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: 550147|回复: 257
go

Regulation of the epithelial Na channel (ENaC) by phosphatidylinositides [复制链接]

Rank: 1

积分
威望
0  
包包
0  
楼主
发表于 2009-4-22 08:43 |只看该作者 |倒序浏览 |打印
作者:Oleh Pochynyuk, Qiusheng Tong, Alexander Staruschenko, He-Ping Ma, and James D. Stockand作者单位:1 Department of Physiology, University of Texas Health Science Center, San Antonio, Texas; and 2 Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 9 M/ d% Z2 B- A& Q3 u
                  
4 c1 A& o1 E0 X1 C" A                    J: l# H1 ~: |# S% p( V
         
/ L( h: P3 J6 @% {* X* S7 n                         7 d  |) x0 j% `' {
            9 M3 U4 v, d* f
            
; b0 t! N* Y  D3 [1 c4 C            
$ G7 G% A% N" _            0 ?" E/ ?- n. [0 N8 V
                      ' l4 I. v* R0 H, Q. O% _
        
, h7 x1 y- _' M& G        
% [( N' }8 |8 j+ N9 W        
) o2 V+ F2 v& _% I" F          【摘要】
7 F+ ?% V) g1 c! z6 w& b0 C      The epithelial Na   channel (ENaC) is an end-effector of diverse cellular signaling cascades, including those with phosphatidylinositide second messengers. Recent evidence also suggests that in some instances, phospatidylinositides can directly interact with ENaC to increase channel activity by increasing channel open probability and/or membrane localization. We review here findings relevant to regulation of ENaC by phosphatidylinositol 4,5- bis phosphate (PIP 2 ) and phosphatidylinositol 3,4,5-triphosphate (PIP 3 ). Similar to its actions on other ion channels, PIP 2 is permissive for ENaC openings having a direct effect on gating. The PIP 2 binding site in ENaC involved in this regulation is most likely localized to the NH 2 terminus of -ENaC. PIP 3 also affects ENaC gating but, rather than being permissive, augments open probability. The PIP 3 binding site in ENaC involved in this regulation is localized to the proximal region of the COOH terminus of -ENaC just following the second transmembrane domain. In complementary pathways, PIP 3 also impacts ENaC membrane levels through both direct actions on the channel and via a signaling cascade involving phosphoinositide 3-OH kinase (PI3-K) and the aldosterone-induced gene product serum and glucocorticoid-inducible kinase. The putative PIP 3 binding site in ENaC involved in direct regulation of channel membrane levels has not yet been identified.2 `3 w3 q9 z% ^& Y* K. W
: m: ]7 M) J; R
5 b$ Z5 x/ g% [# A0 r
! C' c( X: Q7 |" f! _" p9 P
phosphatidylinositol 4,5- bis phosphate; phosphatidylinositol 3,4,5-triphosphate; receptor tyrosine kinase; insulin G protein-coupled receptor* q# ~3 N6 m4 c7 G# `
3 G: o2 E; A% q& |0 v: m2 @8 J

1 L% |7 Y* C" Z. ]6 E5 v/ M; d0 {7 c+ v& k: K% [
ION CHANNELS PLAY CRITICAL roles in every aspect of physiology. Moreover, ion channels have long been recognized to be important end-effectors of diverse cellular signaling cascades, including those having phosphatidylinositide second messengers. Direct regulation of ion channel activity by phosphatidylinositide-signaling molecules, in addition, is now becoming widely appreciated (reviewed in Ref. 33 ). This mechanism for ion channel modulation is recognized to be physiologically important, for its disruption, in some instances, leads to disease (e.g., Bartter's and Andersen's syndromes) ( 16, 43, 56, 65 ). Diverse types of ion channels are directly modulated by phosphatidylinositides ( 3, 17, 42, 58, 74, 84, 96, 97 ). Phosphatidylinositides, such as phosphatidylinositol 4,5- bis phosphate (PIP 2 ) and phosphatidylinositol 3,4,5-triphosphate (PIP 3 ), directly interact with these channels to modulate gating ( 58, 93, 97 ). Several recent studies identify the epithelial Na   channel (ENaC) as being a channel sensitive to direct phosphatidylinositide regulation.8 c( w3 ^' j! s) h, }, y# p# V

. E. N, D  Y7 Z# Q6 n2 F- h' S1 e( N$ q4 m) [
  h" O* I3 q9 s) }+ Z
ENaC is a heteromeric channel composed of three distinct but similar subunits:,, and ( 8, 9 ). All ENaC subunits have NH 2 - and COOH-terminal cytosolic domains separated by two transmembrane domains and a large extracellular region. ENaC serves an essential physiological function, for its activity is limiting for Na   absorption across many epithelia, including that in the distal renal nephron (reviewed in Refs. 27, 37, and 69 ). Thus ENaC is well positioned to influence systemic Na   balance and blood pressure. Indeed, gain and loss of ENaC function lead to inheritable forms of hypertension and hypotension, respectively, associated with inappropriate salt conservation and wasting at the kidney ( 35, 41 ). In addition, ENaC plays a critical role in hydration and fluid reabsorption across many mucosal membranes. Deletion and overexpression of ENaC correspondingly lead to excessively wet and dry air spaces, with associated disease ( 35, 46 ).
' g0 q6 P: G% Y; b$ i/ `7 Q. l8 V6 M  [

4 B  L5 u7 ]% [8 @4 E; ]" u5 L6 O/ p1 V5 M; f5 o$ B3 H0 m. p, S! A& t
Here, we summarize recent findings and current thinking regarding phosphatidylinositide regulation of this important ion channel. Both the regulation of ENaC by signaling cascades utilizing PIP 3 as a second messenger and the direct effects of PIP 2 and PIP 3 on ENaC are discussed. Where possible, the physiological implications of recent findings are addressed.
) w: c* c) w: d- ?) y( e& C          【关键词】 Regulation epithelial phosphatidylinositides
8 d6 J8 K3 J! I& \' ]+ l% n                  REGULATION OF ENaC BY PIP 2
; J0 ~% C1 Y+ u: p/ c+ @. `9 }
# w2 g! {+ x8 \/ V; f- k5 r  VPIP 2 Increases ENaC Open Probability3 l0 A$ M) h9 A: P4 e! w: U  G

) A3 {! g  g$ v2 Y& s, E/ iMost ion channels sensitive to PIP 2, including the first channel identified to be sensitive to this signaling molecule, K ATP (Kir6.2/SUR2) ( 32 ), have a continuing decrease in activity when excised from the cell membrane in an inside-out patch configuration (reviewed in Refs. 33 and 85 ). This hallmark is referred to as channel "run-down." In excised, inside-out patches, ENaC shows run-down typical to PIP 2 -sensitive channels ( 44, 95 ).
3 j1 |6 N: g" L( X- u! g% x0 |3 M% D7 x
Ma and colleagues ( 44 ) provided the first clear evidence that ENaC is directly regulated by phosphatidylinositides. This group demonstrated that scavenging PIP 2 with a specific antibody and poly- L -lysine accelerated ENaC run-down in excised inside-out patches made from the amphibian distal tubule A6 cell line. Similarly, increased PIP 2 hydrolysis in response to activation of endogenous PLC via signaling from purinergic receptors and addition of exogenous PLC accelerated ENaC run-down. Run-down was markedly slowed by addition of PIP 2 to the cytosolic bath solution bathing the intracellular face of the channel. The primary effect of PIP 2 on ENaC was not related to changes in the membrane levels of this channel within 30 min. Thus the authors concluded that acute regulation of ENaC by PIP 2 is independent of channel trafficking. This work suggests that PIP 2 augments ENaC open probability through direct interactions with intracellular/membrane portions of the channel or through interactions with a phosphatidylinositide-sensitive, intracellular regulator of the channel.- P9 q3 H( T, V3 N( v8 p3 k
, }6 w2 i. F0 O  V& y, S* H
Several investigators have subsequently confirmed possible direct effects of PIP 2 on ENaC in excised, inside-out patches ( 39, 88, 95 ). Figure 1 shows a representative finding demonstrating PIP 2 activation of ENaC in an excised, inside-out patch (Tong Q and Stockand JD, unpublished observations).! n7 c  |! |1 l. u/ o

