干细胞之家 - 中国干细胞行业门户第一站

 

 

搜索
朗日生物

免疫细胞治疗专区

欢迎关注干细胞微信公众号

  
查看: 472737|回复: 237
go

Disinhibitory pathways for control of sodium transport: regulation of ENaC by SG [复制链接]

Rank: 1

积分
威望
0  
包包
0  
楼主
发表于 2009-4-22 08:36 |只看该作者 |倒序浏览 |打印
作者:Vivek Bhalla,, Rama Soundararajan,, Alan C. Pao, Hongyan Li, and David Pearce作者单位:1 Division of Nephrology, Department of Medicine, and 2 Department of Dermatology, University of California, San Francisco, California 4 Z/ A4 e2 F: G/ H
                  
- U0 C  L' H8 N6 F                  
+ X% `& @( e4 g5 ~$ b& d  i         
& l7 R$ @: Z; y) A                        
7 \% J  `* T: u" M* Z            
5 r$ s0 v: @. A) K+ i; P            5 H1 K' x$ p' }) G3 {
            7 G2 A. W6 {! ~! @6 A
            
" T9 a/ d& K* {) I3 V                      ( M  z# c/ ?6 |
        8 ?& @7 c6 b1 s) ^1 W
        
9 J9 N8 z- R( ?7 |: o, ]. k        
$ V& J4 s" h; n( Z2 f          【摘要】
  ^. m: f( @6 E( m* M8 c      Regulation of ENaC occurs at several levels. The principal hormonal regulator of ENaC, aldosterone, acts through the mineralocorticoid receptor to modulate ENaC-mediated sodium transport, and considerable attention has focused on defining the components of the early phase of this response. Two genes, SGK1 and GILZ, have now been implicated in this regulation. While the functional significance of SGK1 in mediating aldosterone effects is well established, new evidence has enhanced our understanding of the mechanisms of SGK1 action. In addition, recent work demonstrates a novel role for GILZ in the stimulation of ENaC-mediated sodium transport. Interestingly, both SGK1 and GILZ appear to negatively regulate tonic inhibition of ENaC and thus use disinhibition to propagate the rapid effects of aldosterone to increase sodium reabsorption in tight epithelia.
% H3 H3 Q/ k* t! c; F$ P$ A          【关键词】 epithelial sodium channel Nedd ERK trafficking
4 W# z  o2 t0 B) {                  APPROPRIATE COORDINATION OF signaling pathways that regulate distal nephron ion transport is essential for physiological control of blood pressure and electrolyte composition. Sodium (Na   ), as the principal extracellular cation, is a major target of this regulation, primarily through the control of activity and/or apical plasma membrane abundance of the epithelial sodium channel (ENaC) ( 14, 15 ). ENaC constitutes the rate-limiting step in Na   translocation and represents the convergence of multiple activating and inhibitory regulatory factors. Aldosterone, which is the key hormonal mediator of the response to variation in dietary Na  , chloride (Cl - ), and potassium (K   ), acts primarily in the distal nephron to stimulate transport of these essential ions. The reabsorption of Na   and Cl - in particular establish osmotic gradients, which, in the presence of antidiuretic hormone, drive aquaporin-mediated water reabsorption, thus sustaining extracellular fluid volume and blood pressure ( 61 ). Aldosterone-regulated K   secretion is central to the control of extracellular and intracellular concentrations of this essential ion. Other hormones, including insulin, vasopressin, atrial natriuretic peptide, and -MSH, also act in the distal nephron to modulate ion transport.
/ h' ?4 ~8 i3 K- j
4 r; `# l8 g8 _/ y( VRegulation of ENaC, composed of three genetically distinct subunits (,, and ), occurs at multiple levels, including gene transcription and several stages of posttranslational modification ( 34, 59 ), which alter channel open probability ( 74, 76 ), intracellular trafficking, and degradation ( 13 ). Aldosterone induces early- and late-phase responses to increase transepithelial Na   transport in the collecting duct, which are largely, or possibly solely, mediated through effects on gene transcription. For example, target genes encode the transporters themselves, which tend to be induced relatively slowly, and probably play a role in the consolidation ("late") phase of the response ( 78 ) (e.g., ENaC- and Na   -K   -ATPase -subunit). However, regulatory factors are rapidly induced ( 16, 68 ) and mediate the early phases of the response by modulating the trafficking and possibly the open probability of already synthesized channels and transporters. ENaC, in particular, appears to be strongly regulated by changes in trafficking, although some evidence also supports effects of regulators such as K-Ras on open probability ( 70 ). Aldosterone induces an increase in channel density in the apical membrane within 2 h without altering expression of either the - or -subunit of ENaC, in both cultured cells and intact animals ( 6, 43, 47 ). Moreover, heterologous expression of rapidly induced genes, such as serum and glucocorticoid kinase 1 (SGK1) and glucocorticoid-induced leucine zipper protein (GILZ), increases cell surface expression of ENaC and, concomitantly, Na   transport ( 3, 16, 68 ). This review focuses on recent studies that have shed new light on the complex intracellular signaling cascades by which aldosterone stimulates Na   current and alters the cell surface expression of ENaC.) d( {9 Z: X& `) J' U

2 O% d4 k  V( I2 O6 X$ f' ]$ VSGK1 is an Important Aldosterone-Induced Gene, Which Controls ENaC-Mediated Na   Transport
& g' Q/ r8 x4 H+ a4 ^& a
+ f  W) F9 r" e; SSGK1 was originally identified in a mammary tumor cell line as a serine-threonine kinase of unknown function ( 82 ). It was subsequently independently cloned by several groups in screens for genes induced by osmotic shock ( 80 ), brain injury ( 37 ), and aldosterone ( 16, 53 ). In amphibian collecting duct-like cells (A6) and in native rat kidney, SGK1 mRNA levels are upregulated by aldosterone within 30 min, protein levels increase within 45 min, and maximal protein levels are observed within 6 h ( 16 ). SGK1 is expressed ubiquitously in mammalian tissues but is strongly induced in collecting duct, and heterologous expression of SGK1 with ENaC in Xenopus laevis oocytes markedly stimulates amiloride-sensitive Na   transport ( 3, 16 ). This mechanism has also been demonstrated in several epithelial cell lines, including mammalian collecting duct ( 54 ). In addition, SGK1 knockout mice have elevated aldosterone levels and are unable to conserve Na   appropriately on a low-salt diet ( 84 ). Furthermore, immunohistochemical staining shows that despite ENaC expression levels, which are comparable to those of wild-type, apical membrane expression of ENaC is diminished.
, M4 {+ f& [; ~0 x9 n9 A1 {: L
5 j  S: ]8 f$ ]SGK1 is one of six members of the Sgk1/Akt family of serine-threonine kinases, which are known to be activated in a phosphatidylinositide 3'-kinase (PI3K)-dependent fashion. Insulin signals through PI3K and has been shown in cell culture experiments to activate SGK1 ( 58 ). Conversely, pharmacological inhibition of PI3K deactivates SGK1 and Na   transport in various epithelial cell lines. These data support the paradigm that SGK1 is found at the convergence of two extracellular signals (aldosterone and insulin) and regulates distal nephron Na   transport ( 23, 81 ). Physiological data in SGK1 knockout mice further support this idea. Recent evidence also suggests that the PI3K-dependent activity of SGK1 may be regulated by interaction with WNK1 [with no lysine(K) isoform 1] and that this interaction may lead to increased Na   transport through ENaC ( 85, 86 ). Intronic deletions in WNK1 which lead to upregulation of the gene product have been identified recently in patients with type II pseudohypoaldosteronism (Gordon's syndrome) ( 83 ), and more recently WNK1 gene polymorphisms have been linked to hypertension in the general population ( 73 ). WNK1 may therefore cause hypertension through PI3K and SGK1, but this remains to be fully substantiated.
  o) q; ~8 h/ G) j( f5 H
7 q) q* }% d) F7 p4 o- IWhile no monogenetic forms of hypertension have been linked to SGK1, several human genetic studies have linked single-nucleotide polymorphisms in SGK1 to elevated systolic and diastolic blood pressure, as well as hypertension associated with hyperinsulinemia ( 11, 12, 79 ). It remains to be seen whether mutations that increase SGK1 activity cause hypertension, or whether inhibition of SGK1 activity in select populations (for example, patients with the metabolic syndrome or type II diabetes) could ameliorate hypertension.
/ O# B' O( Y- R' L
/ N: Y2 }& D0 w" }( n% ^8 cSGK1 is the Physiologically Relevant Isoform that Stimulates Aldosterone-Induced Na   Transport- h( X) z7 M' m9 m, q+ P% Y8 b
* |% b, n" Z( }! y8 N- B
The Na   -wasting phenotype of SGK1 knockout mice is clearly milder than the severe Na   wasting observed in mice lacking either the mineralocorticoid receptor ( 7 ), ENaC- ( 35 ), or ENaC- ( 49 ). Potential compensatory elements include two other SGK isoforms, SGK2 and SGK3, which were identified by homology cloning ( 18, 41 ), as well as non-SGK/Akt family mediators. SGK2 and SGK3 share 80% homology with SGK1 in their kinase domains and, like SGK1, are activated through the PI3K signaling pathway ( 40 ). SGK2 and SGK3 are expressed in the kidney ( 18, 41 ), share in vitro substrates with SGK1 ( 41 ), and appear to stimulate ENaC ( 25 ) and Na   -K   -ATPase ( 31 ) in X. laevis oocytes. However, SGK2 and SGK3 are not corticosteroid regulated in cultured cells ( 41, 54 ) or in the kidney ( 2 ), suggesting that they do not play significant roles in aldosterone-regulated Na   reabsorption.
& q* L# u6 L/ G5 M( h! c- i1 E4 j; a2 O
Consistent with this notion, mice lacking SGK3 display normal Na   balance even when Na   restricted and do not display the aldosterone resistance observed in SGK1 knockout mice ( 48 ). Furthermore, SGK1-SGK3 double knockout mice do not show a more severe Na   -wasting phenotype than SGK1 knockouts ( 26 ). Surprisingly, the principal phenotype of SGK3 null mice is a striking defect in postnatal hair follicle development ( 48 ). Moreover, SGK2 mRNA levels are not upregulated in compound SGK1 and SGK3 knockout mice, suggesting that SGK2 does not compensate for SGK1 at the transcriptional level. Taken together, SGK1 appears to be the relevant SGK isoform in the response to Na   restriction. Interestingly, one recent study suggests that SGK1 may also be implicated in mediating the natriferic and hypertensive effects of insulin in states of insulin resistance ( 33 ). It remains to be determined whether other SGK/Akt isoforms mediate these natriferic actions as well. In contrast to SGK2 and SGK3, Akt1 does not stimulate ENaC-mediated Na   transport in model systems ( 4 ).! q. r2 Q  d  n+ R9 _% A% c# {

