1
|
Vrettou CS, Issaris V, Kokkoris S, Poupouzas G, Keskinidou C, Lotsios NS, Kotanidou A, Orfanos SE, Dimopoulou I, Vassiliou AG. Exploring Aquaporins in Human Studies: Mechanisms and Therapeutic Potential in Critical Illness. Life (Basel) 2024; 14:1688. [PMID: 39768394 PMCID: PMC11676363 DOI: 10.3390/life14121688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Aquaporins (AQPs) are membrane proteins facilitating water and other small solutes to be transported across cell membranes. They are crucial in maintaining cellular homeostasis by regulating water permeability in various tissues. Moreover, they regulate cell migration, signaling pathways, inflammation, tumor growth, and metastasis. In critically ill patients, such as trauma, sepsis, and patients with acute respiratory distress syndrome (ARDS), which are frequently encountered in intensive care units (ICUs), water transport regulation is crucial for maintaining homeostasis, as dysregulation can lead to edema or dehydration, with the latter also implicating hemodynamic compromise. Indeed, AQPs are involved in fluid transport in various organs, including the lungs, kidneys, and brain, where their dysfunction can exacerbate conditions like ARDS, acute kidney injury (AKI), or cerebral edema. In this review, we discuss the implication of AQPs in the clinical entities frequently encountered in ICUs, such as systemic inflammation and sepsis, ARDS, AKI, and brain edema due to different types of primary brain injury from a clinical perspective. Current and possible future therapeutic implications are also considered.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Alice G. Vassiliou
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (C.S.V.); (V.I.); (S.K.); (G.P.); (C.K.); (N.S.L.); (A.K.); (S.E.O.); (I.D.)
| |
Collapse
|
2
|
Guo Y, Qiao R, Xie G, Yao Y, Du C, Shao Y, Guan Y, Zhang X. Activation of TGR5 Increases Urine Concentration by Inducing AQP2 and AQP3 Expression in Renal Medullary Collecting Ducts. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:181-192. [PMID: 38835402 PMCID: PMC11149995 DOI: 10.1159/000538107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 02/26/2024] [Indexed: 06/06/2024]
Abstract
Introduction G protein-coupled bile acid receptor (TGR5), the first G protein-coupled receptor for bile acids identified, is capable of activating a variety of intracellular signaling pathways after interacting with bile acids. TGR5 plays an important role in multiple physiological processes and is considered to be a potential target for the treatment of various metabolic diseases, including type 2 diabetes. Evidence has emerged that genetic deletion of TGR5 results in an increase in basal urine output, suggesting that it may play a critical role in renal water and salt reabsorption. The present study aims to elucidate the effect and mechanism of TGR5 activation on urine concentration. Methods Mice were treated with TGR5 agonists (LCA and INT-777) for 3 days. The 24-h urine of mice was collected and analyzed for urine biochemical parameters. The mRNA expressions were detected by real-time PCR, and the protein expressions were detected by western blot. Immunohistochemistry and immunofluorescence were performed to examine the cellular location of proteins. The cultured primary medullary collecting duct cells were pretreated with H89 (a PKA inhibitor) for 1 h, followed by 12-h treatment of LCA and INT-777. Luciferase reporter assays were used to detect the effect of CREB on the gene transcription of AQPs. Gel electrophoretic mobility shift assays were used to analyze DNA-protein interactions. Results Treatment of mice with the TGR5 agonist LCA and INT-777 markedly reduced urine output and increased urine osmolality, accompanied by a marked increase in AQP2 and AQP3 protein expression and membrane translocation. In cultured primary medullary collecting duct cells, LCA and INT-777 dose-dependently upregulated AQP2 and AQP3 expression in a cAMP/PKA-dependent manner. Mechanistically, both AQP2 and AQP3 gene promoter contains a putative CREB-binding site, which can be bound and activated by CREB as assessed by both gene promoter-driven luciferase and gel shift assays. Conclusion Collectively, our findings demonstrate that activation of TGR5 can promote urine concentration by upregulation of AQP2 and AQP3 expression in renal collecting ducts. TGR5 may represent an attractive target for the treatment of patients with urine concentration defect.
Collapse
Affiliation(s)
- Yanlin Guo
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, China
- Health Science Center, East China Normal University, Shanghai, China
| | - Rongfang Qiao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Guixiang Xie
- Health Science Center, East China Normal University, Shanghai, China
| | - Yao Yao
- Division of Nephrology, Affiliated Hospital and Medical School, Nantong University, Nantong, China
| | - Chunxiu Du
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, China
- Health Science Center, East China Normal University, Shanghai, China
| | - Yunxia Shao
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaoyan Zhang
- Division of Nephrology, Wuhu Hospital, East China Normal University, Wuhu, China
- Health Science Center, East China Normal University, Shanghai, China
| |
Collapse
|
3
|
Dugbartey GJ. Physiological role of hydrogen sulfide in the kidney and its therapeutic implications for kidney diseases. Biomed Pharmacother 2023; 166:115396. [PMID: 37647689 DOI: 10.1016/j.biopha.2023.115396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/21/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023] Open
Abstract
For over three centuries, hydrogen sulfide (H2S) has been known as a toxic and deadly gas at high concentrations, with a distinctive smell of rotten eggs. However, studies over the past two decades have shown that H2S has risen above its historically notorious label and has now received significant scientific attention as an endogenously produced gaseous signaling molecule that participates in cellular homeostasis and influences a myriad of physiological and pathological processes at low concentrations. Its endogenous production is enzymatically regulated, and when dysregulated, contributes to pathogenesis of renal diseases. In addition, exogenous H2S administration has been reported to exhibit important therapeutic characteristics that target multiple molecular pathways in common renal pathologies in which reduced levels of renal and plasma H2S were observed. This review highlights functional anatomy of the kidney and renal production of H2S. The review also discusses current understanding of H2S in renal physiology and seeks to lay the foundation as a new targeted therapeutic agent for renal pathologies such as hypertensive nephropathy, diabetic kidney disease and water balance disorders.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana; Accra College of Medicine, Magnolia St, JVX5+FX9, East Legon, Accra, Ghana.