2 _- F  ^/ h! {" I8 L; v; A% f( qFig. 1. Phosphatidylinositol 4,5- bis phosphate (PIP 2 ) activates epithelial Na   channel (ENaC). These current traces are from an excised, inside-out patch made from Chinese hamster ovary (CHO) cells expressing human (h)ENaC. The patched membrane was clamped to 0 mV. Inward Na   currents are downward, with the dashed lines denoting the closed state. The intracellular bath and extracellular pipette solutions contained 5 and 145 mM Na  , respectively, with symmetrical Cl -. The bath also contained 0.3 mM GTP. Over the course of this experiment, ENaC activity ran down and was then reactivated with addition of 30 µM PIP 2 to the bath solution (denoted by 1st arrow). ENaC activity was subsequently decreased on addition of the PIP 2 scavenger poly- L -lysine (20 µg/ml; denoted by 2nd arrow). The complete experiment is shown in the top trace, with the middle and bottom traces showing the areas under the gray bars before and after PIP 2 addition at expanded time and amplitude scales.
2 k, t: H- A  }# ~7 s2 W6 q7 ^. k8 u, N
& j" i# K/ f7 B# x- y/ N3 }An interesting extension to the original finding by Ma and colleagues ( 44 ) was made by Yue and colleagues ( 95 ). This group demonstrated that PIP 2 had only modest effects on ENaC in excised, inside-out patches from A6 cells compared with the effects of this phosphatidylinositide when added in combination with GTP. The primary action of PIP 2  GTP was to reverse run-down by increasing channel open probability to levels approaching starting values.* z' j: N' G- k$ R# c6 b
+ z; m3 a3 k1 [% K& W% k, Y0 s! m
Addition of activated G i-3 and G protein -subunits, in the study by Yue and coauthors, mimicked and reversed the actions of GTP, respectively, implicating involvement of a trimeric G protein in a PIP 2 response. We have reported a similar finding where dialysis of intracellular GTP with GDP S quickly decreases activity of mouse and human ENaC heterologously expressed in Chinese hamster ovary (CHO) cells in the absence and presence of activated phosphoinositide 3-OH kinase (PI3-K) ( 57 ). PI3-K activates ENaC via PIP 3 signaling (see below). We attribute the action of GDP S to inhibition of either a Ras or Rho small G protein ( 57, 77 - 79 ). Thus G proteins in a yet to be fully appreciated manner play a critical role in how ENaC responds to phosphatidylinositides.
& f1 U2 d; g; Q, P, k1 h
' K* s3 E3 L- C! r0 HThere is precedence for interactions between G proteins and phosphatidylinositide regulation of ion channels. For G protein-activated, inwardly rectifying K   channel (GIRK), it is probable that PIP 2 and G protein -subunits bind to the COOH terminus of the channel in close proximity ( 62, 73 ). The closeness of these binding sites provides a platform for PIP 2 and G protein -subunits to interact with respect to regulating the channel. It currently is not clear whether G proteins directly interact with ENaC; however, PIP 2 does (see below).3 U/ t4 l# o( S

- I* d6 w, p. S6 ^Two recent studies investigated regulation of ENaC by PIP 2 using experimental approaches not complicated by channel run-down. In the first, Kunzelmann and colleagues ( 39 ) showed that inhibition in response to purinergic receptor signaling of Na   absorption mediated by ENaC across tracheal epithelia and M1 collecting duct cell monolayers was suppressed by scavenging PIP 2. Recovery of inhibition in response to purinergic receptor signaling, moreover, was abolished by blocking PI4-K and diacylglycerol kinase. Both PI4-K and diacylglycerol kinase are involved in PIP 2 synthesis ( 24 ).5 B& f2 S; T# E! x5 w" \
, r' {* F" g6 A# U3 V# _
We recently demonstrated that ENaC activity in whole cell and excised, outside-out patch configurations decreases on depletion of membrane PIP 2 in response to activation of receptor tyrosine kinase signaling and signaling via a classic G q/11 -coupled seven-transmembrane-spanning receptor ( 88 ). In contrast to the excised, inside-out patch configuration, ENaC does not run down in either of these latter patch configurations ( 6, 79, 87, 88 ). PIP 2 levels were likely depleted by receptor tyrosine kinase- and G q/11 -coupled signaling via PLC- and -, respectively. In excised, outside-out patches, depletion of PIP 2 decreased ENaC activity by decreasing open probability. As expected, buffering PIP 2 to prevent dynamic changes in the levels of this phosphatidylinositide countered decreases in ENaC activity in response to receptor tyrosine kinase signaling. Thus the response of ENaC to PIP 2 depletion mediated by receptor tyrosine kinase-PLC- and G q/11 -PLC- parallels regulation of tryptophan (TRP), P/Q-, and N-type Ca 2  and GIRK channels by receptor tyrosine kinases ( 13, 38, 58, 92 ), and KCNQ and N-type Ca 2  channels by G q/11 -coupled receptors ( 26, 40, 85, 92 ).+ T7 \1 h) O: i# B
/ _0 @+ |; L# V1 I- \( ~$ D& _
We propose that the effects of PIP 2 on ENaC open probability are permissive rather then regulatory. Our rationale is that ENaC quickly runs down in settings such as excised inside-out patches and activation of PLC, where membrane PIP 2 levels rapidly decline in response to uninhibited hydrolysis and/or washout ( 39, 44, 47, 88, 95 ). In contrast, in settings, such as whole cell and excised, outside-out patch configurations, where PIP 2 levels are expected to be relatively constant, ENaC does not run down ( 6, 79, 87, 88 ). Moreover, ENaC activity (and open probability) never exceeds control levels in response to addition of exogenous PIP 2 but rather approaches that observed before run-down. A more succinct way of stating this is that loss of ENaC activity parallels declines in PIP 2 levels.
, O0 _/ a# W! n/ Z
/ n; \! h, X, _% i3 l8 T; z2 o8 yA finding that elevations of membrane PIP 2 levels increase ENaC activity above control values would distinguish whether this phosphatidylinositide is permissive or regulatory. To date, all experiments reporting PIP 2 actions on ENaC open probability have demonstrated a close link between decreases in open probability and declining PIP 2 levels. The converse of increasing (above starting levels) open probability linked to elevations in PIP 2 levels has not been established. Indeed, increasing cellular PIP 2 levels by direct addition of a permeant form of this phosphatidylinositide failed to increase Na   absorption mediated by ENaC in renal epithelia ( 47 ). Moreover, chronic overexpression of PI( 4 )5-K, which is a phospholipid kinase that promotes PIP 2 synthesis, increased membrane levels of ENaC heterologously expressed in CHO cells rather than the channel's open probability ( 77 ) (see below). Thus we propose that active ENaC constitutively interacts with membrane PIP 2, with this interaction being permissive for normal channel gating. Loss of PIP 2 then would decrease open probability, but increases in PIP 2 might not be expected to affect open probability if membrane levels of this phosphatidylinositide with respect to actions on ENaC are saturated at rest. Supporting such a mechanism is the fact that PIP 2 is the most abundant phosphatidylinositide in the membrane, exceeding the levels of PIP 3 by up to 100-fold ( 4, 24, 52 ).
( e! o+ @" A# r% o- {3 K) f  \# q0 ^7 Q, V2 m0 R
PIP 2 Directly Interacts with ENaC
  @; g- {2 A: A$ d2 C
( G: O3 H$ B1 u6 _# zTwo studies have begun to delineate the PIP 2 binding site in ENaC that impacts channel open probability ( 39, 95 ). Both suggest that PIP 2 binds to a region in -ENaC found in the cytosolic NH 2 terminus of this subunit. As shown in Fig. 2 A, this region of the channel contains several well-conserved, positively charged arginines and lysines. In this regard, then, the putative PIP 2 binding site in ENaC is similar to that proposed for Kir and TRP channels, which all have an amino acid track containing several well-conserved, positively charged residues that potentially facilitate electrostatic interactions with the polar head groups of the phosphatidylinositides in the inner leaflet of the plasma membrane ( 17, 58, 66, 73, 97 ). Mutation of this domain in -ENaC reduces ENaC activity without affecting surface expression ( 39 ).
( \( Q8 f$ D8 T% l& Y$ J, @3 [$ ]3 y9 Z+ Q% J) j+ Z
Fig. 2. PIP 2 and phosphatidylinositol 3,4,5-triphosphate (PIP 3 ) binding sites in ENaC. Shown are sequence alignments of the putative PIP 2 ( A ) and PIP 3 ( B ) binding sites in - and -ENaC, respectively. Amino acid number is for mouse (m)ENaC. ch, Chimp; r, rat; ra, rabbit; c, dog; x, Xenopus laevis.- g& ^( l6 j* H
+ d& R+ l( K* {; O) B  q0 n
PIP 2 Also Affects Membrane Levels of ENaC
2 h+ u3 n3 ^* T% ^
- w5 Q2 a# D! ~' _4 R9 qVesicle movement and membrane targeting in eukaryotic cells are intimately tied to PIP 2 ( 14, 19, 67 ). Small G proteins in the Rho family increase ENaC activity when they are overexpressed together in CHO cells ( 77 ). Rho increases channel activity by increasing the membrane levels of ENaC. PI( 4 )5-K is a downstream effector of Rho and Rho-kinase, the Rho effector ( 54, 91, 94 ). Overexpression of activated Rho-kinase and PI( 4 )5-K, similar to Rho, increases ENaC activity. Inhibition of Rho-kinase and PI4-K disrupts the effect of Rho on ENaC. Thus in CHO cells at least, Rho increases ENaC activity by increasing the membrane levels of this channel via a signaling cascade involving Rho-kinase, PI( 4 )5-K, and its product, PIP 2. We speculate that a sustained increase in PIP 2 levels increases membrane ENaC through a mechanism independent of direct phosphatidylinositide binding to the channel. This mechanism likely is a more generalized trafficking response to chronic changes in PIP 2. The physiological importance of PIP 2 regulation of ENaC membrane levels via a trafficking response in epithelial cells has not yet been determined.
! b$ q; W$ q8 j- w' X: Y+ Y/ e
3 O6 D/ M' L* n' E! f- ]! ^4 oIn summary, decreases in ENaC open probability parallel decreases in PIP 2 levels. In contrast, increases in ENaC membrane levels parallel increases in PIP 2 levels. Thus, as shown in the flow chart in Fig. 3, PIP 2 may have multiple actions on ENaC to control channel activity.
- f* c" _$ |) R4 w7 `- s% K" }+ L1 |( |0 F  ?
Fig. 3. PIP 2 and PIP 3 have both direct and indirect effects on ENaC open probability and membrane number. The flow chart shown here describes the effects of PIP 2 and PIP 3 on ENaC open probability ( P o ) and membrane levels ( N ). Up and down arrows indicate increases and decreases in phosphatidylinositide levels, and filled and dashed arrows indicate responses resulting from direct interaction of the phosphatidylinositide with the channel and those resulting from a signaling cascade that utilizes the phosphatidylinositide as a second messenger, respectively.
) K* t9 t1 s* s
8 _& o2 C1 m& ]* J8 L; ]" E5 q0 oREGULATION OF ENaC BY PIP 3: a( C2 m) ~0 `$ N