, k2 V( U7 s5 J  PSGK1 Stimulates ENaC Through Phosphorylation of Nedd4-2- _# i+ Q5 ?" \  l: y( f

8 T) m/ J" i2 W5 a' _$ OSGK1 kinase activity is required for all known SGK1 functions, including ENaC regulation ( 10, 20, 42 ). The best characterized target of SGK1 in the context of Na   transport is the E3 ubiquitin ligase Nedd4-2 (for neural precursor cell expressed, developmentally downregulated protein Nedd4-2), a close relative of the "WW domain"-containing protein Nedd4-1, which was identified by a two-hybrid screen as an interacting partner for the COOH-terminal tail of the -subunit of ENaC ( 71 ). Both Nedd4-1 and Nedd4-2 are expressed in kidney and have both been shown to bind ENaC (through direct interaction of tryptophan-rich WW domains with proline- and tyrosine-containing "PY" motifs within ENaC cytoplasmic tails), but only Nedd4-2 has been shown to alter ENaC-mediated Na   transport in X. laevis oocytes ( 38 ). Overexpression of Nedd4-2 leads to decreased cell surface expression and decreased Na   transport ( 1 ). Lack of interaction between Nedd4-2 and Liddle's variants of ENaC (which lack functional PY motifs) is an important mechanism of increased distal Na   reabsorption in patients with Liddle's syndrome, a rare monogenetic form of hypertension.% p% a$ e; e3 _1 U4 d) e' _

0 N' Z* k! E$ B3 m9 o. {( dNedd4-2 appears to play a physiological role in aldosterone-mediated Na   transport because, in contrast to Nedd4-1, it is a target of SGK1 ( 20, 66 ). Overexpression of SGK1 results in phosphorylation of Nedd4-2, and disruption of Nedd4-2's tonic inhibition of ENaC can indirectly stimulate an increase in Na   transport. Moreover, recent work from Flores et al. ( 23 ) has demonstrated that, after acute treatment with aldosterone, the rise in phosphorylated Nedd4-2 parallels the increase in SGK1 in a cortical collecting duct cell line. In addition, targeted gene (siRNA) knockdown of Nedd4-2 in cultured cells has suggested that Nedd4-2, but not Nedd4-1, is required for aldosterone-mediated Na   transport ( 67 ). However, this issue remains unsettled as Vecchione and colleagues ( 77 ) have shown that specific WW domains, found in both isoforms, may functionally inhibit ENaC, and it seems possible that either or both isoforms may be used in a context-dependent fashion. Interestingly, recent evidence has suggested that Nedd4-2 can also ubiquitinylate SGK1 and induce its proteasome-mediated degradation ( 88 ). Thus SGK1-mediated phosphorylation of Nedd4-2 may shift the ligase's substrate specificity from ENaC to SGK1 itself. This model of codependent regulation may allow for feedback inhibition and more finely tuned regulation of Na   transport.7 ~  [' _8 ?  `- Q; v
" B* }4 ~3 v& [' X
14-3-3 Proteins Inhibit Nedd4-2 to Stimulate Aldosterone-Mediated Na   Transport3 K* ~' {9 ?4 Y! n3 b

. F- Y1 S! h  W2 K9 Q9 hRecent studies have begun to shed light on the mechanistic basis of SGK1-mediated modulation of Nedd4-2, by implicating phosphorylation-induced interaction with 14-3-3 proteins ( 8, 36 ). 14-3-3 Proteins are small scaffolding proteins, which are highly conserved in eukaryotic organisms and modulate a remarkable variety of cellular processes, including nuclear localization of transcription factors such as FKHRL1, the activity of enzymes such as serotonin- N -acetyltransferase, and progression of ion channels such as KCNK3 from the endoplasmic reticulum (ER) to the Golgi apparatus ( 9, 56, 57 ). There are seven known mammalian isoforms of 14-3-3 (,,,,,, and ) encoded by distinct genes, which bind as homo- or heterodimers to single or paired motifs within target proteins ( 52 ). The modulation of target protein function is accomplished through several different types of activities, including exposure of nuclear export signals, alteration of active site conformation, displacement of interacting proteins, or bridging of target proteins to induce interaction ( 75 ).0 ~+ B  P  g5 J# `* m

) m. s3 Z7 a/ jThe functionally critical SGK1 target motif within Nedd4-2, surrounding serine 444 ( X. laevis numbering) ( 20 ), conforms perfectly to the 14-3-3 consensus motif, while two other sites match it partially ( 24, 65 ). One of these, serine 338, has been shown to be of secondary importance in the response to SGK1. The third site has not been well characterized. These SGK1 sites are conserved in vertebrate species, from Xenopus to human, and SGK1-mediated phosphorylation of Nedd4-2 ( 20, 66 ) has been demonstrated in vivo and in vitro at both motifs ( 20 ). In particular, Nedd4-2 mutants with alanine substitution at serine 444 inhibit ENaC as strongly as wild-type but do not respond to SGK1; furthermore, SGK1-mediated phosphorylation is markedly reduced.! e- ~) A. I3 q0 ]$ A( {& f
8 w7 u, G/ \' b
Importantly, 14-3-3 proteins interact in cultured cells and in vitro with wild-type, but not mutant, Nedd4-2 in an SGK1-dependent fashion ( Fig. 1 A ). Moreover, endogenous Nedd4-2 and 14-3-3 interact in collecting duct principal cells, and this interaction is stimulated by mineralocorticoids and blunted by inhibitors of PI3K. 14-3-3 Proteins also play a functional role in SGK1-mediated Na   transport in heterologous systems ( Fig. 1 B ). Thus 14-3-3 proteins may be necessary to coordinate SGK1 regulation of Nedd4-2 and ENaC, but further in vivo studies are needed to determine whether 14-3-3 proteins are necessary and/or sufficient to control Nedd4-2 activity in the context of physiologically relevant Na   transport. Moreover, which 14-3-3 isoform(s) is implicated in Nedd4-2 regulation of ENaC is uncertain; evidence supporting roles for two different isoforms has been adduced ( 8, 36 ). Figure 2 shows the simplest interpretation of the 14-3-3 effect: SGK1 phosphorylation of Nedd4-2 induces binding of a 14-3-3 dimer, resulting in attenuation of the Nedd4-2-ENaC interaction.! h4 L% U5 q( T$ d. n1 ~

) i0 ^( x& B3 z5 i5 s( k; t& lFig. 1. 14-3-3 Induces an inhibitory interaction with SGK1 on phosphorylation of Nedd4-2. A : SGK1 stimulates Nedd4-2-14-3-3 interaction. Wild-type (WT) or mutant FLAG-tagged Nedd4-2 was expressed in HEK 293 cells with or without constitutively active SGK1 (S422D) as shown. Coimmunoprecipitation of endogenous 14-3-3 with Nedd4-2 was markedly stimulated by SGK1 ( lane 3 ). Mutation of Nedd4-2 at Pro446 or Ser444 abrogated the SGK1-dependent interaction of Nedd4-2 and 14-3-3. Kinase-dead SGK1 (K127M) was unable to stimulate interaction. IP, immunoprecipitated; IB, immunoblotted. B : interaction of Nedd4-2 and 14-3-3 augments SGK1-stimulated Na   transport. Nedd4-2 mutations that disrupt 14-3-3 interaction reduce SGK1-mediated epithelial Na   channel (ENaC) activation in Xenopus laevis oocytes ( top ). 14-3-3 Overexpression further stimulates ENaC-mediated Na   current in the presence of SGK1 ( bottom ). ENaC-mediated Na   current was measured by 2-electrode voltage clamp in the presence and absence of phenamil and is depicted relative to ENaC alone. The statistical significance between different conditions was compared as indicated. See Ref. 8 for details. * P ' K7 u. p4 I3 y' M6 O8 r0 F

