1
|
Zhang Y, Yan M, Yu Y, Wang J, Jiao Y, Zheng M, Zhang S. 14-3-3ε: a protein with complex physiology function but promising therapeutic potential in cancer. Cell Commun Signal 2024; 22:72. [PMID: 38279176 PMCID: PMC10811864 DOI: 10.1186/s12964-023-01420-w] [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: 08/25/2023] [Accepted: 12/02/2023] [Indexed: 01/28/2024] Open
Abstract
Over the past decade, the role of the 14-3-3 protein has received increasing interest. Seven subtypes of 14-3-3 proteins exhibit high homology; however, each subtype maintains its specificity. The 14-3-3ε protein is involved in various physiological processes, including signal transduction, cell proliferation, apoptosis, autophagy, cell cycle regulation, repolarization of cardiac action, cardiac development, intracellular electrolyte homeostasis, neurodevelopment, and innate immunity. It also plays a significant role in the development and progression of various diseases, such as cardiovascular diseases, inflammatory diseases, neurodegenerative disorders, and cancer. These immense and various involvements of 14-3-3ε in diverse processes makes it a promising target for drug development. Although extensive research has been conducted on 14-3-3 dimers, studies on 14-3-3 monomers are limited. This review aimed to provide an overview of recent reports on the molecular mechanisms involved in the regulation of binding partners by 14-3-3ε, focusing on issues that could help advance the frontiers of this field. Video Abstract.
Collapse
Affiliation(s)
- Yue Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Man Yan
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Yongjun Yu
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121, People's Republic of China
| | - Jiangping Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Yuqi Jiao
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300071, People's Republic of China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300071, People's Republic of China.
| |
Collapse
|
2
|
14-3-3-protein regulates Nedd4-2 by modulating interactions between HECT and WW domains. Commun Biol 2021; 4:899. [PMID: 34294877 PMCID: PMC8298602 DOI: 10.1038/s42003-021-02419-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/05/2021] [Indexed: 11/16/2022] Open
Abstract
Neural precursor cell expressed developmentally down-regulated 4 ligase (Nedd4-2) is an E3 ubiquitin ligase that targets proteins for ubiquitination and endocytosis, thereby regulating numerous ion channels, membrane receptors and tumor suppressors. Nedd4-2 activity is regulated by autoinhibition, calcium binding, oxidative stress, substrate binding, phosphorylation and 14-3-3 protein binding. However, the structural basis of 14-3-3-mediated Nedd4-2 regulation remains poorly understood. Here, we combined several techniques of integrative structural biology to characterize Nedd4-2 and its complex with 14-3-3. We demonstrate that phosphorylated Ser342 and Ser448 are the key residues that facilitate 14-3-3 protein binding to Nedd4-2 and that 14-3-3 protein binding induces a structural rearrangement of Nedd4-2 by inhibiting interactions between its structured domains. Overall, our findings provide the structural glimpse into the 14-3-3-mediated Nedd4-2 regulation and highlight the potential of the Nedd4-2:14-3-3 complex as a pharmacological target for Nedd4-2-associated diseases such as hypertension, epilepsy, kidney disease and cancer. Pohl et al. investigated the structural basis of Nedd4-2 regulation by 14-3-3 and found that phosphorylated Ser342 and Ser448 are the main residues that facilitate 14-3-3 binding to Nedd4-2. The authors propose that the Nedd4-2:14-3-3 complex then stimulates a structural rearrangement of Nedd4-2 through inhibiting interaction of its structured domains.
Collapse
|
3
|
Abstract
The Epithelial Na+ Channel, ENaC, comprised of 3 subunits (αβγ, or sometimes δβγENaC), plays a critical role in regulating salt and fluid homeostasis in the body. It regulates fluid reabsorption into the blood stream from the kidney to control blood volume and pressure, fluid absorption in the lung to control alveolar fluid clearance at birth and maintenance of normal airway surface liquid throughout life, and fluid absorption in the distal colon and other epithelial tissues. Moreover, recent studies have also revealed a role for sodium movement via ENaC in nonepithelial cells/tissues, such as endothelial cells in blood vessels and neurons. Over the past 25 years, major advances have been made in our understanding of ENaC structure, function, regulation, and role in human disease. These include the recently solved three-dimensional structure of ENaC, ENaC function in various tissues, and mutations in ENaC that cause a hereditary form of hypertension (Liddle syndrome), salt-wasting hypotension (PHA1), or polymorphism in ENaC that contributes to other diseases (such as cystic fibrosis). Moreover, great strides have been made in deciphering the regulation of ENaC by hormones (e.g., the mineralocorticoid aldosterone, glucocorticoids, vasopressin), ions (e.g., Na+ ), proteins (e.g., the ubiquitin-protein ligase NEDD4-2, the kinases SGK1, AKT, AMPK, WNKs & mTORC2, and proteases), and posttranslational modifications [e.g., (de)ubiquitylation, glycosylation, phosphorylation, acetylation, palmitoylation]. Characterization of ENaC structure, function, regulation, and role in human disease, including using animal models, are described in this article, with a special emphasis on recent advances in the field. © 2021 American Physiological Society. Compr Physiol 11:1-29, 2021.
Collapse
Affiliation(s)
- Daniela Rotin
- The Hospital for Sick Children, and The University of Toronto, Toronto, Canada
| | - Olivier Staub
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
4
|
Bukhari AAS, Zhang X, Li M, Zhao A, Dong H, Liang X. Cofilin participates in regulating alpha-epithelial sodium channel by interaction with 14-3-3 isoforms. J Biomed Res 2020; 34:351-360. [PMID: 32981895 PMCID: PMC7540242 DOI: 10.7555/jbr.34.20190155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Renal epithelial sodium channel (ENaC) plays a crucial role in maintaining homeostasis and sodium absorption. While insulin participates in controlling sodium transport across the renal epithelium, the underlying molecular mechanism remain unclear. In this study, we found that insulin increased the expression and function of alpha-epithelial sodium channel (α-ENaC) as well as phosphorylation of cofilin, a family of actin-binding proteins which disassembles actin filaments, in mouse cortical collecting duct (mpkCCDc14) cells. The wild-type (WT) cofilin and its constitutively phosphorylated form (S3D), but not its constitutively non-phosphorylable form (S3A), contributed to the elevated expression on α-ENaC. Overexpression of 14-3-3ε, β, or γ increased the expression of α-ENaC and cofilin phosphorylation, which was blunted by knockdown of 14-3-3ε, β, or γ. Moreover, it was found that insulin increased the interaction between cofilin and 14-3-3 isoforms, which indicated relevance of 14-3-3 isoforms with cofilin. Furthermore, LIMK1/SSH1 pathway was involved in regulation of cofilin and α-ENaC expression by insulin. The results from this work indicate that cofilin participates in the regulation of α-ENaC by interaction with 14-3-3 isoforms.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiubin Liang
- Department of Pathophysiology;Department of Nephrology, the Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
5
|
Zhu J, Lee KY, Jong TT, Tsai NP. C2-lacking isoform of Nedd4-2 regulates excitatory synaptic strength through GluA1 ubiquitination-independent mechanisms. J Neurochem 2019; 151:289-300. [PMID: 31357244 DOI: 10.1111/jnc.14840] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 01/06/2023]
Abstract
Neural precursor cell expressed developmentally downregulated gene 4-like (Nedd4-2) is an epilepsy-associated gene, which encodes a ubiquitin E3 ligase that is highly expressed in the brain. Nedd4-2's substrates include many ion channels and receptors because its N-terminal C2 domain guides Nedd4-2 to the cell membrane. We previously found that Nedd4-2 ubiquitinates the glutamate receptor subunit 1 (GluA1) subunit of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor, which leads to reduction of neuronal excitability and seizures in mice. However, despite awareness of a Nedd4-2 isoform with no C2 domain, the functions of this isoform remain elusive. In this study, we showed that the C2-lacking Nedd4-2 has reduced membrane distribution and exhibits reduced affinity toward ubiquitinating GluA1. However, when expressed in primary cortical neurons, we found that the C2-lacking Nedd4-2 exhibits a similar activity toward reducing excitatory synaptic strength as does the C2-containing Nedd4-2. In an attempt to identify novel Nedd4-2 substrates that could mediate excitatory synaptic strength, we used unbiased proteomic screening and found multiple synaptic regulators that were up-regulated in the brain of conditional Nedd4-2 knockout mice, including protein phosphatase 3 catalytic subunit-α (PPP3CA; alternately called calcineurin A-α). We confirmed PPP3CA as a substrate of the C2-lacking Nedd4-2 and showed that all three epilepsy-associated missense mutations of Nedd4-2 disrupted PPP3CA ubiquitination. Altogether, our results revealed novel potential Nedd4-2 substrates and suggest that the C2-lacking Nedd4-2 represses excitatory synaptic strength most likely through GluA1 ubiquitination-independent mechanisms. These findings provide novel information to further our knowledge about Nedd4-2-dependent neuronal excitability homeostasis and pathological hyperexcitability when Nedd4-2 is compromised.
Collapse
Affiliation(s)
- Jiuhe Zhu
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Tiffany T Jong
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
6
|
Spirli A, Cheval L, Debonneville A, Penton D, Ronzaud C, Maillard M, Doucet A, Loffing J, Staub O. The serine-threonine kinase PIM3 is an aldosterone-regulated protein in the distal nephron. Physiol Rep 2019; 7:e14177. [PMID: 31397090 PMCID: PMC6687858 DOI: 10.14814/phy2.14177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 12/30/2022] Open
Abstract
The mineralocorticoid hormone aldosterone plays a crucial role in the control of Na+ and K+ balance, blood volume, and arterial blood pressure, by acting in the aldosterone-sensitive distal nephron (ASDN) and stimulating a complex transcriptional, translational, and cellular program. Because the complexity of the aldosterone response is still not fully appreciated, we aimed at identifying new elements in this pathway. Here, we demonstrate that the expression of the proto-oncogene PIM3 (Proviral Integration Site of Moloney Murine Leukemia Virus 3), a serine/threonine kinase belonging to the calcium/calmodulin-regulated group of kinases, is stimulated by aldosterone in vitro (mCCDcl1 cells), ex vivo (mouse kidney slices), and in vivo in mice. Characterizing a germline Pim3-/- mouse model, we found that these mice have an upregulated Renin-Angiotensin-Aldosterone System (RAAS), with high circulating aldosterone and plasma renin activity levels on both standard or Na+ -deficient diet. Surprisingly, we did not observe any obvious salt-losing phenotype in Pim3 KO mice as shown by normal blood pressure, plasma and urinary electrolytes, as well as unchanged expression levels of the major Na+ transport proteins. These observations suggest that the potential effects of the loss of the Pim3 gene are physiologically compensated. Indeed, the 2 other family members of the PIM kinase family, PIM1 and PIM2 are upregulated in the kidney of Pim3-/- mice, and may therefore be involved in such compensation. In conclusion, our data demonstrate that the PIM3 kinase is a novel aldosterone-induced protein, but its precise role in aldosterone-dependent renal homeostasis remains to be determined.
Collapse
Affiliation(s)
- Alessia Spirli
- Department of Pharmacology & ToxicologyUniversity of LausanneLausanneSwitzerland
- National Centre of Competence in Research “Kidney.ch”LausanneSwitzerland
| | - Lydie Cheval
- Centre de Recherche des CordeliersINSERM, Sorbonne Universités, USPC, Université Paris Descartes, Université Paris Diderot, Physiologie Rénale et TubulopathiesParisFrance
| | - Anne Debonneville
- Department of Pharmacology & ToxicologyUniversity of LausanneLausanneSwitzerland
- National Centre of Competence in Research “Kidney.ch”LausanneSwitzerland
| | - David Penton
- National Centre of Competence in Research “Kidney.ch”LausanneSwitzerland
- Institute of AnatomyUniversity of ZurichZurichSwitzerland
| | - Caroline Ronzaud
- Department of Pharmacology & ToxicologyUniversity of LausanneLausanneSwitzerland
- National Centre of Competence in Research “Kidney.ch”LausanneSwitzerland
| | - Marc Maillard
- Service of NephrologyLausanne University Hospital (CHUV)LausanneSwitzerland
| | - Alain Doucet
- Centre de Recherche des CordeliersINSERM, Sorbonne Universités, USPC, Université Paris Descartes, Université Paris Diderot, Physiologie Rénale et TubulopathiesParisFrance
| | - Johannes Loffing
- National Centre of Competence in Research “Kidney.ch”LausanneSwitzerland
- Institute of AnatomyUniversity of ZurichZurichSwitzerland
| | - Olivier Staub
- Department of Pharmacology & ToxicologyUniversity of LausanneLausanneSwitzerland
- National Centre of Competence in Research “Kidney.ch”LausanneSwitzerland
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW The review describes studies investigating the role of microRNAs in the signaling pathway of the mineralocorticoid hormone, aldosterone. RECENT FINDINGS Emerging evidence indicates that aldosterone alters the expression of microRNAs in target tissues thereby modulating the expression of key regulatory proteins. SUMMARY The mineralocorticoid hormone aldosterone is released by the adrenal glands in a homeostatic mechanism to regulate blood volume. The long-term renal action of aldosterone is to increase the retrieval of sodium from filtered plasma to restore blood pressure. Emerging evidence indicates aldosterone may alter noncoding RNAs (ncRNAs) to integrate this hormonal response in target tissue. Expression of the best characterized small ncRNAs, microRNAs, is regulated by aldosterone stimulation. MicroRNAs modulate protein expression at all steps in the renin-angiotensin-aldosterone-signaling (RAAS) system. In addition to acting as a rheostat to fine-tune protein levels in aldosterone-responsive cells, there is evidence that microRNAs down-regulate components of the signaling cascade as a feedback mechanism. The role of microRNAs is, therefore, as signal integrator, and damper in aldosterone signaling, which has implications in understating the RAAS system from both a physiological and pathophysiological perspective. Recent evidence for microRNA's role in RAAS signaling will be discussed.