1 _- f: [/ Y4 a: m  z! z- [6 u' _PIP 3 is a Second Messenger of an Aldosterone-Sensitive Cellular Signaling Cascade that Impacts ENaC Activity. }# K% v: Z5 e# C

. N0 r! i  e* d3 R. Q  N8 x5 j* lENaC is a critical end-effector of the renin-angiotensin-aldosterone system involved in negative-feedback control of blood pressure. Aldosterone increases ENaC activity in epithelia (reviewed in Refs. 27, 41, 82, and 89 ). Aldosterone increases both the number of ENaC in the plasma membrane and the open probability of the channel to increase the number of active channels in the membrane. Paunescu and colleagues ( 55 ) demonstrated that aldosterone activates PI3-K to increase PIP 3 levels in renal epithelia and that active PI3-K is required for aldosterone actions on Na   transport mediated by ENaC. Several laboratories have subsequently confirmed the observations that aldosterone activates PI3-K in renal epithelia and that active PI3-K is necessary for increases in Na   transport in response to aldosterone ( 22, 86, 90 ). Two other natriferic factors, insulin and antidiuretic hormone (ADH), also activate PI3-K in epithelia and increase Na   absorption mediated by ENaC in a PI3-K-sensitive manner ( 20, 47, 48, 59 ). Importantly, insulin, while targeting basolateral receptors, quickly increases apical membrane PIP 3 levels to affect activity of ENaC in this latter membrane ( 5 ). Active PI3-K also plays a role in setting basal ENaC activity in the absence of hormone stimulation ( 4, 55, 59 ). These findings suggest that active PI3-K and its phospholipid products are either permissive for ENaC activity or involved in regulation of both basal and hormone-sensitive ENaC activity. As expected, overexpression of active PI3-K with ENaC in CHO cells increased channel activity ( 79, 86, 87 ). Similarly, addition of membrane-permeant PIP 3 increased Na   absorption across renal epithelia, and overexpression of a PIP 3 -phosphatase, phosphatase and TENsin homolog deleted on chromosome 10, attenuated the natriferic effects of insulin and exogenous PIP 3 ( 47 ). Activation of PI3-K, moreover, with H 2 O 2 provoked a rise in Na   transport ( 48 ). Thus PI3-K activity is intimately tied to ENaC activity.5 Y% b% D# o( P2 ]% y5 q/ R

& s+ V5 n& r- W( A6 e0 _- W0 }The mechanism by which aldosterone activates PI3-K in epithelia remains uncertain. We ( 31, 83 ) and others ( 49, 75, 76 ) demonstrated that in renal epithelia, aldosterone via control of gene expression increases the levels and activity of the small G protein K-Ras. PI3-K is a well-documented first effector of Ras ( 2, 60 ). In a recent publication ( 86 ), we argued that activation of PI3-K was a consequence of increased K-Ras expression in response to aldosterone.
. b. w0 l4 H8 F" N: j5 H- g# a3 b; e2 h  q1 T" @
Active PI3-K has been shown to modulate ENaC activity through, at least, three mechanisms (see Fig. 3 ). Two involve increases in the membrane levels of the channel, and the other involves increases in the open probability of the channel. As described further below, the product of PI3-K, PIP 3, affects ENaC membrane levels by both a direct mechanism involving physical association of the phosphatidylinositide with the channel ( 30 ) and an indirect mechanism involving a signaling cascade that impinges on channel retrieval ( 15, 70 ). Regulation of ENaC open probability by PI3-K is a direct consequence of the physical association of PIP 3 with the channel ( 57, 87 ).) X2 Y0 z+ I1 z3 o3 `& ^* z
$ z/ Q) p0 z( E' w) G4 G, o+ G  J9 Z
PIP 3 Signaling Impacts ENaC Membrane Levels by Suppressing Channel Retrieval
5 L' ^( z9 b  b. w0 W! k8 Z; W1 E9 [) V
The most studied and thus best-documented mechanism by which PI3-K signaling impacts ENaC activity involves suppression of channel retrieval by downstream effectors of this kinase. This mechanism is the focus of many excellent reviews ( 36, 63, 68, 69 ) and thus is covered only briefly here.2 A7 v, s$ o6 b) I1 u% `, e
, J. Q. @* i) f/ Q+ ~
Membrane levels of ENaC are set, in part, by mediated channel retrieval. Ubiquitin ligases in the Nedd4 family interact with the conserved PY motifs in the cytosolic COOH-terminal domains of ENaC subunits ( 1, 29, 72, 80, 81 ). This interaction facilitates ubiquitinylation of the channel, targeting it for internalization and subsequent degradation. Sgk is a serine/threonine kinase that is regulated at the level of transcription by corticosteroids, including aldosterone ( 10, 11, 51 ). This kinase also contains a PY motif and physically interacts with Nedd4 ubiquitin ligases ( 15, 70, 98 ). Association of Sgk with Nedd4 ubiquitin ligases leads to phosphorylation of the latter. This compromises the ability of Nedd4 ubiquitin ligases to bind and regulate ENaC. Thus activation of Sgk promotes retention of ENaC in the membrane. Sgk is a downstream effector of PI3-K sensitive to both PIP 3 levels and phosphorylation by another PI3-K effector, PDK1 ( 22, 86, 90 ). Activation of PI3-K promotes activation of Sgk and subsequent protection of membrane ENaC levels. Aldosterone likely increases both Sgk levels and activity, with the former being a transcriptional event and the latter a signaling event in response to prior activation of PI3-K. The physiological importance of this PI3-K-sensitive mechanism controlling ENaC membrane levels is clear. Disruption of the PY motifs in ENaC subunits leads to gain-of-function mutations associated with increased channel activity, improper Na   handling by the kidney and hypertension in humans ( 1, 25, 72, 80, 81 ). Mutation of the genes encoding Nedd4 and Sgk, moreover, has recently been linked to blood pressure imbalances and inappropriate renal electrolyte handling in humans ( 7, 23 ).
6 }% R' I# {, ?+ W% r6 U: X8 v* h, n9 b- e, s- w6 c
Blazer-Yost and colleagues ( 5 ) recently proposed an interesting mechanism for insulin modulation of Na   reabsorption in renal epithelia cells that has many parallels to regulation of ENaC by aldosterone. Both are dependent on increases in PI3-K activity, ultimately increasing ENaC activity. Blazer-Yost and colleagues proposed that activation of basolateral PI3-K in response to insulin signaling stimulates PIP 3 production in this membrane, with the phosphatidylinositide rapidly diffusing within the inner membrane leaflet across tight junctions to the apical membrane. Increases in luminal PIP 3 then increase ENaC activity by increasing the number of functional channels in the membrane. The importance of this signaling pathway is that it contains a possible mechanism for rapidly coupling basolateral signaling events to changes in activity of luminal ion channels. The rapidity of this putative signaling pathway arises from PIP 3 diffusing within a planar lipid surface rather then a three-dimensional cystolic space. It is unclear whether the increase in ENaC activity observed in this study resulted from direct effects of the phosphatidylinositide on ENaC membrane levels and/or gating or indirect effects on ENaC insertion and/or retrieval." }' N2 k& T2 w4 N
9 g- [+ |7 Z( G% c1 r, P
PIP 3 Directly Interacts with ENaC to Increase Membrane Levels of the Channel
- {& G; d1 Z+ K. A3 V( w
2 k0 @* ~. d  X, ~. J, P: _& IIn addition to influencing membrane levels of ENaC through a cell-signaling cascade, impinging on channel retrieval, PI3-K and its product PIP 3 influence membrane levels of this channel through a more direct but less well-understood mechanism. Helms and colleagues ( 30 ) demonstrated that PIP 3 binds ENaC at a yet to be defined domain, or a protein closely associated with the channel, to promote movement of the channel to the membrane. Currently, this mechanism appears to be distinct from that involving Sgk and Nedd4 proteins. However, further investigation is required to better substantiate this. That these experiments were performed in a renal epithelial cell line increases their importance. A domain near the NH 2 terminus of -ENaC that contains several conserved positively charged residues was found to be critical for PIP 3 to increase ENaC activity and targeting of the channel to the membrane; however, this region appears not to be involved in PIP 3 binding to the channel. Interestingly, this domain is similar in locale and charge to the PIP 2 binding domain in -ENaC. The importance of this similarity, as well as the exact function of this domain in -ENaC, however, remains unclear.
0 Q( q; O. ^% G6 Y5 c/ P; @
. N) S* z3 T& J/ v/ o% KPIP 3 Directly Interacts with ENaC to Increase Open Probability+ v" W* a5 d* L  G