4 B/ N" j. ?  j. j6 G4 JFig. 2. Schematic depiction of the role of SGK1 and GILZ in hormonal regulation of ENaC activity in an epithelial cell. SGK1 and GILZ abundance is regulated by aldosterone (Aldo). SGK1 activity is regulated by insulin through phosphatidylinositide 3'-kinase (PI3K). According to our hypothesis, GILZ acts in parallel to inhibit ERK signaling. Together, they coordinately block Nedd4-2 inhibition of ENaC. See text for details. For simplicity, only some of the proposed and/or demonstrated interacting complexes are shown. Small circled P, phosphate; Ins, insulin. See the text for definitions of other abbreviations.
4 g' |) r/ t; A5 P2 {
: ?' @) ~7 u% \, K7 [SGK1 May Increase Cell Surface Expression of ENaC. j# b7 [9 j. M0 [& O$ _6 t
; p8 ]8 |4 e2 U' q% _, j+ E
Taken together, the available data suggest the schematic model shown in Fig. 2. Aldosterone may or may not affect ENaC open probability ( 29, 39 ), and although there is evidence that SGK1 phosphorylates ENaC itself ( 21 ), the majority of evidence favors the hypothesis that SGK1 substrates are regulatory proteins, such as Nedd4-2, or components of the trafficking machinery. In addition, aldosterone and SGK1 clearly have substantial effects on channel plasma membrane abundance ( 20, 43, 47 ), although it remains uncertain whether their effects are primarily of channel insertion, retrieval, or both ( 19 ). The effects of brefeldin A (BFA) on ENaC currents may shed some light on this issue. BFA is a fungal metabolite, which potently inhibits anterograde vesicular transport from the ER to the cis -Golgi, and hence delivery of newly synthesized channels (or other cargo) through the Golgi to the plasma membrane. Regulatory factors that primarily stimulate steady-state transport by inhibiting the endocytic pathway will delay BFA-induced run-down in Na   current, whereas factors that stimulate exocytosis should have little or no effect. One prior study did not find a substantial effect of SGK1 on BFA-induced run-down in ENaC-mediated Na   current in X. laevis oocytes ( 3 ). In this study, Nedd4-2 was not coexpressed, and the SGK1 effect on the baseline current was modest ( 2-fold). Nedd4-2-mediated ubiquitinylation increases ENaC retrieval and degradation ( 44, 45, 72 ), and SGK1 has a substantially more potent ENaC stimulatory effect when it is acting in the presence of Nedd4-2 ( Fig. 3 ) ( 8, 20, 66 ). Interestingly, under conditions of Nedd4-2 coexpression, SGK1 markedly inhibits BFA-induced run-down, consistent with a pronounced effect on channel internalization ( Fig. 3 ). However, its effects may not be limited to this arm of ENaC trafficking; Nedd4-2 ubiquitinylation may also influence trafficking at the level of sorting in endosomes and trans -Golgi ( 28, 32 ). Moreover, SGK1 may have Nedd4-2-independent effects on trafficking of the channel. Thus SGK1 likely acts at multiple points in ENaC trafficking to control its plasma membrane abundance.
' x3 Z# M0 c: _2 _  U) d+ w) D
: ?! X: }/ M# m% a" _  S3 [1 _2 qFig. 3. SGK1 markedly blunts brefeldin A (BFA)-induced rundown of ENaC-mediated Na   current. X. laevis oocytes were injected with cRNA for ENaC subunits and Nedd4-2 with ( top 2 curves, gray) or without ( bottom 2 curves, black) SGK1. Oocytes were treated with BFA (5 µM) or vehicle (arrow), and 2-electrode voltage clamp was performed at various times after BFA exposure. At earlier time periods, washout of BFA brought current back up to pretreatment levels (not shown). Phenamil-sensitive Na   current ( I ) is expressed relative to ENaC Nedd4-2 before injection of BFA.
0 W* Z! I; _) r7 z
5 ^4 Z0 S- R! V* n' tAldosterone-Induced GILZ Negatively Regulates ERK to Stimulate Na   Transport
1 d$ `# f' L7 i6 R/ V! J
! _" o) }  X) S4 G, b, c# \2 \Several lines of evidence suggest that SGK1 is not the only mediator of aldosterone action. Most notably, although SGK1 knockout mice have aldosterone resistance, their phenotype is substantially less severe than that of either mineralocorticoid receptor- or ENaC- knockout mice ( 25, 84 ), or of adrenalectomized wild-type animals ( 43 ), suggesting that renal ENaC function and renal mineralocorticoid action are partially, but not completely, dependent on SGK1. The observation that short-term aldosterone treatment induces an accumulation of SGK1 in the entire aldosterone-sensitive distal nephron, whereas the apical insertion of ENaC takes place only in the connecting tubule and early collecting duct, indicates that other factors are probably required to drive ENaC cell surface expression and activity, and thus Na   reabsorption ( 43 ). It is also notable that inhibitors of PI3K (which is responsible for activation of SGK1) do not completely abolish aldosterone-induced Na   transport ( 81 ), further supporting the idea that aldosterone may act through multiple pathways to stimulate Na   reabsorption, some of which are SGK1 and PI3K independent. Other regulators, including K-Ras ( 69 ) and KS-WNK1 ( 54 ), have been identified and shown to stimulate Na   transport; however, aldosterone regulation has not been demonstrated in mammals, and their physiological importance has not yet been established.2 G' q% D. s, |8 |* `
1 U6 Q  p7 i; W! k5 ?  T: x
Recent evidence supports the theory that GILZ represents an important mediator of aldosterone action, which acts in parallel with SGK1 to increase ENaC plasma membrane localization ( 68 ). GILZ is a member of the TSC22 (TGF- -stimulated clone 22) family of leucine zipper proteins that were initially thought to function as transcription factors ( 17 ). As its name implies, it was originally identified in T lymphocytes as a glucocorticoid early-response gene ( 17 ). It was identified subsequently as a mineralocorticoid-regulated transcript by serial analysis of gene expression and was shown to be rapidly and robustly stimulated by aldosterone in native collecting duct ( 51 ) and cultured collecting duct cells ( 60 ). In cultured T cells, GILZ inhibits IL-2 production by inhibiting ERK signaling, possibly by displacing Ras from the Ras-binding domain of Raf ( 5 ). Interestingly, in collecting duct cells, the ERK pathway has a potent inhibitory effect on ENaC ( 50, 64, 87 ), which may be due, in large measure, to direct ENaC phosphorylation, which stimulates interaction with Nedd4-1 and/or Nedd4-2 ( 64 ). This latter observation suggested a possible mechanism for GILZ stimulation of ENaC, which has been supported by recent evidence in oocytes and cultured collecting duct cells ( 68 ), and is reviewed briefly below.4 f2 s, K. D7 ]0 s5 \7 `

