recycling and subsequent entry into the exocytotic pathway. Am J Physiol Renal Physiol 2016; 311:F1346-F1357. [PMID: 27760768 DOI: 10.1152/ajprenal.00336.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/29/2016] [Accepted: 10/11/2016] [Indexed: 12/20/2022] Open
Abstract
Within the past decade tremendous efforts have been made to understand the mechanism behind aquaporin-2 (AQP2) water channel trafficking and recycling, to open a path toward effective diabetes insipidus therapeutics. A recent study has shown that integrin-linked kinase (ILK) conditional-knockdown mice developed polyuria along with decreased AQP2 expression. To understand whether ILK also regulates AQP2 trafficking in kidney tubular cells, we performed in vitro analysis using LLCPK1 cells stably expressing rat AQP2 (LLC-AQP2 cells). Upon treatment of LLC-AQP2 cells with ILK inhibitor cpd22 and ILK-siRNA, we observed increased accumulation of AQP2 in the perinuclear region, without any significant increase in the rate of endocytosis. This perinuclear accumulation did not occur in cells expressing a serine-256-aspartic acid mutation that retains AQP2 in the plasma membrane. We then examined clathrin-mediated endocytosis after ILK inhibition using rhodamine-conjugated transferrin. Despite no differences in overall transferrin endocytosis, the endocytosed transferrin also accumulated in the perinuclear region where it colocalized with AQP2. These accumulated vesicles also contained the recycling endosome marker Rab11. In parallel, the usual vasopressin-induced AQP2 membrane accumulation was prevented after ILK inhibition; however, ILK inhibition did not measurably affect AQP2 phosphorylation at serine-256 or its dephosphorylation at serine-261. Instead, we found that inhibition of ILK increased F-actin polymerization. When F-actin was depolymerized with latrunculin, the perinuclear located AQP2 dispersed. We conclude that ILK is important in orchestrating dynamic cytoskeletal architecture during recycling of AQP2, which is necessary for its subsequent entry into the exocytotic pathway.
Collapse
Affiliation(s)
- Fahmy A Mamuya
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jose Luis Cano-Peñalver
- Department of Systems Biology, Physiology Unit, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, Madrid, Spain; and
| | - Wei Li
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Diego Rodriguez Puyol
- Biomedical Research Foundation and Nephrology Department, Hospital Príncipe de Asturias, Alcalá de Henares, and Instituto Reina Sofia de Investigación Renal and REDinREN from Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Rodriguez Puyol
- Department of Systems Biology, Physiology Unit, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, Madrid, Spain; and
| | - Dennis Brown
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sergio de Frutos
- Department of Systems Biology, Physiology Unit, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, Madrid, Spain; and
| | - Hua Ann Jenny Lu
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
47
|
Cheung PW, Nomura N, Nair AV, Pathomthongtaweechai N, Ueberdiek L, Lu HAJ, Brown D, Bouley R. EGF Receptor Inhibition by Erlotinib Increases Aquaporin 2-Mediated Renal Water Reabsorption. J Am Soc Nephrol 2016; 27:3105-3116. [PMID: 27694161 PMCID: PMC5042667 DOI: 10.1681/asn.2015080903] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/28/2016] [Indexed: 01/04/2023] Open
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.
Collapse
Affiliation(s)
- Pui W Cheung
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Naohiro Nomura
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Anil V Nair
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nutthapoom Pathomthongtaweechai
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lars Ueberdiek
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hua A Jenny Lu
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dennis Brown
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Richard Bouley
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
48
|
Olesen ETB, Moeller HB, Assentoft M, MacAulay N, Fenton RA. The vasopressin type 2 receptor and prostaglandin receptors EP2 and EP4 can increase aquaporin-2 plasma membrane targeting through a cAMP-independent pathway. Am J Physiol Renal Physiol 2016; 311:F935-F944. [PMID: 27558562 DOI: 10.1152/ajprenal.00559.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 08/18/2016] [Indexed: 01/16/2023] Open
Abstract
Apical membrane targeting of the collecting duct water channel aquaporin-2 (AQP2) is essential for body water balance. As this event is regulated by Gs coupled 7-transmembrane receptors such as the vasopressin type 2 receptor (V2R) and the prostanoid receptors EP2 and EP4, it is believed to be cAMP dependent. However, on the basis of recent reports, it was hypothesized in the current study that increased cAMP levels are not necessary for AQP2 membrane targeting. The role and dynamics of cAMP signaling in AQP2 membrane targeting in Madin-Darby canine kidney and mouse cortical collecting duct (mpkCCD14) cells was examined using selective agonists against the V2R (dDAVP), EP2 (butaprost), and EP4 (CAY10580). During EP2 stimulation, AQP2 membrane targeting continually increased during 80 min of stimulation; whereas cAMP levels reached a plateau after 10 min. EP4 stimulation caused a rapid and transient increase in AQP2 membrane targeting, but did not significantly increase cAMP levels. After washout of the EP2 agonist or dDAVP, AQP2 membrane abundance remained elevated for at least 80 min, whereas cAMP levels rapidly decreased. Similar effects of the EP2 agonist were also observed for AQP2 constitutively nonphosphorylated at ser-269. The adenylyl cyclase inhibitor SQ22536 did not prevent AQP2 targeting during stimulation of each receptor, nor after dDAVP washout. In conclusion, this study demonstrates that although direct stimulation with cAMP causes AQP2 membrane targeting, cAMP is not necessary for receptor-mediated AQP2 membrane targeting and Gs-coupled receptors can also signal through an alternative pathway that increases AQP2 membrane targeting.