) @" {/ O. W) Y$ n6 X% G7 @; h% kAs mentioned above, PI3-K is a first effector of Ras small G proteins ( 2, 60 ). In some epithelia, K-Ras is an aldosterone-induced protein necessary for steroid actions on ENaC open probability ( 49, 75, 83 ). Overexpression of K-Ras with ENaC in CHO cells increases channel activity independently of effects on ENaC membrane levels ( 78 ). K-Ras actions on ENaC are blocked by the PI3-K inhibitor wortmannin and mimicked by overexpression of active PI3-K ( 78, 79, 87 ). Overexpression of an effector-specific mutant of Ras capable of only activating PI3-K also increases ENaC activity in a wortmannin-sensitive manner ( 79 ). These results show that the aldosterone-induced protein, K-Ras, increases ENaC open probability by stimulating PI3-K.
$ ?( Q2 H; P* G) r7 i# J( C) l& g* G8 z9 V3 f: g, x
The first indication that the phosphatidylinositide products of PI3-K, PIP 3 and/or phosphatidylinositol-3,4- bis phosphate [PI( 3, 4 )P 2 ], might directly interact with ENaC to modulate channel open probability came from experiments reporting parallel but fast changes in the active state of PI3-K, membrane PIP 3 /PI( 3, 4 )P 2 levels, and ENaC activity in CHO cells ( 87 ). ENaC activity increased simultaneously with increases in membrane PIP 3 /PI( 3, 4 )P 2 levels in response to both relief of PI3-K from inhibition and stimulation of PI3-K with IGF-I. Moreover, addition of exogenous PIP 3 to the cytosolic face of inside-out patches containing ENaC that had run down increased channel activity ( 87 ). Addition of exogenous PIP 3 and PI( 3, 4 )P 2 to outside-out patches containing ENaC that had not run down increased channel activity above control levels ( 79, 87 ). These increases in activity resulted from definitive changes in open probability. Consistent findings have been reported for ENaC expressed in oocytes, where exogenous PIP 3 increases channel activity without affecting membrane levels ( 44 ). Thus PIP 3 directly regulates ENaC to increase activity and open probability above starting levels. Consistent with a direct interaction with the channel, a PIP 3 /PI( 3, 4 )P 2 binding site in ENaC has now been described ( 57; see below). Disruption of this binding site counters PI3-K actions on ENaC activity, as well as disrupts increases in ENaC open probability in response to addition of exogenous PIP 3. These findings thus demonstrate that similar to its actions on Kir channels ( 21, 45, 61, 62 ), PIP 3 physically associates with ENaC to affect open probability.
9 r- Q% S8 x$ Y- ~" b3 f4 A6 ?# G
# {9 k( b* ~( Z. j% K; o, IWe believe that in contrast to PIP 2, the direct interaction of PIP 3 with ENaC is regulatory rather than permissive. Our rationale is that PI3-K is a central component of several signaling cascades that increase ENaC activity above basal levels, and exogenous PIP 3 increases channel activity above control levels. In addition, one effect of aldosterone and K-Ras is to increase ENaC open probability, an effect possibly mediated by PI3-K and direct interaction of PIP 3 with the channel (as discussed above). Moreover, addition of exogenous PIP 3 has little further effect on ENaC activity in renal epithelial cells pretreated with aldosterone but does significantly increase channel activity in cells not pretreated with steroid ( 30 ). Reports that membrane PIP 3 levels are low compared with those of PIP 2 and that aldosterone increases PIP 3 levels are also consistent with a regulatory role for PIP 3. However, it is also clear that PI3-K plays a role in maintaining basal ENaC activity for inhibition of this kinase rapidly decreases ENaC activity in epithelial cells not treated with steroid ( 55, 59 ). Possible involvement of a direct interaction between PIP 3 and ENaC in setting basal channel activity remains to be fully explored.
2 F; q% W0 k2 H9 Y! {: l8 L5 h( v' d6 V) T, m8 p  j, y/ f0 u
PIP 3 Binding Sites in ENaC
4 \; f3 X8 y8 W$ ]. o3 S. v4 d
! a* K0 ?- F' a1 P7 v) c& N$ sThe PIP 3 /PI( 3, 4 )P 2 binding site in ENaC involved in regulation of channel open probability includes a region in the COOH-terminal cytosolic domain of -ENaC ( 57 ). This region is just distal to the second transmembrane domain in this subunit. Similar to the putative PIP 2 binding site in the NH 2 terminus of -ENaC, this region of ENaC, as show in Fig. 2 B, contains several well-conserved, positively charged arginine and lysine residues. Deletion of this region disrupts physical association of PIP 3 with ENaC, as well as activation of the channel by PIP 3 ( 57 ). Point mutation of the conserved positively charged residues in this region also disrupts regulation of the channel by PI3-K. This PIP 3 binding site in ENaC then is similar to the phosphatidylinositide binding site in other channels in the respect that they all contain several positively charged conserved residues ( 17, 58, 66, 73, 97 ). Moreover, the location of this PIP 3 binding site in ENaC just distal to the second transmembrane domain is ideally suited to modulate open probability. This locale just after a pore-lining transmembrane domain is also similar to the position of the phosphatidylinositide binding site in Kir and TRP channels.% P" c1 |( i' C: J1 F7 U

9 |! m+ L+ g. h/ q2 \5 y% rAn interesting additional observation made in this study, as reillustrated in Fig. 4, was that alanine substitution of the negatively charged aspartic acid and bulky tryptophans within the PIP 3 binding site in -ENaC increased both basal and PI3-K-responsive ENaC activity. This suggests that these bulky and negatively charged residues may impact binding affinity or specificity. This possibility is similar to the role played by some noncharged and negatively charged residues in the binding sites of other phosphatidylinosite-sensitive channels. It has not yet been tested whether PIP 2 also interacts with ENaC at this site. However, deletion of this site does decrease basal open probability, possibly indicating permissive actions of PIP 2 or PIP 3 binding." J5 {& x$ R; R3 X+ d8 n6 y

1 n# Y: A1 i6 ]' w( g8 @Fig. 4. Conserved positively and negatively charged residues, as well as bulky aromatic amino acids in a putative PIP 3 binding site in -ENaC modulate phosphoinositide 3-OH kinase (PI3-K) regulation of ENaC. A : summary graph showing ENaC activity in CHO cells in the absence (black bars) and presence of coexpression of constitutively active PI3-K (gray bars). Cells expressed either wild-type,, -ENaC or wild-type, -ENaC plus -ENaC containing the indicated (bold) alanine-substituted amino acids. *Vs. activity in the absence of PI3-K. **Mutant ENaC PI3-K significantly less than wild-type ENaC PI3-K. B : summary graph showing ENaC activity in the absence (black bars) and presence of coexpression of constitutively active PI3-K (gray bars). Cells expressed wild-type,, -ENaC or wild-type, -ENaC plus -ENaC containing the indicated alanine-substituted aspartic acid and tryptophan residues. *Activity of mutant ENaC in the absence and presence of PI3-K vs. wild-type ENaC in the absence and presence of PI3-K, respectively. All groups had significantly more current in the presence of PI3-K vs. its absence. Reprinted from Ref. 57 with permission.* `$ ?1 b: }% y' s% e0 @
5 r& V# U6 V: o
IMPLICATIONS OF SPATIAL AND TEMPORAL REGULATION OF PHOSPHATIDYLINOSITIDE TURNOVER FOR CONTROL OF ENaC- Y, t0 h' l+ J1 {7 w, i' j

5 S* M( x( D3 I) OIt is becoming clear that similar to other second messengers, such as cAMP and Ca 2 , which regulate myriad diverse cellular activities, PIP 2 and PIP 3 synthesis and metabolism must be both spatially and temporally regulated (reviewed in Refs. 18 and 53 ). Such regulation allows for localized subcellular changes in phosphatidylinositide levels, leading to dynamic and versatile phosphatidylinositide signaling. This is particularly important in polarized cells, which have the potential for distinct phospholipid compositions in specialized membranes and membrane areas. For instance, enzymes that synthesize and metabolize PIP 3 are polarized in macrophages during chemotaxis, leading to a membrane PIP 3 gradient ( 34 ). During fibroblast migration, moreover, membrane PIP 3 levels show localized changes in response to PDGF ( 64 ). Similarly, the subcellular localization of PI3-K and PIP 3 play critical roles in axon specification during neurite elongation and polarization ( 50 ). In response to insulin signaling, PIP 3 synthesized in the basolateral membrane quickly diffuses to the apical membrane in renal epithelia containing ENaC ( 5 ). The translocation and development of a PIP 3 gradient correlate with increases in channel activity. Similar to PIP 3, PIP 2 accumulates in plasmalemmal microdomains ( 28 ). These membrane patches exhibit locally regulated PIP 2 turnover and reduced diffusion, as well as accumulation of specialized proteins involved in membrane trafficking, signaling, and polarization. Localized rather then global changes in membrane phosphatidylinositide levels are presently thought to underlie many forms of ion channel regulation, as exemplified by G q -coupled receptor regulation of GIRK ( 12 ).% }3 S6 j( I# B( x/ ]2 w
8 f2 ?5 l- d, g4 b  w% ^
Spatial organization of phosphatidylinositides within cellular membranes arises from the localized expression and/or activation of the kinases and phosphatases involved in synthesis and metabolism, as well as the localization of specific phospholipid precursors ( 18, 53 ). Temporal regulation arises from the activity of upstream regulators and the structure of these kinases and phosphatases. Recent investigation of phosphatidylinositide regulation of ENaC is consistent with temporal and spatial regulation of PIP 2 and PIP 3 playing important roles in modulation of this channel.1 r1 `0 Y) g- Q& s