# N' r3 P! V% [' l+ c7 V$ i# e4 k8 E: ?It is now increasingly clear that ERK activation, for example by EGF (in cultured collecting duct cells) ( 22, 27, 63 ) or by progesterone (in ENaC-expressing X. laevis oocytes) ( 55 ), inhibits Na   current, thereby serving as a late brake, which provides feedback inhibition to limit aldosterone-stimulatory effects ( 27 ). However, there is constitutive ERK activation in collecting duct cells ( 46, 68 ), which could also influence basal levels of Na   transport, as well as electrical potential gradients. GILZ expression in progesterone-treated oocytes (to activate ERK), markedly inhibits phospho-ERK2 formation and stimulates ENaC-mediated Na   current, in a manner that is similar to, and nonadditive with, MEK inhibition by U-0126 ( 68 ) ( Fig. 4 ). Similarly, in EGF-treated mpkCCD c14 (murine collecting duct) cells, heterologous expression of GILZ induces parallel downregulation of activated ERK and upregulation of transepithelial Na   transport. As in oocytes, MEK inhibition has a nonadditive effect, similar to GILZ expression, further suggesting that they act in the same pathway, upstream of ERK1/2. As in oocytes, most of the stimulatory effect of GILZ in mpkCCD c14 cells appears to reflect inhibition of ERK signaling, and hence disinhibition of Na   transport. Moreover, EGF, which is known to act through EGFR to stimulate ERK1/2 in HEK 293 cells, markedly decreases cell surface ENaC expression, and heterologous expression of GILZ in EGF-treated HEK 293 cells completely reverses this effect. Together, these data strongly support the view that GILZ contributes to mineralocorticoid stimulation of ENaC-mediated Na   transport by inhibiting the ERK cascade ( Fig. 2 ).+ H8 n" L: w; B8 z$ Z, ^
9 b1 Q3 k' L$ ]. X
Fig. 4. GILZ stimulates ENaC activity in X. laevis oocytes by inhibiting ERK. A : X. laevis oocytes were coinjected with cRNA for all 3 ENaC subunits (3 ng each) either alone or in combination with GILZ cRNA (3 ng). Twenty-four hours after incubation in a low-Na   medium, they were treated with 2 µg/ml progesterone (or an equal volume of ethanol/DMSO vehicle control) to activate ERK. As a control, 1 group of oocytes was pretreated with the MEK inhibitor U-0126 for 2 h before progesterone treatment. ENaC activity was measured using 2-electrode voltage clamp and expressed relative to ENaC alone vehicle. B : GILZ prevents activation of ERK in a manner similar to U-0126. Shown is Western blot analysis for active/phosphorylated (Phospho-) ERK (p42/ERK2) in GILZ-expressing oocytes (or controls) treated with progesterone or ethanol/DMSO (vehicle control). Blots were stripped and reprobed for total ERK as shown. The blot was probed with anti-Myc antibody to detect Myc-tagged GILZ expression. Adapted from Ref. 68.
3 r$ d4 n/ Z/ p' N' v
" I% G# Z2 I0 I0 n% p6 ZInterestingly, these results involving GILZ and ERK are reminiscent of the mechanism by which SGK1 stimulates ENaC activity, which also involves "inhibiting an inhibitor": Nedd4-2 ( 20 ). ERK-mediated ENaC inhibition appears to proceed through phosphorylation of the channel, which stimulates interaction with Nedd4 proteins ( 64 ), leading to a decrease in channel surface expression and subsequent degradation ( 30 ). The latter may be due to a late effect that is secondary to enhanced internalization. Although this mechanism remains to be further elucidated, it is notable that in HEK 293 cells, 1-h treatment with EGF markedly diminished cell surface ENaC without any detectable change in the total cellular pool. Here, too, the effect of GILZ on ENaC appears to be largely, if not solely, mediated by its inhibition of ERK1/2. These data support the view that the effects of ERK and GILZ, like those of Nedd4-2 and SGK1 ( 20, 43 ), are primarily on levels of plasma membrane channel expression. During the early phase of aldosterone action, the parallel effects of SGK1 and GILZ converge on the ENaC-Nedd4-2 interaction, abrogating it by increasing Nedd4-2 phosphorylation (and hence recruitment of 14-3-3) and decreasing channel phosphorylation, respectively. This study highlights the importance of "disinhibition" as a key mechanism in the control of ENaC function, probably through modulation of trafficking and degradation ( 62 ).
  l0 m1 G6 X' w* V
- }0 X5 ~0 F" PPerspectives
, Q% v/ J9 M9 S
5 l4 y) E3 Z# H0 J' QThe rapid effects of aldosterone on ENaC-mediated Na   transport modulate existing PI3K-dependent and ERK-dependent signaling cascades, acting, at least in part, to disinhibit the tonic inhibition of ENaC by ubiquitin ligase Nedd4-2 ( Fig. 2 ). Several investigations have provided further insight into the mechanisms by which aldosterone-induced SGK1 modulates Nedd4-2 to stimulate ENaC. In addition, elucidation of mechanisms by which aldosterone-induced GILZ negatively regulates both ERK activation and the Nedd4-2-ENaC interaction suggests a parallel mechanism to SGK1 in the control of epithelial Na   transport. We suggest that SGK1 and GILZ, in concert, mediate cross talk between steroid and growth factor signaling pathways, thereby providing context-appropriate regulation of Na   transport., ]+ ~' Z$ N% y1 W$ @, C' Z
" e0 ~+ M0 v% C% c$ \
NOTE ADDED IN PROOF
9 l+ F+ g. ]% h+ z7 @3 G  K/ t* O% F& f
Since submission of this review, Liang et al. have shown that 14-3-3 may specifically regulate the interaction between phosphorylated Nedd4-2 and ENaC ( J Biol Chem. In press.).0 d% J& d, a1 U2 f

! C) Q7 c5 z1 U1 fGRANTS
+ P6 V2 i! K6 g" ~3 I; T" g) I) Q: a! o
This work was supported by National Institutes of Health Grants K08-DK-071648 (to V. Bhalla), K08-DK-073487 (to A. C. Pao), and R01-DK-51151 (to D. Pearce) and by an American Heart Association Western States Affiliate Grant 0525234Y (to R. Soundararajan).
4 c9 \, l, y+ Q" S7 n          【参考文献】
  h- v7 B" Q+ j1 a6 c3 q* Q' C& U' d Abriel H, Loffing J, Rebhun JF, Pratt JH, Schild L, Horisberger JD, Rotin D, and Staub O. Defective regulation of the epithelial Na   channel by Nedd4 in Liddle's syndrome. J Clin Invest 103: 667-673, 1999.$ X1 e2 ?+ N- o0 [" G

5 T4 f1 A( {, S: `1 ^+ f) G3 [- C# v1 e* P0 u  ?8 _
1 Q2 \) k( K. t: v* [5 X, E
Alvarez de la Rosa D and Canessa CM. Role of SGK in hormonal regulation of epithelial sodium channel in A6 cells. Am J Physiol Cell Physiol 284: C404-C414, 2003.7 G) v5 U$ J' p) h$ y7 u. {
+ v$ C4 h+ t7 U) \9 ]: c, a

0 w6 x& I7 o) @+ ]! Z& n2 @9 o8 W
Alvarez de la Rosa D, Zhang P, Naray-Fejes-Toth A, Fejes-Toth G, and Canessa CM. The serum and glucocorticoid kinase sgk increases the abundance of epithelial sodium channels in the plasma membrane of Xenopus oocytes. J Biol Chem 274: 37834-37839, 1999.
" J& a$ T( M& |& h/ Z1 r1 z' v+ J" j% F* ~3 g: R

/ E/ f* j8 ^3 K4 H8 Q; B3 o3 [- T. z" o1 U# s5 c
Arteaga MF and Canessa CM. Functional specificity of Sgk1 and Akt1 on ENaC activity. Am J Physiol Renal Physiol 289: F90-F96, 2005.
5 W) A% q0 }& \6 Q& {% v0 Q
" ~5 O; F+ k% Y
5 Y7 u6 W) ?' C$ O7 ?& l6 O; h
9 ]" l" ~6 c( z, t/ k+ P0 vAyroldi E, Zollo O, Macchiarulo A, Di Marco B, Marchetti C, and Riccardi C. Glucocorticoid-induced leucine zipper inhibits the Raf-extracellular signal-regulated kinase pathway by binding to Raf-1. Mol Cell Biol 22: 7929-7941, 2002.
  t! r9 O  |8 Q4 u: q9 g" M$ @( S/ `& y2 ]9 t0 B9 D5 a

; n" H7 ~6 A. b% ]7 a! v9 Y/ i* ]7 f5 r
Bens M, Vallet V, Cluzeaud F, Pascual-Letallec L, Kahn A, Rafestin-Oblin ME, Rossier BC, and Vandewalle A. Corticosteroid-dependent sodium transport in a novel immortalized mouse collecting duct principal cell line. J Am Soc Nephrol 10: 923-934, 1999.
& |9 O+ |0 I% _- |+ E4 `- U. f( l) X& R

9 f" [! V4 b# `; o# s( u4 ^$ o8 m8 X: v* |. Z" [+ [
Berger S, Bleich M, Schmid W, Cole TJ, Peters J, Watanabe H, Kriz W, Warth R, Greger R, and Schutz G. Mineralocorticoid receptor knockout mice: pathophysiology of Na   metabolism. Proc Natl Acad Sci USA 95: 9424-9429, 1998.
) J) @0 ^; H& X0 Z" S* ~: o; `
( ]- b! v, H  D( y
) o4 k) ?. g6 c5 e% P! L
! `6 w- U  y9 K0 U  eBhalla V, Daidie D, Li H, Pao AC, LaGrange LP, Wang J, Vandewalle A, Stockand JD, Staub O, and Pearce D. Serum- and glucocorticoid-regulated kinase 1 regulates ubiquitin ligase neural precursor cell-expressed, developmentally down-regulated protein 4-2 by inducing interaction with 14-3-3. Mol Endocrinol 19: 3073-3084, 2005.- h1 `  u  D5 q+ P8 P+ U
7 k5 _# H) T: c" \! R& a8 O

# A9 b# H: {( c
$ u, J/ T. j- I0 A) m7 N" ~Brunet A, Kanai F, Stehn J, Xu J, Sarbassova D, Frangioni JV, Dalal SN, DeCaprio JA, Greenberg ME, and Yaffe MB. 14-3-3 Transits to the nucleus and participates in dynamic nucleocytoplasmic transport. J Cell Biol 156: 817-828, 2002.
6 _& U9 P& S3 O0 H% I- G
% b' b- c& V7 v# \3 w3 E  c/ N) e1 e. K+ x& r( }. z

; `, @: W' y5 ?  b' ]/ b3 v7 BBrunet A, Park J, Tran H, Hu LS, Hemmings BA, and Greenberg ME. Protein kinase SGK mediates survival signals by phosphorylating the forkhead transcription factor FKHRL1 (FOXO3a). Mol Cell Biol 21: 952-965, 2001.' d* l, ^: t  _. d# W$ x7 F

& T4 l4 `  [' n4 v* r" E2 c( [- G1 @% p, z( H' b% ]2 D- N
7 E: ?( q# b: w5 l1 H
Busjahn A, Aydin A, Uhlmann R, Krasko C, Bahring S, Szelestei T, Feng Y, Dahm S, Sharma AM, Luft FC, and Lang F. Serum- and glucocorticoid-regulated kinase (SGK1) gene and blood pressure. Hypertension 40: 256-260, 2002.9 x) Z  P$ \6 [. x# L/ H" ~
2 r# v+ z+ [! c7 e0 I2 \
0 O6 B% G. L. q! I: L/ q9 \6 W
$ f% R6 F0 d7 K" b# v
Busjahn A and Luft FC. Twin studies in the analysis of minor physiological differences between individuals. Cell Physiol Biochem 13: 51-58, 2003.
( D* [* _" n# q& }
2 j0 z( |3 z  ?6 |7 e, T
- ?. N/ L+ e2 p; `( u$ _5 D8 T  U& [
Butterworth MB, Edinger RS, Johnson JP, and Frizzell RA. Acute ENaC stimulation by cAMP in a kidney cell line is mediated by exocytic insertion from a recycling channel pool. J Gen Physiol 125: 81-101, 2005.( O: n; O- [$ E: j% \/ Y& w* L