Collapse
|
8
|
Ho PY, Li H, Pavlov TS, Tuerk RD, Tabares D, Brunisholz R, Neumann D, Staruschenko A, Hallows KR. β 1Pix exchange factor stabilizes the ubiquitin ligase Nedd4-2 and plays a critical role in ENaC regulation by AMPK in kidney epithelial cells. J Biol Chem 2018; 293:11612-11624. [PMID: 29858246 DOI: 10.1074/jbc.ra118.003082] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/22/2018] [Indexed: 12/22/2022] Open
Abstract
Our previous work has established that the metabolic sensor AMP-activated protein kinase (AMPK) inhibits the epithelial Na+ channel (ENaC) by promoting its binding to neural precursor cell-expressed, developmentally down-regulated 4-2, E3 ubiquitin protein ligase (Nedd4-2). Here, using MS analysis and in vitro phosphorylation, we show that AMPK phosphorylates Nedd4-2 at the Ser-444 (Xenopus Nedd4-2) site critical for Nedd4-2 stability. We further demonstrate that the Pak-interacting exchange factor β1Pix is required for AMPK-mediated inhibition of ENaC-dependent currents in both CHO and murine kidney cortical collecting duct (CCD) cells. Short hairpin RNA-mediated knockdown of β1Pix expression in CCD cells attenuated the inhibitory effect of AMPK activators on ENaC currents. Moreover, overexpression of a β1Pix dimerization-deficient mutant unable to bind 14-3-3 proteins (Δ602-611) increased ENaC currents in CCD cells, whereas overexpression of WT β1Pix had the opposite effect. Using additional immunoblotting and co-immunoprecipitation experiments, we found that treatment with AMPK activators promoted the binding of β1Pix to 14-3-3 proteins in CCD cells. However, the association between Nedd4-2 and 14-3-3 proteins was not consistently affected by AMPK activation, β1Pix knockdown, or overexpression of WT β1Pix or the β1Pix-Δ602-611 mutant. Moreover, we found that β1Pix is important for phosphorylation of the aforementioned Nedd4-2 site critical for its stability. Overall, these findings elucidate novel molecular mechanisms by which AMPK regulates ENaC. Specifically, they indicate that AMPK promotes the assembly of β1Pix, 14-3-3 proteins, and Nedd4-2 into a complex that inhibits ENaC by enhancing Nedd4-2 binding to ENaC and its degradation.
Collapse
Affiliation(s)
- Pei-Yin Ho
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Hui Li
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Tengis S Pavlov
- the Division of Hypertension and Vascular Research, Henry Ford Health System, Detroit, Michigan 48202; Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Roland D Tuerk
- Department of Biology, Institute of Cell Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Diego Tabares
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - René Brunisholz
- Functional Genomics Center, ETH Zurich, 8097 Zurich, Switzerland
| | - Dietbert Neumann
- Department of Biology, Institute of Cell Biology, ETH Zurich, 8093 Zurich, Switzerland; Department of Pathology, School for Cardiovascular Diseases, Maastricht University, 6200 MD Maastricht, The Netherlands
| | | | - Kenneth R Hallows
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California 90033.
| |
Collapse
|
9
|
Huang Y, Sun Y, Cao Y, Sun H, Li M, You H, Su D, Li Y, Liang X. HRD1 prevents apoptosis in renal tubular epithelial cells by mediating eIF2α ubiquitylation and degradation. Cell Death Dis 2017; 8:3202. [PMID: 29233968 PMCID: PMC5870601 DOI: 10.1038/s41419-017-0002-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/09/2017] [Accepted: 09/21/2017] [Indexed: 12/30/2022]
Abstract
Apoptosis of renal tubular epithelial cells is a key feature of the pathogenicity associated with tubulointerstitial fibrosis and other kidney diseases. One factor that regulates important cellular processes like apoptosis and cell proliferation is HRD1, an E3 ubiquitin ligase that acts by promoting ubiquitylation and degradation of its target protein. However, the detailed mechanisms by which HRD1 acts as a regulator of apoptosis in renal tubular epithelial cells have not been established. In our previous liquid chromatography-tandem mass spectrometry (LC-MS/MS) study (Mol Endocrinol. 2016;30:600–613), we demonstrated that one substrate of HRD1 was eIF2α, a critical protein in the PERK-eIF2α-ATF4-CHOP signaling pathway of endoplasmic reticulum (ER) stress. Here, we show that eIF2α expression was increased and HRD1 expression decreased when apoptosis was induced in HKC-8 cells by palmitic acid (PA) or high glucose (HG). HRD1 expression was also lower in kidney tissues from mice with diabetic nephropathy (DN) than in control mice. Forced expression of HRD1 also inhibited apoptosis in HKC-8 cells, while HRD1 overexpression decreased the expression of phosphorylated eIF2α and eIF2α. Further analysis indicated that HRD1 interacted with eIF2α and promoted its ubiquitylation and degradation by the proteasome. Moreover, the HRD1 protection of PA-treated HKC-8 cells was blunted by transfection with Myc-eIF2α. Thus, eIF2α ubiquitylation by HRD1 protects tubular epithelial cells from apoptosis caused by HG and PA, indicating a novel upstream target for therapeutic prevention of renal tubulointerstitial injury.
Collapse
Affiliation(s)
- Yujie Huang
- Renal Division, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.,Department of Pathology, Jiangsu Province Hospital of TCM, Affiliated Hospital of Nanjing University of TCM, Nanjing, 210026, Jiangsu Province, China
| | - Yifei Sun
- Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yizhi Cao
- Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Hui Sun
- Renal Division, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Min Li
- Renal Division, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.,Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Hui You
- Renal Division, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Dongming Su
- Center of Pathology and Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Yanjiao Li
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China
| | - Xiubin Liang
- Renal Division, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu, China. .,Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
10
|
Direct interaction with 14-3-3γ promotes surface expression of Best1 channel in astrocyte. Mol Brain 2017; 10:51. [PMID: 29121962 PMCID: PMC5679146 DOI: 10.1186/s13041-017-0331-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/28/2017] [Indexed: 01/01/2023] Open
Abstract
Background Bestrophin-1 (Best1) is a calcium-activated anion channel (CAAC) that is expressed broadly in mammalian tissues including the brain. We have previously reported that Best1 is expressed in hippocampal astrocytes at the distal peri-synaptic regions, called microdomains, right next to synaptic junctions, and that it disappears from the microdomains in Alzheimer’s disease mouse model. Although Best1 appears to be dynamically regulated, the mechanism of its regulation and modulation is poorly understood. It has been reported that a regulatory protein, 14-3-3 affects the surface expression of numerous membrane proteins in mammalian cells. Methods The protein-protein interaction between Best1 and 14-3-3γ was confirmed by yeast-two hybrid assay and BiFC method. The effect of 14-3-3γ on Best1-mediated current was measured by whole-cell patch clamp technique. Results We identified 14-3-3γ as novel binding partner of Best1 in astrocytes: among 7 isoforms of 14-3-3 protein, only 14-3-3γ was found to bind specifically. We determined a binding domain on the C-terminus of Best1 which is critical for an interaction with 14-3-3γ. We also revealed that interaction between Best1 and 14-3-3γ was mediated by phosphorylation of S358 in the C-terminus of Best1. We confirmed that surface expression of Best1 and Best1-mediated whole-cell current were significantly decreased after a gene-silencingof 14-3-3γ without a significant change in total Best1 expression in cultured astrocytes. Furthermore, we discovered that 14-3-3γ-shRNA reduced Best1-mediated glutamate release from hippocampal astrocyte by recording a PAR1 receptor-induced NMDA receptor-mediated current from CA1 pyramidal neurons in hippocampal slices injected with adenovirus carrying 14-3-3γ-shRNA. Finally, through a structural modeling, we found critical amino acid residues containing S358 of Best1 exhibiting binding affinities to 14-3-3γ. Conclusions 14-3-3γ promotes surface expression of Best1 channel in astrocytes through direct interaction.
Collapse
|
11
|
Dang VD, Jella KK, Ragheb RRT, Denslow ND, Alli AA. Lipidomic and proteomic analysis of exosomes from mouse cortical collecting duct cells. FASEB J 2017; 31:5399-5408. [PMID: 28821634 DOI: 10.1096/fj.201700417r] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 07/25/2017] [Indexed: 12/13/2022]
Abstract
Exosomes are endosome-derived nanovesicles that are involved in cellular communication and signaling. Exosomes are produced by epithelial cells and are found in biologic fluids including blood and urine. The packaged material within exosomes includes proteins and lipids, but the molecular comparison within exosome subtypes is largely unknown. The purpose of this study was to investigate differences between exosomes derived from the apical plasma membrane and basolateral plasma membrane of polarized murine cortical collecting duct principal cells. Nanoparticle tracking analysis showed that the size and concentration of apical and basolateral exosomes remained relatively stable across 3 different temperatures (23, 37, and 42°C). Liquid chromatography-tandem mass spectrometry analysis revealed marked differences between the proteins packaged within the two types of exosomes from the same cells. Several proteins expressed at the inner leaflet of the plasma membrane, including α-actinin-1, moesin, 14-3-3 protein ζ/δ, annexin A1/A3/A4/A5/A6, clathrin heavy chain 1, glyceraldehyde-3-phosphate dehydrogenase, α-enolase, filamin-A, and heat shock protein 90, were identified in samples of apical plasma membrane-derived exosomes, but not in basolateral plasma membrane exosomes from mouse cortical collecting duct cells. In addition to differences at the protein level, mass spectrometry-based shotgun lipidomics analysis showed significant differences in the lipid classes and fatty acid composition of the two types of exosomes. We found higher levels of sphingomyelin and lower levels of cardiolipin, among other phospholipids in the apical plasma membrane compared to the basolateral plasma membrane exosomes. The molecular analyses of exosome subtypes presented herein will contribute to our understanding of exosome biogenesis, and the results may have potential implications for biomarker discovery.-Dang, V. D., Jella, K. K., Ragheb, R. R. T., Denslow, N. D., Alli, A. A. Lipidomic and proteomic analysis of exosomes from mouse cortical collecting duct cells.