  r+ s) ?7 \$ D: F( m; TAlthough few studies, as yet, have specifically tested the effects of localized phosphatidylinositide turnover on ENaC activity, many predictions can be made from available results. For instance, regulation of ENaC open probability by PIP 2 and PIP 3 is predicted to be a localized rather than global response. Our rationale is that ENaC is found only in the apical membrane, and PIP 2 and PIP 3 likely directly interact with the channel to affect gating. Thus it ultimately is the phosphatidylinositide levels in the luminal membrane and more specifically near the channel that are important for phosphatidylinositide modulation of gating. The finding that PIP 3 synthesized in response to insulin translocates to the luminal membrane is consistent with a localized response ( 5 ). Our finding that the aldosterone-induced protein K-Ras localizes PI3-K near ENaC is also consistent with tight special coupling between the channel and localized changes in membrane phosphatidylinositide levels ( 79 ). Findings in excised patches from both native epithelial and reconstituted systems documenting decreases in ENaC activity and open probability on activation of receptors coupled to PIP 2 hydrolysis mandate, at least in these experimental settings, a localized response ( 39, 44, 88 ). Thus the cellular machinery apparently necessary for regulation of ENaC by local phosphatidylinositide turnover is present in many of the cases studied. Moreover, the tight temporal coupling of membrane phosphatidylinositide levels with changes in ENaC gating also suggests a localized rather than a global response ( 39, 44, 88 ).
8 }" ?9 j, b. J* N/ z/ R. c. c( |2 i( k! W8 K4 b6 h
It is more difficult to make predictions about whether local or global changes in PIP 2 and PIP 3 affect ENaC membrane levels. However, it is becoming clear that the effects of these phosphatidylinositides on cellular trafficking are tightly controlled both spatially and temporally ( 18, 53 ). Thus it is also likely that PIP 2 and PIP 3 regulation of ENaC membrane levels is a response with specific subcellular check points, leading to a targeted and localized effect. However, compared with regulation of gating, regulation of ENaC membrane levels by phosphatidylinositides appears to develop more slowly. We excitedly await further determination of the physiological relevance of spatial and temporal control of cellular phosphatidylinositide levels with respect to control of ENaC activity.
9 X6 a! G; ~2 i# F0 K
+ ?+ y! r* \3 L( f7 HGRANTS
/ ~' m% X! a6 v% Y6 g0 s% B
! i8 q' ?5 A0 A, o7 \* xResearch in our laboratories on regulation of ENaC by phosphatidylinositides has been supported by National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) Grant RO1-DK-59594, American Heart Association-Texas Affiliate Grant 0355012Y, and the American Society of Nephrology Carl W. Gottschalk Research Scholar Grant (to J. D. Stockand); a National Kidney Foundation Research Fellowship (to A. Staruschenko); and NIDDK Grant RO1-DK-067110 (to H.-P. Ma).
9 x% m+ X) g' E1 D6 O' `' `3 |
& C! c8 ]" ?+ X1 EACKNOWLEDGMENTS
) _3 H7 C& U! Q, Y2 k5 T- P: s3 }7 j5 F; L) a# p, X* N2 P, z
We thank J. Medina and P. Patel for excellent technical support.4 Z* I  Y7 ]9 n7 D
          【参考文献】$ |' ]9 D7 ?) {
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.  m* l# a/ _) a" r
0 _+ a" t3 }9 _8 I( Z7 K, l0 s

6 z, i9 a  D$ [9 @; {  n7 e" j' k: y) o9 ^; q: V6 n" S
Bar-Sagi D. A Ras by any other name. Mol Cell Biol 21: 1441-1443, 2001.+ b2 |9 L* z8 d% h6 o8 n6 W

$ ~5 W' E  V3 V; y& N2 s! q
7 @0 b9 A% E2 c' S, A. r. _/ Y1 ^& U' f8 y. e* @, G# g" I2 ?  O+ V
Bian JS, Kagan A, and McDonald TV. Molecular analysis of PIP 2 regulation of HERG and I Kr. Am J Physiol Heart Circ Physiol 287: H2154-H2163, 2004.8 G5 ]6 l1 G9 o, Q3 p$ k# J
% k0 p6 T$ P( f+ Q% ?
% a6 c% _% b, Z/ M9 n

& R2 g7 N1 z+ W/ A8 cBlazer-Yost BL, Paunescu TG, Helman SI, Lee KD, and Vlahos CJ. Phosphoinositide 3-kinase is required for aldosterone-regulated sodium reabsorption. Am J Physiol Cell Physiol 277: C531-C536, 1999.
5 r+ I4 J: R) E. v7 z7 Y7 \- l+ E
! f/ ^" f) \) {  d7 D

1 V9 f) k1 A9 B: D( |9 {; N- XBlazer-Yost BL, Vahle JC, Byars JM, and Bacallao RL. Real-time three-dimensional imaging of lipid signal transduction: apical membrane insertion of epithelial Na   channels. Am J Physiol Cell Physiol 287: C1569-C1576, 2004.( g6 B0 W- k4 P) E( p" q
8 E; p9 O# r- W8 F7 \

5 x/ [/ q5 j% ^$ p% g0 B1 ?0 t* u5 _0 x8 N, Z6 l
Booth RE, Tong Q, Medina J, Snyder PM, Patel P, and Stockand JD. A region directly following the second transmembrane domain in gamma ENaC is required for normal channel gating. J Biol Chem 278: 41367-41379, 2003.5 M9 |5 `# t1 r$ P- a

1 @* \+ O, g* J7 }! e1 H7 \' ^" p; K7 e6 |- }
. S# O* j! I4 y, ~- a4 j7 ^
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.- J8 D9 B6 h, v8 a

/ X$ C. A  N) Q. Z0 U) l  P6 }
  V/ L! D% K3 s) B& f4 U6 v: v) U; ]8 U
Canessa CM, Horisberger JD, and Rossier BC. Epithelial sodium channel related to proteins involved in neurodegeneration. Nature 361: 467-470, 1993.
, ?" `# d, T0 a. k. }
4 }6 e, G) q4 j3 k7 D" b' l, }$ f- Q5 r9 C
  [8 X& O. u; m$ H/ p
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.
5 y/ I+ @; u$ A, |" r4 O2 h
" P1 \, `$ H# ]' U& s' R$ i9 V5 M$ i8 z4 h4 |

. I' N/ e( @5 j( Y* }Chen S, Bhargava A, Meijer O, Rozansky D, Spindler B, Verrey F, and Pearce D. Identification and characterization of novel mineralocorticoid-regulated genes in tight epithelia (Abstract). J Am Soc Nephrol 9: 398A, 1998.
$ ]9 q% J& M, H% m2 n7 H
2 n% N" c3 i6 m% ?
: [% E; N- `3 f1 U+ I$ J. [
. D" W0 R: ]5 _( b. i9 a' CChen S, Bhargava S, Mastroberardino L, Meijer OC, Wang J, Firestone P, Verrey F, and Pearce D. Epithelial sodium channel regulated by aldosterone-induced protein sgk. Proc Natl Acad Sci USA 96: 2514-2519, 1999.
: ?9 I7 |3 }  W' _, U3 u
8 _4 U" ^; E+ T( E/ \& L/ P- J; L9 G0 B
9 x  e/ e$ \" M. n+ k8 p7 g+ G- E9 R
Cho H, Kim YA, Yoon JY, Lee D, Kim JH, Lee SH, and Ho WK. Low mobility of phosphatidylinositol 4,5-bisphosphate underlies receptor specificity of Gq-mediated ion channel regulation in atrial myocytes. Proc Natl Acad Sci USA 102: 15241-15246, 2005.
) a3 g* }5 b5 B6 a6 ^3 T& G8 {. D. F5 a  V- k

' j; R; O6 U) y% G; j7 y
! K9 {  ~7 {/ Q  G+ T& e* m/ UChuang H, Prescott E, Kong H, Shields S, Jordt S, Basbaum A, Chao M, and Julius D. Bradykinin and nerve growth factor release the capsaicin receptor from PtdIns(4,5)P 2 -mediated inhibition. Nature 411: 957-962, 2001.2 k" F) L6 |7 z8 Z& q! M* ~. Q3 d
9 L- h/ d: }8 R) B5 A- F% z# Q
" V, J1 g& g) g$ o( T$ L

. j& q) E7 M  E% O6 w' BCullen PJ, Cozier GE, Banting G, and Mellor H. Modular phosphoinositide-binding domains-their role in signalling and membrane trafficking. Curr Biol 11: R882-R893, 2001.
) R5 c0 u( N/ d. |9 ~( M% ?9 i
2 k( Q; D1 N* j6 ^8 R
4 a+ A- n2 g% f6 K+ }+ t' {. Z# A) O/ O
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.
& G! D: W- f! [" m# Y
" g' u+ S$ \2 i( V+ ^: c+ |8 C0 ~- Z" I- [
+ X- X2 C7 ^: V# I: d& I
Donaldson MR, Jensen JL, Tristani-Firouzi M, Tawil R, Bendahhou S, Suarez WA, Cobo AM, Poza JJ, Behr E, Wagstaff J, Szepetowski P, Pereira S, Mozaffar T, Escolar DM, Fu YH, and Ptacek LJ. IP 2 binding residues of Kir2.1 are common targets of mutations causing Andersen syndrome. Neurology 60: 1811-1816, 2003.
6 h& W, U1 j6 v# \  d4 h/ M! ]0 Y- u$ s+ [1 U