| |
Collapse
|
4
|
Wang Y, LaRocque LM, Ruiz JA, Rodriguez EL, Sands JM, Klein JD. Aldosterone Contributes to Vasopressin Escape through Changes in Water and Urea Transport. Biomedicines 2023; 11:1844. [PMID: 37509484 PMCID: PMC10376660 DOI: 10.3390/biomedicines11071844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Hyponatremia (hypo-osmolality) is a disorder of water homeostasis due to abnormal renal diluting capacity. The body limits the degree to which serum sodium concentration falls through a mechanism called "vasopressin escape". Vasopressin escape is a process that prevents the continuous decrease in serum sodium concentration even under conditions of sustained high plasma vasopressin levels. Previous reports suggest that aldosterone may be involved in the vasopressin escape mechanism. The abilities of aldosterone synthase (Cyp11b2) knockout and wild-type mice to escape from vasopressin were compared. Wild-type mice escaped while the aldosterone synthase knockout mice did not. Both the water channel aquaporin 2 (AQP2) and the urea transporter UT-A1 protein abundances were higher in aldosterone synthase knockout than in wild-type mice at the end of the escape period. Vasopressin escape was also blunted in rats given spironolactone, a mineralocorticoid receptor blocker. Next, the role of the phosphatase, calcineurin (protein phosphatase 2B, PP2B), in vasopressin escape was studied since aldosterone activates calcineurin in rat cortical collecting ducts. Tacrolimus, a calcineurin inhibitor, blunted vasopressin escape in rats compared with the control rats, increased UT-A1, AQP2, and pS256-AQP2, and decreased pS261-AQP2 protein abundances. Our results indicate that aldosterone regulates vasopressin escape through calcineurin-mediated protein changes in UT-A1 and AQP2.
Collapse
Affiliation(s)
- Yanhua Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Lauren M LaRocque
- Renal Division, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Joseph A Ruiz
- Renal Division, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Eva L Rodriguez
- Renal Division, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Jeff M Sands
- Renal Division, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Janet D Klein
- Renal Division, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
5
|
Wang Y, Klein JD, Sands JM. Phosphatases Decrease Water and Urea Permeability in Rat Inner Medullary Collecting Ducts. Int J Mol Sci 2023; 24:ijms24076537. [PMID: 37047509 PMCID: PMC10095045 DOI: 10.3390/ijms24076537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
We previously showed that the phosphatases PP1/PP2A and PP2B dephosphorylate the water channel, AQP2, suggesting their role in water reabsorption. In this study, we investigated whether protein phosphatase 2A (PP2A) and protein phosphatase 2B (PP2B or calcineurin), which are present in the inner medullary collecting duct (IMCD), are regulators of urea and water permeability. Inhibition of calcineurin by tacrolimus increased both basal and vasopressin-stimulated osmotic water permeability in perfused rat IMCDs. However, tacrolimus did not affect osmotic water permeability in the presence of aldosterone. Inhibition of PP2A by calyculin increased both basal and vasopressin-stimulated osmotic water permeability, and aldosterone reversed the increase by calyculin. Previous studies showed that adrenomedullin (ADM) activates PP2A and decreases osmotic water permeability. Inhibition of PP2A by calyculin prevented the ADM-induced decrease in water reabsorption. ADM reduced the phosphorylation of AQP2 at serine 269 (pSer269 AQP2). Urea is linked to water reabsorption by building up hyperosmolality in the inner medullary interstitium. Calyculin increased urea permeability and phosphorylated UT-A1. Our results indicate that phosphatases regulate water reabsorption. Aldosterone and adrenomedullin decrease urea or osmotic water permeability by acting through calcineurin and PP2A, respectively. PP2A may regulate water reabsorption by dephosphorylating pSer269, AQP2, and UT-A1.
Collapse
|
6
|
Identification of AGXT2, SHMT1, and ACO2 as important biomarkers of acute kidney injury by WGCNA. PLoS One 2023; 18:e0281439. [PMID: 36735737 PMCID: PMC9897545 DOI: 10.1371/journal.pone.0281439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/23/2023] [Indexed: 02/04/2023] Open
Abstract
Acute kidney injury (AKI) is a serious and frequently observed disease associated with high morbidity and mortality. Weighted gene co-expression network analysis (WGCNA) is a research method that converts the relationship between tens of thousands of genes and phenotypes into the association between several gene sets and phenotypes. We screened potential target genes related to AKI through WGCNA to provide a reference for the diagnosis and treatment of AKI. Key biomolecules of AKI were investigated based on transcriptome analysis. RNA sequencing data from 39 kidney biopsy specimens of AKI patients and 9 normal subjects were downloaded from the GEO database. By WGCNA, the top 20% of mRNAs with the largest variance in the data matrix were used to construct a gene co-expression network with a p-value < 0.01 as a screening condition, showing that the blue module was most closely associated with AKI. Thirty-two candidate biomarker genes were screened according to the threshold values of |MM|≥0.86 and |GS|≥0.4, and PPI and enrichment analyses were performed. The top three genes with the most connected nodes, alanine-glyoxylate aminotransferase 2(AGXT2), serine hydroxymethyltransferase 1(SHMT1) and aconitase 2(ACO2), were selected as the central genes based on the PPI network. A rat AKI model was constructed, and the mRNA and protein expression levels of the central genes in the model and control groups were verified by PCR and immunohistochemistry experiments. The results showed that the relative mRNA expression and protein levels of AGXT2, SHMT1 and ACO2 showed a decrease in the model group. In conclusion, we inferred that there is a close association between AGXT2, SHMT1 and ACO2 genes and the development of AKI, and the down-regulation of their expression levels may induce AKI.