Collapse
Affiliation(s)
- Viet D Dang
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA.,Center for Environmental and Human Toxicology, University of Florida, Gainesville, Florida, USA.,Department of Veterinary Diagnostic and Production Animal Production, Iowa State University, Ames, Iowa, USA
| | - Kishore Kumar Jella
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Nancy D Denslow
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, USA.,Center for Environmental and Human Toxicology, University of Florida, Gainesville, Florida, USA
| | - Abdel A Alli
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA; .,Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
12
|
Epilepsy-associated gene Nedd4-2 mediates neuronal activity and seizure susceptibility through AMPA receptors. PLoS Genet 2017; 13:e1006634. [PMID: 28212375 PMCID: PMC5338825 DOI: 10.1371/journal.pgen.1006634] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 03/06/2017] [Accepted: 02/14/2017] [Indexed: 12/11/2022] Open
Abstract
The neural precursor cell expressed developmentally down-regulated gene 4–2, Nedd4-2, is an epilepsy-associated gene with at least three missense mutations identified in epileptic patients. Nedd4-2 encodes a ubiquitin E3 ligase that has high affinity toward binding and ubiquitinating membrane proteins. It is currently unknown how Nedd4-2 mediates neuronal circuit activity and how its dysfunction leads to seizures or epilepsies. In this study, we provide evidence to show that Nedd4-2 mediates neuronal activity and seizure susceptibility through ubiquitination of GluA1 subunit of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor, (AMPAR). Using a mouse model, termed Nedd4-2andi, in which one of the major forms of Nedd4-2 in the brain is selectively deficient, we found that the spontaneous neuronal activity in Nedd4-2andi cortical neuron cultures, measured by a multiunit extracellular electrophysiology system, was basally elevated, less responsive to AMPAR activation, and much more sensitive to AMPAR blockade when compared with wild-type cultures. When performing kainic acid-induced seizures in vivo, we showed that elevated seizure susceptibility in Nedd4-2andi mice was normalized when GluA1 is genetically reduced. Furthermore, when studying epilepsy-associated missense mutations of Nedd4-2, we found that all three mutations disrupt the ubiquitination of GluA1 and fail to reduce surface GluA1 and spontaneous neuronal activity when compared with wild-type Nedd4-2. Collectively, our data suggest that impaired GluA1 ubiquitination contributes to Nedd4-2-dependent neuronal hyperactivity and seizures. Our findings provide critical information to the future development of therapeutic strategies for patients who carry mutations of Nedd4-2. Many patients with neurological disorders suffer from an imbalance in neuronal and circuit excitability and present with seizure or epilepsy as the common comorbidity. Human genetic studies have identified many epilepsy-associated genes, but the pathways by which those genes are connected to brain circuit excitability are largely unknown. Our study focused on one of the epilepsy-associated genes, Nedd4-2, and aimed to dissect the molecular mechanism underlying Nedd4-2-associated epilepsy. Nedd4-2 encodes a ubiquitin E3 ligase. Several neuronal ion channels have been identified as its substrates, including the GluA1 subunit of AMPAR. Our results first demonstrate up-regulation of spontaneous neuronal activity and seizure susceptibility when Nedd4-2 is reduced in a mouse model. These deficits can be corrected when GluA1/AMPAR is pharmacologically or genetically inhibited. In addition, we found that three epilepsy-associated missense mutations of Nedd4-2 inhibit the ubiquitination of GluA1 and fail to reduce GluA1 surface expression or spontaneous neuronal activity when compared to wild-type Nedd4-2. These findings suggest the reduction of GluA1 ubiquitination as a crucial deficit underlying insufficient function of Nedd4-2 and provide critical information to the development of therapies for patients who carry mutations of Nedd4-2.
Collapse
|
13
|
You H, Ge Y, Zhang J, Cao Y, Xing J, Su D, Huang Y, Li M, Qu S, Sun F, Liang X. Derlin-1 promotes ubiquitylation and degradation of the epithelial Na + channel, ENaC. J Cell Sci 2017; 130:1027-1036. [PMID: 28137758 DOI: 10.1242/jcs.198242] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/24/2017] [Indexed: 11/20/2022] Open
Abstract
Ubiquitylation of the epithelial Na+ channel (ENaC) plays a critical role in cellular functions, including transmembrane transport of Na+, Na+ and water balance, and blood pressure stabilization. Published studies have suggested that ENaC subunits are targets of ER-related degradation (ERAD) in yeast systems. However, the molecular mechanism underlying proteasome-mediated degradation of ENaC subunits remains to be established. Derlin-1, an E3 ligase mediator, links recognized target proteins to ubiquitin-mediated proteasomal degradation in the cytosol. In the present study, we found that derlin-1 suppressed the expression of ENaC at the protein level and that the subunit α-ENaC (also known as SCNN1A) physically interacted with derlin-1 at the membrane-anchored domains or the loop regions, and that derlin-1 initiated α-ENaC retrotranslocation. In addition, HUWE1, an endoplasmic reticulum (ER)-resident E3 ubiquitin ligase, was recruited and promoted K11-linked polyubiquitylation of α-ENaC and, hence, formation of an α-ENaC ubiquitin-mediated degradation complex. These findings suggest that derlin-1 promotes ENaC ubiquitylation and enhances ENaC ubiquitin- mediated proteasome degradation. The derlin-1 pathway therefore may represent a significant early checkpoint in the recognition and degradation of ENaC in mammalian cells.
Collapse
Affiliation(s)
- Hui You
- Renal Division, Sir Run Run Hospital, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, 211166, China.,Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tong-Ji University, Shanghai, 200072, China
| | - Yamei Ge
- Renal Division, Sir Run Run Hospital, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, 211166, China
| | - Jian Zhang
- Renal Division, Sir Run Run Hospital, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, 211166, China
| | - Yizhi Cao
- Renal Division, Sir Run Run Hospital, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, 211166, China
| | - Jing Xing
- Renal Division, Sir Run Run Hospital, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, 211166, China
| | - Dongming Su
- Center of Pathology and Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, 211166, China
| | - Yujie Huang
- Renal Division, Sir Run Run Hospital, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, 211166, China
| | - Min Li
- Renal Division, Sir Run Run Hospital, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, 211166, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tong-Ji University, Shanghai, 200072, China
| | - Fei Sun
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Xiubin Liang
- Renal Division, Sir Run Run Hospital, Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, 211166, China
| |
Collapse
|
14
|
Yan C, Xu W, Huang Y, Li M, Shen Y, You H, Liang X. HRD1-Mediated IGF-1R Ubiquitination Contributes to Renal Protection of Resveratrol in db/db Mice. Mol Endocrinol 2016; 30:600-13. [PMID: 27082896 DOI: 10.1210/me.2015-1277] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Many studies have provided evidence to demonstrate the beneficial renal effects of resveratrol (RESV) due to its antioxidant character and its capacity for activation of surtuin 1. However, the molecular mechanisms underlying the protective role of RESV against kidney injury are still incompletely understood. The present study used Lepr db/db (db/db) and Lepr db/m (db/m) mice as models to evaluate the effect of RESV on diabetic nephropathy (DN). RESV reduced proteinuria and attenuated the progress of renal fibrosis in db/db mice. Treatment with RESV markedly attenuated the diabetes-induced changes in renal superoxide dismutase copper/zinc, superoxide dismutase manganese, catalase, and malonydialdehyde as well as the renal expression of nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4), α-smooth muscle actin (α-SMA), and E-cadherin in db/db mice. The kidney expression of the IGF-1 receptor (IGF-1R) was increased in db/db mice, but the expression of 3-hydroxy-3-methylglutaryl reductase degradation (HRD1), a ubiquitin E3 ligase, was significantly decreased in the DN model. RESV treatment dramatically decreased IGF-1R and increased HRD1 expressions, consistent with data obtained with HKC-8 cells. HRD1 physically interacted with IGF-1R in HKC-8 cells and liquid chromatography and tandem mass spectrometry (LC-MS/MS) data supported the concept that IGF-1R is one of the HRD1 substrates. HRD1 promoted the IGF-1R ubiquitination for degradation in HKC-8 cells, and the down-regulation of HRD1 reversed the protective effects of RESV in HKC-8 cells. In summary, we have demonstrated that RESV reduces proteinuria and attenuates the progression of renal fibrosis in db/db mice. These protective effects of RESV on DN were associated with the up-regulation of HRD1, induced by RESV, and the promotion of IGF-1R ubiquitination and degradation.
Collapse
Affiliation(s)
- Caifeng Yan
- Department of Endocrinology (C.Y.), Clinical Medical College of Yangzhou University, Yangzhou 225001, Jiangsu, China; Jiangsu Key Laboratory of Xenotransplantation (C.Y., W.X., Y.H., M.L., Y.S., H.Y., X.L.), Department of Pathophysiology, Nanjing Medical University, Nanjing 210029, Jiangsu, China. and Renal Division (W.X.), Zhuji People Hospital, Zhuji 311800, Zhejiang, China
| | - Weifeng Xu
- Department of Endocrinology (C.Y.), Clinical Medical College of Yangzhou University, Yangzhou 225001, Jiangsu, China; Jiangsu Key Laboratory of Xenotransplantation (C.Y., W.X., Y.H., M.L., Y.S., H.Y., X.L.), Department of Pathophysiology, Nanjing Medical University, Nanjing 210029, Jiangsu, China. and Renal Division (W.X.), Zhuji People Hospital, Zhuji 311800, Zhejiang, China
| | - Yujie Huang
- Department of Endocrinology (C.Y.), Clinical Medical College of Yangzhou University, Yangzhou 225001, Jiangsu, China; Jiangsu Key Laboratory of Xenotransplantation (C.Y., W.X., Y.H., M.L., Y.S., H.Y., X.L.), Department of Pathophysiology, Nanjing Medical University, Nanjing 210029, Jiangsu, China. and Renal Division (W.X.), Zhuji People Hospital, Zhuji 311800, Zhejiang, China
| | - Min Li
- Department of Endocrinology (C.Y.), Clinical Medical College of Yangzhou University, Yangzhou 225001, Jiangsu, China; Jiangsu Key Laboratory of Xenotransplantation (C.Y., W.X., Y.H., M.L., Y.S., H.Y., X.L.), Department of Pathophysiology, Nanjing Medical University, Nanjing 210029, Jiangsu, China. and Renal Division (W.X.), Zhuji People Hospital, Zhuji 311800, Zhejiang, China
| | - Yachen Shen
- Department of Endocrinology (C.Y.), Clinical Medical College of Yangzhou University, Yangzhou 225001, Jiangsu, China; Jiangsu Key Laboratory of Xenotransplantation (C.Y., W.X., Y.H., M.L., Y.S., H.Y., X.L.), Department of Pathophysiology, Nanjing Medical University, Nanjing 210029, Jiangsu, China. and Renal Division (W.X.), Zhuji People Hospital, Zhuji 311800, Zhejiang, China
| | - Hui You
- Department of Endocrinology (C.Y.), Clinical Medical College of Yangzhou University, Yangzhou 225001, Jiangsu, China; Jiangsu Key Laboratory of Xenotransplantation (C.Y., W.X., Y.H., M.L., Y.S., H.Y., X.L.), Department of Pathophysiology, Nanjing Medical University, Nanjing 210029, Jiangsu, China. and Renal Division (W.X.), Zhuji People Hospital, Zhuji 311800, Zhejiang, China
| | - Xiubin Liang
- Department of Endocrinology (C.Y.), Clinical Medical College of Yangzhou University, Yangzhou 225001, Jiangsu, China; Jiangsu Key Laboratory of Xenotransplantation (C.Y., W.X., Y.H., M.L., Y.S., H.Y., X.L.), Department of Pathophysiology, Nanjing Medical University, Nanjing 210029, Jiangsu, China. and Renal Division (W.X.), Zhuji People Hospital, Zhuji 311800, Zhejiang, China
| |
Collapse
|
15
|
Al-Qusairi L, Basquin D, Roy A, Stifanelli M, Rajaram RD, Debonneville A, Nita I, Maillard M, Loffing J, Subramanya AR, Staub O. Renal tubular SGK1 deficiency causes impaired K+ excretion via loss of regulation of NEDD4-2/WNK1 and ENaC. Am J Physiol Renal Physiol 2016; 311:F330-42. [PMID: 27009335 DOI: 10.1152/ajprenal.00002.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 03/21/2016] [Indexed: 11/22/2022] Open
Abstract
The stimulation of postprandial K(+) clearance involves aldosterone-independent and -dependent mechanisms. In this context, serum- and glucocorticoid-induced kinase (SGK)1, a ubiquitously expressed kinase, is one of the primary aldosterone-induced proteins in the aldosterone-sensitive distal nephron. Germline inactivation of SGK1 suggests that this kinase is fundamental for K(+) excretion under conditions of K(+) load, but the specific role of renal SGK1 remains elusive. To avoid compensatory mechanisms that may occur during nephrogenesis, we used inducible, nephron-specific Sgk1(Pax8/LC1) mice to assess the role of renal tubular SGK1 in K(+) regulation. Under a standard diet, these animals exhibited normal K(+) handling. When challenged by a high-K(+) diet, they developed severe hyperkalemia accompanied by a defect in K(+) excretion. Molecular analysis revealed reduced neural precursor cell expressed developmentally downregulated protein (NEDD)4-2 phosphorylation and total expression. γ-Epithelial Na(+) channel (ENaC) expression and α/γENaC proteolytic processing were also decreased in mutant mice. Moreover, with no lysine kinase (WNK)1, which displayed in control mice punctuate staining in the distal convoluted tubule and diffuse distribution in the connecting tubule/cortical colleting duct, was diffused in the distal convoluted tubule and less expressed in the connecting tubule/collecting duct of Sgk(Pax8/LC1) mice. Moreover, Ste20-related proline/alanine-rich kinase phosphorylation, and Na(+)-Cl(-) cotransporter phosphorylation/apical localization were reduced in mutant mice. Consistent with the altered WNK1 expression, increased renal outer medullary K(+) channel apical localization was observed. In conclusion, our data suggest that renal tubular SGK1 is important in the regulation of K(+) excretion via the control of NEDD4-2, WNK1, and ENaC.