- u  x4 X. |2 o" q
0 z( i  Y7 ^' m& `Dong K, Tang L, Macgregor GG, and Hebert SC. Localization of the ATP/phosphatidylinositol 4,5 diphosphate-binding site to a 39-amino acid region of the carboxyl terminus of the ATP-regulated K   channel Kir1.1. J Biol Chem 277: 49366-49373, 2002.
4 `* R& ]4 X" L* l0 N' K1 y- b- |  `0 G5 g
1 l) U- }6 F) L
: q7 `' Q% w% r' [5 t, E/ ^
Doughman RL, Firestone AJ, and Anderson RA. Phosphatidylinositol phosphate kinases put PI4,5P 2 in its place. J Membr Biol 194: 77-89, 2003.: o. a8 k  J! @# ^5 [1 B

. j  }, |, N+ C# P* S) _) B7 k* E8 R( a- f. F0 N) k' N
. E0 D; w. ?# f/ |2 D5 M
Downes CP, Gray A, and Lucocq JM. Probing phosphoinositide functions in signaling and membrane trafficking. Trends Cell Biol 15: 259-268, 2005.
/ [5 ?. E1 j4 Q& B$ w: M/ e/ Q
: {" ~% h! d/ T  u' W( N# u! v' V1 A6 ~8 t2 S* j

8 L3 z# I1 r! Y4 \. `Edinger RS, Rokaw MD, and Johnson JP. Vasopressin stimulates sodium transport in A6 cells via a phosphatidylinositide 3-kinase-dependent pathway. Am J Physiol Renal Physiol 277: F575-F579, 1999.  K& |5 [' q4 w  \3 L  m! _, T( ^
/ ^: H# z* V4 K

, f3 ]; ~. ?/ R" R; e/ n3 v8 Q0 ?/ b: G1 C9 [
Enkvetchakul D, Jeliazkova I, and Nichols CG. Direct modulation of Kir channel gating by membrane phosphatidylinositol 4,5-bisphosphate. J Biol Chem 280: 35785-35788, 2005.
; R5 ~9 i% G3 l% K( m/ I/ `+ r! m9 N1 |* l$ ]6 o

! A( x3 f$ Z* ]4 u: N+ j, [5 @4 n% _) p
Faletti CJ, Perrotti N, Taylor SI, and Blazer-Yost BL. sgk : An essential convergence point for peptide and steroid hormone regulation of ENaC-mediated Na   transport. Am J Physiol Cell Physiol 282: C494-C500, 2002.! l8 T5 v" ?' R# D

  L/ z3 w9 @- X" @0 `% o  Y) x; B- I* J6 F5 q" m. f+ d) \
# s% b) p7 @, P! ]) {' z( [
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., Y8 n. d$ x/ ~; g) B: ^" a
- f6 P8 G" y8 R! L) e  l
, ~' g$ Z$ P0 r: @! T, C3 ~
/ [+ ^) s% `8 s+ ?
Fruman D, Meyers R, and Cantley L. Phosphoinositide kinases (Abstract). Annu Rev Biochem 67: 507, 1998.
6 g; r; r5 A: [+ E( Y. e, k
4 l4 l% V2 j; W4 {* t  e6 w9 B" {
6 m5 I) F( \4 i6 [0 y: H
Fyfe GK and Canessa CM. Subunit composition determines the single channel kinetics of the epithelial sodium channel. J Gen Physiol 112: 423-432, 1998.4 \% d9 p( c$ y1 V- }; h; d
$ a. S+ f$ [5 s  l+ H
9 S1 S5 k* ~/ i* t  W
2 d$ z& H4 u" h, ?% y5 \$ C
Gamper N, Reznikov V, Yamada Y, Yang J, and Shapiro MS. Phosphatidylinositol 4,5-bisphosphate signals underlie receptor-specific Gq/11-mediated modulation of N-type Ca 2  channels. J Neurosci 24: 10980-10992, 2004.7 e  O! ^1 k' z- n9 x* ^' Z2 z

& d9 w+ r. U: S6 l+ p7 D# I
8 }' B1 B2 I9 E& Y8 N* D6 D9 u/ u7 e. N/ g
Garty H and Palmer LG. Epithelial sodium channels: function, structure, and regulation. Physiol Rev 77: 359-396, 1997.  o* s% q9 _! O% g

7 m. U# `/ L" O. j' r$ O4 j2 n( P8 v1 }, }6 l& `/ i% X' I3 c, G

- c$ G/ l$ ~, I- _; W" |. YGolub T and Caroni P. PI(4,5)P 2 -dependent microdomain assemblies capture microtubules to promote and control leading edge motility. J Cell Biol 169: 151-165, 2005.  M# O) A. D: w# H1 D! z. |3 z6 I

5 w8 p: V9 U; h3 D1 G9 X, S& [) k% G% O' r
, o+ E/ i# v+ w: {0 Y
Goulet CC, Volk KA, Adams CM, Prince LS, Stokes JB, and Snyder PM. Inhibition of the epithelial Na   channel by interaction of Nedd4 with a PY motif deleted in Liddle's syndrome. J Biol Chem 273: 30012-30017, 1998.
: v4 w2 i: R6 K2 h$ ~, K' p" _- I% _; ~) H1 h( A4 T
1 V# p- i0 b, \; o- P
3 P. N9 A: e0 o: I( {& D8 e
Helms MN, Liu L, Liang YY, Al-Khalili O, Vandewalle A, Saxena S, Eaton DC, and Ma HP. Phosphatidylinositol 3,4,5-trisphosphate (PIP 3 ) mediates aldosterone stimulation of epithelial sodium channel (ENaC) and interacts with -ENaC. J Biol Chem 280: 40885-40891, 2005.
% h% r8 J9 L' r
2 a' j9 e' G1 [1 D$ [* O; \3 X* q$ H
) j$ p* B; m0 ?/ N
Hendron E and Stockand JD. Activation of mitogen-activated protein kinase (mitogen-activated protein kinase/extracellular signal-regulated kinase) cascade by aldosterone. Mol Biol Cell 13: 3042-3054, 2002.* ~: L' V/ ?' A9 {  n2 A6 a5 l

+ o- X" @) W  E* |. a7 g  I0 x$ x: D% T, _
1 p1 J. I! Q5 K5 \& h, R
Hilgemann D and Ball R. Regulation of cardiac Na  ,Ca 2  exchange and KATP potassium channels by PIP 2. Science 273: 956-959, 1996.5 O/ B1 [6 P: q. B6 P% y9 B4 P
  e2 B/ t1 R0 M2 O3 b2 O6 v* Z4 M* ^* |3 U
' b  V+ r5 H6 x
, u6 r: L. W1 h" F, m7 K
Hilgemann D, Feng S, and Nasuhoglu C. The complex and intriguing lives of PIP 2 with ion channels and transporters. Sci STKE 111: RE19, 2001.5 G, Q4 O0 _, x$ w( H7 s/ t' ]1 `( A# {

& \8 l$ ]1 r. D( o- A( h/ N; u( n0 ?1 z; G( c/ U; ]

- Q* w$ O+ N8 _5 B& g( \1 j4 FHuang YE, Iijima M, Parent CA, Funamoto S, Firtel RA, and Devreotes P. Receptor-mediated regulation of PI3Ks confines PI(3,4,5)P 3 to the leading edge of chemotaxing cells. Mol Biol Cell 14: 1913-1922, 2003." J- Q; U! J" R

2 J! D; g+ G, r' y
2 ^2 X. S! b- E9 o* g9 }& z: m6 H( [
Hummler E and Horisberger JD. Genetic disorders of membrane transport. V. The epithelial sodium channel and its implication in human diseases. Am J Physiol Gastrointest Liver Physiol 276: G567-G571, 1999.4 t) }" z2 `4 R/ p
# s  ?" f3 {. ]% ]3 U' g2 S
6 ?) e- ]/ L- J  W
& a- Y5 |2 O; }+ X, ?  E, M
Kamynina E and Staub O. Concerted action of ENaC, Nedd4-2, and Sgk1 in transepithelial Na   transport. Am J Physiol Renal Physiol 283: F377-F387, 2002./ W. F  @8 S2 u
2 _  G- ]9 ~: m, o1 Z
" g6 Y( u8 [# J8 K9 i

% y, A. ?; _$ J4 k; N) `Kellenberger S and Schild L. Epithelial sodium channel/degenerin family of ion channels: a variety of functions for a shared structure. Physiol Rev 82: 735-767, 2002.
" f& }2 x$ o, T# q- G2 }: q
) G$ y1 ?' z9 I6 R
9 p- B1 J4 t" L$ b9 `5 e0 g! B) u! Q# m8 v# J
Kobrinsky E, Mirshahi T, Zhang H, Jin T, and Logothetis D. Receptor-mediated hydrolysis of plasma membrane messenger PIP 2 leads to K-current desensitization. Nat Cell Biol 2: 507-514, 2000.7 T% w- B, ?! x! O% Y* B) r( H
$ E6 t9 }3 a( l5 w# v# s
, \% w7 C8 h7 Z( y% n5 q

