1
|
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] [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
|
2
|
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
|
3
|
Nagase H, Shitara A, Ohno Y, Satoh K, Kashimata M. Loss of Cdc42 in Exocrine Acini Decreases Saliva Secretion but Increases Tear Secretion-A Potential Model of Exocrine Gland Senescence. Int J Mol Sci 2023; 24:17220. [PMID: 38139048 PMCID: PMC10743476 DOI: 10.3390/ijms242417220] [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: 10/19/2023] [Revised: 11/17/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Cdc42 is a small GTPase essential for the cell cycle, morphogenesis, and cell adhesion, and it is involved in the polarity of epithelial cells. However, the functional roles of Cdc42 in exocrine glands, such as the maintenance of acini and water secretion, are not yet well understood. In this study, we generated acinar-cell-specific Cdc42 conditional knockout (Cdc42cKO) mice to assess their maintenance of acinar cells and physiological functions in the salivary glands (SGs) and lacrimal glands (LGs). Our data revealed that the loss of Cdc42 altered the luminal structures to bulging structures and induced acinar cell apoptosis in both the parotid glands (PGs) and LGs of Cdc42cKO mice. Interestingly, saliva secretion in response to pilocarpine stimulation was decreased in the Cdc42cKO group, whereas tear secretion was increased. Consistent with the water secretion results, protein expression of the water channel AQP5 in acinar cells was also decreased in the PGs but conversely increased in the LGs. Moreover, the changes that increased AQP5 expression in LGs occurred in the acinar cells rather than the duct cells. The present study demonstrates that Cdc42 is involved in the structural and survival maintenance of acinar cells in SGs and LGs. On the other hand, depletion of Cdc42 caused the opposite physiological phenomena between PGs and LGs.
Collapse
Affiliation(s)
- Haruna Nagase
- Department of Pharmacology, Asahi University School of Dentistry, 1851-1 Hozumi, Mizuho, Gifu 501-0296, Japan
| | - Akiko Shitara
- Department of Pharmacology, Asahi University School of Dentistry, 1851-1 Hozumi, Mizuho, Gifu 501-0296, Japan
| | - Yuta Ohno
- Department of Pharmacology, Asahi University School of Dentistry, 1851-1 Hozumi, Mizuho, Gifu 501-0296, Japan
| | - Keitaro Satoh
- Department of Pharmacology, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado, Saitama 350-0283, Japan
| | - Masanori Kashimata
- Department of Pharmacology, Asahi University School of Dentistry, 1851-1 Hozumi, Mizuho, Gifu 501-0296, Japan
| |
Collapse
|
4
|
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
|
5
|
Zhao X, Liang B, Li C, Wang W. Expression Regulation and Trafficking of Aquaporins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1398:39-51. [PMID: 36717485 DOI: 10.1007/978-981-19-7415-1_3] [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) mediate the bidirectional water flow driven by an osmotic gradient. Either gating or trafficking allows for rapid and specific AQP regulation in a tissue-dependent manner. The regulatory mechanisms of AQP2 are discussed mainly in this chapter, as the mechanisms controlling the regulation and trafficking of AQP2 have been very well studied. The targeting of AQP2 to the apical plasma membrane of collecting duct principal cells is mainly regulated by the action of arginine vasopressin (AVP) on the type 2 AVP receptor (V2R), which cause increased intracellular cAMP or elevated intracellular calcium levels. Activation of these intracellular signaling pathways results in vesicles bearing AQP2 transport, docking and fusion with the apical membrane, which increase density of AQP2 on the membrane. The removal of AQP2 from the membrane requires dynamic cytoskeletal remodeling. AQP2 is degraded through the ubiquitin proteasome pathway and lysosomal proteolysis pathway. Finally, we review updated findings in transcriptional and epigenetic regulation of AQP2.
Collapse
Affiliation(s)
- Xiaoduo Zhao
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Baien Liang
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Chunling Li
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Weidong Wang
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
6
|
Lu HAJ, He J. Aquaporins in Diabetes Insipidus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1398:267-279. [PMID: 36717500 DOI: 10.1007/978-981-19-7415-1_18] [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
Disruption of water and electrolyte balance is frequently encountered in clinical medicine. Regulating water metabolism is critically important. Diabetes insipidus (DI) presented with excessive water loss from the kidney is a major disorder of water metabolism. To understanding the molecular and cellular mechanisms and pathophysiology of DI and rationales of clinical management of DI is important for both research and clinical practice. This chapter will first review various forms of DI focusing on central diabetes insipidus (CDI) and nephrogenic diabetes insipidus (NDI). This is followed by a discussion of regulatory mechanisms underlying CDI and NDI, with a focus on the regulatory axis of vasopressin, vasopressin receptor 2 (V2R) and the water channel molecule, aquaporin 2 (AQP2). The clinical manifestation, diagnosis, and management of various forms of DI will also be discussed with highlights of some of the latest therapeutic strategies that are developed from in vitro experiments and animal studies.
Collapse
Affiliation(s)
- H A Jenny Lu
- Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Jinzhao He
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
7
|
Soh JF, Bodenstein K, Yu OHY, Linnaranta O, Renaud S, Mahdanian A, Su CL, Mucsi I, Mulsant B, Herrmann N, Rajji T, Beaulieu S, Sekhon H, Rej S. Atorvastatin lowers serum calcium levels in lithium-users: results from a randomized controlled trial. BMC Endocr Disord 2022; 22:238. [PMID: 36153583 PMCID: PMC9508741 DOI: 10.1186/s12902-022-01145-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 08/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although lithium is considered the gold-standard treatment for bipolar disorder (BD), it is associated with a variety of major endocrine and metabolic side effects, including parathyroid hormone (PTH) dependent hypercalcemia. Aside from surgery and medication discontinuation, there are limited treatments for hypercalcemia. This paper will assess data from a randomized controlled trial (RCT). METHODS This is a secondary analysis of an RCT that explored the effects of atorvastatin (n = 27) versus placebo (n = 33) on lithium-induced nephrogenic diabetes insipidus (NDI) in patients with BD and major depressive disorder (MDD) using lithium (n = 60), over a 12-week period. This secondary analysis will explore serum calcium levels and thyroid stimulating hormone (TSH) measured at baseline, week 4, and week 12. RESULTS At 12-weeks follow-up while adjusting results for baseline, linear regression analyses found that corrected serum calcium levels were significantly lower in the treatment group (mean (M) = 2.30 mmol/L, standard deviation (SD) = 0.07) compared to the placebo group (M = 2.33 mmol/L, SD = 0.07) (β = - 0.03 (95% C.I.; - 0.0662, - 0.0035), p = 0.03) for lithium users. There were no significant changes in TSH. CONCLUSION In lithium users with relatively normal calcium levels, receiving atorvastatin was associated with a decrease in serum calcium levels. Although exciting, this is a preliminary finding that needs further investigation with hypercalcemic patients. Future RCTs could examine whether atorvastatin can treat PTH dependent hypercalcemia due to lithium and other causes.
Collapse
Affiliation(s)
- Jocelyn Fotso Soh
- GeriPARTy Research Group, Jewish General Hospital, Montreal, Canada
- Department of Psychology, Concordia University, Montreal, Canada
| | - Katie Bodenstein
- GeriPARTy Research Group, Jewish General Hospital, Montreal, Canada
- Department of Psychiatry, McGill University, 1033 Avenue des Pins, Montreal, H3A 1A1, Canada
| | - Oriana Hoi Yun Yu
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada
- Division of Endocrinology and Metabolism, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Outi Linnaranta
- Department of Psychiatry, McGill University, 1033 Avenue des Pins, Montreal, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
- National Institute for Health and Welfare, Helsinki, Finland
| | - Suzane Renaud
- Department of Psychiatry, McGill University, 1033 Avenue des Pins, Montreal, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Artin Mahdanian
- Department of Psychiatry, McGill University, 1033 Avenue des Pins, Montreal, H3A 1A1, Canada
- Department of Psychiatry, Jewish General Hospital, Montreal, Canada
| | - Chien-Lin Su
- Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada
| | - Istvan Mucsi
- Multiorgan Transplant Program, University Health Network and Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Canada
| | - Benoit Mulsant
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Department of Psychiatry, Centre for Addictions and Mental Health, University of Toronto, Toronto, Canada
| | - Nathan Herrmann
- Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Tarek Rajji
- Department of Psychiatry, Centre for Addictions and Mental Health, University of Toronto, Toronto, Canada
| | - Serge Beaulieu
- Department of Psychiatry, McGill University, 1033 Avenue des Pins, Montreal, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Harmehr Sekhon
- GeriPARTy Research Group, Jewish General Hospital, Montreal, Canada.