Collapse
Affiliation(s)
- Lama Al-Qusairi
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; National Centre of Competence in Research "Kidney.ch," Lausanne, Switzerland
| | - Denis Basquin
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; National Centre of Competence in Research "Kidney.ch," Lausanne, Switzerland
| | - Ankita Roy
- Department of Medicine, University of Pittsburgh School of Medicine and Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Matteo Stifanelli
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Renuga Devi Rajaram
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Anne Debonneville
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Izabela Nita
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Marc Maillard
- Service of Nephrology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Johannes Loffing
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; and National Centre of Competence in Research "Kidney.ch," Lausanne, Switzerland
| | - Arohan R Subramanya
- Department of Medicine, University of Pittsburgh School of Medicine and Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Olivier Staub
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; National Centre of Competence in Research "Kidney.ch," Lausanne, Switzerland
| |
Collapse
|
16
|
Xu W, Huang Y, Li L, Sun Z, Shen Y, Xing J, Li M, Su D, Liang X. Hyperuricemia induces hypertension through activation of renal epithelial sodium channel (ENaC). Metabolism 2016; 65:73-83. [PMID: 26892518 DOI: 10.1016/j.metabol.2015.10.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 10/19/2015] [Accepted: 10/21/2015] [Indexed: 11/24/2022]
Abstract
OBJECTIVES The mechanisms leading to hypertension associated with hyperuricemia are still unclear. The activity of the distal nephron epithelial sodium channel (ENaC) is an important determinant of sodium balance and blood pressure. Our aim was to investigate whether the effect of hyperuricemia on blood pressure is related to ENaC activation. METHODS A hyperuricemic model was induced in rats by 2% oxonic acid and 6 mg/dl uric acid (UA). The hyperuricemic rats were co-treated with either 10mg/kg/d benzbromarone (Ben) or 1 mg/kg/d amiloride (Ami). Blood pressure was monitored using a tail-cuff, and blood, urine, and kidney samples were taken. Western blotting and immunohistochemical staining were performed to determine the expressions of ENaC subunits and components of the ENaC Regulatory Complex (ERC) in kidney tissue or mCCD cells. RESULTS Serum uric acid (SUA) was increased 2.5-3.5 times above normal in hyperuricemic rats after 3 weeks and remained at these high levels until 6 weeks. The in vivo rise in SUA was followed by elevated blood pressure, renal tubulointerstitial injury, and increased expressions of ENaC subunits, SGK1, and GILZ1, which were prevented by Ben treatment. The decrease in urinary Na(+) excretion in hyperuricemic rats was blunted by Ami. UA induced the expression of all three ENaC subunits, SGK1, and GILZ1, and increased Na(+) transport in mCCD cells. Phosphorylation of ERK was significantly decreased in both UA-treated mCCD cells and hyperuricemic rat kidney; this effect was prevented by Ben co-treatment. CONCLUSION Our findings suggest that elevated serum uric acid could induce hypertension by activation of ENaC and regulation of ERC expression.
Collapse
Affiliation(s)
- Weifeng Xu
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China; Zhuji people Hospital, Zhuji, Zhejiang Province, China
| | - Yujie Huang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lei Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhen Sun
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yachen Shen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jing Xing
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Min Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Dongming Su
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu Province, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiubin Liang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu Province, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW NEDD4-2 is an ubiquitin-protein ligase that was originally identified as an interactor of the epithelial Na+ channel (ENaC); this interaction is defective in Liddle's syndrome, causing elevated ENaC activity and salt-sensitive hypertension. In this review we aim to highlight progress achieved in recent years demonstrating that NEDD4-2 is involved in the control of Na+ transporters that are different from ENaC, but which also play a role in salt-sensitive hypertension. RECENT FINDINGS It has been shown that NEDD4-2 interacts with ubiquitylates and negatively regulates the thiazide-sensitive NCC (Na+,Cl- -cotransporter), both in vitro and in vivo in inducible, nephron-specific Nedd4-2 knockout mice. Moreover, evidence has been provided that NEDD4-2 is also involved in the regulation of human NHE3 (Na+,H+-exchanger 3) and NKCC2 (Na+,K+,2Cl- -cotransporter 2). SUMMARY The emerging role of NEDD4-2 in the regulation of different Na+ transporters along the nephron and the identification of human polymorphisms in the NEDD4-2 gene (Nedd4L) related to salt-sensitive hypertension makes this ubiquitin-protein ligase an interesting target for the development of antihypertensive drugs.
Collapse
|
18
|
Shen Y, Xu W, You H, Su D, Xing J, Li M, Li L, Liang X. FoxO1 inhibits transcription and membrane trafficking of epithelial Na+ channel. J Cell Sci 2015; 128:3621-30. [PMID: 26272921 DOI: 10.1242/jcs.171876] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/02/2015] [Indexed: 01/06/2023] Open
Abstract
The epithelial Na(+) channel (ENaC), regulated by insulin, is of fundamental importance in the control of Na(+) reabsorption in the distal nephron. The potential role of Forkhead box O1 (FoxO1), downstream of insulin signaling, in the regulation of ENaC remains to be investigated. Here, we found that the overexpression of a constitutively active form of FoxO1 (ADA-FoxO1) suppressed the mRNA level of the ENaC α subunit (α-ENaC; also known as SCCN1A) and the apical density of ENaC in mouse cortical collecting duct (mCCD) cells. Conversely, knockdown of FoxO1 increased the apical membrane levels of α-ENaC and Na(+) transport under basal conditions. Insulin elevated α-ENaC expression and induced FoxO1 phosphorylation; however, the increase in α-ENaC and phosphorylated FoxO1 expression observed with insulin treatment was blunted ∼ 60% in cells expressing ADA-FoxO1. Moreover, insulin induced the interaction between phosphorylated FoxO1 and 14-3-3ε, indicating that FoxO1 phosphorylation promotes ENaC membrane trafficking by binding to 14-3-3ε. FoxO1 also suppressed activity of the α-ENaC promoter, and the putative FoxO1 target site is located in the -500 to -200 nt region of the α-ENaC promoter. These findings indicate that FoxO1 is a key negative regulatory factor in the insulin-dependent control of ENaC expression and forward trafficking in mCCD epithelia.
Collapse
Affiliation(s)
- Yachen Shen
- Center of Metabolic Disease Research, Nanjing Medical University, 210029 Nanjing, Jiangsu Province, China
| | - Weifeng Xu
- Center of Metabolic Disease Research, Nanjing Medical University, 210029 Nanjing, Jiangsu Province, China
| | - Hui You
- Center of Metabolic Disease Research, Nanjing Medical University, 210029 Nanjing, Jiangsu Province, China
| | - Dongming Su
- Department of Pathology, Nanjing Medical University, 210029 Nanjing, Jiangsu Province, China State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 210029 Nanjing, Jiangsu Province, China
| | - Jing Xing
- Center of Metabolic Disease Research, Nanjing Medical University, 210029 Nanjing, Jiangsu Province, China
| | - Min Li
- Center of Metabolic Disease Research, Nanjing Medical University, 210029 Nanjing, Jiangsu Province, China
| | - Lei Li
- Center of Metabolic Disease Research, Nanjing Medical University, 210029 Nanjing, Jiangsu Province, China
| | - Xiubin Liang
- Center of Metabolic Disease Research, Nanjing Medical University, 210029 Nanjing, Jiangsu Province, China State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 210029 Nanjing, Jiangsu Province, China
| |
Collapse
|
19
|
Feng X, Li Z, Du Y, Fu H, Klein JD, Cai H, Sands JM, Chen G. Downregulation of urea transporter UT-A1 activity by 14-3-3 protein. Am J Physiol Renal Physiol 2015; 309:F71-8. [PMID: 25995111 PMCID: PMC4490382 DOI: 10.1152/ajprenal.00546.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 05/13/2015] [Indexed: 11/22/2022] Open
Abstract
Urea transporter (UT)-A1 in the kidney inner medulla plays a critical role in the urinary concentrating mechanism and thereby in the regulation of water balance. The 14-3-3 proteins are a family of seven isoforms. They are multifunctional regulatory proteins that mainly bind to phosphorylated serine/threonine residues in target proteins. In the present study, we found that all seven 14-3-3 isoforms were detected in the kidney inner medulla. However, only the 14-3-3 γ-isoform was specifically and highly associated with UT-A1, as demonstrated by a glutathione-S-transferase-14-3-3 pulldown assay. The cAMP/adenylyl cyclase stimulator forskolin significantly enhanced their binding. Coinjection of 14-3-3γ cRNA into oocytes resulted in a decrease of UT-A1 function. In addition, 14-3-3γ increased UT-A1 ubiquitination and protein degradation. 14-3-3γ can interact with both UT-A1 and mouse double minute 2, the E3 ubiquitin ligase for UT-A1. Thus, activation of cAMP/PKA increases 14-3-3γ interactions with UT-A1 and stimulates mouse double minute 2-mediated UT-A1 ubiquitination and degradation, thereby forming a novel regulatory mechanism of urea transport activity.
Collapse
Affiliation(s)
- Xiuyan Feng
- Renal Division, Department of Medicine, Emory University, School of Medicine, Atlanta, Georgia; Section of Nephrology, Atlanta Veterans Administration Medical Center, Decatur, Georgia
| | - Zenggang Li
- Department of Pharmacology, Emory University, School of Medicine, Atlanta, Georgia
| | - Yuhong Du
- Department of Pharmacology, Emory University, School of Medicine, Atlanta, Georgia
| | - Haian Fu
- Department of Pharmacology, Emory University, School of Medicine, Atlanta, Georgia
| | - Janet D Klein
- Renal Division, Department of Medicine, Emory University, School of Medicine, Atlanta, Georgia; Department of Physiology, Emory University, School of Medicine, Atlanta, Georgia; and
| | - Hui Cai
- Renal Division, Department of Medicine, Emory University, School of Medicine, Atlanta, Georgia; Department of Physiology, Emory University, School of Medicine, Atlanta, Georgia; and Section of Nephrology, Atlanta Veterans Administration Medical Center, Decatur, Georgia
| | - Jeff M Sands
- Renal Division, Department of Medicine, Emory University, School of Medicine, Atlanta, Georgia; Department of Physiology, Emory University, School of Medicine, Atlanta, Georgia; and
| | - Guangping Chen
- Renal Division, Department of Medicine, Emory University, School of Medicine, Atlanta, Georgia; Department of Physiology, Emory University, School of Medicine, Atlanta, Georgia; and
| |
Collapse
|
20
|
Li Y, Shen Y, Li M, Su D, Xu W, Liang X, Li R. Inhibitory effects of peroxisome proliferator-activated receptor γ agonists on collagen IV production in podocytes. Mol Cell Biochem 2015; 405:233-41. [PMID: 25920446 DOI: 10.1007/s11010-015-2414-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/18/2015] [Indexed: 12/14/2022]
Abstract
Peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists have beneficial effects on the kidney diseases through preventing microalbuminuria and glomerulosclerosis. However, the mechanisms underlying these effects remain to be fully understood. In this study, we investigate the effects of PPAR-γ agonist, rosiglitazone (Rosi) and pioglitazone (Pio), on collagen IV production in mouse podocytes. The endogenous expression of PPAR-γ was found in the primary podocytes and can be upregulated by Rosi and Pio, respectively, detected by RT-PCR and Western blot. PPAR-γ agonist markedly blunted the increasing of collagen IV expression and extraction in podocytes induced by TGF-β. In contrast, adding PPAR-γ antagonist, GW9662, to podocytes largely prevented the inhibition of collagen IV expression from Pio treatment. Our data also showed that phosphorylation of Smad2/3 enhanced by TGF-β in a time-dependent manner was significantly attenuated by adding Pio. The promoter region of collagen IV gene contains one putative consensus sequence of Smad-binding element (SBE) by promoter analysis, Rosi and Pio significantly ameliorated TGF-β-induced SBE4-luciferase activity. In conclusion, PPAR-γ activation by its agonist, Rosi or Pio, in vitro directly inhibits collagen IV expression and synthesis in primary mouse podocytes. The suppression of collagen IV production was related to the inhibition of TGF-β-driven phosphorylation of Smad2/3 and decreased response activity of SBEs of collagen IV in PPAR-γ agonist-treated mouse podocytes. This represents a novel mechanistic support regarding PPAR-γ agonists as podocyte protective agents.
Collapse
Affiliation(s)
- Yanjiao Li
- Department of Nephrology, Shanxi Provincial People's Hospital, The Affiliated People's Hospital of Shanxi Medical University, Taiyuan, 030012, Shanxi, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
The amiloride-sensitive epithelial Na(+) channel (ENaC) is a key player in the regulation of Na(+) homeostasis. Its functional activity is under continuous control by a variety of signaling molecules, including bioactive peptides of endothelin family. Since ENaC dysfunction is causative for disturbances in total body Na(+) levels associated with the abnormal regulation of blood volume, blood pressure, and lung fluid balance, uncovering the molecular mechanisms of inhibitory modulation or inappropriate activation of ENaC is crucial for the successful treatment of a variety of human diseases including hypertension. The precise regulation of ENaC is particularly important for normal Na(+) and fluid homeostasis in organs where endothelins are known to act: the kidneys, lung, and colon. Inhibition of ENaC by endothelin-1 (ET-1) has been established in renal cells, and several molecular mechanisms of inhibition of ENaC by ET-1 are proposed and will be reviewed in this chapter.