Collapse
|
7
|
Qiu Z, Jiang T, Li Y, Wang W, Yang B. Aquaporins in Urinary System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1398:155-177. [PMID: 36717493 DOI: 10.1007/978-981-19-7415-1_11] [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
There are at least eight aquaporins (AQPs) expressed in the kidney. Including AQP1 expressed in proximal tubules, thin descending limb of Henle and vasa recta; AQP2, AQP3, AQP4, AQP5, and AQP6 expressed in collecting ducts; AQP7 expressed in proximal tubules; AQP8 expressed in proximal tubules and collecting ducts; and AQP11 expressed in the endoplasmic reticulum of proximal tubular epithelial cells. Over years, researchers have constructed different AQP knockout mice and explored the effect of AQP knockout on kidney function. Thus, the roles of AQPs in renal physiology are revealed, providing very useful information for addressing fundamental questions about transepithelial water transport and the mechanism of near isoosmolar fluid reabsorption. This chapter introduces the localization and function of AQPs in the kidney and their roles in different kidney diseases to reveal the prospects of AQPs in further basic and clinical studies.
Collapse
Affiliation(s)
- Zhiwei Qiu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Tao Jiang
- College of Basic Medicine, Beihua University, Jilin, China
| | - Yingjie Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Weiling Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, P.R. China
| | - Baoxue Yang
- School of Basic Medical Sciences, Peking University, Beijing, China.
| |
Collapse
|
8
|
Ma W, Liang X, Su Z. Effects of a Chinese herbal extract on the intestinal tract and aquaporin in Adriamycin-induced nephropathy. Bioengineered 2022; 13:2732-2745. [PMID: 35068345 PMCID: PMC8973663 DOI: 10.1080/21655979.2021.2014620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Wuling Decoction is a traditional Chinese medicine that has been used to open knots, benefit water, transform Qi, return fluid, and has a significant effect on strengthening the spleen and removing dampness. To explore the effects of Wuling Decoction on the intestinal tract and aquaporin in Adriamycin-induced nephropathy, 45 specific pathogen free (SPF) Wistar rats were randomly divided into a blank control group (5 rats), Dosing control group (10 rats), Adriamycin nephropathy model group (10 rats), diarrhea group (10 rats), and an Adriamycin nephropathy diarrhea model group (10 rats). The tissue localization of aquaporin (AQP) was determined by immunohistochemistry. The expression of AQP mRNA and protein was measured by RT-PCR and western blot analysis, respectively. The results indicated that Wuling Decoction causes excretion of AQP2 through the urine, regulates AQP2 levels, and exerts diuretic and anti-diarrheal effects. It also regulates the levels of antidiuretic hormone (ADH) and arginine vasopressin (AVP), affects water absorption rate, and reduces the level of cyclic adenosine monophosphate (cAMP) in each tissue, thus reducing the absorption of AQP2 to water. Wuling Decoction promoted AQP2 expression in the nephropathy model group and inhibited AQP2 expression in the diarrhea group. Wuling Decoction increased the expression of aquaporin in the intestinal tract, reduced the water content of stool by promoting the absorption of water in the intestinal tract, inhibited the expression of aquaporin and its regulatory factors in nephridia tissue, and reduced the reabsorption of water to increase urine volume, to decrease the occurrence of diarrhea.