- Postdoctoral Research Fellow, Department of Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Soham Rej
- GeriPARTy Research Group, Jewish General Hospital, Montreal, Canada
- Department of Psychiatry, McGill University, 1033 Avenue des Pins, Montreal, H3A 1A1, Canada
- McGill Meditation and Mind-Body Medicine Research Clinic (MMMM-RC), Montreal, Canada
| |
Collapse
|
8
|
Takata T, Hamada S, Mae Y, Iyama T, Ogihara R, Seno M, Nakamura K, Takata M, Sugihara T, Isomoto H. Uromodulin Regulates Murine Aquaporin-2 Activity via Thick Ascending Limb-Collecting Duct Cross-Talk during Water Deprivation. Int J Mol Sci 2022; 23:ijms23169410. [PMID: 36012675 PMCID: PMC9408883 DOI: 10.3390/ijms23169410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 12/03/2022] Open
Abstract
Uromodulin, a urinary protein synthesized and secreted from the thick ascending limb (TAL) of the loop of Henle, is associated with hypertension through the activation of sodium reabsorption in the TAL. Uromodulin is a potential target for hypertension treatment via natriuresis. However, its biological function in epithelial cells of the distal nephron segment, particularly the collecting duct, remains unknown. Herein, we examined the regulation of uromodulin production during water deprivation in vivo as well as the effect of uromodulin on the activity of the water channel aquaporin−2 (AQP2) in vitro and in vivo using transgenic mice. Water deprivation upregulated uromodulin production; immunofluorescence experiments revealed uromodulin adhesion on the apical surface of the collecting duct. Furthermore, the activation of AQP2 was attenuated in mice lacking uromodulin. Uromodulin enhanced the phosphorylation and apical trafficking of AQP2 in mouse collecting duct cells treated with the vasopressin analog dDAVP. The uromodulin-induced apical trafficking of AQP2 was attenuated via endocytosis inhibitor treatment, suggesting that uromodulin activates AQP2 through the suppression of endocytosis. This study provides novel insights into the cross−talk between TAL and the collecting duct, and indicates that the modulation of uromodulin is a promising approach for diuresis and hypertension treatment.
Collapse
Affiliation(s)
- Tomoaki Takata
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
- Correspondence: ; Tel.: +81-859-38-6527
| | - Shintaro Hamada
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Yukari Mae
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Takuji Iyama
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Ryohei Ogihara
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Misako Seno
- Advanced Medicine & Translational Research Center, Organization for Research Initiative and Promotion, Tottori University, Yonago 683-8504, Japan
| | - Kazuomi Nakamura
- Advanced Medicine, Innovation and Clinical Research Center, Tottori University Hospital, Yonago 683-8504, Japan
| | - Miki Takata
- Division of Respiratory Medicine and Rheumatology, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Takaaki Sugihara
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Hajime Isomoto
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| |
Collapse
|
9
|
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]
|
10
|
Vaz de Castro PAS, Bitencourt L, de Oliveira Campos JL, Fischer BL, Soares de Brito SBC, Soares BS, Drummond JB, Simões E Silva AC. Nephrogenic diabetes insipidus: a comprehensive overview. J Pediatr Endocrinol Metab 2022; 35:421-434. [PMID: 35146976 DOI: 10.1515/jpem-2021-0566] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/26/2022] [Indexed: 12/14/2022]
Abstract
Nephrogenic diabetes insipidus (NDI) is characterized by the inability to concentrate urine that results in polyuria and polydipsia, despite having normal or elevated plasma concentrations of arginine vasopressin (AVP). In this study, we review the clinical aspects and diagnosis of NDI, the various etiologies, current treatment options and potential future developments. NDI has different clinical manifestations and approaches according to the etiology. Hereditary forms of NDI are mainly caused by mutations in the genes that encode key proteins in the AVP signaling pathway, while acquired causes are normally associated with specific drug exposure, especially lithium, and hydroelectrolytic disorders. Clinical manifestations of the disease vary according to the degree of dehydration and hyperosmolality, being worse when renal water losses cannot be properly compensated by fluid intake. Regarding the diagnosis of NDI, it is important to consider the symptoms of the patient and the diagnostic tests, including the water deprivation test and the baseline plasma copeptin measurement, a stable surrogate biomarker of AVP release. Without proper treatment, patients may developcomplications leading to high morbidity and mortality, such as severe dehydration and hypernatremia. In that sense, the treatment of NDI consists in decreasing the urine output, while allowing appropriate fluid balance, normonatremia, and ensuring an acceptable quality of life. Therefore, therapeutic options include nonpharmacological interventions, including sufficient water intake and a low-sodium diet, and pharmacological treatment. The main medications used for NDI are thiazide diuretics, nonsteroidal anti-inflammatory drugs (NSAIDs), and amiloride, used isolated or in combination.
Collapse
Affiliation(s)
- Pedro Alves Soares Vaz de Castro
- Interdisciplinary Laboratory of Medical Investigation, Unit of Pediatric Nephrology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Letícia Bitencourt
- Interdisciplinary Laboratory of Medical Investigation, Unit of Pediatric Nephrology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Juliana Lacerda de Oliveira Campos
- Interdisciplinary Laboratory of Medical Investigation, Unit of Pediatric Nephrology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Bruna Luisa Fischer
- Interdisciplinary Laboratory of Medical Investigation, Unit of Pediatric Nephrology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Stephanie Bruna Camilo Soares de Brito
- Interdisciplinary Laboratory of Medical Investigation, Unit of Pediatric Nephrology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Beatriz Santana Soares
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, UFMG, Belo Horizonte, Brazil
| | - Juliana Beaudette Drummond
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, UFMG, Belo Horizonte, Brazil
| | - Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, Unit of Pediatric Nephrology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
11
|
Steichen C, Hervé C, Hauet T, Bourmeyster N. Rho GTPases in kidney physiology and diseases. Small GTPases 2022; 13:141-161. [PMID: 34138686 PMCID: PMC9707548 DOI: 10.1080/21541248.2021.1932402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Rho family GTPases are molecular switches best known for their pivotal role in dynamic regulation of the actin cytoskeleton, but also of cellular morphology, motility, adhesion and proliferation. The prototypic members of this family (RhoA, Rac1 and Cdc42) also contribute to the normal kidney function and play important roles in the structure and function of various kidney cells including tubular epithelial cells, mesangial cells and podocytes. The kidney's vital filtration function depends on the structural integrity of the glomerulus, the proximal portion of the nephron. Within the glomerulus, the architecturally actin-based cytoskeleton podocyte forms the final cellular barrier to filtration. The glomerulus appears as a highly dynamic signalling hub that is capable of integrating intracellular cues from its individual structural components. Dynamic regulation of the podocyte cytoskeleton is required for efficient barrier function of the kidney. As master regulators of actin cytoskeletal dynamics, Rho GTPases are therefore of critical importance for sustained kidney barrier function. Dysregulated activities of the Rho GTPases and of their effectors are implicated in the pathogenesis of both hereditary and idiopathic forms of kidney diseases. Diabetic nephropathy is a progressive kidney disease that is caused by injury to kidney glomeruli. High glucose activates RhoA/Rho-kinase in mesangial cells, leading to excessive extracellular matrix production (glomerulosclerosis). This RhoA/Rho-kinase pathway also seems involved in the post-transplant hypertension frequently observed during treatment with calcineurin inhibitors, whereas Rac1 activation was observed in post-transplant ischaemic acute kidney injury.
Collapse
Affiliation(s)
- Clara Steichen
- Inserm UMR-1082 Irtomit, Poitiers, France
- Faculté De Médecine Et De Pharmacie, Université De Poitiers, Poitiers, France
| | | | - Thierry Hauet
- Inserm UMR-1082 Irtomit, Poitiers, France
- Faculté De Médecine Et De Pharmacie, Université De Poitiers, Poitiers, France
- Department of Medical Biology, Service De Biochimie, CHU De Poitiers, Poitiers, France
| | - Nicolas Bourmeyster
- Faculté De Médecine Et De Pharmacie, Université De Poitiers, Poitiers, France
- Department of Medical Biology, Service De Biochimie, CHU De Poitiers, Poitiers, France
- Laboratoire STIM CNRS ERL 7003, Université de Poitiers, Poitiers Cédex, France
| |
Collapse
|
12
|
Kim S, Jo CH, Kim GH. The Role of Vasopressin V2 Receptor in Drug-Induced Hyponatremia. Front Physiol 2021; 12:797039. [PMID: 34955900 PMCID: PMC8703040 DOI: 10.3389/fphys.2021.797039] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022] Open
Abstract
Hyponatremia is frequently encountered in clinical practice and usually induced by renal water retention. Many medications are considered to be among the various causes of hyponatremia, because they either stimulate the release of arginine vasopressin (AVP) or potentiate its action in the kidney. Antidepressants, anticonvulsants, antipsychotics, diuretics, and cytotoxic agents are the major causes of drug-induced hyponatremia. However, studies addressing the potential of these drugs to increase AVP release from the posterior pituitary gland or enhance urine concentration through intrarenal mechanisms are lacking. We previously showed that in the absence of AVP, sertraline, carbamazepine, haloperidol, and cyclophosphamide each increased vasopressin V2 receptor (V2R) mRNA and aquaporin-2 (AQP2) protein and mRNA expression in primary cultured inner medullary collecting duct cells. The upregulation of AQP2 was blocked by the V2R antagonist tolvaptan or protein kinase A (PKA) inhibitors. These findings led us to conclude that the nephrogenic syndrome of inappropriate antidiuresis (NSIAD) is the main mechanism of drug-induced hyponatremia. Previous studies have also shown that the V2R has a role in chlorpropamide-induced hyponatremia. Several other agents, including metformin and statins, have been found to induce antidiuresis and AQP2 upregulation through various V2R-independent pathways in animal experiments but are not associated with hyponatremia despite being frequently used clinically. In brief, drug-induced hyponatremia can be largely explained by AQP2 upregulation from V2R-cAMP-PKA signaling in the absence of AVP stimulation. This paper reviews the central and nephrogenic mechanisms of drug-induced hyponatremia and discusses the importance of the canonical pathway of AQP2 upregulation in drug-induced NSIAD.