- E+ Q4 {0 \3 b' F" Q) p
/ Z- i4 [5 `0 c; A2 ^9 Y
1 e/ {# J7 D- D  o  e9 x, N% T: N7 pCanessa CM, Horisberger JD, and Rossier BC. Epithelial sodium channel related to proteins involved in neurodegeneration. Nature 361: 467-470, 1993.
4 S0 i# z* @) k( x! G0 x" z( b% C% M# D% Y5 s/ z
# R* O1 A: f& v: D  G$ V
- s% c0 F7 J1 O) D  ~
Canessa CM, Schild L, Buell G, Thorens B, Gautschi I, Horisberger JD, and Rossier BC. Amiloride-sensitive epithelial Na   channel is made of three homologous subunits. Nature 367: 463-467, 1994.
/ z1 i2 \! j0 [! e  I6 y- {9 O+ O4 p) S& [. p( t
3 h3 {) @3 J9 j: }8 V) t. D
7 l: O* }& j6 J. X. R- F5 d* ~4 o
Chen SY, Bhargava A, Mastroberardino L, Meijer OC, Wang J, Buse P, Firestone GL, Verrey F, and Pearce D. Epithelial sodium channel regulated by aldosterone-induced protein sgk. Proc Natl Acad Sci USA 96: 2514-2519, 1999.4 ]: J$ U7 }* D, `3 v
, G& E; A  _8 R3 ]- J* [4 h
& I7 f: @6 n" d4 b. B
. G/ C& t: p* A1 w3 i1 R* A
D'Adamio F, Zollo O, Moraca R, Ayroldi E, Bruscoli S, Bartoli A, Cannarile L, Migliorati G, and Riccardi C. A new dexamethasone-induced gene of the leucine zipper family protects T lymphocytes from TCR/CD3-activated cell death. Immunity 7: 803-812, 1997.& i8 s4 }2 Y' X
8 `. C5 l4 C# M6 |% o: _
9 T; z) B$ r' v* b; c1 ?+ W$ F( r5 g
& y% M5 ^1 v' j3 `
Dai F, Yu L, He H, Zhao Y, Yang J, Zhang X, and Zhao S. Cloning and mapping of a novel human serum/glucocorticoid regulated kinase-like gene, SGKL, to chromosome 8q12.3-q131. Genomics 62: 95-97, 1999.
( T: I& s, s0 Y" y3 r& z9 H3 @- M' C: i
# l; L: ?! Y* m7 ~1 s  y9 X* Y) k
( s9 {5 W1 {! u5 F0 V
De La Rosa DA, Paunescu TG, Els WJ, Helman SI, and Canessa CM. Mechanisms of regulation of epithelial sodium channel by SGK1 in A6 cells. J Gen Physiol 124: 395-407, 2004.& ?) m) ^+ J; W' k/ L" a1 p
1 ~( A( r% E3 Q0 `( t/ V
+ B& j$ C" i+ l( P* ?  S
/ S# j% w. v$ Q
Debonneville C, Flores SY, Kamynina E, Plant PJ, Tauxe C, Thomas MA, Munster C, Chraibi A, Pratt JH, Horisberger JD, Pearce D, Loffing J, and Staub O. Phosphorylation of Nedd4-2 by Sgk1 regulates epithelial Na   channel cell surface expression. EMBO J 20: 7052-7059, 2001.
8 M' x/ f9 s1 B. ~( c( C: |
- d: E7 P$ C* D) F' I
, ^5 T) ~. y+ d' U, Z6 l7 I
8 @1 [# k3 E" {6 i5 @1 [4 qDiakov A and Korbmacher C. A novel pathway of epithelial sodium channel activation involves a serum- and glucocorticoid-inducible kinase consensus motif in the C terminus of the channel's alpha-subunit. J Biol Chem 279: 38134-38142, 2004.
; G2 y/ H" ^8 a% Z3 {7 t$ `% s+ z+ x
- B# N5 N; @( V3 [) ^

3 C" {4 k& F  `- H5 W7 m& u$ IFalin R, Veizis IE, and Cotton CU. A role for ERK1/2 in EGF- and ATP-dependent regulation of amiloride-sensitive sodium absorption. Am J Physiol Cell Physiol 288: C1003-C1011, 2005.
1 p" {6 ]8 b* t! x0 G& X$ S
6 C* d5 n/ `( ?" g( m
- H& T' C. Q. a/ |$ i' y
3 }3 t$ G. N+ R2 XFlores SY, Loffing-Cueni D, Kamynina E, Daidie D, Gerbex C, Chabanel S, Dudler J, Loffing J, and Staub O. Aldosterone-induced serum and glucocorticoid-induced kinase 1 expression is accompanied by Nedd4-2 phosphorylation and increased Na   transport in cortical collecting duct cells. J Am Soc Nephrol 16: 2279-2287, 2005.
; q7 b4 ?' j6 Q* |- X3 Q/ _# D, V" e& N+ u6 z
5 X4 g" O0 T. j) M' j( d, G+ o* A
( v" D  t* r- I' M: ?* m1 P  _' |8 O
Fouladkou F, Alikhani-Koopaei R, Vogt B, Flores SY, Malbert-Colas L, Lecomte MC, Loffing J, Frey FJ, Frey BM, and Staub O. A naturally occurring human Nedd4-2 variant displays impaired ENaC regulation in Xenopus laevis oocytes. Am J Physiol Renal Physiol 287: F550-F561, 2004.2 J! }2 l0 m$ ?2 K9 _/ s
, s% z: |1 q" {8 K( w
2 i0 A3 z5 b7 d. U" F
6 J) u' J% r) R/ B5 d$ N
Friedrich B, Feng Y, Cohen P, Risler T, Vandewalle A, Broer S, Wang J, Pearce D, and Lang F. The serine/threonine kinases SGK2 and SGK3 are potent stimulators of the epithelial Na   channel alpha,beta,gamma-ENaC. Pflügers Arch 445: 693-696, 2003.3 L: B) C, w4 }) G! `( ]

0 h% W- t" d, C$ }' R# `4 S; j
% k/ p5 T; p6 e' Z) o3 o1 Y2 U
6 \$ S: g, `& W; {, XGrahammer F, Artunc F, Sandulache D, Rexhepaj R, Friedrich B, Risler T, McCormick JA, Dawson K, Wang J, Pearce D, Wulff P, Kuhl D, and Lang F. Renal function of gene-targeted mice lacking both SGK1 and SGK3. Am J Physiol Regul Integr Comp Physiol 290: R945-R950, 2006.
0 A3 t% L* ^! A6 n( l: R- `% i
3 b: M8 @  [7 h5 t  N% p5 n0 b+ g  J
1 V8 [0 [6 R+ I6 v9 B" {6 s
Grossmann C, Freudinger R, Mildenberger S, Krug AW, and Gekle M. Evidence for epidermal growth factor receptor as negative-feedback control in aldosterone-induced Na   reabsorption. Am J Physiol Renal Physiol 286: F1226-F1231, 2004.  x& [6 j. F) i6 z9 f; z0 m

' |' |( f; w1 ?0 I9 F0 g
$ N4 s8 w% I- ?( M
( T9 d7 Y$ u) v( w7 W3 DHelliwell SB, Losko S, and Kaiser CA. Components of a ubiquitin ligase complex specify polyubiquitination and intracellular trafficking of the general amino acid permease. J Cell Biol 153: 649-662, 2001.
; _" f# s2 M1 z; D; z# f/ }, U8 Q
7 w4 w1 }0 {& v+ e& t# i# ^$ Y+ ~
4 p  k; e1 e& k4 B2 D- o
Helman SI, Liu X, Baldwin K, Blazer-Yost BL, and Els WJ. Time-dependent stimulation by aldosterone of blocker-sensitive ENaCs in A6 epithelia. Am J Physiol Cell Physiol 274: C947-C957, 1998.
  g8 K1 C$ q2 T6 |" w% a- G; B! T+ O0 B; T

1 V$ @% G) O1 |  Z2 v
5 q( |7 k3 h' O% QHendron E, Patel P, Hausenfluke M, Gamper N, Shapiro MS, Booth RE, and Stockand JD. Identification of cytoplasmic domains within the epithelial Na   channel reactive at the plasma membrane. J Biol Chem 277: 34480-34488, 2002.5 ]8 u7 P8 w0 }, o1 l
/ W% |; z2 c3 Q% B0 j3 C" b# O
2 \- l; W- `! W  Y
3 N( `7 _" B3 p4 n
Henke G, Setiawan I, Bohmer C, and Lang F. Activation of Na   /K   -ATPase by the serum and glucocorticoid-dependent kinase isoforms. Kidney Blood Press Res 25: 370-374, 2002.
5 x/ p. c6 D4 r& Z4 m
7 }; n  i6 R& T5 D  V! ]" h  F) R" B: G0 ?) @$ G! \' s/ B

; G& y2 O; u9 q) d! p. c+ QHicke L and Dunn R. Regulation of membrane protein transport by ubiquitin and ubiquitin-binding proteins. A new ticket for entry into budding vesicles-ubiquitin. Annu Rev Cell Dev Biol 19: 141-172, 2003.- P, x  g+ X) Z  F1 ~6 v
2 M  B6 N, _+ W
& Q( e7 p6 G; `: t# l