Collapse
Affiliation(s)
- Weizhong Ma
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xing Liang
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhuowei Su
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
9
|
Kemp BA, Howell NL, Gildea JJ, Keller SR, Brautigan DL, Carey RM. Renal AT 2 Receptors Mediate Natriuresis via Protein Phosphatase PP2A. Circ Res 2022; 130:96-111. [PMID: 34794320 PMCID: PMC8741733 DOI: 10.1161/circresaha.121.319519] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND How signals from activated angiotensin type-2 receptors (AT2R) mediate inhibition of sodium ion (Na+) reabsorption in renal proximal tubule cells is currently unknown. Protein phosphatases including PP2A (protein phosphatase 2A) have been implicated in AT2R signaling in tissues other than kidney. We investigated whether inhibition of protein phosphatase PP2A reduced AT2R-mediated natriuresis and evaluated changes in PP2A activity and localization after renal AT2R activation in normal 4- and 10-week-old control Wistar-Kyoto rats and 4-week-old prehypertensive and 10-week-old hypertensive spontaneously hypertensive rats. METHODS AND RESULTS In Wistar-Kyoto rats, direct renal interstitial administration of selective AT2R nonpeptide agonist Compound-21 (C-21) increased renal interstitial cyclic GMP (cGMP) levels, urine Na+ excretion, and simultaneously increased PP2A activity ≈2-fold in homogenates of renal cortical tubules. The cyclic GMP and natriuretic responses were abolished by concurrent renal interstitial administration of protein phosphatase inhibitor calyculin A. In renal proximal tubule cells in response to C-21, PP2A subunits A, B55α and C, but not B56γ, were recruited to apical plasma membranes together with AT2Rs. Calyculin A treatment abolished C-21-induced translocation of both AT2R and PP2A regulatory subunit B55α to apical plasma membranes. Immunoprecipitation of AT2R solubilized from renal cortical homogenates demonstrated physical association of AT2R with PP2A A, B55α, and C but not B56γ subunits. In contrast, in spontaneously hypertensive rats, administration of C-21 did not alter urine Na+ excretion or PP2A activity and failed to translocate AT2Rs and PP2A subunits to apical plasma membranes. CONCLUSIONS In renal proximal tubule cells of Wistar-Kyoto rats, PP2A is activated and PP2A subunits AB55αC are recruited to C-21-activated AT2Rs during induction of natriuresis. This response is defective in prehypertensive and hypertensive spontaneously hypertensive rats, presenting a potential novel therapeutic target for treating renal Na+ retention and hypertension.
Collapse
Affiliation(s)
- Brandon A. Kemp
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
| | - Nancy L. Howell
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
| | - John J. Gildea
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA
| | - Susanna R. Keller
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
| | - David L. Brautigan
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA
| | - Robert M. Carey
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA
| |
Collapse
|
10
|
Torres-Pinzon DL, Ralph DL, Veiras LC, McDonough AA. Sex-specific adaptations to high-salt diet preserve electrolyte homeostasis with distinct sodium transporter profiles. Am J Physiol Cell Physiol 2021; 321:C897-C909. [PMID: 34613843 PMCID: PMC8616593 DOI: 10.1152/ajpcell.00282.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 02/04/2023]
Abstract
Kidneys continuously filter an enormous amount of sodium and adapt kidney Na+ reabsorption to match Na+ intake to maintain circulatory volume and electrolyte homeostasis. Males (M) respond to high-salt (HS) diet by translocating proximal tubule Na+/H+ exchanger isoform 3 (NHE3) to the base of the microvilli, reducing activated forms of the distal NaCl cotransporter (NCC) and epithelial Na+ channel (ENaC). Males (M) and females (F) on normal-salt (NS) diet present sex-specific profiles of "transporters" (cotransporters, channels, pumps, and claudins) along the nephron, e.g., F exhibit 40% lower NHE3 and 200% higher NCC abundance than M. We tested the hypothesis that adaptations to HS diet along the nephron will, likewise, exhibit sexual dimorphisms. C57BL/6J mice were fed for 15 days with 4% NaCl diet (HS) versus 0.26% NaCl diet (NS). On HS, M and F exhibited normal plasma [Na+] and [K+], similar urine volume, Na+, K+, and osmolal excretion rates normalized to body weight. In F, like M, HS lowered abundance of distal NCC, phosphorylated NCC, and cleaved (activated) forms of ENaC. The adaptations associated with achieving electrolyte homeostasis exhibit sex-dependent and independent mechanisms. Sex differences in baseline "transporters" abundance persist during HS diet, yet the fold changes during HS diet (normalized to NS) are similar along the distal nephron and collecting duct. Sex-dependent differences observed along the proximal tubule during HS show that female kidneys adapt differently from patterns reported in males, yet achieve and maintain fluid and electrolyte homeostasis.
Collapse
Affiliation(s)
- Diana L Torres-Pinzon
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Donna L Ralph
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Luciana C Veiras
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Alicia A McDonough
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| |
Collapse
|
11
|
Shao L, Ma Y, Fang Q, Huang Z, Wan S, Wang J, Yang L. Role of protein phosphatase 2A in kidney disease (Review). Exp Ther Med 2021; 22:1236. [PMID: 34539832 PMCID: PMC8438693 DOI: 10.3892/etm.2021.10671] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
Kidney disease affects millions of people worldwide and is a financial burden on the healthcare system. Protein phosphatase 2A (PP2A), which is involved in renal development and the function of ion-transport proteins, aquaporin-2 and podocytes, is likely to serve an important role in renal processes. PP2A is associated with the pathogenesis of a variety of different kidney diseases including podocyte injury, inflammation, tumors and chronic kidney disease. The current review aimed to discuss the structure and function of PP2A subunits in the context of kidney diseases. How dysregulation of PP2A in the kidneys causes podocyte death and the inactivation of PP2A in renal carcinoma tissues is discussed. Inhibition of PP2A activity prevents epithelial-mesenchymal transition and attenuates renal fibrosis, creating a favorable inflammatory microenvironment and promoting the initiation and progression of tumor pathogenesis. The current review also indicates that PP2A serves an important role in protection against renal inflammation. Understanding the detailed mechanisms of PP2A provides information that can be utilized in the design and application of novel therapeutics for the treatment and prevention of renal diseases.