Collapse
Affiliation(s)
- Andrey Sorokin
- Division of Nephrology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| | | |
Collapse
|
22
|
Li L, Shen Y, Ding Y, Liu Y, Su D, Liang X. Hrd1 participates in the regulation of collagen I synthesis in renal fibrosis. Mol Cell Biochem 2013; 386:35-44. [DOI: 10.1007/s11010-013-1843-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 09/26/2013] [Indexed: 10/26/2022]
|
23
|
Pouly D, Debonneville A, Ruffieux-Daidié D, Maillard M, Abriel H, Loffing J, Staub O. Mice carrying ubiquitin-specific protease 2 (Usp2) gene inactivation maintain normal sodium balance and blood pressure. Am J Physiol Renal Physiol 2013; 305:F21-30. [PMID: 23552861 DOI: 10.1152/ajprenal.00012.2013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Ubiquitylation plays an important role in the control of Na⁺ homeostasis by the kidney. It is well established that the epithelial Na⁺ channel ENaC is regulated by the ubiquitin-protein ligase NEDD4-2, limiting ENaC cell surface expression and activity. Ubiquitylation can be reversed by the action of deubiquitylating enzymes (DUBs). One such DUB, USP2-45, was identified previously as an aldosterone-induced protein in the kidney and is also a circadian output gene. In heterologous expression systems, USP2-45 binds to ENaC, deubiquitylates it, and enhances channel density and activity at the cell surface. Because the role of USP2-45 in renal Na⁺ transport had not been studied in vivo, we investigated here the effect of Usp2 gene inactivation in this process. We demonstrate first that USP2-45 protein has a rhythmic expression with a peak at ZT12. Usp2-KO mice did not show any differences from wild-type littermates with respect to the diurnal control of Na⁺ or K⁺ urinary excretion and plasma levels either on a standard diet or after acute and chronic changes to low- and high-Na⁺ diets, respectively. Moreover, they had similar aldosterone levels on either a low- or high-Na⁺ diet. Blood pressure measurements using telemetry did not reveal variations compared with control mice. Usp2-KO mice did not display alterations in expression of genes involved in sodium homeostasis or the ubiquitin system, as evidenced by transcriptome analysis in the kidney. Our data suggest that USP2 does not play a primary role in the control of Na⁺ balance or blood pressure.
Collapse
Affiliation(s)
- Daniel Pouly
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
24
|
Ito M, Urano T, Hiroi H, Momoeda M, Saito M, Hosokawa Y, Tsutsumi R, Zenri F, Koizumi M, Nakae H, Horie-Inoue K, Fujii T, Yano T, Kozuma S, Inoue S, Taketani Y. The progesterone-responsive gene 14-3-3τ enhances the transcriptional activity of progesterone receptor in uterine cells. J Mol Endocrinol 2012; 49:193-202. [PMID: 22967481 DOI: 10.1530/jme-12-0112] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Members of the 14-3-3 family are intracellular dimeric phosphoserine-binding proteins that can associate with and modulate the activities of many proteins. In our efforts to isolate the genes regulated by progesterone (P(4)) using suppressive subtractive hybridization, we previously found that 14-3-3τ is one of the genes upregulated by P(4). In this study, we demonstrated by quantitative RT-PCR (qRT-PCR), western blot analyses, and immunohistochemistry that 14-3-3τ mRNA and protein levels were increased in the rat uterus after P(4) treatment. Furthermore, qRT-PCR indicated that P(4) increased 14-3-3τ mRNA levels in human endometrial epithelial cells and endometrial stromal cells (ESCs). Western blot and qRT-PCR analyses revealed that in vitro decidualization using cAMP and medroxyprogesterone 17-acetate increased levels of 14-3-3τ mRNA and protein in ESCs. We have shown by qRT-PCR and western blot analyses that P(4) increased the mRNA and protein levels of 14-3-3τ in Ishikawa cells that stably express P(4) receptor-B (PR-B). Immunocytochemistry revealed that 14-3-3τ colocalizes with PR and translocates from the cytoplasm to the nucleus in response to P(4). Moreover, by luciferase reporter assay, we demonstrated that 14-3-3τ enhances the transcriptional activity of PR-B. Taken together, we propose that 14-3-3τ is a P(4)-responsive gene in uterine cells that modulates P(4) signaling.
Collapse
Affiliation(s)
- Masanori Ito
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Edinger RS, Bertrand CA, Rondandino C, Apodaca GA, Johnson JP, Butterworth MB. The epithelial sodium channel (ENaC) establishes a trafficking vesicle pool responsible for its regulation. PLoS One 2012; 7:e46593. [PMID: 23029554 PMCID: PMC3460899 DOI: 10.1371/journal.pone.0046593] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 09/05/2012] [Indexed: 12/16/2022] Open
Abstract
The epithelial sodium channel (ENaC) is the rate-limiting step for sodium reabsorption across tight epithelia. Cyclic-AMP (cAMP) stimulation promotes ENaC trafficking to the apical surface to increase channel number and transcellular Na(+) transport. Removal of corticosteroid supplementation in a cultured cortical collecting duct cell line reduced ENaC expression. Concurrently, the number of vesicles trafficked in response to cAMP stimulation, as measured by a change in membrane capacitance, also decreased. Stimulation with aldosterone restored both the basal and cAMP-stimulated ENaC activity and increased the number of exocytosed vesicles. Knocking down ENaC directly decreased both the cAMP-stimulated short-circuit current and capacitance response in the presence of aldosterone. However, constitutive apical recycling of the Immunoglobulin A receptor was unaffected by alterations in ENaC expression or trafficking. Fischer Rat Thyroid cells, transfected with α,β,γ-mENaC had a significantly greater membrane capacitance response to cAMP stimulation compared to non-ENaC controls. Finally, immunofluorescent labeling and quantitation revealed a smaller number of vesicles in cells where ENaC expression was reduced. These findings indicate that ENaC is not a passive passenger in regulated epithelial vesicle trafficking, but plays a role in establishing and maintaining the pool of vesicles that respond to cAMP stimulation.
Collapse
Affiliation(s)
- Robert S. Edinger
- Department of Medicine, Renal-Electrolyte Division, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Carol A. Bertrand
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Christine Rondandino
- Department of Medicine, Renal-Electrolyte Division, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Gerard A. Apodaca
- Department of Medicine, Renal-Electrolyte Division, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - John P. Johnson
- Department of Medicine, Renal-Electrolyte Division, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Michael B. Butterworth
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
26
|
Lang F, Shumilina E. Regulation of ion channels by the serum- and glucocorticoid-inducible kinase SGK1. FASEB J 2012; 27:3-12. [PMID: 23012321 DOI: 10.1096/fj.12-218230] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The ubiquitously expressed serum- and glucocorticoid-inducible kinase-1 (SGK1) is genomically regulated by cell stress (including cell shrinkage) and several hormones (including gluco- and mineralocorticoids). SGK1 is activated by insulin and growth factors through PI3K and 3-phosphoinositide-dependent kinase PDK1. SGK1 activates a wide variety of ion channels (e.g., ENaC, SCN5A, TRPV4-6, ROMK, Kv1.3, Kv1.5, Kv4.3, KCNE1/KCNQ1, KCNQ4, ASIC1, GluR6, ClCKa/barttin, ClC2, CFTR, and Orai/STIM), which participate in the regulation of transport, hormone release, neuroexcitability, inflammation, cell proliferation, and apoptosis. SGK1-sensitive ion channels participate in the regulation of renal Na(+) retention and K(+) elimination, blood pressure, gastric acid secretion, cardiac action potential, hemostasis, and neuroexcitability. A common (∼3-5% prevalence in Caucasians and ∼10% in Africans) SGK1 gene variant is associated with increased blood pressure and body weight as well as increased prevalence of type II diabetes and stroke. SGK1 further contributes to the pathophysiology of allergy, peptic ulcer, fibrosing disease, ischemia, tumor growth, and neurodegeneration. The effect of SGK1 on channel activity is modest, and the channels do not require SGK1 for basic function. SGK1-dependent ion channel regulation may thus become pathophysiologically relevant primarily after excessive (pathological) expression. Therefore, SGK1 may be considered an attractive therapeutic target despite its broad range of functions.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology, University of Tuebingen, Gmelinstrasse 5, 72076 Tuebingen, Germany.
| | | |
Collapse
|
27
|
Gabriel L, Lvov A, Orthodoxou D, Rittenhouse AR, Kobertz WR, Melikian HE. The acid-sensitive, anesthetic-activated potassium leak channel, KCNK3, is regulated by 14-3-3β-dependent, protein kinase C (PKC)-mediated endocytic trafficking. J Biol Chem 2012; 287:32354-66. [PMID: 22846993 DOI: 10.1074/jbc.m112.391458] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The acid-sensitive neuronal potassium leak channel, KCNK3, is vital for setting the resting membrane potential and is the primary target for volatile anesthetics. Recent reports demonstrate that KCNK3 activity is down-regulated by PKC; however, the mechanisms responsible for PKC-induced KCNK3 down-regulation are undefined. Here, we report that endocytic trafficking dynamically regulates KCNK3 activity. Phorbol esters and Group I metabotropic glutamate receptor (mGluR) activation acutely decreased both native and recombinant KCNK3 currents with concomitant KCNK3 surface losses in cerebellar granule neurons and cell lines. PKC-mediated KCNK3 internalization required the presence of both 14-3-3β and a novel potassium channel endocytic motif, because depleting either 14-3-3β protein levels or ablating the endocytic motif completely abrogated PKC-regulated KCNK3 trafficking. These results demonstrate that neuronal potassium leak channels are not static membrane residents but are subject to 14-3-3β-dependent regulated trafficking, providing a straightforward mechanism to modulate neuronal excitability and synaptic plasticity by Group I mGluRs.
Collapse
Affiliation(s)
- Luke Gabriel
- Graduate Program in Neuroscience, University of Massachusetts Medical School, Worcester, Massachusetts 01604, USA
| | | | | | | | | | | |
Collapse
|
28
|
Rotin D, Staub O. Nedd4-2 and the regulation of epithelial sodium transport. Front Physiol 2012; 3:212. [PMID: 22737130 PMCID: PMC3380336 DOI: 10.3389/fphys.2012.00212] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 05/30/2012] [Indexed: 12/31/2022] Open
Abstract
Nedd4-2 is a ubiquitin ligase previously demonstrated to regulate endocytosis and lysosomal degradation of the epithelial Na(+) channel (ENaC) and other ion channels and transporters. Recent studies using Nedd4-2 knockout mice specifically in kidney or lung epithelia has revealed a critical role for this E3 ubiquitin ligase in regulating salt and fluid transport in these tissues/organs and in maintaining homeostasis of body blood pressure. Interestingly, the primary targets for Nedd4-2 may differ in these two organs: in the lung Nedd4-2 targets ENaC, and loss of Nedd4-2 leads to excessive ENaC function and to cystic fibrosis - like lung disease, whereas in the kidney, Nedd4-2 targets the Na(+)/Cl(-) cotransporter (NCC) in addition to targeting ENaC. In accord, loss of Nedd4-2 in the distal nephron leads to increased NCC abundance and function. The aldosterone-responsive kinase, Sgk1, appears to be involved in the regulation of NCC by Nedd4-2 in the kidney, similar to its regulation of ENaC. Collectively, these new findings underscore the physiological importance of Nedd4-2 in regulating epithelial salt and fluid transport and balance.
Collapse
Affiliation(s)
- Daniela Rotin
- Program in Cell Biology, The Hospital for Sick Children, Biochemistry Department, University of Toronto Toronto, ON, Canada
| | | |
Collapse
|
29
|
Abstract
Small GTPases function as molecular switches in cell signaling, alternating between an inactive, GDP-bound state, and active GTP-bound state. βPix is one of guanine nucleotide exchange factors (GEFs) that catalyze the exchange of bound GDP for ambient GTP. The central goal of this review article is to summarize recent findings on βPix and the role it plays in kidney pathology and physiology. Recent studies shed new light on several key questions concerning the signaling mechanisms mediated by βPix. This manuscript provides a review of the various mechanisms whereby βPix has been shown to function within the kidney through a wide range of actions. Both canonical GEF activity and non-canonical signaling pathways mediated by βPix are discussed. Distribution patterns of βPix in the kidney will be also covered. Much has yet to be discerned, but it is clear that βPix plays a significant role in the kidney.