# l. R3 S6 J& W3 ~9 V  }Huang DY, Boini KM, Friedrich B, Metzger M, Just L, Osswald H, Wulff P, Kuhl D, Vallon V, and Lang F. Blunted hypertensive effect of combined fructose and high-salt diet in gene-targeted mice lacking functional serum- and glucocorticoid-inducible kinase Sgk1. Am J Physiol Regul Integr Comp Physiol 290: R935-R944, 2006./ T+ ?0 U* q: l0 Y. t$ ]6 r6 y

+ b9 Z) x8 M7 F' V8 r; `  [1 l2 V  O$ W( s* ~
: ]' i) b$ {, ?6 h/ f7 y: r$ C" u
Hughey RP, Bruns JB, Kinlough CL, Harkleroad KL, Tong Q, Carattino MD, Johnson JP, Stockand JD, and Kleyman TR. Epithelial sodium channels are activated by furin-dependent proteolysis. J Biol Chem 279: 18111-18114, 2004.
8 s- F* Q$ k7 z7 U  n
7 k7 ?" o  O# ~: b0 B: j5 O( ~7 ?1 |! D3 S

2 U8 P$ I7 C1 r+ ^Hummler E, Barker P, Gatzy J, Beermann F, Verdumo C, Schmidt A, Boucher R, and Rossier BC. Early death due to defective neonatal lung liquid clearance in alpha-ENaC-deficient mice. Nat Genet 12: 325-328, 1996.
7 C/ P; m( U/ E( e- [
. n7 K- q) w- i$ r
2 {+ J$ \% Z  ]9 ]" p, q
4 n' G; q, a3 n1 j7 }Ichimura T, Yamamura H, Sasamoto K, Tominaga Y, Taoka M, Kakiuchi K, Shinkawa T, Takahashi N, Shimada S, and Isobe T. 14-3-3 Proteins modulate the expression of epithelial Na   channels by phosphorylation-dependent interaction with Nedd4-2 ubiquitin ligase. J Biol Chem 280: 13187-13194, 2005.
$ G1 D6 ^  u1 i! \8 F0 n( a; Q% a6 F0 c0 V8 K; e; y/ c
! o! H5 i7 E( R! W
: f( j/ B9 [6 |4 _- e
Imaizumi K, Tsuda M, Wanaka A, Tohyama M, and Takagi T. Differential expression of sgk mRNA, a member of the Ser/Thr protein kinase gene family, in rat brain after CNS injury. Brain Res Mol Brain Res 26: 189-196, 1994." Q3 a9 L% x9 t/ W6 R2 Z
6 H, {! h. R/ T
! F5 e. V6 L# O. C
7 m# x6 B* W  q4 x! V$ m
Kamynina E, Tauxe C, and Staub O. Distinct characteristics of two human Nedd4 proteins with respect to epithelial Na   channel regulation. Am J Physiol Renal Physiol 281: F469-F477, 2001.4 f2 j7 [/ k- S% W1 z

+ C5 h$ D. R  Z8 ]- }3 Q( _
3 v5 S& S' k. R1 V
# ]9 Y& m! R7 r: |1 o/ {Kleyman TR, Zuckerman JB, Middleton P, McNulty KA, Hu B, Su X, An B, Eaton DC, and Smith PR. Cell surface expression and turnover of the alpha-subunit of the epithelial sodium channel. Am J Physiol Renal Physiol 281: F213-F221, 2001.# B4 v. Z0 i( i% N  s. Q
1 w( H9 {4 g1 I$ _  }& M. ~) d* e# }

- P8 ^3 N* m! N" W: m- W+ D
% u5 c6 y; X( [6 W0 ^  i$ B. xKobayashi T and Cohen P. Activation of serum- and glucocorticoid-regulated protein kinase by agonists that activate phosphatidylinositide 3-kinase is mediated by 3-phosphoinositide-dependent protein kinase-1 (PDK1) and PDK2. Biochem J 339: 319-328, 1999.
" X% F) _6 q7 T$ S/ n) h  e$ t: J/ F- j: S8 X9 ~
# \7 S. A% _9 [. M6 S, b, }$ U
  v; q" c, }: k
Kobayashi T, Deak M, Morrice N, and Cohen P. Characterization of the structure and regulation of two novel isoforms of serum- and glucocorticoid-induced protein kinase. Biochem J 344: 189-197, 1999.% p9 n4 K; r  `# Y5 X8 ?7 ]

! l, q/ U) `) \0 A8 R2 Z$ p; Q6 I5 s- ~+ m, O+ G* s' {

) L& I: O$ U- L7 G3 B: R& MLeong ML, Maiyar AC, Kim B, O'Keeffe BA, and Firestone GL. Expression of the serum- and glucocorticoid-inducible protein kinase, Sgk, is a cell survival response to multiple types of environmental stress stimuli in mammary epithelial cells. J Biol Chem 278: 5871-5882, 2003.
" |: C, [- K* H0 s' p( A; G  B4 p, j# k9 L( w( X3 `- A3 C

0 `8 |1 r$ S5 d6 f: d, e/ J: v1 i2 V0 d6 f1 o5 O6 [
Loffing J, Zecevic M, Feraille E, Kaissling B, Asher C, Rossier BC, Firestone GL, Pearce D, and Verrey F. Aldosterone induces rapid apical translocation of ENaC in early portion of renal collecting system: possible role of SGK. Am J Physiol Renal Physiol 280: F675-F682, 2001.
9 T4 H) T2 a2 ?6 b# m0 E2 _9 P" B8 b( ]0 _) _* }( e

2 \4 u; F5 [, L+ F3 P
- b- \- _! U. D, c3 xMalik B, Schlanger L, Al-Khalili O, Bao HF, Yue G, Price SR, Mitch WE, and Eaton DC. ENaC degradation in A6 cells by the ubiquitin-proteosome proteolytic pathway. J Biol Chem 276: 12903-12910, 2001.8 J8 O% ?; \8 P
- m4 M) A$ ?' Q" s

2 A* ^: f, O/ d# B0 D) _2 E# `' N2 S% {: `/ l9 l& g1 g5 I% P  n7 l
Malik B, Yue Q, Yue G, Chen XJ, Price SR, Mitch WE, and Eaton DC. Role of Nedd4-2 and polyubiquitination in epithelial sodium channel degradation in untransfected renal A6 cells expressing endogenous ENaC subunits. Am J Physiol Renal Physiol 289: F107-F116, 2005.
, n' q3 u, R$ f  s$ @" ]0 I0 X: h9 ?+ F) }- \+ {* X* G8 C( L- q

  U5 T4 l: A0 d% T2 J5 e2 Q' g
Masaki T, Stambe C, Hill PA, Dowling J, Atkins RC, and Nikolic-Paterson DJ. Activation of the extracellular-signal regulated protein kinase pathway in human glomerulopathies. J Am Soc Nephrol 15: 1835-1843, 2004./ G& H6 K, ~) \+ ~( s# w
7 D& B& E% Z# M: J- d; G& V; K

1 U9 w' O' B; h) N
0 @4 j/ u# G! H+ Z7 x: T; RMasilamani S, Kim GH, Mitchell C, Wade JB, and Knepper MA. Aldosterone-mediated regulation of ENaC alpha, beta, and gamma subunit proteins in rat kidney. J Clin Invest 104: R19-R23, 1999.
! j+ K+ J( u9 ?0 Q6 Y' q
8 q0 I3 V# o) b6 w4 `8 d, _& X
$ p9 [+ x$ c9 }# h/ m5 y. D4 t
7 S1 a# O( v" I; j% F$ }  }McCormick JA, Feng Y, Dawson K, Behne MJ, Yu B, Wang J, Wyatt AW, Henke G, Grahammer F, Mauro TM, Lang F, and Pearce D. Targeted disruption of the protein kinase SGK3/CISK impairs postnatal hair follicle development. Mol Biol Cell 15: 4278-4288, 2004.
& w5 i, {; E6 u5 A
  L/ m5 C2 B. J7 t+ E' L
( ^8 C$ \3 j0 U0 Q4 _6 h& F8 p5 [* |: o3 K/ w, U
McDonald FJ, Yang B, Hrstka RF, Drummond HA, Tarr DE, McCray PB Jr, Stokes JB, Welsh MJ, and Williamson RA. Disruption of the beta subunit of the epithelial Na   channel in mice: hyperkalemia and neonatal death associated with a pseudohypoaldosteronism phenotype. Proc Natl Acad Sci USA 96: 1727-1731, 1999.
8 F1 @2 `" M: `3 L/ ?7 L# {5 m, |: K  X8 i" h
8 z- ^+ V5 h/ P4 L" y4 ~7 p
) a* _1 s" A) a- n9 Q$ D! v
Michlig S, Harris M, Loffing J, Rossier BC, and Firsov D. Progesterone down-regulates the open probability of the amiloride-sensitive epithelial sodium channel via a Nedd4-2-dependent mechanism. J Biol Chem 280: 38264-38270, 2005.
" d/ m+ C1 {8 x9 z) _4 e$ U2 c* S' e+ W, H
7 M( @# W3 y! S

4 i9 }6 D4 D6 E$ F+ R7 sMuller OG, Parnova RG, Centeno G, Rossier BC, Firsov D, and Horisberger JD. Mineralocorticoid effects in the kidney: correlation between alphaENaC, GILZ, and Sgk-1 mRNA expression and urinary excretion of Na   and K  . J Am Soc Nephrol 14: 1107-1115, 2003.
( S0 ?/ F' b! d+ L/ s+ F% Y# m) r  F1 w8 _; ?