Collapse
Affiliation(s)
- Lishi Shao
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Yiqun Ma
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Qixiang Fang
- Department of Urology, The First Affiliated Hospital of the Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P.R. China
| | - Ziye Huang
- Department of Urology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Shanshan Wan
- Department of Radiology, Yunnan Kun-Gang Hospital, Anning, Yunnan 650300, P.R. China
| | - Jiaping Wang
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Li Yang
- Department of Anatomy, Histology and Embryology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
12
|
Ranieri M, Di Mise A, Centrone M, D'Agostino M, Tingskov SJ, Venneri M, Pellegrino T, Difonzo G, Caponio F, Norregaard R, Valenti G, Tamma G. Olive Leaf Extract (OLE) impaired vasopressin-induced aquaporin-2 trafficking through the activation of the calcium-sensing receptor. Sci Rep 2021; 11:4537. [PMID: 33633156 PMCID: PMC7907100 DOI: 10.1038/s41598-021-83850-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Vasopressin (AVP) increases water permeability in the renal collecting duct through the regulation of aquaporin-2 (AQP2) trafficking. Several disorders, including hypertension and inappropriate antidiuretic hormone secretion (SIADH), are associated with abnormalities in water homeostasis. It has been shown that certain phytocompounds are beneficial to human health. Here, the effects of the Olive Leaf Extract (OLE) have been evaluated using in vitro and in vivo models. Confocal studies showed that OLE prevents the vasopressin induced AQP2 translocation to the plasma membrane in MCD4 cells and rat kidneys. Incubation with OLE decreases the AVP-dependent increase of the osmotic water permeability coefficient (Pf). To elucidate the possible effectors of OLE, intracellular calcium was evaluated. OLE increases the intracellular calcium through the activation of the Calcium Sensing Receptor (CaSR). NPS2143, a selective CaSR inhibitor, abolished the inhibitory effect of OLE on AVP-dependent water permeability. In vivo experiments revealed that treatment with OLE increases the expression of the CaSR mRNA and decreases AQP2 mRNA paralleled by an increase of the AQP2-targeting miRNA-137. Together, these findings suggest that OLE antagonizes vasopressin action through stimulation of the CaSR indicating that this extract may be beneficial to attenuate disorders characterized by abnormal CaSR signaling and affecting renal water reabsorption.
Collapse
Affiliation(s)
- Marianna Ranieri
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125, Bari, Italy.
| | - Annarita Di Mise
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125, Bari, Italy
| | - Mariangela Centrone
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125, Bari, Italy
| | - Mariagrazia D'Agostino
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125, Bari, Italy
| | | | - Maria Venneri
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125, Bari, Italy
| | - Tommaso Pellegrino
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125, Bari, Italy
| | - Graziana Difonzo
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Caponio
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Rikke Norregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Giovanna Valenti
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125, Bari, Italy
| | - Grazia Tamma
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125, Bari, Italy.
| |
Collapse
|
13
|
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
|
14
|
Abstract
Aquaporins (AQPs) are transmembrane channel proteins that mainly facilitate the water translocation through the plasma cell membrane. For several years these proteins have been extensively examined for their biologic role in health and their potential implication in different diseases. Technological improvements associated with the methods employed to evaluate the functions of the AQPs have provided us with significant new knowledge. In this chapter, we will examine the role of AQPs in health and disease based on the latest currently available evidence.
Collapse
Affiliation(s)
- Dimitrios E Magouliotis
- Division of Surgery and Interventional Sciences, UCL, London, United Kingdom; Department of Surgery, University of Thessaly, Biopolis, Larissa, Greece.
| | | | - Alexis A Svokos
- Geisinger Lewisburg-Women's Health, Lewisburg, PA, United States
| | - Konstantina A Svokos
- The Warren Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
15
|
Wang Y, Ma F, Rodriguez EL, Klein JD, Sands JM. Aldosterone Decreases Vasopressin-Stimulated Water Reabsorption in Rat Inner Medullary Collecting Ducts. Cells 2020; 9:cells9040967. [PMID: 32295252 PMCID: PMC7226978 DOI: 10.3390/cells9040967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 11/27/2022] Open
Abstract
Aldosterone indirectly regulates water reabsorption in the distal tubule by regulating sodium reabsorption. However, the direct effect of aldosterone on vasopressin-regulated water and urea permeability in the rat inner medullary collecting duct (IMCD) has not been tested. We investigated whether aldosterone regulates osmotic water permeability in isolated perfused rat IMCDs. Adding aldosterone (500 nM) to the bath significantly decreased osmotic water permeability in the presence of vasopressin (50 pM) in both male and female rat IMCDs. Aldosterone significantly decreased aquaporin-2 (AQP2) phosphorylation at S256 but did not change it at S261. Previous studies show that aldosterone can act both genomically and non-genomically. We tested the mechanism by which aldosterone attenuates osmotic water permeability. Blockade of gene transcription with actinomycin D did not reverse aldosterone-attenuated osmotic water permeability. In addition to AQP2, the urea transporter UT-A1 contributes to vasopressin-regulated urine concentrating ability. We tested aldosterone-regulated urea permeability in vasopressin-treated IMCDs. Blockade of gene transcription did not reverse aldosterone-attenuated urea permeability. In conclusion, aldosterone directly regulates water reabsorption through a non-genomic mechanism. Aldosterone-attenuated water reabsorption may be related to decreased trafficking of AQP2 to the plasma membrane. There may be a sex difference apparent in the inhibitory effect of aldosterone on water reabsorption in the inner medullary collecting duct. This study is the first to show a direct effect of aldosterone to inhibit vasopressin-stimulated osmotic water permeability and urea permeability in perfused rat IMCDs.