5 C1 b4 ]* \: s4 B4 w0 d& YKunzelmann K, Bachhuber T, Regeer R, Markovich D, Sun J, and Schreiber R. Purinergic inhibition of the epithelial Na   transport via hydrolysis of PIP 2. FASEB J 19: 142-143, 2005., l& r' a3 `; M

! O* r0 i: u+ ~" W- s% ~
; A) j8 L  ~# B$ ^1 j; T" v
/ y& Y, ?3 s6 o5 ^Li Y, Gamper N, Hilgemann D, and Shapiro MS. Regulation of Kv7 (KCNQ) K   channel open probability by phosphatidylinositol (4,5)- bis phosphate. J Neurosci 25: 9825-9835, 2005.
* ^5 b. V2 ]9 r- {2 f. `- J; O1 V! _4 p# J3 Z3 R( m
6 G( S/ D: u- H+ Q$ A& K
6 M( g* i4 c% k+ S3 i% h8 y
Lifton RP, Gharavi AG, and Geller DS. Molecular mechanisms of human hypertension. Cell 104: 545-556, 2001.
- u9 m" X! g3 U, B4 y0 S, Z5 K, ^: h
( @$ i- h  g2 i. p+ w) D4 {

3 u  H6 H4 ^/ g0 eLopes CM, Rohacs T, Czirjak G, Balla T, Enyedi P, and Logothetis DE. PIP 2 hydrolysis underlies agonist-induced inhibition and regulates voltage gating of 2-pore domain K   channels. J Physiol 564: 117-129, 2005., T/ b( T5 O! i- ^+ V9 e! D" f
# R; Z8 B; I  Y. \
8 ?. E5 s+ |0 L# d$ B9 h
' Z9 I$ t0 Y. r% ?* V( G) T
Lopes C, Zhang H, Rohacs T, Jin T, and Logothetis D. Alterations in conserved Kir channel-PIP 2 interactions underlie channelopathies. Neuron 34: 933-944, 2002.' `' S, H0 A+ k  o; X" U

1 V9 O; I, P1 O" |  `: t- G! v; B8 P( K4 ~

1 u$ E# j. ?, R! j* O: K; ZMa HP, Saxena S, and Warnock DG. Anionic phospholipids regulate native and expressed epithelial sodium channel (ENaC). J Biol Chem 277: 7641-7644, 2002.
& M/ r4 \) j) w9 l" B. D* [7 u2 Y3 P! B  {3 |

5 _( O/ R" |) C  Y$ F/ P, [9 k- Y1 x
  T" ?; D6 K+ ]2 P, r9 \6 {% GMacGregor G, Dong K, Vanoye C, Tang L, Giebisch G, and Hebert S. Nucleotides and phospholipids compete for binding to the C terminus of K-ATP channels. Proc Natl Acad Sci USA 99: 2726-2731, 2002.9 B1 L, w+ O8 {  z
0 d. ~0 ?7 B% l/ K/ w

. f" P/ ?$ p% R" L/ N
4 }  {0 C' T1 K- ~1 b1 F7 wMall M, Grubb B, Harkema J, O'Neal W, and Boucher R. Increased airway epithelial Na   absorption produces cystic fibrosis-like lung disease in mice. Nat Med 10: 487-493, 2004.6 Y4 A8 o! I7 c. u+ z

' P, t( u7 d, Z7 i' l
3 X4 B5 [" N  _! w. D5 ^" E
+ b7 _* C+ ]4 FMarkadieu N, Blero D, Boom A, Erneux C, and Beauwens R. Phosphatidylinositol 3,4,5- tris phosphate: an early mediator of insulin-stimulated sodium transport in A6 cells. Am J Physiol Renal Physiol 287: F319-F328, 2004.
1 q0 V1 z; T3 V% L  f8 b( o( n! e0 e  z. Z2 M( p5 t

* F" u" d" L. B3 l6 X- g9 y' d
Markadieu N, Crutzen R, Blero D, Erneux C, and Beauwens R. Hydrogen peroxide and epidermal growth factor activate phosphatidylinositol 3-kinase and increase sodium transport in A6 cell monolayers. Am J Physiol Renal Physiol 288: F1201-F1212, 2005.
/ `, `, Z4 Z! }. j" r
( [) q3 F  ~( s9 i. w4 N, g8 W! q6 H8 [: x+ M8 v

0 U' F# ?) v' n5 gMastroberardino L, Spindler B, Forster I, Loffing J, Assandri R, May A, and Verrey F. Ras pathway activates epithelial Na channel and decreases its surface expression in Xenopus oocytes. Mol Biol Cell 9: 3417-3427, 1998.  u+ h! r8 }" I8 c2 k
$ U3 p* n% k  u: N+ b# a' ]5 g8 l/ J
: Q; w4 i) H1 ~( p

" R2 }& y( H) M. ?Menager C, Arimura N, Fukata Y, and Kaibuchi K. PIP 3 is involved in neuronal polarization and axon formation. J Neurochem 89: 109-118, 2004.' O0 c7 c9 B' g6 ~! U6 J
" Z, D# o  o& E+ k2 z& e$ q

# G7 l1 h: y8 q- L2 M
# p" V2 G/ H: |. k2 ~/ `Naray-Fejes-Toth A., Canessa C., Cleaveland E.S, 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.
  b& d' a4 _- H9 z1 g) _( x
2 t4 g7 `# O* u7 U! N+ ~! ~9 s& ]$ q( O  J, W6 O

( {0 O$ r) j7 a1 aNasuhoglu C, Feng S, Mao J, Yamamoto M, Yin HL, Earnest S, Barylko B, Albanesi JP, and Hilgemann DW. Nonradioactive analysis of phosphatidylinositides and other anionic phospholipids by anion-exchange high-performance liquid chromatography with suppressed conductivity detection. Anal Biochem 301: 243-254, 2002.
' E. O8 Z8 V+ H3 b% x/ d0 a! M  d% W1 ]# H* g2 U2 x" p( U
* O& q! r3 x5 E- g- I7 n
3 y% W% v# K  |$ v5 x
Oude Weernink PA, Schmidt M, and Jakobs KH. Regulation and cellular roles of phosphoinositide 5-kinases. Eur J Pharmacol 500: 87-99, 2004.3 ^$ h0 Z# \6 {. |
- m0 Y% U* C; t1 r  Z1 k
5 r. _, D2 D$ y' x) p, y. J

( j/ d6 @  A4 \6 Q7 i  Z6 rOude Weernink PA, Schulte P, Guo Y, Wetzel J, Amano M, Kaibuchi K, Haverland S, Voss M, Schmidt M, Mayr GW, and Jakobs KH. Stimulation of phosphatidylinositol-4-phosphate 5-kinase by Rho-kinase. J Biol Chem 275: 10168-10174, 2000.
' V/ t( X: }# ^/ T9 a$ V$ Q; l& B. h6 g9 I

( J2 [9 Z* d+ t* @
' ?- Z- l1 x& W4 [1 X+ a' ?Paunescu TG, Blazer-Yost BL, Vlahos CJ, and Helman SI. LY-294002-inhibitable PI 3-kinase and regulation of baseline rates of Na   transport in A6 epithelia. Am J Physiol Cell Physiol 279: C236-C247, 2000.
# D9 G. Z- ]2 _( {4 i8 }1 W% G2 O0 C! d/ K

7 J7 M4 ^+ G# Y: a, B& S( v3 @5 i
, l7 x! }+ o8 X( w  D$ b  x! c$ }1 vPlaster NM, Tawil R, Tristani-Firouzi M, Canun S, Bendahhou S, Tsunoda A, Donaldson MR, Iannaccone ST, Brunt E, Barohn R, Clark J, Deymeer F, George AL Jr, Fish FA, Hahn A, Nitu A, Ozdemir C, Serdaroglu P, Subramony SH, Wolfe G, Fu YH, and Ptacek LJ. Mutations in Kir21 cause the developmental and episodic electrical phenotypes of Andersen's syndrome. Cell 105: 511-519, 2001.
) W& r6 ]3 W7 S! |5 H; y5 ?
7 L/ ^3 c: s1 K( k& W8 n, |* |
  e9 i; O6 Q* }% G# T! B9 E0 }6 g
Pochynyuk O, Staruschenko A, Tong Q, Medina J, and Stockand JD. Identification of a functional phosphatidylinositol 3,4,5-trisphosphate binding site in the epithelial Na   channel. J Biol Chem 280: 37565-37571, 2005.
$ Q- r/ m  @' x9 P  j! `1 P/ `3 e$ e7 |2 c0 x
' U; L2 _7 U) D. W  W

) e* ]9 a  T' h9 r; yPrescott E and Julius D. A modular PIP 2 binding site as a determinant of capsaicin receptor sensitivity. Science 300: 1284-1288, 2003.
# ~; X0 J; |% b7 k) J# v5 C# u
6 G) |2 }/ c& W, T( G/ R" _
- h9 z  Z! D0 k1 W* b$ x( j3 y4 ?7 p+ r' [, p+ |; H  R4 L
Record RD, Froelich LL, Vlahos CJ, and Blazer-Yost BL. Phosphatidylinositol 3-kinase activation is required for insulin- stimulated sodium transport in A6 cells. Am J Physiol Endocrinol Metab 274: E611-E617, 1998.) Y2 E6 Y- |5 B% c$ @