Collapse
|
30
|
Faresse N, Lagnaz D, Debonneville A, Ismailji A, Maillard M, Fejes-Toth G, Náray-Fejes-Tóth A, Staub O. Inducible kidney-specific Sgk1 knockout mice show a salt-losing phenotype. Am J Physiol Renal Physiol 2012; 302:F977-85. [PMID: 22301619 DOI: 10.1152/ajprenal.00535.2011] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The expression of the serum- and glucocorticoid-regulated kinase 1 (Sgk1) is induced by mineralocorticoids and, in turn, upregulates the renal epithelial Na(+) channel (ENaC). Total inactivation of Sgk1 has been associated with transient urinary Na(+) wasting with a low-Na(+) diet, while the aldosterone-mediated ENaC channel activation was unchanged in the collecting duct. Since Sgk1 is ubiquitously expressed, we aimed to study the role of renal Sgk1 and generated an inducible kidney-specific knockout (KO) mouse. We took advantage of the previously described TetOn/CreLoxP system, in which rtTA is under the control of the Pax8 promotor, allowing inducible inactivation of the floxed Sgk1 allele in the renal tubules (Sgk1fl/fl/Pax8/LC1 mice). We found that under a standard Na(+) diet, renal water and Na(+)/K(+) excretion had a tendency to be higher in doxycycline-treated Sgk1 KO mice compared with control mice. The impaired ability of Sgk1 KO mice to retain Na(+) increased significantly with a low-salt diet despite higher plasma aldosterone levels. On a low-Na(+) diet, the Sgk1 KO mice were also hyperkaliuric and lost body weight. This phenotype was accompanied by a decrease in systolic and diastolic blood pressure. At the protein level, we observed a reduction in phosphorylation of the ubiquitin protein-ligase Nedd4-2 and a decrease in the expression of the Na(+)-Cl(-)-cotransporter (NCC) and to a lesser extent of ENaC.
Collapse
Affiliation(s)
- Nourdine Faresse
- Department of Pharmacology and Toxicology, University of Lausanne, Rue du Bugnon 27, Lausanne, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Liang X, Da Paula AC, Bozóky Z, Zhang H, Bertrand CA, Peters KW, Forman-Kay JD, Frizzell RA. Phosphorylation-dependent 14-3-3 protein interactions regulate CFTR biogenesis. Mol Biol Cell 2012; 23:996-1009. [PMID: 22278744 PMCID: PMC3302758 DOI: 10.1091/mbc.e11-08-0662] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
cAMP/PKA stimulation elicited posttranslational increases in CFTR expression and the interaction of specific 14-3-3 proteins with phosphorylated sites within the R region. This improved the efficiency of nascent CFTR biogenesis and reduced its interaction with the COPI retrograde retrieval mechanism, making more CFTR available for anion secretion. Cystic fibrosis transmembrane conductance regulator (CFTR) is a cAMP/protein kinase A (PKA)–regulated chloride channel whose phosphorylation controls anion secretion across epithelial cell apical membranes. We examined the hypothesis that cAMP/PKA stimulation regulates CFTR biogenesis posttranslationally, based on predicted 14-3-3 binding motifs within CFTR and forskolin-induced CFTR expression. The 14-3-3β, γ, and ε isoforms were expressed in airway cells and interacted with CFTR in coimmunoprecipitation assays. Forskolin stimulation (15 min) increased 14-3-3β and ε binding to immature and mature CFTR (bands B and C), and 14-3-3 overexpression increased CFTR bands B and C and cell surface band C. In pulse-chase experiments, 14-3-3β increased the synthesis of immature CFTR, reduced its degradation rate, and increased conversion of immature to mature CFTR. Conversely, 14-3-3β knockdown decreased CFTR B and C bands (70 and 55%) and elicited parallel reductions in cell surface CFTR and forskolin-stimulated anion efflux. In vitro, 14-3-3β interacted with the CFTR regulatory region, and by nuclear magnetic resonance analysis, this interaction occurred at known PKA phosphorylated sites. In coimmunoprecipitation assays, forskolin stimulated the CFTR/14-3-3β interaction while reducing CFTR's interaction with coat protein complex 1 (COP1). Thus 14-3-3 binding to phosphorylated CFTR augments its biogenesis by reducing retrograde retrieval of CFTR to the endoplasmic reticulum. This mechanism permits cAMP/PKA stimulation to make more CFTR available for anion secretion.
Collapse
Affiliation(s)
- Xiubin Liang
- Department of Cell Biology and Physiology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Grant MP, Stepanchick A, Cavanaugh A, Breitwieser GE. Agonist-Driven Maturation and Plasma Membrane Insertion of Calcium-Sensing Receptors Dynamically Control Signal Amplitude. Sci Signal 2011; 4:ra78. [DOI: 10.1126/scisignal.2002208] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
33
|
Serum- and glucocorticoid-inducible kinase 1 in the regulation of renal and extrarenal potassium transport. Clin Exp Nephrol 2011; 16:73-80. [DOI: 10.1007/s10157-011-0488-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 04/08/2010] [Indexed: 01/24/2023]
|
34
|
Thomas SV, Kathpalia PP, Rajagopal M, Charlton C, Zhang J, Eaton DC, Helms MN, Pao AC. Epithelial sodium channel regulation by cell surface-associated serum- and glucocorticoid-regulated kinase 1. J Biol Chem 2011; 286:32074-85. [PMID: 21784856 PMCID: PMC3173222 DOI: 10.1074/jbc.m111.278283] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 07/20/2011] [Indexed: 11/06/2022] Open
Abstract
Serum- and glucocorticoid-regulated kinase 1 (sgk1) participates in diverse biological processes, including cell growth, apoptosis, and sodium homeostasis. In the cortical collecting duct of the kidney, sgk1 regulates sodium transport by stimulating the epithelial sodium channel (ENaC). Control of subcellular localization of sgk1 may be an important mechanism for modulating specificity of sgk1 function; however, which subcellular locations are required for sgk1-regulated ENaC activity in collecting duct cells has yet to be established. Using cell surface biotinylation studies, we detected endogenous sgk1 at the apical cell membrane of aldosterone-stimulated mpkCCD(c14) collecting duct cells. The association of sgk1 with the cell membrane was enhanced when ENaC was co-transfected with sgk1 in kidney cells, suggesting that ENaC brings sgk1 to the cell surface. Furthermore, association of endogenous sgk1 with the apical cell membrane of mpkCCD(c14) cells could be modulated by treatments that increase or decrease ENaC expression at the apical membrane; forskolin increased the association of sgk1 with the apical surface, whereas methyl-β-cyclodextrin decreased the association of sgk1 with the apical surface. Single channel recordings of excised inside-out patches from the apical membrane of aldosterone-stimulated A6 collecting duct cells revealed that the open probability of ENaC was sensitive to the sgk1 inhibitor GSK650394, indicating that endogenous sgk1 is functionally active at the apical cell membrane. We propose that the association of sgk1 with the apical cell membrane, where it interacts with ENaC, is a novel means by which sgk1 specifically enhances ENaC activity in aldosterone-stimulated collecting duct cells.
Collapse
Affiliation(s)
- Sheela V. Thomas
- From the Department of Medicine, Division of Nephrology, Stanford University, Stanford, California 94305
| | - Paru P. Kathpalia
- From the Department of Medicine, Division of Nephrology, Stanford University, Stanford, California 94305
| | - Madhumitha Rajagopal
- From the Department of Medicine, Division of Nephrology, Stanford University, Stanford, California 94305
| | - Carol Charlton
- From the Department of Medicine, Division of Nephrology, Stanford University, Stanford, California 94305
| | - Jianning Zhang
- the Department of Medicine, Division of Nephrology, University of Texas, Southwestern Medical Center, Dallas, Texas 75390, and
| | - Douglas C. Eaton
- the Department of Physiology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - My N. Helms
- the Department of Physiology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Alan C. Pao
- From the Department of Medicine, Division of Nephrology, Stanford University, Stanford, California 94305
| |
Collapse
|
35
|
Chandran S, Li H, Dong W, Krasinska K, Adams C, Alexandrova L, Chien A, Hallows KR, Bhalla V. Neural precursor cell-expressed developmentally down-regulated protein 4-2 (Nedd4-2) regulation by 14-3-3 protein binding at canonical serum and glucocorticoid kinase 1 (SGK1) phosphorylation sites. J Biol Chem 2011; 286:37830-40. [PMID: 21900244 DOI: 10.1074/jbc.m111.293233] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Regulation of epithelial Na(+) channel (ENaC)-mediated transport in the distal nephron is a critical determinant of blood pressure in humans. Aldosterone via serum and glucocorticoid kinase 1 (SGK1) stimulates ENaC by phosphorylation of the E3 ubiquitin ligase Nedd4-2, which induces interaction with 14-3-3 proteins. However, the mechanisms of SGK1- and 14-3-3-mediated regulation of Nedd4-2 are unclear. There are three canonical SGK1 target sites on Nedd4-2 that overlap phosphorylation-dependent 14-3-3 interaction motifs. Two of these are termed "minor," and one is termed "major," based on weak or strong binding to 14-3-3 proteins, respectively. By mass spectrometry, we found that aldosterone significantly stimulates phosphorylation of a minor, relative to the major, 14-3-3 binding site on Nedd4-2. Phosphorylation-deficient minor site Nedd4-2 mutants bound less 14-3-3 than did wild-type (WT) Nedd4-2, and minor site Nedd4-2 mutations were sufficient to inhibit SGK1 stimulation of ENaC cell surface expression. As measured by pulse-chase and cycloheximide chase assays, a major binding site Nedd4-2 mutant had a shorter cellular half-life than WT Nedd4-2, but this property was not dependent on binding to 14-3-3. Additionally, a dimerization-deficient 14-3-3ε mutant failed to bind Nedd4-2. We conclude that whereas phosphorylation at the Nedd4-2 major site is important for interaction with 14-3-3 dimers, minor site phosphorylation by SGK1 may be the relevant molecular switch that stabilizes Nedd4-2 interaction with 14-3-3 and thus promotes ENaC cell surface expression. We also propose that major site phosphorylation promotes cellular Nedd4-2 protein stability, which potentially represents a novel form of regulation for turnover of E3 ubiquitin ligases.
Collapse
Affiliation(s)
- Sindhu Chandran
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Yang G, Hamacher J, Gorshkov B, White R, Sridhar S, Verin A, Chakraborty T, Lucas R. The Dual Role of TNF in Pulmonary Edema. J Cardiovasc Dis Res 2011; 1:29-36. [PMID: 21188088 PMCID: PMC3004168 DOI: 10.4103/0975-3583.59983] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
—Pulmonary edema, a major manifestation of left ventricular heart failure, renal insufficiency, shock, diffuse alveolar damage and lung hypersensitivity states, is a significant medical problem worldwide and can be life-threatening. The proinflammatory cytokine tumor necrosis factor (TNF) has been shown to contribute to the pathogenesis and development of pulmonary edema. However, some recent studies have demonstrated surprisingly that TNF can also promote alveolar fluid reabsorption in vivo and in vitro. This protective effect of the cytokine is mediated by the lectin-like domain of the cytokine, which is spatially distinct from the TNF receptor binding sites. The TIP peptide, a synthetic mimic of the lectin-like domain of TNF, can significantly increase alveolar fluid clearance and improve lung compliance in pulmonary edema models. In this review, we will discuss the dual role of TNF in pulmonary edema.
Collapse
Affiliation(s)
- Guang Yang
- Vascular Biology Center & Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, GA, 30912, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Eylenstein A, Gehring EM, Heise N, Shumilina E, Schmidt S, Szteyn K, Münzer P, Nurbaeva MK, Eichenmüller M, Tyan L, Regel I, Föller M, Kuhl D, Soboloff J, Penner R, Lang F. Stimulation of Ca2+-channel Orai1/STIM1 by serum- and glucocorticoid-inducible kinase 1 (SGK1). FASEB J 2011; 25:2012-21. [PMID: 21385992 DOI: 10.1096/fj.10-178210] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ca(2+) signaling includes store-operated Ca(2+) entry (SOCE) following depletion of endoplasmic reticulum (ER) Ca(2+) stores. On store depletion, the ER Ca(2+) sensor STIM1 activates Orai1, the pore-forming unit of Ca(2+)-release-activated Ca(2+) (CRAC) channels. Here, we show that Orai1 is regulated by serum- and glucocorticoid-inducible kinase 1 (SGK1), a growth factor-regulated kinase. Membrane Orai1 protein abundance, I(CRAC), and SOCE in human embryonic kidney (HEK293) cells stably expressing Orai1 and transfected with STIM1 were each significantly enhanced by coexpression of constitutively active (S422D)SGK1 (by+81, +378, and+136%, respectively) but not by inactive (K127N)SGK1. Coexpression of the ubiquitin ligase Nedd4-2, an established negatively regulated SGK1 target, down-regulated SOCE (by -48%) and I(CRAC) (by -60%), an effect reversed by expression of (S422D)SGK1 (by +175 and +173%, respectively). Orai1 protein abundance and SOCE were significantly lower in mast cells from SGK1-knockout (sgk1(-/-)) mice (by -37% and -52%, respectively) than in mast cells from wild-type (sgk1(+/+)) littermates. Activation of SOCE by sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase-inhibitor thapsigargin (2 μM) stimulated migration, an effect significantly higher (by +306%) in (S422D)SGK1-expressing than in (K127N)SGK1-expressing HEK293 cells, and also significantly higher (by +108%) in sgk1(+/+) than in sgk1(-/-) mast cells. SGK1 is thus a novel key player in the regulation of SOCE.