Collapse
Affiliation(s)
| | | | | | | | - Jeff M. Sands
- Correspondence: ; Tel.: +1-404-7272-525; Fax: +1-404-7273-425
| |
Collapse
|
16
|
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
|
17
|
Törnroth-Horsefield S. Phosphorylation of human AQP2 and its role in trafficking. VITAMINS AND HORMONES 2019; 112:95-117. [PMID: 32061351 DOI: 10.1016/bs.vh.2019.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human Aquaporin 2 (AQP2) is a membrane-bound water channel found in the kidney collecting duct whose regulation by trafficking plays a key role in regulating urine volume. AQP2 trafficking is tightly controlled by the pituitary hormone arginine vasopressin (AVP), which stimulates translocation of AQP2 residing in storage vesicles to the apical membrane. The AVP-dependent translocation of AQP2 to and from the apical membrane is controlled by multiple phosphorylation sites in the AQP2 C-terminus, the phosphorylation of which alters its affinity to proteins within the cellular membrane protein trafficking machinery. The aim of this chapter is to provide a summary of what is currently known about AVP-mediated AQP2 trafficking, dissecting the roles of individual phosphorylation sites, kinases and phosphatases and interacting proteins. From this, the picture of an immensely complex process emerges, of which many structural and molecular details remains to be elucidated.
Collapse
|
18
|
Nooh MM, Kale A, Bahouth SW. Involvement of PDZ-SAP97 interactions in regulating AQP2 translocation in response to vasopressin in LLC-PK 1 cells. Am J Physiol Renal Physiol 2019; 317:F375-F387. [PMID: 31141395 PMCID: PMC6732448 DOI: 10.1152/ajprenal.00228.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 11/22/2022] Open
Abstract
Arginine-vasopressin (AVP)-mediated translocation of aquaporin-2 (AQP2) protein-forming water channels from storage vesicles to the membrane of renal collecting ducts is critical for the renal conservation of water. The type-1 PDZ-binding motif (PBM) in AQP2, "GTKA," is a critical barcode for its translocation, but its precise role and that of its interacting protein partners in this process remain obscure. We determined that synapse-associated protein-97 (SAP97), a membrane-associated guanylate kinase protein involved in establishing epithelial cell polarity, was an avid binding partner to the PBM of AQP2. The role of PBM and SAP97 on AQP2 redistribution in response to AVP was assessed in LLC-PK1 renal collecting cells by confocal microscopy and cell surface biotinylation techniques. These experiments indicated that distribution of AQP2 and SAP97 overlapped in the kidneys and LLC-PK1 cells and that knockdown of SAP97 inhibited the translocation of AQP2 in response to AVP. Binding between AQP2 and SAP97 was mediated by specific interactions between the second PDZ of SAP97 and PBM of AQP2. Mechanistically, inactivation of the PBM of AQP2, global delocalization of PKA, or knockdown of SAP97 inhibited AQP2 translocation as well as AVP- and forskolin-mediated phosphorylation of Ser256 in AQP2, which serves as the major translocation barcode of AQP2. These results suggest that the targeting of PKA to the microdomain of AQP2 via SAP97-AQP2 interactions in association with cross-talk between two barcodes in AQP2, namely, the PBM and phospho-Ser256, plays an important role in the translocation of AQP2 in the kidney.
Collapse
Affiliation(s)
- Mohammed M Nooh
- Department of Pharmacology, The University of Tennessee Health Sciences Center, Memphis, Tennessee
- Department of Biochemistry, Faculty of Pharmacy Cairo University, Cairo, Egypt
| | - Ajay Kale
- Department of Pharmacology, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana
| | - Suleiman W Bahouth
- Department of Pharmacology, The University of Tennessee Health Sciences Center, Memphis, Tennessee
| |
Collapse
|
19
|
Oshikawa-Hori S, Yokota-Ikeda N, Sonoda H, Ikeda M. Urinary extracellular vesicular release of aquaporins in patients with renal transplantation. BMC Nephrol 2019; 20:216. [PMID: 31185935 PMCID: PMC6580655 DOI: 10.1186/s12882-019-1398-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 05/28/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Diuresis has been observed within a week following renal transplantation, suggesting that the procedure causes acute disturbance of renal water homeostasis. Aquaporin (AQP) 1 and AQP2, important proteins for renal water reabsorption, have been identified in urinary extracellular vesicles (uEV-AQP1 and -AQP2), and experimental studies have shown that the presence of uEV-AQP1 and -AQP2 may be an indicator of their levels of expression in the kidney. However, the release patterns of uEV-AQP1 and -AQP2 during the acute phase following renal transplantation are largely unknown. METHODS In this study, we examined the release of uEV-AQP1 and -AQP2 in recipients until 6 days (day 6) after renal transplantation. At Miyazaki prefectural Miyazaki Hospital, Japan, uEVs were obtained from 7 recipients, all of whom had received renal allografts from living donors. uEVs were isolated by differential centrifugation. RESULTS Immunoblotting analysis showed that the release of uEV-AQP2 was significantly decreased on day 1 in comparison with a control sample (from 3 healthy volunteers), accompanied by high urine output and low urine osmolality. Thereafter, the level increased gradually to the control level by day 6. The release pattern of uEV-AQP1 was similar to that of uEV-AQP2, but the levels did not reach statistical significance in comparison with the control level at any of the time points examined. Evaluation of the relationship between urinary osmolality and uEV-AQPs revealed a significant correlation for uEV-AQP2, but not for uEV-AQP1. CONCLUSION These results indicate that acute diuresis after renal transplantation might be due to a decrease in the renal expression of AQP2, whose level can be estimated from the amount released in uEVs.