  ]( U* o* c* q: n' ^0 t9 B+ Q9 k
2 A- G# S7 q5 i1 T* @) m- PMuslin AJ, Tanner JW, Allen PM, and Shaw AS. Interaction of 14-3-3 with signaling proteins is mediated by the recognition of phosphoserine. Cell 84: 889-897, 1996.
+ h! D: w; `. r: @' d6 [# e  X6 l1 y: e9 B7 n
9 G7 C0 A5 O, H) H; B6 H

) y) K% m" w0 P' W8 _Naray-Fejes-Toth A, Canessa C, Cleaveland ES, Aldrich G, and Fejes-Toth G. sgk Is an aldosterone-induced kinase in the renal collecting duct. Effects on epithelial Na   channels. J Biol Chem 274: 16973-16978, 1999.
9 a0 \5 n" K6 [) C: c1 |0 E
4 }1 R' m! r. X; D; D! [3 f
9 t/ @. V! u" u6 b" x1 p- p$ |" z
9 @: c. c3 h) \. w& _Naray-Fejes-Toth A, Helms MN, Stokes JB, and Fejes-Toth G. Regulation of sodium transport in mammalian collecting duct cells by aldosterone-induced kinase, SGK1: structure/function studies. Mol Cell Endocrinol 217: 197-202, 2004.4 C+ A) P- K- T; E' u! r
/ \! k/ D4 L% R* m  Y8 u0 y
8 i* k# Q3 W% @4 u) n# C0 |$ U$ T* l, w

( }/ m/ w* @$ E/ z; l5 s% R9 ^Nicod M, Michlig S, Flahaut M, Salinas M, Fowler Jaeger N, Horisberger JD, Rossier BC, and Firsov D. A novel vasopressin-induced transcript promotes MAP kinase activation and ENaC downregulation. EMBO J 21: 5109-5117, 2002.
% a" a" P$ S5 D$ e( K5 J
  V3 R, y0 @" G0 N! \" k( X+ k2 s( {5 r1 ]
. _2 P$ t9 O8 w. @0 Q' m
O'Kelly I, Butler MH, Zilberberg N, and Goldstein SA. Forward transport 14-3-3 binding overcomes retention in endoplasmic reticulum by dibasic signals. Cell 111: 577-588, 2002.2 q$ M" W7 \( s  D

) d3 q2 j1 r: [+ l
6 r4 m% o0 F$ @6 k: ~  o$ k: G! u
. q$ Y' X% \: ^Obsil T, Ghirlando R, Klein DC, Ganguly S, and Dyda F. Crystal structure of the 14-3-3zeta:serotonin N-acetyltransferase complex. A role for scaffolding in enzyme regulation. Cell 105: 257-267, 2001.: `) R; f8 t8 c% w- }

5 I9 [1 t  m1 j8 B/ @/ ~: L# x- }/ ?4 p$ T2 _

: P# X: w6 k6 J0 @# z7 o* LPark J, Leong ML, Buse P, Maiyar AC, Firestone GL, and Hemmings BA. Serum and glucocorticoid-inducible kinase (SGK) is a target of the PI 3-kinase-stimulated signaling pathway. EMBO J 18: 3024-3033, 1999.$ i; [2 I8 o, f* ^* o4 q
7 A4 C' Y) e! Z/ R
' a* ?2 I1 }' N& u% J- n- I$ `6 I
) E" k4 x5 j% b$ ?2 |& w6 i
Prince LS and Welsh MJ. Cell surface expression and biosynthesis of epithelial Na   channels. Biochem J 336: 705-710, 1998.
4 p! r: q5 `) G, @7 I0 e6 v! d7 b$ [1 n# H2 R
( T5 ?6 m* m  K6 m; ~  }6 u% B

8 v9 J/ d0 }3 s" ]Robert-Nicoud M, Flahaut M, Elalouf JM, Nicod M, Salinas M, Bens M, Doucet A, Wincker P, Artiguenave F, Horisberger JD, Vandewalle A, Rossier BC, and Firsov D. Transcriptome of a mouse kidney cortical collecting duct cell line: effects of aldosterone and vasopressin. Proc Natl Acad Sci USA 98: 2712-2716, 2001.
' |; i, g( {  L7 @$ m/ x' O3 j" \
/ D; t. G  L( ]" e4 ]
8 M4 x2 |$ E) _9 ]1 {
7 o! q, k* N( xRose B. Clinical Physiology of Acid-Base and Electrolyte Disorders. New York: McGraw-Hill, 1994, p. 916.) w9 r+ L6 s; j" D4 g

/ `* c. ~; g: h: F
$ d) u9 ^* f& T. x/ v1 j
1 f7 _( Q7 A# |- |* R+ h; ]6 C' J* CRossier BC. Negative regulators of sodium transport in the kidney: key factors in understanding salt-sensitive hypertension? J Clin Invest 111: 947-950, 2003.
1 F% R6 ?$ ]# ~( V7 t( P
& o- I3 G! X7 p. z# p5 f" t# }' w& Q

# w) L5 q$ |8 Y6 j6 z+ H3 {7 CShen JP and Cotton CU. Epidermal growth factor inhibits amiloride-sensitive sodium absorption in renal collecting duct cells. Am J Physiol Renal Physiol 284: F57-F64, 2003.$ y9 ^; o+ G( W7 K. B
7 z5 E% [1 f( e" S
' C* y- k5 E* s( {* m

2 ~$ m% `; U2 P/ f2 d; Z4 O) KShi H, Asher C, Chigaev A, Yung Y, Reuveny E, Seger R, and Garty H. Interactions of beta and gamma ENaC with Nedd4 can be facilitated by an ERK-mediated phosphorylation. J Biol Chem 277: 13539-13547, 2002.1 B) k. P' R8 w; T% J, b( a

% Z# I7 ^7 H" K1 u3 C$ A
- c! d$ g2 h3 K8 X9 b; i: F4 F3 y& w. J8 ^3 \
Snyder PM, Olson DR, Kabra R, Zhou R, and Steines JC. cAMP and serum and glucocorticoid-inducible kinase (SGK) regulate the epithelial Na   channel through convergent phosphorylation of Nedd4-2. J Biol Chem 279: 45753-45758, 2004.0 O+ L& `# g( \! O* ^8 F

1 @; R; x' S4 G" d" d
7 @: I- b7 N/ L+ J% F- C( K
& e4 Y5 x/ f$ }3 ~, K% ~! A! }Snyder PM, Olson DR, and Thomas BC. Serum and glucocorticoid-regulated kinase modulates Nedd4-2-mediated inhibition of the epithelial Na   channel. J Biol Chem 277: 5-8, 2002.
. M5 N% \; g% G1 o4 S. V) t8 I$ ~1 ^: p  l1 T* b* \

( R% o- y+ ]4 v
; t  }' e1 J" vSnyder PM, Steines JC, and Olson DR. Relative contribution of Nedd4 and Nedd4-2 to ENaC regulation in epithelia determined by RNA interference. J Biol Chem 279: 5042-5046, 2004.
7 D) P& \4 y! W0 a# \+ N
; X8 q/ Z. t# f% S* B# L
7 Z; x3 \( d' Z4 s+ H. L0 E5 _0 Y% d# D, W
Soundararajan R, Zhang TT, Wang J, Vandewalle A, and Pearce D. A novel role for glucocorticoid-induced leucine zipper protein in epithelial sodium channel-mediated sodium transport. J Biol Chem 280: 39970-39981, 2005.
3 I6 S% I# ]6 a  b8 E, V% P& ]# G6 P' ]( w' V  ^3 D

- J1 f; T) _: i0 p3 K4 R* ^
; H, k) D% E0 G0 Y8 WStaruschenko A, Patel P, Tong Q, Medina JL, and Stockand JD. Ras activates the epithelial Na   channel through phosphoinositide 3-OH kinase signaling. J Biol Chem 279: 37771-37778, 2004." a6 l( D' B; m0 ]

* V/ L+ E) M* {. A) u" L
* j/ R+ i, u% r2 O
  z5 J, w5 Q$ T% H( s! q- ]) nStaruschenko A, Pochynyuk OM, Tong Q, and Stockand JD. Ras couples phosphoinositide 3-OH kinase to the epithelial Na   channel. Biochim Biophys Acta 1669: 108-115, 2005.
9 M( o, W& }; }- V+ v9 D* D) }) a3 J' p( E
' q3 `# p7 ~+ q
: `7 Q8 \4 n9 p- d! e; V5 R
Staub O, Dho S, Henry P, Correa J, Ishikawa T, McGlade J, and Rotin D. WW domains of Nedd4 bind to the proline-rich PY motifs in the epithelial Na   channel deleted in Liddle's syndrome. EMBO J 15: 2371-2380, 1996.
- n+ n, c3 S9 P. H7 B7 `% K6 X$ v7 k; N

* ^1 g, P6 u5 ~9 P* P
+ o) X( T: T" i& d6 u% BStaub O, Gautschi I, Ishikawa T, Breitschopf K, Ciechanover A, Schild L, and Rotin D. Regulation of stability and function of the epithelial Na   channel (ENaC) by ubiquitination. EMBO J 16: 6325-6336, 1997.
4 M  ^% l0 M1 ~3 M! R: ]: r& r# w8 @! `8 u) M) ^1 G