Collapse
Affiliation(s)
- Anja Eylenstein
- Department of Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Role of the ubiquitin system in regulating ion transport. Pflugers Arch 2010; 461:1-21. [PMID: 20972579 DOI: 10.1007/s00424-010-0893-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Revised: 10/04/2010] [Accepted: 10/04/2010] [Indexed: 12/22/2022]
Abstract
Ion channels and transporters play a critical role in ion and fluid homeostasis and thus in normal animal physiology and pathology. Tight regulation of these transmembrane proteins is therefore essential. In recent years, many studies have focused their attention on the role of the ubiquitin system in regulating ion channels and transporters, initialed by the discoveries of the role of this system in processing of Cystic Fibrosis Transmembrane Regulator (CFTR), and in regulating endocytosis of the epithelial Na(+) channel (ENaC) by the Nedd4 family of ubiquitin ligases (mainly Nedd4-2). In this review, we discuss the role of the ubiquitin system in ER Associated Degradation (ERAD) of ion channels, and in the regulation of endocytosis and lysosomal sorting of ion channels and transporters, focusing primarily in mammalian cells. We also briefly discuss the role of ubiquitin like molecules (such as SUMO) in such regulation, for which much less is known so far.
Collapse
|
39
|
Interaction of serum- and glucocorticoid regulated kinase 1 (SGK1) with the WW-domains of Nedd4-2 is required for epithelial sodium channel regulation. PLoS One 2010; 5:e12163. [PMID: 20730100 PMCID: PMC2921341 DOI: 10.1371/journal.pone.0012163] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 07/15/2010] [Indexed: 11/25/2022] Open
Abstract
Background The epithelial sodium channel (ENaC) is an integral component of the pathway for Na+ absorption in epithelial cells. The ubiquitin ligases Nedd4 and Nedd4-2 bind to ENaC and decrease its activity. Conversely, Serum- and Glucocorticoid regulated Kinase-1 (SGK1), a downstream mediator of aldosterone, increases ENaC activity. This effect is at least partly mediated by direct interaction between SGK and Nedd4-2. SGK binds both Nedd4 and Nedd4-2, but it is only able to phosphorylate Nedd4-2. Phosphorylation of Nedd4-2 reduces its ability to bind to ENaC, due to the interaction of phosphorylated Nedd4-2 with 14-3-3 proteins, and hence increases ENaC activity. WW-domains in Nedd4-like proteins bind PY-motifs (PPXY) present in ENaC subunits, and SGK also has a PY-motif. Principal Finding Here we show that single or tandem WW-domains of Nedd4 and Nedd4-2 mediate binding to SGK and that different WW-domains of Nedd4 and Nedd4-2 are involved. Our data also show that WW-domains 2 and 3 of Nedd4-2 mediate the interaction with SGK in a cooperative manner, that activated SGK has increased affinity for the WW-domains of Nedd4-2 in vitro, and a greater stimulatory effect on ENaC Na+ transport compared to wildtype SGK. Further, SGK lacking a PY motif failed to stimulate ENaC activity in the presence of Nedd4-2. Conclusions Binding of Nedd4-2 WW-domains to SGK is necessary for SGK-induced ENaC activity.
Collapse
|
40
|
Hallows KR, Bhalla V, Oyster NM, Wijngaarden MA, Lee JK, Li H, Chandran S, Xia X, Huang Z, Chalkley RJ, Burlingame AL, Pearce D. Phosphopeptide screen uncovers novel phosphorylation sites of Nedd4-2 that potentiate its inhibition of the epithelial Na+ channel. J Biol Chem 2010; 285:21671-8. [PMID: 20466724 PMCID: PMC2898378 DOI: 10.1074/jbc.m109.084731] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 04/11/2010] [Indexed: 11/06/2022] Open
Abstract
The E3 ubiquitin ligase Nedd4-2 regulates several ion transport proteins, including the epithelial Na(+) channel (ENaC). Nedd4-2 decreases apical membrane expression and activity of ENaC. Although it is subject to tight hormonal control, the mechanistic basis of Nedd4-2 regulation remains poorly understood. To characterize regulatory inputs to Nedd4-2 function, we screened for novel sites of Nedd4-2 phosphorylation using tandem mass spectrometry. Three of seven identified Xenopus Nedd4-2 Ser/Thr phosphorylation sites corresponded to previously identified target sites for SGK1, whereas four were novel, including Ser-293, which matched the consensus for a MAPK target sequence. Further in vitro and in vivo phosphorylation experiments revealed that Nedd4-2 serves as a target of JNK1, but not of p38 MAPK or ERK1/2. Additional rounds of tandem mass spectrometry identified two other phosphorylated residues within Nedd4-2, including Thr-899, which is present within the catalytic domain. Nedd4-2 with mutations at these sites had markedly inhibited JNK1-dependent phosphorylation, virtually no ENaC inhibitory activity, and significantly reduced ubiquitin ligase activity. These data identify phosphorylatable residues that activate Nedd4-2 and may work together with residues targeted by inhibitory kinases (e.g. SGK1 and protein kinase A) to govern Nedd4-2 regulation of epithelial ion transport.
Collapse
Affiliation(s)
- Kenneth R. Hallows
- From the Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Vivek Bhalla
- the Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California 94305
| | - Nicholas M. Oyster
- From the Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Marjolein A. Wijngaarden
- the Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Jeffrey K. Lee
- From the Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Hui Li
- From the Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Sindhu Chandran
- the Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California 94305
| | - Xiaoyu Xia
- the Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California 94305
| | - Zhirong Huang
- the Division of Nephrology, Department of Medicine, University of California San Francisco School of Medicine, San Francisco, California 94107, and
| | - Robert J. Chalkley
- the Department of Pharmaceutical Chemistry, University of California San Francisco School of Pharmacy, San Francisco, California 94158
| | - Alma L. Burlingame
- the Department of Pharmaceutical Chemistry, University of California San Francisco School of Pharmacy, San Francisco, California 94158
| | - David Pearce
- the Division of Nephrology, Department of Medicine, University of California San Francisco School of Medicine, San Francisco, California 94107, and
| |
Collapse
|
41
|
Liang X, Butterworth MB, Peters KW, Frizzell RA. AS160 modulates aldosterone-stimulated epithelial sodium channel forward trafficking. Mol Biol Cell 2010; 21:2024-33. [PMID: 20410134 PMCID: PMC2883946 DOI: 10.1091/mbc.e10-01-0042] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AS160 assists in defining an intracellular compartment in which ENaC accumulates under basal conditions, and this compartment is accessed by aldosterone, via SGK-mediated phosphorylation of AS160, to permit the forward trafficking of ENaC to the apical membrane. Aldosterone-induced increases in apical membrane epithelial sodium channel (ENaC) density and Na transport involve the induction of 14-3-3 protein expression and their association with Nedd4-2, a substrate of serum- and glucocorticoid-induced kinase (SGK1)-mediated phosphorylation. A search for other 14-3-3 binding proteins in aldosterone-treated cortical collecting duct (CCD) cells identified the Rab-GAP, AS160, an Akt/PKB substrate whose phosphorylation contributes to the recruitment of GLUT4 transporters to adipocyte plasma membranes in response to insulin. In CCD epithelia, aldosterone (10 nM, 24 h) increased AS160 protein expression threefold, with a time-course similar to increases in SGK1 expression. In the absence of aldosterone, AS160 overexpression increased total ENaC expression 2.5-fold but did not increase apical membrane ENaC or amiloride-sensitive Na current (Isc). In AS160 overexpressing epithelia, however, aldosterone increased apical ENaC and Isc 2.5-fold relative to aldosterone alone, thus recruiting the accumulated ENaC to the apical membrane. Conversely, AS160 knockdown increased apical membrane ENaC and Isc under basal conditions to ∼80% of aldosterone-stimulated values, attenuating further steroid effects. Aldosterone induced AS160 phosphorylation at five sites, predominantly at the SGK1 sites T568 and S751, and evoked AS160 binding to the steroid-induced 14-3-3 isoforms, β and ε. AS160 mutations at SGK1 phospho-sites blocked its selective interaction with 14-3-3β and ε and suppressed the ability of expressed AS160 to augment aldosterone action. These findings indicate that the Rab protein regulator, AS160, stabilizes ENaC in a regulated intracellular compartment under basal conditions, and that aldosterone/SGK1-dependent AS160 phosphorylation permits ENaC forward trafficking to the apical membrane to augment Na absorption.
Collapse
Affiliation(s)
- Xiubin Liang
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | |
Collapse
|
42
|
Pavlov TS, Chahdi A, Ilatovskaya DV, Levchenko V, Vandewalle A, Pochynyuk O, Sorokin A, Staruschenko A. Endothelin-1 inhibits the epithelial Na+ channel through betaPix/14-3-3/Nedd4-2. J Am Soc Nephrol 2010; 21:833-43. [PMID: 20338996 DOI: 10.1681/asn.2009080885] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Epithelial Na+ channels (ENaCs) mediate sodium reabsorption in the cortical collecting duct (CCD), but the regulatory pathways that modulate the activity of these channels are incompletely understood. Here, we observed that endothelin-1 (ET-1) attenuates ENaC activity acutely by reducing the channel's open probability and chronically by decreasing the number of channels in the plasma membrane. To investigate whether beta1Pix, a signaling protein activated by ET-1, mediates ENaC activity, we reconstituted ENaC in CHO cells with or without coexpressed beta1Pix and found that beta1Pix negatively regulates ENaC. Knockdown of betaPix in native principal cells abolished the ET-1-induced decrease in ENaC channel number. Furthermore, we found that betaPix does not decrease ENaC activity through its guanine nucleotide exchange factor (GEF) activity for Rac1 and Cdc42. Instead, coexpression of beta1Pix mutant constructs revealed that beta1Pix affects ENaC activity through binding 14-3-3 proteins. Coimmunoprecipitation experiments supported a physical interaction between beta1Pix and 14-3-3beta in cultured principal cells. Coexpression of 14-3-3beta increased ENaC activity in CHO cells, but concomitant expression of beta1Pix attenuated this increase. Recruitment of 14-3-3beta by beta1Pix impaired the interaction of 14-3-3beta with the ubiquitin ligase Nedd4-2, thereby promoting ubiquitination and degradation of ENaC. Taken together, these results suggest that the inhibitory effects of chronic ET-1 on ENaC result from betaPix interacting with the 14-3-3/Nedd4-2 pathway.
Collapse
Affiliation(s)
- Tengis S Pavlov
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Ke Y, Butt AG, Swart M, Liu YF, McDonald FJ. COMMD1 downregulates the epithelial sodium channel through Nedd4-2. Am J Physiol Renal Physiol 2010; 298:F1445-56. [PMID: 20237237 DOI: 10.1152/ajprenal.00257.2009] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The epithelial sodium channel (ENaC) is important for the long-term control of Na(+) homeostasis and blood pressure. Our previous studies demonstrated that Copper Metabolism Murr1 Domain-containing protein 1 (COMMD1; previously known as Murr1), a protein involved in copper metabolism, inhibited amiloride-sensitive current in Xenopus laevis oocytes expressing ENaC (J Biol Chem 279: 5429, 2004). In this study, we report that COMMD1 inhibits amiloride-sensitive current in mammalian epithelial cells expressing ENaC, that the COMM domain of COMMD1 is sufficient for this effect, and that knockdown of COMMD1 increases amiloride-sensitive current. COMMD1 is coexpressed with ENaC in rat kidney medulla cells. COMMD1 increased ubiquitin modification of ENaC and decreased its cell surface expression. COMMD1 abolished insulin-stimulated amiloride-sensitive current and attenuated the stimulation of current by activated serum and glucocorticoid-regulated kinase (SGK1). COMMD1 was found to interact with both SGK1 and Akt1/protein kinase B, and knockdown of COMMD1 enhanced the stimulatory effect of both SGK1 and Akt1 on amiloride-sensitive current. COMMD1's effects were reduced in the presence of ENaC proteins containing PY motif mutations, abolished in the presence of a dominant negative form of Nedd4-2, and knockdown of COMMD1 reduced the inhibitory effect of Nedd4-2 on ENaC, but did not enhance current when Nedd4-2 was knocked down. These data suggest that COMMD1 modulates Na(+) transport in epithelial cells through regulation of ENaC cell surface expression and this effect is likely mediated via Nedd4-2.