Collapse
Affiliation(s)
- Sua Kim
- Institute of Biomedical Science, Hanyang University College of Medicine, Seoul, South Korea
| | - Chor Ho Jo
- Institute of Biomedical Science, Hanyang University College of Medicine, Seoul, South Korea
| | - Gheun-Ho Kim
- Institute of Biomedical Science, Hanyang University College of Medicine, Seoul, South Korea.,Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea
| |
Collapse
|
13
|
Ho CH, Yang HH, Su SH, Yeh AH, Yu MJ. α-Actinin 4 Links Vasopressin Short-Term and Long-Term Regulation of Aquaporin-2 in Kidney Collecting Duct Cells. Front Physiol 2021; 12:725172. [PMID: 34925053 PMCID: PMC8674656 DOI: 10.3389/fphys.2021.725172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 10/31/2021] [Indexed: 12/18/2022] Open
Abstract
Water permeability of the kidney collecting ducts is regulated by the peptide hormone vasopressin. Between minutes and hours (short-term), vasopressin induces trafficking of the water channel protein aquaporin-2 to the apical plasma membrane of the collecting duct principal cells to increase water permeability. Between hours and days (long-term), vasopressin induces aquaporin-2 gene expression. Here, we investigated the mechanisms that bridge the short-term and long-term vasopressin-mediated aquaporin-2 regulation by α-actinin 4, an F-actin crosslinking protein and a transcription co-activator of the glucocorticoid receptor. Vasopressin induced F-actin depolymerization and α-actinin 4 nuclear translocation in the mpkCCD collecting duct cell model. Co-immunoprecipitation followed by immunoblotting showed increased interaction between α-actinin 4 and glucocorticoid receptor in response to vasopressin. ChIP-PCR showed results consistent with α-actinin 4 and glucocorticoid receptor binding to the aquaporin-2 promoter. α-actinin 4 knockdown reduced vasopressin-induced increases in aquaporin-2 mRNA and protein expression. α-actinin 4 knockdown did not affect vasopressin-induced glucocorticoid receptor nuclear translocation, suggesting independent mechanisms of vasopressin-induced nuclear translocation of α-actinin 4 and glucocorticoid receptor. Glucocorticoid receptor knockdown profoundly reduced vasopressin-induced increases in aquaporin-2 mRNA and protein expression. In the absence of glucocorticoid analog dexamethasone, vasopressin-induced increases in glucocorticoid receptor nuclear translocation and aquaporin-2 mRNA were greatly reduced. α-actinin 4 knockdown further reduced vasopressin-induced increase in aquaporin-2 mRNA in the absence of dexamethasone. We conclude that glucocorticoid receptor plays a major role in vasopressin-induced aquaporin-2 gene expression that can be enhanced by α-actinin 4. In the absence of vasopressin, α-actinin 4 crosslinks F-actin underneath the apical plasma membrane, impeding aquaporin-2 membrane insertion. Vasopressin-induced F-actin depolymerization in one hand facilitates aquaporin-2 apical membrane insertion and in the other hand frees α-actinin 4 to enter the nucleus where it binds glucocorticoid receptor to enhance aquaporin-2 gene expression.
Collapse
Affiliation(s)
- Cheng-Hsuan Ho
- College of Medicine, Institute of Biochemistry and Molecular Biology, National Taiwan University, Taipei, Taiwan
| | - Hsiu-Hui Yang
- College of Medicine, Institute of Biochemistry and Molecular Biology, National Taiwan University, Taipei, Taiwan
| | - Shih-Han Su
- College of Medicine, Institute of Biochemistry and Molecular Biology, National Taiwan University, Taipei, Taiwan
| | - Ai-Hsin Yeh
- College of Medicine, Institute of Biochemistry and Molecular Biology, National Taiwan University, Taipei, Taiwan
| | - Ming-Jiun Yu
- College of Medicine, Institute of Biochemistry and Molecular Biology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
14
|
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: 21] [Impact Index Per Article: 5.3] [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
|
15
|
Thomsen ML, Grønkjær C, Iervolino A, Rej S, Trepiccione F, Christensen BM. Atorvastatin does not ameliorate nephrogenic diabetes insipidus induced by lithium or potassium depletion in mice. Physiol Rep 2021; 9:e15111. [PMID: 34762363 PMCID: PMC8582289 DOI: 10.14814/phy2.15111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/15/2021] [Accepted: 10/15/2021] [Indexed: 11/30/2022] Open
Abstract
Acquired forms of nephrogenic diabetes insipidus (NDI) include lithium (Li)-induced and hypokalemia-induced NDI. Both forms are associated with AQP2 downregulation and collecting duct (CD) cellular remodeling. Statins are cholesterol-lowering drugs appearing to increase AQP2 membrane-translocation and improve urine concentration in other NDI models. We have investigated if statins are able to prevent or rescue the Li-induced changes in mice and in a mouse cortical CD cell line (mCCDc1l ). Biotinylation assays showed that acute (1hr) atorvastatin, simvastatin, or fluvastatin increased AQP2 membrane accumulation in mCCDc1l cells showing that the cell line responds to acute statin treatment. To see whether chronic statin treatment abolish the Li effects, mCCDc1l cells were treated with 48 h Li, combined Li/atorvastatin or combined Li/simvastatin. Li reduced AQP2, but combined Li/atorvastatin or Li/simvastatin did not prevent AQP2 downregulation. In mice, chronic (21 days) Li increased urine output and reduced urine osmolality, but combined Li/atorvastatin did not prevent these effects. In inner medulla (IM), Li reduced total AQP2 and increased pS261-AQP2. Combined Li/atorvastatin did not abolish these changes. Atorvastatin did not prevent a Li-induced increase in intercalated cells and proliferation in IM. In mice with already established NDI, atorvastatin had no effect on the Li-induced changes either. Mice subjected to 14 days of potassium-deficient diet developed polyuria and AQP2 downregulation in IM. Co-treatment with atorvastatin did not prevent this. In conclusion, atorvastatin does not appear to be able to prevent or rescue Li-NDI or to prevent hypokalemic-induced NDI.
Collapse
Affiliation(s)
| | | | - Anna Iervolino
- Department of Translational Medical SciencesUniversity of Campania “L. Vanvitelli”NaplesItaly
- Biogem Institute of Molecular Biology and GeneticsAriano IrpinoItaly
| | - Soham Rej
- Jewish General Hospital/Lady Davis Institute/Department of PsychiatryMcGill UniversityMontrealQuebecCanada
| | - Francesco Trepiccione
- Department of Translational Medical SciencesUniversity of Campania “L. Vanvitelli”NaplesItaly
- Biogem Institute of Molecular Biology and GeneticsAriano IrpinoItaly
| | | |
Collapse
|
16
|
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
|
17
|
Valenti G, Tamma G. The vasopressin-aquaporin-2 pathway syndromes. HANDBOOK OF CLINICAL NEUROLOGY 2021; 181:249-259. [PMID: 34238461 DOI: 10.1016/b978-0-12-820683-6.00018-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Vasopressin is the key hormone involved in water conservation and regulation of water balance, essential for life. In the renal collecting duct, vasopressin binds to the V2 receptor, increasing water permeability through activation of aquaporin-2 redistribution to the luminal membrane. This mechanism promotes rapid water reabsorption, important for immediate survival; however, only recently it has become clear that long-term adverse effects are associated with alterations of the vasopressin-aquaporin-2 pathway, leading to several syndromes associated with water balance disorders. The kidney resistance to the vasopressin action may cause severe dehydration for patients and, conversely, nonosmotic release of vasopressin is associated with water retention and increasing the circulatory blood volume. This chapter discusses the relevance of the altered vasopressin-aquaporin-2 pathway in some diseases associated with water balance disorders, including congenital nephrogenic diabetes insipidus, syndrome of inappropriate secretion of antidiuretic hormone, nephrogenic syndrome of inappropriate antidiuresis, and autosomal dominant polycystic kidney disease. The emerging picture suggests that targeting the vasopressin-AQP2 axis can provide therapeutic benefits in those patients.
Collapse
Affiliation(s)
- Giovanna Valenti
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy.
| | - Grazia Tamma
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy
| |
Collapse
|
18
|
Skov J, Falhammar H, Calissendorff J, Lindh JD, Mannheimer B. Association between lipid-lowering agents and severe hyponatremia: a population-based case-control study. Eur J Clin Pharmacol 2021; 77:747-755. [PMID: 33215235 PMCID: PMC8032630 DOI: 10.1007/s00228-020-03006-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/18/2020] [Indexed: 11/17/2022]
Abstract
PURPOSE Drug-induced hyponatremia is common, with medications from many drug-classes implicated. Lipid-lowering agents are among the most prescribed drugs. Limited evidence suggests an inverse association between statins and hyponatremia, while data on other lipid-lowering agents is absent. The objective of this investigation was to study the association between lipid-lowering drugs and hospitalization due to hyponatremia. METHODS This was a register-based case-control study of the general Swedish population. Those hospitalized with a main diagnosis of hyponatremia (n = 11,213) were compared with matched controls (n = 44,801). Multivariable logistic regression adjusting for co-medication, diseases, previous hospitalizations, and socioeconomic factors was used to explore the association between severe hyponatremia and the use of lipid-lowering drugs. RESULTS Unadjusted ORs (95% CI) for hospitalization due to hyponatremia were 1.28 (1.22-1.35) for statins, 1.09 (0.79-1.47) for ezetimibe, 1.38 (0.88-2.12) for fibrates, and 2.12 (1.31-3.35) for resins. After adjustment for confounding factors the adjusted odds ratios (95% CI) compared with controls were 0.69 (0.64-0.74) for statins, 0.60 (0.41-0.86) for ezetimibe, 0.87 (0.51-1.42) for fibrates, and 1.21 (0.69-2.06) for resins. CONCLUSIONS Use of statins and ezetimibe was inversely correlated with severe hyponatremia. Consequently, these drugs are unlikely culprits in patients with hyponatremia, and they appear safe to initiate in hyponatremic patients. A potential protective effect warrants further studies on how statins and other lipid-lowering drugs are linked to dysnatremias.