Collapse
Affiliation(s)
- Sayaka Oshikawa-Hori
- Department of Veterinary Pharmacology, Faculty of Agriculture, University of Miyazaki, Gakuenkibanadai-Nishi 1-1, Miyazaki, 889-2192 Japan
| | - Naoko Yokota-Ikeda
- Nephrology, Miyazaki Prefectural Miyazaki Hospital, Kitatakamatsu 5-30, Miyazaki, 880-8510 Japan
| | - Hiroko Sonoda
- Department of Veterinary Pharmacology, Faculty of Agriculture, University of Miyazaki, Gakuenkibanadai-Nishi 1-1, Miyazaki, 889-2192 Japan
| | - Masahiro Ikeda
- Department of Veterinary Pharmacology, Faculty of Agriculture, University of Miyazaki, Gakuenkibanadai-Nishi 1-1, Miyazaki, 889-2192 Japan
| |
Collapse
|
20
|
Nesverova V, Törnroth-Horsefield S. Phosphorylation-Dependent Regulation of Mammalian Aquaporins. Cells 2019; 8:cells8020082. [PMID: 30678081 PMCID: PMC6406877 DOI: 10.3390/cells8020082] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/17/2019] [Accepted: 01/21/2019] [Indexed: 12/26/2022] Open
Abstract
Water homeostasis is fundamental for cell survival. Transport of water across cellular membranes is governed by aquaporins—tetrameric integral membrane channels that are highly conserved throughout the prokaryotic and eukaryotic kingdoms. In eukaryotes, specific regulation of these channels is required and is most commonly carried out by shuttling the protein between cellular compartments (trafficking) or by opening and closing the channel (gating). Structural and functional studies have revealed phosphorylation as a ubiquitous mechanism in aquaporin regulation by both regulatory processes. In this review we summarize what is currently known about the phosphorylation-dependent regulation of mammalian aquaporins. Focusing on the water-specific aquaporins (AQP0–AQP5), we discuss how gating and trafficking are controlled by phosphorylation and how phosphorylation affects the binding of aquaporins to regulatory proteins, thereby highlighting structural details and dissecting the contribution of individual phosphorylated residues when possible. Our aim is to provide an overview of the mechanisms behind how aquaporin phosphorylation controls cellular water balance and to identify key areas where further studies are needed.
Collapse
Affiliation(s)
- Veronika Nesverova
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, Box 124, 221 00 Lund, Sweden.
| | - Susanna Törnroth-Horsefield
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, Box 124, 221 00 Lund, Sweden.
| |
Collapse
|
21
|
Aquaporins in Renal Diseases. Int J Mol Sci 2019; 20:ijms20020366. [PMID: 30654539 PMCID: PMC6359174 DOI: 10.3390/ijms20020366] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 12/16/2022] Open
Abstract
Aquaporins (AQPs) are a family of highly selective transmembrane channels that mainly transport water across the cell and some facilitate low-molecular-weight solutes. Eight AQPs, including AQP1, AQP2, AQP3, AQP4, AQP5, AQP6, AQP7, and AQP11, are expressed in different segments and various cells in the kidney to maintain normal urine concentration function. AQP2 is critical in regulating urine concentrating ability. The expression and function of AQP2 are regulated by a series of transcriptional factors and post-transcriptional phosphorylation, ubiquitination, and glycosylation. Mutation or functional deficiency of AQP2 leads to severe nephrogenic diabetes insipidus. Studies with animal models show AQPs are related to acute kidney injury and various chronic kidney diseases, such as diabetic nephropathy, polycystic kidney disease, and renal cell carcinoma. Experimental data suggest ideal prospects for AQPs as biomarkers and therapeutic targets in clinic. This review article mainly focuses on recent advances in studying AQPs in renal diseases.
Collapse
|
22
|
Cheung PW, Ueberdiek L, Day J, Bouley R, Brown D. Protein phosphatase 2C is responsible for VP-induced dephosphorylation of AQP2 serine 261. Am J Physiol Renal Physiol 2017; 313:F404-F413. [PMID: 28381458 PMCID: PMC5582913 DOI: 10.1152/ajprenal.00004.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/28/2017] [Accepted: 04/03/2017] [Indexed: 12/11/2022] Open
Abstract
Aquaporin 2 (AQP2) trafficking is regulated by phosphorylation and dephosphorylation of serine residues in the AQP2 COOH terminus. Vasopressin (VP) binding to its receptor (V2R) leads to a cascade of events that result in phosphorylation of serine 256 (S256), S264, and S269, but dephosphorylation of S261. To identify which phosphatase is responsible for VP-induced S261 dephosphorylation, we pretreated cells with different phosphatase inhibitors before VP stimulation. Sanguinarine, a specific protein phosphatase (PP) 2C inhibitor, but not inhibitors of PP1, PP2A (okadaic acid), or PP2B (cyclosporine), abolished VP-induced S261 dephosphorylation. However, sanguinarine and VP significantly increased phosphorylation of ERK, a kinase that can phosphorylate S261; inhibition of ERK by PD98059 partially decreased baseline S261 phosphorylation. These data support a role of ERK in S261 phosphorylation but suggest that, upon VP treatment, increased phosphatase activity overcomes the increase in ERK activity, resulting in overall dephosphorylation of S261. We also found that sanguinarine abolished VP-induced S261 dephosphorylation in cells expressing mutated AQP2 S256A, suggesting that the phosphorylation state of S261 is independent of S256. Sanguinarine alone did not induce AQP2 membrane trafficking, nor did it inhibit VP-induced AQP2 membrane accumulation in cells and kidney tissues, suggesting that S261 does not play an observable role in acute AQP2 membrane accumulation. In conclusion, PP2C activity is required for S261 AQP2 dephosphorylation upon VP stimulation, which occurs independently of S256 phosphorylation. Understanding the pathways involved in modulating PP2C will help elucidate the role of S261 in cellular events involving AQP2.