Collapse
Affiliation(s)
- Ying Ke
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | | | | | | | | |
Collapse
|
44
|
Yang B, Kumar S. Nedd4 and Nedd4-2: closely related ubiquitin-protein ligases with distinct physiological functions. Cell Death Differ 2010; 17:68-77. [PMID: 19557014 PMCID: PMC2818775 DOI: 10.1038/cdd.2009.84] [Citation(s) in RCA: 183] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The Nedd4 (neural precursor cell-expressed developmentally downregulated gene 4) family of ubiquitin ligases (E3s) is characterized by a distinct modular domain architecture, with each member consisting of a C2 domain, 2-4 WW domains, and a HECT-type ligase domain. Of the nine mammalian members of this family, Nedd4 and its close relative, Nedd4-2, represent the ancestral ligases with strong similarity to the yeast, Rsp5. In Saccharomyces cerevisiae Rsp5 has a key role in regulating the trafficking, sorting, and degradation of a large number of proteins in multiple cellular compartments. However, in mammals the Nedd4 family members, including Nedd4 and Nedd4-2, appear to have distinct functions, thereby suggesting that these E3s target specific proteins for ubiquitylation. In this article we focus on the biology and emerging functions of Nedd4 and Nedd4-2, and review recent in vivo studies on these E3s.
Collapse
Affiliation(s)
- B Yang
- Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - S Kumar
- Centre for Cancer Biology, SA Pathology and Hanson Institute, Adelaide, SA 5000, Australia
| |
Collapse
|
45
|
Abstract
Compelling evidence is accumulating indicating a pathophysiological role of the serum-and-glucocorticoid-inducible-kinase-1 (SGK1) in the development and complications of diabetes. SGK1 is ubiquitously expressed with exquisitely high transcriptional volatility. Stimulators of SGK1 expression include hyperglycemia, cell shrinkage, ischemia, glucocorticoids and mineralocorticoids. SGK1 is activated by insulin and growth factors via PI3K, 3-phosphoinositide dependent kinase PDK1 and mTOR. SGK1 activates ion channels (including ENaC, TRPV5, ROMK, KCNE1/KCNQ1 and CLCKa/Barttin), carriers (including NCC, NKCC, NHE3, SGLT1 and EAAT3), and the Na(+)/K(+)-ATPase. It regulates the activity of several enzymes (e.g., glycogen-synthase-kinase-3, ubiquitin-ligase Nedd4-2, phosphomannose-mutase-2), and transcription factors (e.g., forkhead-transcription-factor FOXO3a, beta-catenin and NF-kappaB). A common SGK1 gene variant ( approximately 3 - 5% prevalence in Caucasians, approximately 10% in Africans) is associated with increased blood pressure, obesity and type 2 diabetes. In patients suffering from type 2 diabetes, SGK1 presumably contributes to fluid retention and hypertension, enhanced coagulation and increased deposition of matrix proteins leading to tissue fibrosis such as diabetic nephropathy. Accordingly, targeting SGK1 may favourably influence occurrence and course of type 2 diabetes.
Collapse
Affiliation(s)
- Florian Lang
- Eberhard-Karls-University of Tuebingen, Department of Physiology, Gmelinstrasse 5, Tuebingen 72076, Germany.
| | | | | |
Collapse
|
46
|
Lang F, Artunc F, Vallon V. The physiological impact of the serum and glucocorticoid-inducible kinase SGK1. Curr Opin Nephrol Hypertens 2009; 18:439-48. [PMID: 19584721 PMCID: PMC2883450 DOI: 10.1097/mnh.0b013e32832f125e] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW The role of serum and glucocorticoid-inducible kinase 1 (SGK1) in renal physiology and pathophysiology is reviewed with particular emphasis on recent advances. RECENT FINDINGS The mammalian target of rapamycin complex 2 has been shown to phosphorylate SGK1 at Ser422 (the so-called hydrophobic motif). Ser397 and Ser401 are two additional SGK1-phosphorylation sites required for maximal SGK1 activity. A 5' variant alternate transcript of human Sgk1 has been identified that is widely expressed and shows improved stability, enhanced membrane association, and greater stimulation of epithelial Na+ transport. SGK1 is essential for optimal processing of the epithelial sodium channel and also regulates the expression of the Na+-Cl- cotransporter. With regard to pathophysiology, SGK1 participates in the stimulation of renal tubular glucose transport in diabetes, the renal profibrotic effect of both angiotensin II and aldosterone, and in fetal programing of arterial hypertension. SUMMARY The outlined recent findings advanced our understanding of the molecular regulation of SGK1 as well as the role of the kinase in renal physiology and the pathophysiology of renal disease and hypertension. Future studies using pharmacological inhibitors of SGK1 will reveal the utility of the kinase as a new therapeutic target.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology, University of Tuebingen, Gmelinstrasse 5, Tuebingen 72076, Germany.
| | | | | |
Collapse
|
47
|
Wang J, Lou H, Pedersen CJ, Smith AD, Perez RG. 14-3-3zeta contributes to tyrosine hydroxylase activity in MN9D cells: localization of dopamine regulatory proteins to mitochondria. J Biol Chem 2009; 284:14011-9. [PMID: 19289463 DOI: 10.1074/jbc.m901310200] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The 14-3-3 proteins stimulate the activation of tyrosine hydroxylase (TH), the rate-limiting catecholamine biosynthetic enzyme. To explore if particular endogenous 14-3-3 isoforms specifically affected TH activity and dopamine synthesis, we utilized rodent nigrostriatal tissues and midbrain-derived MN9D dopaminergic cells. Extracts from ventral midbrain and MN9D cells contained similar pools of 14-3-3 mRNAs, with 14-3-3zeta being relatively abundant in both. Protein levels of 14-3-3zeta were also abundant. [(32)P]Orthophosphate labeling of MN9D cells, followed by co-immunoprecipitation with pan-TH and pan-14-3-3 antibodies brought down similar amounts of phosphorylated TH in each, confirming that 14-3-3-bound phosphorylated TH in our cells. Co-immunoprecipitation of striatal tissues with a pan-TH antibody precipitated 14-3-3zeta but not another potential TH regulatory isoform, 14-3-3eta. In whole cell extracts from MN9D cells after 14-3-3 small interfering RNA treatments, we found that 14-3-3zeta knockdown significantly reduced TH activity and dopamine synthesis whereas knockdown of 14-3-3eta had no effect. 14-3-3zeta was found co-localized on mitochondria with TH, and its knockdown by small interfering RNA reduced TH phosphorylation and TH activity in the mitochondrial pool. Together the data support a role for 14-3-3zeta as an endogenous activator of TH in midbrain dopaminergic neurons and furthermore, identify mitochondria as a potential novel site for dopamine synthesis, with implications for Parkinson disease.
Collapse
Affiliation(s)
- Jian Wang
- Departments of Neurology and Pharmacology & Chemical Biology
| | | | | | | | | |
Collapse
|
48
|
Loffing J, Korbmacher C. Regulated sodium transport in the renal connecting tubule (CNT) via the epithelial sodium channel (ENaC). Pflugers Arch 2009; 458:111-35. [PMID: 19277701 DOI: 10.1007/s00424-009-0656-0] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Revised: 02/18/2009] [Accepted: 02/22/2009] [Indexed: 12/29/2022]
Abstract
The aldosterone-sensitive distal nephron (ASDN) includes the late distal convoluted tubule 2, the connecting tubule (CNT) and the collecting duct. The appropriate regulation of sodium (Na(+)) absorption in the ASDN is essential to precisely match urinary Na(+) excretion to dietary Na(+) intake whilst taking extra-renal Na(+) losses into account. There is increasing evidence that Na(+) transport in the CNT is of particular importance for the maintenance of body Na(+) balance and for the long-term control of extra-cellular fluid volume and arterial blood pressure. Na(+) transport in the CNT critically depends on the activity and abundance of the amiloride-sensitive epithelial sodium channel (ENaC) in the luminal membrane of the CNT cells. As a rate-limiting step for transepithelial Na(+) transport, ENaC is the main target of hormones (e.g. aldosterone, angiotensin II, vasopressin and insulin/insulin-like growth factor 1) to adjust transepithelial Na(+) transport in this tubular segment. In this review, we highlight the structural and functional properties of the CNT that contribute to the high Na(+) transport capacity of this segment. Moreover, we discuss some aspects of the complex pathways and molecular mechanisms involved in ENaC regulation by hormones, kinases, proteases and associated proteins that control its function. Whilst cultured cells and heterologous expression systems have greatly advanced our knowledge about some of these regulatory mechanisms, future studies will have to determine the relative importance of the various pathways in the native tubule and in particular in the CNT.
Collapse
|
49
|
Edinger RS, Lebowitz J, Li H, Alzamora R, Wang H, Johnson JP, Hallows KR. Functional regulation of the epithelial Na+ channel by IkappaB kinase-beta occurs via phosphorylation of the ubiquitin ligase Nedd4-2. J Biol Chem 2009; 284:150-157. [PMID: 18981174 PMCID: PMC2610498 DOI: 10.1074/jbc.m807358200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Revised: 10/28/2008] [Indexed: 12/31/2022] Open
Abstract
We have previously shown that IkappaB kinase-beta (IKKbeta) interacts with the epithelial Na+ channel (ENaC) beta-subunit and enhances ENaC activity by increasing its surface expression in Xenopus oocytes. Here, we show that the IKKbeta-ENaC interaction is physiologically relevant in mouse polarized kidney cortical collecting duct (mpkCCDc14) cells, as RNA interference-mediated knockdown of endogenous IKKbeta in these cells by approximately 50% resulted in a similar reduction in transepithelial ENaC-dependent equivalent short circuit current. Although IKKbeta binds to ENaC, there was no detectable phosphorylation of ENaC subunits by IKKbeta in vitro. Because IKKbeta stimulation of ENaC activity occurs through enhanced channel surface expression and the ubiquitin-protein ligase Nedd4-2 has emerged as a central locus for ENaC regulation at the plasma membrane, we tested the role of Nedd4-2 in this regulation. IKKbeta-dependent phosphorylation of Xenopus Nedd4-2 expressed in HEK-293 cells occurred both in vitro and in vivo, suggesting a potential mechanism for regulation of Nedd4-2 and thus ENaC activity. 32P labeling studies utilizing wild-type or mutant forms of Xenopus Nedd4-2 demonstrated that Ser-444, a key SGK1 and protein kinase A-phosphorylated residue, is also an important IKKbeta phosphorylation target. ENaC stimulation by IKKbeta was preserved in oocytes expressing wild-type Nedd4-2 but blocked in oocytes expressing either a dominant-negative (C938S) or phospho-deficient (S444A) Nedd4-2 mutant, suggesting that Nedd4-2 function and phosphorylation by IKKbeta are required for IKKbeta regulation of ENaC. In summary, these results suggest a novel mode of ENaC regulation that occurs through IKKbeta-dependent Nedd4-2 phosphorylation at a recognized SGK1 and protein kinase A target site.
Collapse
Affiliation(s)
- Robert S Edinger
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Jonathan Lebowitz
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Hui Li
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Rodrigo Alzamora
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Huamin Wang
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - John P Johnson
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261; Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Kenneth R Hallows
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261; Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261.
| |
Collapse
|
50
|
Abstract
Hypertension is a serious medical problem affecting a large population worldwide. Liddle syndrome is a hereditary form of early onset hypertension caused by mutations in the epithelial Na+ channel (ENaC). The mutated region, called the PY (Pro-Pro-x-Tyr) motif, serves as a binding site for Nedd4-2, an E3 ubiquitin ligase from the HECT family. Nedd4-2 binds the ENaC PY motif via its WW domains, normally leading to ENaC ubiquitylation and endocytosis, reducing the number of active channels at the plasma membrane. In Liddle syndrome, this endocytosis is impaired due to the inability of the mutated PY motif in ENaC to properly bind Nedd4-2. This leads to accumulation of active channels at the cell surface and increased Na+ (and fluid) absorption in the distal nephron, resulting in elevated blood volume and blood pressure. Small molecules/compounds that destabilize cell surface ENaC, or enhance Nedd4-2 activity in the kidney, could potentially serve to alleviate hypertension. Republished from Current BioData's Targeted Proteins database (TPdb; ).
Collapse
Affiliation(s)
- Daniela Rotin
- Program in Cell Biology, The Hospital for Sick Children, and Biochemistry Department, University of Toronto, Ontario, M5G 1X8, Canada.
| |
Collapse
|