Collapse
Affiliation(s)
- Jakob Skov
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, Karlstad Central Hospital, Karlstad, Sweden
| | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden
| | - Jan Calissendorff
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden
| | - Jonatan D Lindh
- Department of Laboratory Medicine, Division of Clinical Pharmacology, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Buster Mannheimer
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
19
|
Kim S, Jo CH, Kim GH. Psychotropic drugs upregulate aquaporin-2 via vasopressin-2 receptor/cAMP/protein kinase A signaling in inner medullary collecting duct cells. Am J Physiol Renal Physiol 2021; 320:F963-F971. [PMID: 33843270 DOI: 10.1152/ajprenal.00576.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 04/07/2021] [Indexed: 12/17/2022] Open
Abstract
Psychotropic drugs may be associated with hyponatremia, but an understanding of how they induce water retention in the kidney remains elusive. Previous studies have postulated that they may increase vasopressin production in the hypothalamus without supporting evidence. In this study, we investigated the possibility of drug-induced nephrogenic syndrome of inappropriate antidiuresis using haloperidol, sertraline, and carbamazepine. Haloperidol, sertraline, or carbamazepine were treated in inner medullary collecting duct (IMCD) suspensions and primary cultured IMCD cells prepared from male Sprague-Dawley rats. The responses of intracellular cAMP production, aquaporin-2 (AQP2) protein expression and localization, vasopressin-2 receptor (V2R) and AQP2 mRNA, and cAMP-responsive element-binding protein (CREB) were tested with and without tolvaptan and the protein kinase A (PKA) inhibitors H89 and Rp-cAMPS. In IMCD suspensions, cAMP production was increased by haloperidol, sertraline, or carbamazepine and was relieved by tolvaptan cotreatment. In primary cultured IMCD cells, haloperidol, sertraline, or carbamazepine treatment increased total AQP2 and decreased phosphorylated Ser261-AQP2 protein expression. Notably, these responses were reversed by cotreatment with tolvaptan or a PKA inhibitor. AQP2 membrane trafficking was induced by haloperidol, sertraline, or carbamazepine and was also blocked by cotreatment with tolvaptan or a PKA inhibitor. Furthermore, upregulation of V2R and AQP2 mRNA and phosphorylated CREB was induced by haloperidol, sertraline, or carbamazepine and was blocked by tolvaptan cotreatment. We conclude that, in the rat IMCD, psychotropic drugs upregulate AQP2 via V2R-cAMP-PKA signaling in the absence of vasopressin stimulation. The vasopressin-like action on the kidney appears to accelerate AQP2 transcription and dephosphorylate AQP2 at Ser261.NEW & NOTEWORTHY It is unclear whether antipsychotic drugs can retain water in the kidney in the absence of vasopressin. This study demonstrates that haloperidol, sertraline, and carbamazepine can produce nephrogenic syndrome of inappropriate antidiuresis because they directly upregulate vasopressin-2 receptor and aquaporin-2 (AQP2) via cAMP/PKA signaling. We showed that, in addition to AQP2 trafficking, AQP2 protein abundance was rapidly increased by treatment with antipsychotic drugs in association with dephosphorylation of AQP2 at Ser261 and accelerated AQP2 transcription.
Collapse
Affiliation(s)
- Sua Kim
- Institute of Biomedical Science, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Chor Ho Jo
- Institute of Biomedical Science, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Gheun-Ho Kim
- Institute of Biomedical Science, Hanyang University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
20
|
Klein JD, Khanna I, Pillarisetti R, Hagan RA, LaRocque LM, Rodriguez EL, Sands JM. An AMPK activator as a therapeutic option for congenital nephrogenic diabetes insipidus. JCI Insight 2021; 6:146419. [PMID: 33724959 PMCID: PMC8119225 DOI: 10.1172/jci.insight.146419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/10/2021] [Indexed: 12/27/2022] Open
Abstract
Nephrogenic diabetes insipidus (NDI) patients produce large amounts of dilute urine. NDI can be congenital, resulting from mutations in the type-2 vasopressin receptor (V2R), or acquired, resulting from medications such as lithium. There are no effective treatment options for NDI. Activation of PKA is disrupted in both congenital and acquired NDI, resulting in decreased aquaporin-2 phosphorylation and water reabsorption. We show that adenosine monophosphate–activated protein kinase (AMPK) also phosphorylates aquaporin-2. We identified an activator of AMPK, NDI-5033, and we tested its ability to increase urine concentration in animal models of NDI. NDI-5033 increased AMPK phosphorylation by 2.5-fold, confirming activation. It increased urine osmolality in tolvaptan-treated NDI rats by 30%–50% and in V2R-KO mice by 50%. Metformin, another AMPK activator, can cause hypoglycemia, which makes it a risky option for treating NDI patients, especially children. Rats with NDI receiving NDI-5033 showed no hypoglycemia in a calorie-restricted, exercise protocol. Congenital NDI therapy needs to be effective long-term. We administered NDI-5033 for 3 weeks and saw no reduction in efficacy. We conclude that NDI-5033 can improve urine concentration in animals with NDI and holds promise as a potential therapy for patients with congenital NDI due to V2R mutations.
Collapse
Affiliation(s)
- Janet D Klein
- Emory University, Department of Medicine, Renal Division, Atlanta, Georgia, USA.,NephroDI Therapeutics, Philadelphia, Pennsylvania, USA
| | - Ish Khanna
- NephroDI Therapeutics, Philadelphia, Pennsylvania, USA
| | | | | | - Lauren M LaRocque
- Emory University, Department of Medicine, Renal Division, Atlanta, Georgia, USA
| | - Eva L Rodriguez
- Emory University, Department of Medicine, Renal Division, Atlanta, Georgia, USA
| | - Jeff M Sands
- Emory University, Department of Medicine, Renal Division, Atlanta, Georgia, USA.,NephroDI Therapeutics, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Fotso Soh J, Beaulieu S, Trepiccione F, Linnaranta O, Torres-Platas G, Platt RW, Renaud S, Su CL, Mucsi I, D'Apolito L, Mulsant BH, Levinson A, Saury S, Müller D, Schaffer A, Dols A, Low N, Cervantes P, Christensen BM, Herrmann N, Rajji T, Rej S. A double-blind, randomized, placebo-controlled pilot trial of atorvastatin for nephrogenic diabetes insipidus in lithium users. Bipolar Disord 2021; 23:66-75. [PMID: 32621644 DOI: 10.1111/bdi.12973] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 06/17/2020] [Accepted: 06/24/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Lithium remains an important treatment for mood disorders but is associated with kidney disease. Nephrogenic diabetes insipidus (NDI) is associated with up to 3-fold risk of incident chronic kidney disease among lithium users. There are limited randomized controlled trials (RCT) for treatments of lithium-induced NDI, and existing therapies can be poorly tolerated. Therefore, novel treatments are needed for lithium-induced NDI. METHOD We conducted a 12-week double-blind pilot RCT to assess the feasibility and efficacy of 20 mg/d atorvastatin vs placebo in the treatment of NDI in chronic lithium users. Patients, recruited between September 2017 and October 2018, were aged 18 to 85, currently on a stable dose of lithium, and determined to have NDI. RESULTS Urinary osmolality (UOsm) at 12 weeks adjusted for baseline was not statistically different between groups (+39.6 mOsm/kg [95% CI, -35.3, 114.5] in atorvastatin compared to placebo groups). Secondary outcomes of fluid intake and aquaporin-2 excretions at 12 weeks adjusted for baseline were -0.13 L [95% CI, -0.54, 0.28] and 98.68 [95% CI, -190.34, 387.70], respectively. A moderate effect size was observed for improvements in baseline UOsm by ≥100 mOsm/kg at 12 weeks in patients who received atorvastatin compared to placebo (38.45% (10/26) vs 22.58% (7/31); Cohen's d = 0.66). CONCLUSION Among lithium users with NDI, atorvastatin 20 mg/d did not significantly improve urinary osmolality compared to placebo over a 12-week period. Larger confirmatory trials with longer follow-up periods may help to further assess the effects of statins on NDI, especially within patients with more severe NDI.