Collapse
Affiliation(s)
- Emma T B Olesen
- Department of Biomedicine, InterPrET Center, Health, Aarhus University, Aarhus C, Denmark; .,Department of Neuroscience and Pharmacology, Faculty of Health Sciences, Copenhagen University, Copenhagen, Denmark; and.,Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hanne B Moeller
- Department of Biomedicine, InterPrET Center, Health, Aarhus University, Aarhus C, Denmark
| | - Mette Assentoft
- Department of Neuroscience and Pharmacology, Faculty of Health Sciences, Copenhagen University, Copenhagen, Denmark; and
| | - Nanna MacAulay
- Department of Neuroscience and Pharmacology, Faculty of Health Sciences, Copenhagen University, Copenhagen, Denmark; and
| | - Robert A Fenton
- Department of Biomedicine, InterPrET Center, Health, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
49
|
Al-Bataineh MM, Li H, Ohmi K, Gong F, Marciszyn AL, Naveed S, Zhu X, Neumann D, Wu Q, Cheng L, Fenton RA, Pastor-Soler NM, Hallows KR. Activation of the metabolic sensor AMP-activated protein kinase inhibits aquaporin-2 function in kidney principal cells. Am J Physiol Renal Physiol 2016; 311:F890-F900. [PMID: 27534994 PMCID: PMC5130465 DOI: 10.1152/ajprenal.00308.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/15/2016] [Indexed: 11/22/2022] Open
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-β-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.
Collapse
Affiliation(s)
- Mohammad M Al-Bataineh
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hui Li
- Department of Medicine, University of Southern California/University Kidney Research Organization, Kidney Research Center, Division of Nephrology and Hypertension, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Kazuhiro Ohmi
- Department of Medicine, University of Southern California/University Kidney Research Organization, Kidney Research Center, Division of Nephrology and Hypertension, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Fan Gong
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Allison L Marciszyn
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sajid Naveed
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xiaoqing Zhu
- Department of Molecular Genetics, School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands; and
| | - Dietbert Neumann
- Department of Molecular Genetics, School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands; and
| | - Qi Wu
- Department of Biomedicine, InterPrET Center, Aarhus University, Aarhus, Denmark
| | - Lei Cheng
- Department of Biomedicine, InterPrET Center, Aarhus University, Aarhus, Denmark
| | - Robert A Fenton
- Department of Biomedicine, InterPrET Center, Aarhus University, Aarhus, Denmark
| | - Núria M Pastor-Soler
- Department of Medicine, University of Southern California/University Kidney Research Organization, Kidney Research Center, Division of Nephrology and Hypertension, Keck School of Medicine of University of Southern California, Los Angeles, California;
| | - Kenneth R Hallows
- Department of Medicine, University of Southern California/University Kidney Research Organization, Kidney Research Center, Division of Nephrology and Hypertension, Keck School of Medicine of University of Southern California, Los Angeles, California
| |
Collapse
|
50
|
TRPV4 is associated with central rather than nephrogenic osmoregulation. Pflugers Arch 2016; 468:1595-607. [PMID: 27364478 DOI: 10.1007/s00424-016-1850-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 12/28/2022]
Abstract
TRPV4 is a polymodal cation channel expressed in osmosensitive neurons of the hypothalamus and in the mammalian nephron. The segmental distribution and role(s) of TRPV4 in osmoregulation remain debated. We investigated the renal distribution pattern of TRPV4 and the functional consequences of its disruption in mouse models. Using qPCR on microdissected segments, immunohistochemistry, and a LacZ reporter mouse, we found that TRPV4 is abundantly expressed in the proximal tubule, the late distal convoluted tubule, and throughout the connecting tubule and collecting duct, including principal and intercalated cells. TRPV4 was undetectable in the glomeruli and thick ascending limb and weakly abundant in the early distal convoluted tubule. Metabolic studies in Trpv4 (+/+) and Trpv4 (-/-) littermates revealed that the lack of TRPV4 did not influence activity, food and water intake, renal function, and urinary concentration at baseline. The mice showed a similar response to furosemide, water loading and deprivation, acid loading, and dietary NaCl restriction. However, Trpv4 (-/-) mice showed a significantly lower vasopressin synthesis and release after water deprivation, with a loss of the positive correlation between plasma osmolality and plasma vasopressin levels, and a delayed water intake upon acute administration of hypertonic saline. Specific activation of TRPV4 in primary cultures of proximal tubule cells increased albumin uptake, whereas no effect of TRPV4 deletion could be observed at baseline. These data reveal that, despite its abundant expression in tubular segments, TRPV4 does not play a major role in the kidney or is efficiently compensated when deleted. Instead, TRPV4 is critical for the release of vasopressin, the sensation of thirst, and the central osmoregulation.
Collapse
|