Collapse
Affiliation(s)
- Pui W Cheung
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lars Ueberdiek
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jack Day
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Richard Bouley
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dennis Brown
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
23
|
Li Y, Wang W, Jiang T, Yang B. Aquaporins in Urinary System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 969:131-148. [PMID: 28258571 DOI: 10.1007/978-94-024-1057-0_9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Several aquaporin (AQP )-type water channels are expressed in kidney: AQP1 in the proximal tubule, thin descending limb of Henle, and vasa recta; AQP2 -6 in the collecting duct; AQP7 in the proximal tubule; AQP8 in the proximal tubule and collecting duct; and AQP11 in the endoplasmic reticulum of proximal tubule cells. AQP2 is the vasopressin-regulated water channel that is important in hereditary and acquired diseases affecting urine-concentrating ability. The roles of AQPs in renal physiology and transepithelial water transport have been determined using AQP knockout mouse models. This chapter describes renal physiologic insights revealed by phenotypic analysis of AQP knockout mice and the prospects for further basic and clinical studies.
Collapse
Affiliation(s)
- Yingjie Li
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Weiling Wang
- State Key Laboratory of Natural and Biomimetic Drugs, and Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Tao Jiang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China.
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, 100191, China.
| |
Collapse
|
24
|
LeMaire SM, Raghuram V, Grady CR, Pickering CM, Chou CL, Umejiego EN, Knepper MA. Serine/threonine phosphatases and aquaporin-2 regulation in renal collecting duct. Am J Physiol Renal Physiol 2016; 312:F84-F95. [PMID: 27784696 DOI: 10.1152/ajprenal.00455.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/05/2016] [Accepted: 10/19/2016] [Indexed: 11/22/2022] Open
Abstract
Phosphorylation of the aquaporin-2 (AQP2) water channel at four COOH-terminal serines plays a central role in the regulation of water permeability of the renal collecting duct. The level of phosphorylation at these sites is determined by a balance between phosphorylation by protein kinases and dephosphorylation by phosphatases. The phosphatases that dephosphorylate AQP2 have not been identified. Here, we use large-scale data integration techniques to identify serine-threonine phosphatases likely to interact with AQP2 in renal collecting duct principal cells. As a first step, we have created a comprehensive list of 38 S/T phosphatase catalytic subunits present in the mammalian genome. Then we used Bayes' theorem to integrate available information from large-scale data sets from proteomic and transcriptomic studies to rank the known S/T phosphatases with regard to the likelihood that they interact with AQP2 in renal collecting duct cells. To broaden the analysis, we have generated new proteomic data (LC-MS/MS) identifying 4538 distinct proteins including 22 S/T phosphatases in cytoplasmic fractions from native inner medullary collecting duct cells from rats. The official gene symbols corresponding to the top-ranked phosphatases (common names in parentheses) were: Ppp1cb (PP1-β), Ppm1g (PP2C), Ppp1ca (PP1-α), Ppp3ca (PP2-B or calcineurin), Ppp2ca (PP2A-α), Ppp1cc (PP1-γ), Ppp2cb (PP2A-β), Ppp6c (PP6C), and Ppp5c (PP5). This ranking correlates well with results of prior reductionist studies of ion and water channels in renal collecting duct cells.
Collapse
Affiliation(s)
- Sophia M LeMaire
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and.,Howard University College of Medicine, Washington, District of Columbia
| | - Viswanathan Raghuram
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Cameron R Grady
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Christina M Pickering
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Ezigbobiara N Umejiego
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| |
Collapse
|
25
|
Jung HJ, Kwon TH. Molecular mechanisms regulating aquaporin-2 in kidney collecting duct. Am J Physiol Renal Physiol 2016; 311:F1318-F1328. [PMID: 27760771 DOI: 10.1152/ajprenal.00485.2016] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/11/2016] [Accepted: 10/11/2016] [Indexed: 01/04/2023] Open
Abstract
The kidney collecting duct is an important renal tubular segment for regulation of body water homeostasis and urine concentration. Water reabsorption in the collecting duct principal cells is controlled by vasopressin, a peptide hormone that induces the osmotic water transport across the collecting duct epithelia through regulation of water channel proteins aquaporin-2 (AQP2) and aquaporin-3 (AQP3). In particular, vasopressin induces both intracellular translocation of AQP2-bearing vesicles to the apical plasma membrane and transcription of the Aqp2 gene to increase AQP2 protein abundance. The signaling pathways, including AQP2 phosphorylation, RhoA phosphorylation, intracellular calcium mobilization, and actin depolymerization, play a key role in the translocation of AQP2. This review summarizes recent data demonstrating the regulation of AQP2 as the underlying molecular mechanism for the homeostasis of water balance in the body.
Collapse
Affiliation(s)
- Hyun Jun Jung
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea
| |
Collapse
|