Collapse
Affiliation(s)
- Jocelyn Fotso Soh
- Geri-PARTy Research Group, Jewish General Hospital, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Serge Beaulieu
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | | | - Outi Linnaranta
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | | | - Robert W Platt
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University Health Centre, Montreal, QC, Canada
| | - Suzane Renaud
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Chien-Lin Su
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University Health Centre, Montreal, QC, Canada
| | - Istvan Mucsi
- Division of Nephrology, University Health Network, University of Toronto (UofT), Toronto, ON, Canada
| | - Luciano D'Apolito
- Biogem S.c.a.r.l., Istituto di Ricerche Genetiche "Gaetano Salvatore", Ariano Irpino, Italy
| | - Benoit H Mulsant
- Department of Psychiatry, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Andrea Levinson
- Department of Psychiatry, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Sybille Saury
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Daniel Müller
- Department of Psychiatry, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Ayal Schaffer
- Department of Psychiatry, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Annemiek Dols
- Amsterdam UMC, Department of Psychiatry, GGZinGeest, Neuroscience, Amsterdam, The Netherlands
| | - Nancy Low
- Department of Psychiatry, McGill University Health Centre, Montreal, QC, Canada
| | - Pablo Cervantes
- Department of Psychiatry, McGill University Health Centre, Montreal, QC, Canada
| | | | - Nathan Herrmann
- Department of Psychiatry, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Tarek Rajji
- Department of Psychiatry, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Soham Rej
- Geri-PARTy Research Group, Jewish General Hospital, Montreal, QC, Canada.,Douglas Mental Health University Institute, Montreal, QC, Canada
| |
Collapse
|
22
|
Abstract
The hormone arginine vasopressin (AVP) is a nonapeptide synthesized by hypothalamic magnocellular nuclei and secreted from the posterior pituitary into the bloodstream. It binds to AVP receptor 2 in the kidney to promote the insertion of aquaporin channels (AQP2) and antidiuretic responses. AVP secretion deficits produce central diabetes insipidus (CDI), while renal insensitivity to the antidiuretic effect of AVP causes nephrogenic diabetes insipidus (NDI). Hereditary and acquired forms of CDI and NDI generate hypotonic polyuria, polydipsia, hyperosmolality, and hypernatremia. The AVP mutant (Brattleboro) rat is the principal animal model of hereditary CDI, while neurohypophysectomy, pituitary stalk compression, hypophysectomy, and mediobasal hypothalamic lesions produce acquired CDI. In animals, hereditary NDI is mainly caused by mutations in AVP2R or AQP2 genes, while acquired NDI is most frequently induced by lithium. We report here on the determinants of the intake and excretion of water and mineral salts and on the different types of DI in humans. We then describe the hydromineral characteristics of these animal models and the responses observed after administration of hypertonic NaCl or when they are fed with low-sodium diets. Finally, we report on the effects of drugs such as AVP analogues and/or oxytocin, another neuropeptide that increases sodium excretion in animal models and humans with CDI, and sildenafil, a compound that increases the expression and function of AQP2 channels in animal models and humans with NDI.
Collapse
Affiliation(s)
- Javier Mahía
- Department of Psychobiology, and Mind, Brain and Behavior Research Center, University of Granada, Granada, Spain
| | - Antonio Bernal
- Department of Psychobiology, and Mind, Brain and Behavior Research Center, University of Granada, Granada, Spain
| |
Collapse
|
23
|
Kong Y, Feng W, Zhao X, Zhang P, Li S, Li Z, Lin Y, Liang B, Li C, Wang W, Huang H. Statins ameliorate cholesterol-induced inflammation and improve AQP2 expression by inhibiting NLRP3 activation in the kidney. Am J Cancer Res 2020; 10:10415-10433. [PMID: 32929357 PMCID: PMC7482822 DOI: 10.7150/thno.49603] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/07/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Chronic kidney diseases (CKD) are usually associated with dyslipidemia. Statin therapy has been primarily recommended for the prevention of cardiovascular risk in patients with CKD; however, the effects of statins on kidney disease progression remain controversial. This study aims to investigate the effects of statin treatment on renal handling of water in patients and in animals on a high-fat diet. Methods: Retrospective cohort patient data were reviewed and the protein expression levels of aquaporin-2 (AQP2) and NLRP3 inflammasome adaptor ASC were examined in kidney biopsy specimens. The effects of statins on AQP2 and NLRP3 inflammasome components were examined in nlrp3-/- mice, 5/6 nephroectomized (5/6Nx) rats with a high-fat diet (HFD), and in vitro. Results: In the retrospective cohort study, serum cholesterol was negatively correlated to eGFR and AQP2 protein expression in the kidney biopsy specimens. Statins exhibited no effect on eGFR but abolished the negative correlation between cholesterol and AQP2 expression. Whilst nlrp3+/+ mice showed an increased urine output and a decreased expression of AQP2 protein after a HFD, which was moderately attenuated in nlrp3 deletion mice with HFD. In 5/6Nx rats on a HFD, atorvastatin markedly decreased the urine output and upregulated the protein expression of AQP2. Cholesterol stimulated the protein expression of NLRP3 inflammasome components ASC, caspase-1 and IL-1β, and decreased AQP2 protein abundance in vitro, which was markedly prevented by statins, likely through the enhancement of ASC speck degradation via autophagy. Conclusion: Serum cholesterol level has a negative correlation with AQP2 protein expression in the kidney biopsy specimens of patients. Statins can ameliorate cholesterol-induced inflammation by promoting the degradation of ASC speck, and improve the expression of aquaporin in the kidneys of animals on a HFD.
Collapse
|
24
|
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
|
25
|
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
|
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
|
Makita N, Manaka K, Sato J, Iiri T. V2 vasopressin receptor mutations. VITAMINS AND HORMONES 2019; 113:79-99. [PMID: 32138955 DOI: 10.1016/bs.vh.2019.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
V2 vasopressin receptor (V2R) is a member of the G protein-coupled receptor (GPCR) family in which many disease-causing mutations have been identified and thus generated much interest. Loss-of-function V2R mutations cause nephrogenic diabetes insipidus (NDI) whereas gain-of-function mutations cause nephrogenic syndrome of inappropriate antidiuresis (NSIAD). The mechanisms underlying a V2R loss-of-function can be theoretically classified as either protein expression, localization (ER retention) or functional disorders. Functional analyses have revealed however that these mechanisms are likely to be complex. Strikingly, V2R mutations at the same site can result in opposite phenotypes, e.g., R137H and R137L/C cause NDI and NSIAD, respectively. These findings support the notion that the constitutive activation of GPCRs might be often associated with their instability and denaturation. Thus, functional analysis of disease-causing V2R mutations may not only reveal potential new treatment strategies using pharmacochaperones for NDI and inverse agonists for NSIAD, but also provide a greater understanding of the physiological functions of GPCRs and highlight the new paradigms, i.e., biased agonism and protean agonism.
Collapse
Affiliation(s)
- Noriko Makita
- Department of Endocrinology and Nephrology, The University of Tokyo, Tokyo, Japan.
| | - Katsunori Manaka
- Department of Endocrinology and Nephrology, The University of Tokyo, Tokyo, Japan
| | - Junichiro Sato
- Department of Endocrinology and Nephrology, The University of Tokyo, Tokyo, Japan
| | - Taroh Iiri
- Department of Endocrinology and Nephrology, The University of Tokyo, Tokyo, Japan; Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki, Japan.
| |
Collapse
|
28
|
Jung HJ, Kwon TH. New insights into the transcriptional regulation of aquaporin-2 and the treatment of X-linked hereditary nephrogenic diabetes insipidus. Kidney Res Clin Pract 2019; 38:145-158. [PMID: 31189221 PMCID: PMC6577206 DOI: 10.23876/j.krcp.19.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/09/2019] [Accepted: 02/11/2019] [Indexed: 12/18/2022] Open
Abstract
The kidney collecting duct (CD) is a tubular segment of the kidney where the osmolality and final flow rate of urine are established, enabling urine concentration and body water homeostasis. Water reabsorption in the CD depends on the action of arginine vasopressin (AVP) and a transepithelial osmotic gradient between the luminal fluid and surrounding interstitium. AVP induces transcellular water reabsorption across CD principal cells through associated signaling pathways after binding to arginine vasopressin receptor 2 (AVPR2). This signaling cascade regulates the water channel protein aquaporin-2 (AQP2). AQP2 is exclusively localized in kidney connecting tubules and CDs. Specifically, AVP stimulates the intracellular translocation of AQP2-containing vesicles to the apical plasma membrane, increasing the osmotic water permeability of CD cells. Moreover, AVP induces transcription of the Aqp2 gene, increasing AQP2 protein abundance. This review provides new insights into the transcriptional regulation of the Aqp2 gene in the kidney CD with an overview of AVP and AQP2. It summarizes current therapeutic approaches for X-linked nephrogenic diabetes insipidus caused by AVPR2 gene mutations.
Collapse
Affiliation(s)
- Hyun Jun Jung
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Korea
| |
Collapse
|
29
|
Bech AP, Wetzels JFM, Nijenhuis T. Effects of sildenafil, metformin, and simvastatin on ADH-independent urine concentration in healthy volunteers. Physiol Rep 2019; 6:e13665. [PMID: 29611351 PMCID: PMC5880873 DOI: 10.14814/phy2.13665] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 11/24/2022] Open
Abstract
Nephrogenic diabetes insipidus (NDI) is a rare disorder characterized by resistance of the kidney to the action of antidiuretic hormone (ADH), resulting in a decrease in the capacity of the kidney to concentrate the urine. NDI can be inherited or acquired due to, for example, chronic lithium therapy. Current treatment options are limited to attempts to lower urine output by a low-solute diet and the use of diuretics or anti-inflammatory drugs. These measures are only partially effective. Recent reports suggested that sildenafil, metformin, and simvastatin might improve ADH-independent urine concentration. If confirmed, this would provide interesting additional therapeutic options for patients with NDI. We, therefore, tested the effect of these drugs on ADH-independent urine concentrating capacity in healthy volunteers. We included 36 healthy volunteers who received sildenafil 20 mg thrice daily, metformin 500 mg thrice daily or simvastatin 40 mg once daily during 1 week. At baseline and at the end of treatment, a water loading test was performed. No significant increase in lowest urine osmolality was seen after the use of metformin or sildenafil (P = 0.66 and P = 0.09 respectively). Lowest urine osmolality increased modestly but significantly after the use of simvastatin (70 mOsm/kg to 85 mOsm/kg, P = 0.05). Our data suggest that only simvastatin has an effect on urine osmolality in healthy volunteers. Validation studies are needed and, most importantly, these drugs should be tested in patients with NDI.