; W: p0 v+ w0 _: ~& g, h: o# j1 B( M. E5 e+ X# x
Tobin MD, Raleigh SM, Newhouse S, Braund P, Bodycote C, Ogleby J, Cross D, Gracey J, Hayes S, Smith T, Ridge C, Caulfield M, Sheehan NA, Munroe PB, Burton PR, and Samani NJ. Association of WNK1 gene polymorphisms and haplotypes with ambulatory blood pressure in the general population. Circulation 112: 3423-3429, 2005.
, q& o" f7 {& |8 w9 u- w7 p9 a1 y) P6 |9 ^" c  ~
9 n8 @0 z8 k' B( |' Z2 |# r
1 }% l/ b! n3 m
Tong Q, Gamper N, Medina JL, Shapiro MS, and Stockand JD. Direct activation of the epithelial Na   channel by phosphatidylinositol 3,4,5-trisphosphate and phosphatidylinositol 3,4-bisphosphate produced by phosphoinositide 3-OH kinase. J Biol Chem 279: 22654-22663, 2004.
! h( T& q  ^" }6 X8 y# M/ L
: J( _5 S- D% U% e1 M1 h- W. R* Y, v8 [) m
' u1 ^7 s8 p! k1 j" e' U
Tzivion G and Avruch J. 14-3-3 Proteins: active cofactors in cellular regulation by serine/threonine phosphorylation. J Biol Chem 277: 3061-3064, 2002.$ D" u/ c# ?- h( b

9 t8 }3 \% \& T' F
2 h( w# b( Z$ N6 P5 l) \) {  E8 T! P, G+ ^: \0 M( f
Vallet V, Pfister C, Loffing J, and Rossier BC. Cell-surface expression of the channel activating protease xCAP-1 is required for activation of ENaC in the Xenopus oocyte. J Am Soc Nephrol 13: 588-594, 2002.7 c/ k9 ?2 s2 ~! `; y
# r( l6 ]4 |4 Q2 d$ I

, A  j( _( q2 r  o5 j+ A2 R8 y% f& {; F( L8 M' J) o7 k) T
Vecchione A, Marchese A, Henry P, Rotin D, and Morrione A. The Grb10/Nedd4 complex regulates ligand-induced ubiquitination and stability of the insulin-like growth factor I receptor. Mol Cell Biol 23: 3363-3372, 2003." _( M( E% z. W: g$ A

0 t2 X6 N; V! ]6 q" m7 E, h, n7 L% U6 E' ?1 S, x

* V# `" |- a: H# PVerrey F. Early aldosterone action: toward filling the gap between transcription and transport. Am J Physiol Renal Physiol 277: F319-F327, 1999.1 n0 @) E' t5 g/ _- N6 u

( b- G( S9 k# s
% O% J( F3 s' z/ F  \3 Q" \" X- z/ r1 L3 O$ z! e  [
Von Wowern F, Berglund G, Carlson J, Mansson H, Hedblad B, and Melander O. Genetic variance of SGK-1 is associated with blood pressure, blood pressure change over time and strength of the insulin-diastolic blood pressure relationship. Kidney Int 68: 2164-2172, 2005.
% Q1 O, B1 Q8 l7 b" i
3 f7 i) @& @5 Y$ i8 |, f
& L4 G7 G* T2 ~7 B2 A  a$ `4 o! s5 e, X0 w
Waldegger S, Barth P, Raber G, and Lang F. Cloning and characterization of a putative human serine/threonine protein kinase transcriptionally modified during anisotonic and isotonic alterations of cell volume. Proc Natl Acad Sci USA 94: 4440-4445, 1997.
  a# j- j3 }( ]( j( @7 i$ ^: r8 M6 J- }  G8 q0 m6 H
1 ^, N) W  N& K7 w3 T, u' S5 i
3 [" c* b' v5 l4 {
Wang J, Barbry P, Maiyar AC, Rozansky DJ, Bhargava A, Leong M, Firestone GL, and Pearce D. SGK integrates insulin and mineralocorticoid regulation of epithelial sodium transport. Am J Physiol Renal Physiol 280: F303-F313, 2001.
' R+ h0 u. K8 n" N
: O' C! c$ A  [6 P* x0 G- {2 B4 n# h2 p6 {$ U3 r; L1 L0 i
: L7 a2 m2 M4 e( E' h+ _3 N
Webster MK, Goya L, Ge Y, Maiyar AC, and Firestone GL. Characterization of sgk, a novel member of the serine/threonine protein kinase gene family which is transcriptionally induced by glucocorticoids and serum. Mol Cell Biol 13: 2031-2040, 1993.
3 K, |) O0 T( I3 o$ P8 W: q1 `9 @$ l- B1 a* L( q
/ O) R* I& f7 P9 O: B. ?7 x9 V
) [7 L  H. G6 \8 i& k
Wilson FH, Disse-Nicodeme S, Choate KA, Ishikawa K, Nelson-Williams C, Desitter I, Gunel M, Milford DV, Lipkin GW, Achard JM, Feely MP, Dussol B, Berland Y, Unwin RJ, Mayan H, Simon DB, Farfel Z, Jeunemaitre X, and Lifton RP. Human hypertension caused by mutations in WNK kinases. Science 293: 1107-1112, 2001.' J+ `$ d+ x, V) J( _
  J- P2 m. `+ O5 V. o

9 ?) ?6 `. P) p3 B+ g$ D4 z( k
0 }7 W2 U8 W8 V$ qWulff P, Vallon V, Huang DY, Volkl H, Yu F, Richter K, Jansen M, Schlunz M, Klingel K, Loffing J, Kauselmann G, Bosl MR, Lang F, and Kuhl D. Impaired renal Na   retention in the sgk1-knockout mouse. J Clin Invest 110: 1263-1268, 2002.: [- k; v" u% M$ h  \, f7 ?

" ]; d/ X* ?, Z+ t/ c4 [/ p' |& C, q3 q" M8 y
) l* i" c6 M- h' B
Xu BE, Stippec S, Chu PY, Lazrak A, Li XJ, Lee BH, English JM, Ortega B, Huang CL, and Cobb MH. WNK1 activates SGK1 to regulate the epithelial sodium channel. Proc Natl Acad Sci USA 102: 10315-10320, 2005.% c+ t$ Z* c* A
6 |& y4 U0 C3 D2 [/ T9 L5 i7 g

4 e) F- n5 U) ?. y4 C: L& X+ t2 i  O, B0 p  @! \
Xu BE, Stippec S, Lazrak A, Huang CL, and Cobb MH. WNK1 activates SGK1 by a phosphatidylinositol 3-kinase-dependent and non-catalytic mechanism. J Biol Chem 280: 34218-34223, 2005.
, e/ A" j1 q( `: q# _$ d
8 f4 Q) T' r& x, e1 q" C4 C+ [& E; w. p) \0 r
0 ~0 E) x4 `4 }% q! u' b
Zentner MD, Lin HH, Wen X, Kim KJ, and Ann DK. The amiloride-sensitive epithelial sodium channel alpha-subunit is transcriptionally down-regulated in rat parotid cells by the extracellular signal-regulated protein kinase pathway. J Biol Chem 273: 30770-30776, 1998.6 c  J+ x% e0 w; h/ _

: Y2 N$ g) [) u* G5 d; w0 O+ k9 t, r3 I, I2 t+ ?% F9 E
8 k! _- A5 m: F. o, I; b9 X- {
Zhou R and Snyder PM. Nedd4-2 phosphorylation induces serum and glucocorticoid-regulated kinase (SGK) ubiquitination and degradation. J Biol Chem 280: 4518-4523, 2005.

Rank: 2

积分
132 
威望
132  
包包
1727  
沙发
发表于 2015-5-23 09:18 |只看该作者
免疫细胞疗法治疗肿瘤有效  

Rank: 2

积分
98 
威望
98  
包包
1756  
藤椅
发表于 2015-6-3 08:10 |只看该作者
支持你加分  

Rank: 2

积分
163 
威望
163  
包包
1852  
板凳
发表于 2015-6-5 13:28 |只看该作者
干细胞之家微信公众号
不要等到人人都说你丑时才发现自己真的丑。  

Rank: 2

积分
68 
威望
68  
包包
1752  
报纸
发表于 2015-7-1 22:42 |只看该作者
干细胞研究非常有前途

Rank: 2

积分
77 
威望
77  
包包
1964  
地板
发表于 2015-8-5 09:10 |只看该作者
加油啊!!!!顶哦!!!!!  

Rank: 2

积分
79 
威望
79  
包包
1769  
7
发表于 2015-8-11 10:43 |只看该作者
…没我说话的余地…飘走  

Rank: 2

积分
88 
威望
88  
包包
1897  
8
发表于 2015-8-15 18:08 |只看该作者
慢慢来,呵呵  

Rank: 2

积分
162 
威望
162  
包包
1724  
9
发表于 2015-9-3 18:22 |只看该作者
加油啊!!!!顶哦!!!!!  

Rank: 2

积分
64 
威望
64  
包包
1734  
10
发表于 2015-9-21 15:09 |只看该作者
顶一个先  
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册
验证问答 换一个

Archiver|干细胞之家 ( 吉ICP备2021004615号-3 )

GMT+8, 2024-5-18 08:32

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.