Collapse
Affiliation(s)
- Anneke P Bech
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jack F M Wetzels
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tom Nijenhuis
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
30
|
Vukićević T, Hinze C, Baltzer S, Himmerkus N, Quintanova C, Zühlke K, Compton F, Ahlborn R, Dema A, Eichhorst J, Wiesner B, Bleich M, Schmidt-Ott KM, Klussmann E. Fluconazole Increases Osmotic Water Transport in Renal Collecting Duct through Effects on Aquaporin-2 Trafficking. J Am Soc Nephrol 2019; 30:795-810. [PMID: 30988011 DOI: 10.1681/asn.2018060668] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 02/13/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Arginine-vasopressin (AVP) binding to vasopressin V2 receptors promotes redistribution of the water channel aquaporin-2 (AQP2) from intracellular vesicles into the plasma membrane of renal collecting duct principal cells. This pathway fine-tunes renal water reabsorption and urinary concentration, and its perturbation is associated with diabetes insipidus. Previously, we identified the antimycotic drug fluconazole as a potential modulator of AQP2 localization. METHODS We assessed the influence of fluconazole on AQP2 localization in vitro and in vivo as well as the drug's effects on AQP2 phosphorylation and RhoA (a small GTPase, which under resting conditions, maintains F-actin to block AQP2-bearing vesicles from reaching the plasma membrane). We also tested fluconazole's effects on water flow across epithelia of isolated mouse collecting ducts and on urine output in mice treated with tolvaptan, a VR2 blocker that causes a nephrogenic diabetes insipidus-like excessive loss of hypotonic urine. RESULTS Fluconazole increased plasma membrane localization of AQP2 in principal cells independent of AVP. It also led to an increased AQP2 abundance associated with alterations in phosphorylation status and ubiquitination as well as inhibition of RhoA. In isolated mouse collecting ducts, fluconazole increased transepithelial water reabsorption. In mice, fluconazole increased collecting duct AQP2 plasma membrane localization and reduced urinary output. Fluconazole also reduced urinary output in tolvaptan-treated mice. CONCLUSIONS Fluconazole promotes collecting duct AQP2 plasma membrane localization in the absence of AVP. Therefore, it might have utility in treating forms of diabetes insipidus (e.g., X-linked nephrogenic diabetes insipidus) in which the kidney responds inappropriately to AVP.
Collapse
Affiliation(s)
- Tanja Vukićević
- Max Delbrück Center for Molecular Medicine Berlin, (MDC), Research area Cardiovascular & Metabolic Disease, Berlin, Germany
| | - Christian Hinze
- Max Delbrück Center for Molecular Medicine Berlin, (MDC), Research area Cardiovascular & Metabolic Disease, Berlin, Germany.,Department of Nephrology and Medical Intensive Care and.,Berlin Institute of Health, Berlin, Germany
| | - Sandrine Baltzer
- Max Delbrück Center for Molecular Medicine Berlin, (MDC), Research area Cardiovascular & Metabolic Disease, Berlin, Germany
| | - Nina Himmerkus
- Institute of Physiology, Christian Albrechts University Kiel, Kiel, Germany
| | | | - Kerstin Zühlke
- Max Delbrück Center for Molecular Medicine Berlin, (MDC), Research area Cardiovascular & Metabolic Disease, Berlin, Germany
| | - Friederike Compton
- Department of Nephrology and Medical Intensive Care and.,Berlin Institute of Health, Berlin, Germany
| | - Robert Ahlborn
- Information Technology Department, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Alessandro Dema
- Max Delbrück Center for Molecular Medicine Berlin, (MDC), Research area Cardiovascular & Metabolic Disease, Berlin, Germany
| | - Jenny Eichhorst
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Cellular Imaging, Berlin, Germany
| | - Burkhard Wiesner
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Cellular Imaging, Berlin, Germany
| | - Markus Bleich
- Institute of Physiology, Christian Albrechts University Kiel, Kiel, Germany
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine Berlin, (MDC), Research area Cardiovascular & Metabolic Disease, Berlin, Germany; .,Department of Nephrology and Medical Intensive Care and.,Berlin Institute of Health, Berlin, Germany
| | - Enno Klussmann
- Max Delbrück Center for Molecular Medicine Berlin, (MDC), Research area Cardiovascular & Metabolic Disease, Berlin, Germany; .,German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany; and.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Vegetative Physiology, Berlin, Germany
| |
Collapse
|
31
|
Ranieri M, Di Mise A, Tamma G, Valenti G. Vasopressin-aquaporin-2 pathway: recent advances in understanding water balance disorders. F1000Res 2019; 8. [PMID: 30800291 PMCID: PMC6364380 DOI: 10.12688/f1000research.16654.1] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/23/2019] [Indexed: 12/11/2022] Open
Abstract
The alteration of water balance and related disorders has emerged as being strictly linked to the state of activation of the vasopressin–aquaporin-2
(vasopressin–AQP2) pathway. The lack of responsiveness of the kidney to the vasopressin action impairs its ability to concentrate the urine, resulting in polyuria, polydipsia, and risk of severe dehydration for patients. Conversely, non-osmotic release of vasopressin is associated with an increase in water permeability in the renal collecting duct, producing water retention and increasing the circulatory blood volume. This review highlights some of the new insights and recent advances in therapeutic intervention targeting the dysfunctions in the vasopressin–AQP2 pathway causing diseases characterized by water balance disorders such as congenital nephrogenic diabetes insipidus, syndrome of inappropriate antidiuretic hormone secretion, nephrogenic syndrome of inappropriate antidiuresis, and autosomal dominant polycystic kidney disease. The recent clinical data suggest that targeting the vasopressin–AQP2 axis can provide therapeutic benefits in patients with water balance disorders.
Collapse
Affiliation(s)
- Marianna Ranieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy, 70125, Italy
| | - Annarita Di Mise
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy, 70125, Italy
| | - Grazia Tamma
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy, 70125, Italy.,Istituto Nazionale di Biostrutture e Biosistemi, Rome, Roma, Italy, 00136, Italy
| | - Giovanna Valenti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy, 70125, Italy.,Istituto Nazionale di Biostrutture e Biosistemi, Rome, Roma, Italy, 00136, Italy.,Center of Excellence in Comparative Genomics (CEGBA), University of Bari, Bari, Italy, 70125, Italy
| |
Collapse
|
32
|
Yang KT, Yang T, Symons JD. Soluble (pro)renin receptor as a potential therapy for diabetes insipidus. Am J Physiol Renal Physiol 2018; 315:F1416-F1421. [PMID: 30019932 DOI: 10.1152/ajprenal.00266.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The antidiuretic hormone vasopressin (VP) is produced by the hypothalamus and is stored and secreted from the posterior pituitary. VP acts via VP type 2 receptors (V2Rs) on the basolateral membrane of principal cells of the collecting duct (CD) to regulate fluid permeability. The VP-evoked endocrine pathway is essential in determining urine concentrating capability. For example, a defect in any component of the VP signaling pathway can result in polyuria, polydipsia, and hypotonic urine, collectively termed diabetes insipidus (DI). A lack of VP production precipitates central diabetes insipidus (CDI), which can be managed effectively by VP supplementation. A majority of cases of nephrogenic diabetes insipidus (NDI) result from V2R mutations that impair receptor sensitivity. No specific therapy is currently available for management of NDI. Evidence is evolving that (pro)renin receptor (PRR), a newly identified member of the renin-angiotensin system, is capable of regulating VP production and action. As such, PRR should be considered strongly as a therapeutic target for treating CDI and NDI. The current review will summarize recent advances in understanding the physiology of renal and central PRR as it relates to the two types of DI.
Collapse
Affiliation(s)
- Kevin T Yang
- Department of Internal Medicine, University of Utah , Salt Lake City, Utah.,College of Health, University of Utah , Salt Lake City, Utah.,Molecular Medicine Program, University of Utah , Salt Lake City, Utah
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah , Salt Lake City, Utah.,Research Service, Veterans Affairs Medical Center , Salt Lake City, Utah.,Institute of Hypertension, Sun Yat-sen University Zhongshan School of Medicine , Guangzhou , China
| | - J David Symons
- Department of Internal Medicine, University of Utah , Salt Lake City, Utah.,College of Health, University of Utah , Salt Lake City, Utah.,Molecular Medicine Program, University of Utah , Salt Lake City, Utah
| |
Collapse
|
33
|
Fotso Soh J, Torres-Platas SG, Beaulieu S, Mantere O, Platt R, Mucsi I, Saury S, Renaud S, Levinson A, Andreazza AC, Mulsant BH, Müller D, Schaffer A, Dols A, Cervantes P, Low NCP, Herrmann N, Christensen BM, Trepiccione F, Rajji T, Rej S. Atorvastatin in the treatment of Lithium-induced nephrogenic diabetes insipidus: the protocol of a randomized controlled trial. BMC Psychiatry 2018; 18:227. [PMID: 30012135 PMCID: PMC6048831 DOI: 10.1186/s12888-018-1793-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 06/14/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Lithium is the gold-standard treatment for bipolar disorder, is highly effective in treating major depressive disorder, and has anti-suicidal properties. However, clinicians are increasingly avoiding lithium largely due to fears of renal toxicity. Nephrogenic Diabetes Insipidus (NDI) occurs in 15-20% of lithium users and predicts a 2-3 times increased risk of chronic kidney disease (CKD). We recently found that use of statins is associated with lower NDI risk in a cross-sectional study. In this current paper, we describe the methodology of a randomized controlled trial (RCT) to treat lithium-induced NDI using atorvastatin. METHODS We will conduct a 12-week, double-blind placebo-controlled RCT of atorvastatin for lithium-induced NDI at McGill University, Montreal, Canada. We will recruit 60 current lithium users, aged 18-85, who have indicators of NDI, which we defined as urine osmolality (UOsm) < 600 mOsm/kg after 10-h fluid restriction. We will randomize patients to atorvastatin (20 mg/day) or placebo for 12 weeks. We will examine whether this improves measures of NDI: UOsm and aquaporin (AQP2) excretion at 12-week follow-up, adjusted for baseline. RESULTS Not applicable. CONCLUSION The aim of this clinical trial is to provide preliminary data about the efficacy of atorvastatin in treating NDI. If successful, lithium could theoretically be used more safely in patients with a reduced subsequent risk of CKD, hypernatremia, and acute kidney injury (AKI). If future definitive trials confirm this, this could potentially allow more patients to benefit from lithium, while minimizing renal risk. TRIAL REGISTRATION ClinicalTrials.gov NCT02967653 . Registered in February 2017.
Collapse
Affiliation(s)
- Jocelyn Fotso Soh
- Geri-PARTy Research Group, Jewish General Hospital/Lady Davis Institute, McGill University, 4333 Cote Ste-Catherine, Montreal, QC, H3T, 1E4, Montreal, Canada.
| | - Susana G. Torres-Platas
- 0000 0004 1936 8649grid.14709.3bGeri-PARTy Research Group, Jewish General Hospital/Lady Davis Institute, McGill University, 4333 Cote Ste-Catherine, Montreal, QC, H3T, 1E4, Montreal, Canada
| | - Serge Beaulieu
- 0000 0004 1936 8649grid.14709.3bDouglas Mental Health University Institute and Department of Psychiatry, McGill University, Montreal, Canada
| | - Outi Mantere
- 0000 0004 1936 8649grid.14709.3bDouglas Mental Health University Institute and Department of Psychiatry, McGill University, Montreal, Canada
| | - Robert Platt
- 0000 0000 9064 4811grid.63984.30Department of Epidemiology, Biostatistics and Occupational Health, McGill University Health Centre, Montreal, Canada
| | - Istvan Mucsi
- 0000 0001 2157 2938grid.17063.33Division of Nephrology, University Health Network, University of Toronto (UofT), Toronto, Canada
| | - Sybille Saury
- 0000 0004 1936 8649grid.14709.3bDouglas Mental Health University Institute and Department of Psychiatry, McGill University, Montreal, Canada
| | - Suzane Renaud
- 0000 0004 1936 8649grid.14709.3bDouglas Mental Health University Institute and Department of Psychiatry, McGill University, Montreal, Canada
| | - Andrea Levinson
- 0000 0001 2157 2938grid.17063.33Department of Psychiatry, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Ana C. Andreazza
- 0000 0001 2157 2938grid.17063.33Department of Psychiatry, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Benoit H. Mulsant
- 0000 0001 2157 2938grid.17063.33Department of Psychiatry, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Daniel Müller
- 0000 0001 2157 2938grid.17063.33Department of Psychiatry, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Ayal Schaffer
- 0000 0001 2157 2938grid.17063.33Department of Psychiatry, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Annemiek Dols
- Department of Psychiatry, GGZ, Geest, Amsterdam, the Netherlands
| | - Pablo Cervantes
- 0000 0000 9064 4811grid.63984.30Department of Psychiatry, McGill University Health Centre, Montreal, Canada
| | - Nancy CP Low
- 0000 0000 9064 4811grid.63984.30Department of Psychiatry, McGill University Health Centre, Montreal, Canada
| | - Nathan Herrmann
- 0000 0001 2157 2938grid.17063.33Department of Psychiatry, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Birgitte M. Christensen
- 0000 0001 1956 2722grid.7048.bDepartment of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Francesco Trepiccione
- 0000 0001 0790 385Xgrid.4691.aDivision of Nephrology, University of Naples, Naples, Italy
| | - Tarek Rajji
- 0000 0001 2157 2938grid.17063.33Department of Psychiatry, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Soham Rej
- 0000 0004 1936 8649grid.14709.3bGeri-PARTy Research Group, Jewish General Hospital/Lady Davis Institute, McGill University, 4333 Cote Ste-Catherine, Montreal, QC, H3T, 1E4, Montreal, Canada ,0000 0004 1936 8649grid.14709.3bDouglas Mental Health University Institute and Department of Psychiatry, McGill University, Montreal, Canada
| |
Collapse
|
34
|
Schrade K, Tröger J, Eldahshan A, Zühlke K, Abdul Azeez KR, Elkins JM, Neuenschwander M, Oder A, Elkewedi M, Jaksch S, Andrae K, Li J, Fernandes J, Müller PM, Grunwald S, Marino SF, Vukićević T, Eichhorst J, Wiesner B, Weber M, Kapiloff M, Rocks O, Daumke O, Wieland T, Knapp S, von Kries JP, Klussmann E. An AKAP-Lbc-RhoA interaction inhibitor promotes the translocation of aquaporin-2 to the plasma membrane of renal collecting duct principal cells. PLoS One 2018; 13:e0191423. [PMID: 29373579 PMCID: PMC5786306 DOI: 10.1371/journal.pone.0191423] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 01/04/2018] [Indexed: 01/13/2023] Open
Abstract
Stimulation of renal collecting duct principal cells with antidiuretic hormone (arginine-vasopressin, AVP) results in inhibition of the small GTPase RhoA and the enrichment of the water channel aquaporin-2 (AQP2) in the plasma membrane. The membrane insertion facilitates water reabsorption from primary urine and fine-tuning of body water homeostasis. Rho guanine nucleotide exchange factors (GEFs) interact with RhoA, catalyze the exchange of GDP for GTP and thereby activate the GTPase. However, GEFs involved in the control of AQP2 in renal principal cells are unknown. The A-kinase anchoring protein, AKAP-Lbc, possesses GEF activity, specifically activates RhoA, and is expressed in primary renal inner medullary collecting duct principal (IMCD) cells. Through screening of 18,431 small molecules and synthesis of a focused library around one of the hits, we identified an inhibitor of the interaction of AKAP-Lbc and RhoA. This molecule, Scaff10-8, bound to RhoA, inhibited the AKAP-Lbc-mediated RhoA activation but did not interfere with RhoA activation through other GEFs or activities of other members of the Rho family of small GTPases, Rac1 and Cdc42. Scaff10-8 promoted the redistribution of AQP2 from intracellular vesicles to the periphery of IMCD cells. Thus, our data demonstrate an involvement of AKAP-Lbc-mediated RhoA activation in the control of AQP2 trafficking.
Collapse
Affiliation(s)
- Katharina Schrade
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | - Jessica Tröger
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | - Adeeb Eldahshan
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | - Kerstin Zühlke
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | | | - Jonathan M. Elkins
- Structural Genomics Consortium, University of Oxford, Oxford, United Kingdom
| | | | - Andreas Oder
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Mohamed Elkewedi
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | - Sarah Jaksch
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | | | - Jinliang Li
- University of Miami Miller School of Medicine, Miami, United States of America
| | - Joao Fernandes
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | - Paul Markus Müller
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | - Stephan Grunwald
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | - Stephen F. Marino
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | - Tanja Vukićević
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | - Jenny Eichhorst
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Burkhard Wiesner
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | | | - Michael Kapiloff
- University of Miami Miller School of Medicine, Miami, United States of America
| | - Oliver Rocks
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | - Oliver Daumke
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
| | - Thomas Wieland
- Institute of Experimental Pharmacology and Toxicology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Stefan Knapp
- Structural Genomics Consortium, University of Oxford, Oxford, United Kingdom
- Institute for Pharmaceutical Chemistry and Buchmann Institute, Goethe University, Frankfurt, Germany
- DKTK (German Cancer Center Network), partner site Frankfurt/Main, Germany
| | | | - Enno Klussmann
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
- * E-mail:
| |
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: 55] [Impact Index Per Article: 6.9] [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
|
Li W, Jin WW, Tsuji K, Chen Y, Nomura N, Su L, Yui N, Arthur J, Cotecchia S, Paunescu TG, Brown D, Lu HAJ. Ezrin directly interacts with AQP2 and promotes its endocytosis. J Cell Sci 2017; 130:2914-2925. [PMID: 28754689 DOI: 10.1242/jcs.204842] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/21/2017] [Indexed: 11/20/2022] Open
Abstract
The water channel aquaporin-2 (AQP2) is a major regulator of water homeostasis in response to vasopressin (VP). Dynamic trafficking of AQP2 relies on its close interaction with trafficking machinery proteins and the actin cytoskeleton. Here, we report the identification of ezrin, an actin-binding protein from the ezrin/radixin/moesin (ERM) family as an AQP2-interacting protein. Ezrin was first detected in a co-immunoprecipitation (co-IP) complex using an anti-AQP2 antibody in a proteomic analysis. Immunofluorescence staining revealed the co-expression of ezrin and AQP2 in collecting duct principal cells, and VP treatment caused redistribution of both proteins to the apical membrane. The ezrin-AQP2 interaction was confirmed by co-IP experiments with an anti-ezrin antibody, and by pulldown assays using purified full-length and FERM domain-containing recombinant ezrin. By using purified recombinant proteins, we showed that ezrin directly interacts with AQP2 C-terminus through its N-terminal FERM domain. Knocking down ezrin expression with shRNA resulted in increased membrane accumulation of AQP2 and reduced AQP2 endocytosis. Therefore, through direct interaction with AQP2, ezrin facilitates AQP2 endocytosis, thus linking the dynamic actin cytoskeleton network with AQP2 trafficking.
Collapse
Affiliation(s)
- Wei Li
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - William W Jin
- Washington University in St. Louis, College of Arts and Sciences, St Louis, MO 63130, USA
| | - Kenji Tsuji
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Ying Chen
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Naohiro Nomura
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Limin Su
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA.,Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Naofumi Yui
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Julian Arthur
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Susanna Cotecchia
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne 1005, Switzerland
| | - Teodor G Paunescu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Dennis Brown
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| | - Hua A J Lu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
40
|
Abstract
Disruption of water and electrolyte balance is frequently encountered in clinical medicine. Regulating water metabolism is critically important. Diabetes insipidus (DI) presented with excessive water loss from the kidney is a major disorder of water metabolism. To understand the molecular and cellular mechanisms and pathophysiology of DI and rationales of clinical management of DI is important for both research and clinical practice. This chapter will first review various forms of DI focusing on central diabetes insipidus (CDI) and nephrogenic diabetes insipidus (NDI ) . This is followed by a discussion of regulatory mechanisms underlying CDI and NDI , with a focus on the regulatory axis of vasopressin, vasopressin receptor 2 (V2R ) and the water channel molecule, aquaporin 2 (AQP2 ). The clinical manifestation, diagnosis and management of various forms of DI will also be discussed with highlights of some of the latest therapeutic strategies that are developed from in vitro experiments and animal studies.
Collapse
Affiliation(s)
- H A Jenny Lu
- Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA.
| |
Collapse
|
41
|
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
|
42
|
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
|
43
|
Rahman SS, Boesen EI. Outside the mainstream: novel collecting duct proteins regulating water balance. Am J Physiol Renal Physiol 2016; 311:F1341-F1345. [PMID: 27784697 DOI: 10.1152/ajprenal.00488.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/06/2016] [Accepted: 10/18/2016] [Indexed: 12/26/2022] Open
Abstract
Body water balance is critical to survival and, therefore, very tightly regulated by the hypothalamus and kidney. A key mechanism involved in this process, the arginine vasopressin-mediated phosphorylation and apical membrane insertion of aquaporin 2 in the collecting duct, has been extensively studied; however, with the increased availability of conditional knockout animals, several novel collecting duct proteins have recently been implicated in water homeostasis. In this Mini-Review, we briefly discuss these novel proteins and their roles in the regulation of water homeostasis.
Collapse
Affiliation(s)
- Shamma S Rahman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Erika I Boesen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
Sands JM, Klein JD. Physiological insights into novel therapies for nephrogenic diabetes insipidus. Am J Physiol Renal Physiol 2016; 311:F1149-F1152. [PMID: 27534996 DOI: 10.1152/ajprenal.00418.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 08/16/2016] [Indexed: 12/11/2022] Open
Abstract
Fundamental kidney physiology research can provide important insight into how the kidney works and suggest novel therapeutic opportunities to treat human diseases. This is especially true for nephrogenic diabetes insipidus (NDI). Over the past decade, studies elucidating the molecular physiology and signaling pathways regulating water transport have suggested novel therapeutic possibilities. In patients with congenital NDI due to mutations in the type 2 vasopressin receptor (V2R) or acquired NDI due to lithium (or other medications), there are no functional abnormalities in the aquaporin-2 (AQP2) water channel, or in another key inner medullary transport protein, the UT-A1 urea transporter. If it is possible to phosphorylate and/or increase the apical membrane accumulation of these proteins, independent of vasopressin or cAMP, one may be able to treat NDI. Sildenifil (through cGMP), erlotinib, and simvastatin each stimulate AQP2 insertion into the apical plasma membrane. Some recent human data suggest that sildenafil and simvastatin may improve urine concentrating ability. ONO-AE1-329 (ONO) stimulates the EP4 prostanoid receptor (EP4), which stimulates kinases that in turn phosphorylate AQP2 and UT-A1. Clopidogrel is a P2Y12-R antagonist that potentiates the effect of vasopressin and increases AQP2 abundance. Metformin stimulates AMPK to phosphorylate and activate AQP2 and UT-A1, and it increases urine concentrating ability in two rodent models of NDI. Since metformin, sildenafil, and simvastatin are commercially available and have excellent safety records, the potential for rapidly advancing them into clinical trials is high.
Collapse
Affiliation(s)
- Jeff M Sands
- Renal Division, Department of Medicine, and Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Janet D Klein
- Renal Division, Department of Medicine, and Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
47
|
Efe O, Klein JD, LaRocque LM, Ren H, Sands JM. Metformin improves urine concentration in rodents with nephrogenic diabetes insipidus. JCI Insight 2016; 1:88409. [PMID: 27478876 DOI: 10.1172/jci.insight.88409] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Urine concentration is regulated by vasopressin. Congenital nephrogenic diabetes insipidus (NDI) is caused by vasopressin type 2 receptor (V2R) mutations. We studied whether metformin could improve urine concentration in rodent models of congenital NDI by stimulating AMPK. To block the V2R in rats, tolvaptan (10 mg/kg/d) was given by oral gavage with or without metformin (800 mg/ kg/d). Control rats received vehicle with or without metformin. Tamoxifen-induced V2R KO mice were given metformin (600 mg/kg) or vehicle twice daily. Urine osmolality in tolvaptan-treated rats (1,303 ± 126 mOsM) was restored to control levels by metformin (2,335 ± 273 mOsM) within 3 days and was sustained for up to 10 days. Metformin increased protein abundance of inner medullary urea transporter UT-A1 by 61% and aquaporin 2 (AQP2) by 44% in tolvaptan-treated rats, and immunohistochemistry showed increased membrane accumulation of AQP2 with acute and chronic AMPK stimulation. Outer medullary Na+-K+-2Cl- cotransporter 2 (NKCC2) abundance increased (117%) with AMPK stimulation in control rats but not in V2R-blocked rats. Metformin increased V2R KO mouse urine osmolality within 3 hours, and the increase persisted for up to 12 hours. Metformin increased AQP2 in the V2R KO mice similar to the tolvaptan-treated rats. These results indicate that AMPK activators, such as metformin, might provide a promising treatment for congenital NDI.
Collapse
Affiliation(s)
- Orhan Efe
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Janet D Klein
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of Physiology, Emory University, Atlanta, Georgia, USA
| | - Lauren M LaRocque
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Huiwen Ren
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jeff M Sands
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of Physiology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
48
|
Molecular mechanisms in lithium-associated renal disease: a systematic review. Int Urol Nephrol 2016; 48:1843-1853. [DOI: 10.1007/s11255-016-1352-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/21/2016] [Indexed: 02/07/2023]
|
49
|
Abstract
Aquaporins (AQPs) are a 13 member family (AQP0-12) of proteins that act as channels, through which water and, for some family members, glycerol, urea and other small solutes can be transported. Aquaporins are highly abundant in kidney epithelial cells where they play a critical role with respect to water balance. In this review we summarize the current knowledge with respect to the localization and function of AQPs within the kidney tubule, and their role in mammalian water homeostasis and the water balance disorders. Overviews of practical aspects with regard to differential diagnosis for some of these disorders, alongside treatment strategies are also discussed.
Collapse
Affiliation(s)
- Hanne B Moeller
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport, Aarhus University, Denmark
| | - Cecilia H Fuglsang
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport, Aarhus University, Denmark
| | - Robert A Fenton
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport, Aarhus University, Denmark.
| |
Collapse
|
50
|
Vukićević T, Schulz M, Faust D, Klussmann E. The Trafficking of the Water Channel Aquaporin-2 in Renal Principal Cells-a Potential Target for Pharmacological Intervention in Cardiovascular Diseases. Front Pharmacol 2016; 7:23. [PMID: 26903868 PMCID: PMC4749865 DOI: 10.3389/fphar.2016.00023] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/25/2016] [Indexed: 01/13/2023] Open
Abstract
Arginine-vasopressin (AVP) stimulates the redistribution of water channels, aquaporin-2 (AQP2) from intracellular vesicles into the plasma membrane of renal collecting duct principal cells. By this AVP directs 10% of the water reabsorption from the 170 L of primary urine that the human kidneys produce each day. This review discusses molecular mechanisms underlying the AVP-induced redistribution of AQP2; in particular, it provides an overview over the proteins participating in the control of its localization. Defects preventing the insertion of AQP2 into the plasma membrane cause diabetes insipidus. The disease can be acquired or inherited, and is characterized by polyuria and polydipsia. Vice versa, up-regulation of the system causing a predominant localization of AQP2 in the plasma membrane leads to excessive water retention and hyponatremia as in the syndrome of inappropriate antidiuretic hormone secretion (SIADH), late stage heart failure or liver cirrhosis. This article briefly summarizes the currently available pharmacotherapies for the treatment of such water balance disorders, and discusses the value of newly identified mechanisms controlling AQP2 for developing novel pharmacological strategies. Innovative concepts for the therapy of water balance disorders are required as there is a medical need due to the lack of causal treatments.
Collapse
Affiliation(s)
- Tanja Vukićević
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association Berlin, Germany
| | - Maike Schulz
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association Berlin, Germany
| | - Dörte Faust
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association Berlin, Germany
| | - Enno Klussmann
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz AssociationBerlin, Germany; German Centre for Cardiovascular ResearchBerlin, Germany
| |
Collapse
|