" ~+ K- O6 R, I- K0 E' n- r9 C! f# ~# I' y+ ?3 \$ x4 [3 L

* b3 M! k$ S2 K( U6 q2 _0 FRodriguez-Viciana P, Warne PH, Khwaja A, Marte BM, Pappin D, Das P, Waterfield MD, Ridley A, and Downward J. Role of phosphoinositide 3-OH kinase in cell transformation and control of the actin cytoskeleton by Ras. Cell 89: 457-467, 1997.
" R& X! C- n& ^3 a
9 d) i8 T/ {; k5 @6 l
! ]7 r* g- q0 t3 W/ z9 B
2 O$ \0 z- r. R  y. ]Rohacs T, Lopes C, Jin T, Ramdya P, Molnar Z, and Logothetis D. Specificity of activation by phosphoinositides determines lipid regulation of Kir channels. Proc Natl Acad Sci USA 100: 745-750, 2003.) t; }* G. i5 Q# O

: k7 x# W/ A  x8 {. t+ ^+ R. M  j# V' S& l
/ O! m) H/ n$ U; x3 O/ i  H- P# W
Rohacs T, Chen J, Prestwich GD, and Logothetis DE. Distinct specificities of inwardly rectifying K   channels for phosphoinositides. J Biol Chem 274: 36065-36072, 1999., e6 t+ l2 q7 E9 s# \

( l4 Z1 g1 G/ m! f% t( X, k/ Z: S+ }; ~1 u0 l$ |
2 [% M. @/ o* T: H
Rotin D, Kanelis V, and Schild L. Trafficking and cell surface stability of ENaC. Am J Physiol Renal Physiol 281: F391-F399, 2001.
8 }9 v# h4 L" a2 F, d# @; g* }6 u% f3 @, P! H, y) g

% ?# v: I: e- K5 M, l$ R) T& m7 G5 h2 G+ X0 m2 o; e$ v6 M
Schneider IC and Haugh JM. Spatial analysis of 3' phosphoinositide signaling in living fibroblasts. II. Parameter estimates for individual cells from experiments. Biophys J 86: 599-608, 2004.! Z3 f. M1 ]1 n2 t8 g9 X
$ L8 q; e. e, y3 l: d" \
" R* l. X- c5 \

6 c8 n9 m; O# L( y: e3 uSchulte U, Hahn H, Konrad M, Jeck N, Derst C, Wild K, Weidemann S, Ruppersberg JP, Fakler B, and Ludwig J. pH gating of ROMK [K(ir)1.1] channels: control by an Arg-Lys-Arg triad disrupted in antenatal Bartter syndrome. Proc Natl Acad Sci USA 96: 15298-15303, 1999.
$ G+ w6 @  u5 G7 Z: ]3 z  m. G' B, b. O0 Y: {- Z0 O

: H+ M! J3 Z  [0 z  O- [) q
* V4 m3 N  @+ [4 ^* S% n/ zShyng SL, Cukras CA, Harwood J, and Nichols CG. Structural determinants of PIP 2 regulation of inward rectifier K ATP channels. J Gen Physiol 116: 599-608, 2000.
% W3 |9 f$ P: K7 h5 M
2 ~$ G. B6 c8 ~- z: P/ f* h& E$ u5 ^$ \
4 a, o' y2 L, ^+ u
" b% z' `* p% e4 @% ?' HSimonsen A, Wurmser AE, Emr SD, and Stenmark H. The role of phosphoinositides in membrane transport. Curr Opin Cell Biol 13: 485-492, 2001.
# L* x2 U9 F, d+ R5 H  ^% z- S: a# \' n# v- r0 h9 }
7 [; b3 L* X4 H- H0 _7 g: V

( I* T9 ]: y1 W* }, v& p: b2 JSnyder PM. Regulation of epithelial Na   channel trafficking. Endocrinology 146: 5079-5085, 2005.
' e+ M( o. c, H4 a# _* x  f6 d% {% b5 M) T
; K/ V/ e0 d# k( W% x% }& J# D: X
5 |! l/ `2 W6 Z) {4 O/ z
Snyder PM. The epithelial Na   channel: cell surface insertion and retrieval in Na   homeostasis and hypertension. Endocr Rev 23: 258-275, 2002.
' Q8 u" n9 l, t' ]' u* W5 X# R2 E# Y+ b6 i. V: Y/ O2 V, M
. g7 Q; Y1 C/ X6 L0 l/ p* n
/ b9 i" D3 g9 }/ ~9 n: K! y
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.: n* u3 s3 s9 t7 N3 s

" d! h0 i7 w) U; w
4 x' ?: h  w! ?
5 ^9 @1 A  W5 E3 V. {# K* pSnyder PM, Price MP, McDonald FJ, Adams CM, Volk KA, Zeiher BG, Stokes JB, and Welsh MJ. Mechanism by which Liddle's syndrome mutations increase activity of a human epithelial Na   channel. Cell 83: 969-978, 1995.7 `8 ]* I. F7 L9 t/ H

1 o4 R: M- e6 ^9 R( u. n
1 F8 K+ Z4 b: J+ }( n) Y; R* T# c2 A$ F$ W5 B1 X1 q
Soom M, Schonherr R, Kubo Y, Kirsch C, Klinger R, and Heinemann SH. Multiple PIP 2 binding sites in Kir2.1 inwardly rectifying potassium channels. FEBS Lett 490: 49-53, 2001.. z' \; [4 O4 j5 ]8 I7 N& o

9 A0 O! D$ |- {5 C4 z
( x. w/ ?7 o  }
( K" W  y) K6 bSpehr M, Wetzel CH, Hatt H, and Ache BW. 3-Phosphoinositides modulate cyclic nucleotide signaling in olfactory receptor neurons. Neuron 33: 731-739, 2002.3 Y9 r3 Y4 E* p/ ]6 v

& c# v* U' z6 u0 i0 F% g; e' \! V- L0 B  V) d
6 P/ N* e2 F% D/ b/ a# W( m
Spindler B, Mastroberardino L, Custer M, and Verrey F. Characterization of early aldosterone-induced RNAs identified in A6 kidney epithelia. Pflügers Arch 434: 323-331, 1997.
9 e; z& o" z" _. [+ p! d+ k" y
+ M5 T* O: q9 y- ^  A$ p. I$ m
( Z' d; w% [$ M9 b- d4 @! j. j& m  _8 a7 D6 P: n
Spindler B and Verrey F. Aldosterone action: induction of p21 ras and fra -2 and transcription-independent decrease in myc, jun, and fos. Am J Physiol Cell Physiol 276: C1154-C1161, 1999.
7 P. A, N6 _7 e) I3 a9 M) ?3 W1 ?2 h

+ q, O& a7 g! O; a& E8 s: D
+ _& R6 d5 x6 sStaruschenko A, Nichols A, Medina JL, Camacho P, Zheleznova NN, and Stockand JD. Rho small GTPases activate the epithelial Na   channel. J Biol Chem 279: 49989-49994, 2004.% W4 l4 b8 a# {$ m( [% r

5 P+ t1 g  H2 _7 v  b' U
& \0 K2 @9 M5 H, J: g5 [" i: _* H& {$ T6 C( }2 W8 o; ]' l
Staruschenko 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, 2004.9 [- q% ^( K+ A3 r8 p, s2 [

; j8 ?) P" h6 {+ a* @+ j6 z- a
- Q: C2 o/ m8 `$ A1 ^7 T, b/ ^5 X
Staruschenko 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, 2005.5 H# |, b( V& P7 o6 @2 S9 F& D

# N0 P5 A4 Z: J. I
8 ^$ [( S4 T. P8 e6 W9 a( E8 e1 N2 M1 U8 j! b% h* D: X7 @9 Y
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.
+ I: [7 z: E6 D
9 b, L- d3 W) K3 R0 B2 p' i1 {
# x1 b- U2 D! D; G7 v( H
* Z: k* K2 k" t; cStaub 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.
& l. O! x3 S/ _5 z: g9 I7 |4 r4 C( Z: @

; t$ M8 {" ~# J! I$ C+ E* f6 y) w4 G$ w" o0 s- E+ G
Stockand JD. New ideas about aldosterone signaling in epithelia. Am J Physiol Renal Physiol 282: F559-F576, 2002.& c7 n/ n" ?& X
/ @- O2 E! x& A- \5 V0 _# N1 T, d
4 T* c, @/ q/ E$ @
% a8 H$ @  U& f5 r
Stockand JD, Spier BJ, Worrell RT, Yue G, Al-Baldawi N, and Eaton DC. Regulation of Na reabsorption by the aldosterone-induced, small G protein K-Ras2A. J Biol Chem 274: 35449-35454, 1999.
/ o% K3 Z2 V$ f' n& w4 a' g- ^, `1 u4 N" J& P+ a* y
% d# P5 y, Z& G4 p- ]5 ^& Q
0 g! C" t1 T9 J" j5 h
Suh BC and Hille B. Recovery from muscarinic modulation of M current channels requires phosphatidylinositol 4,5- bis phosphate synthesis. Neuron 35: 507-520, 2002.
. P1 @/ |( h" h; z
* K% U3 P3 ^2 D5 }- a  D
: j' t6 Z6 t/ l* |- _
/ l# E; V5 x$ D2 m7 }Suh BC and Hille B. Regulation of ion channels by phosphatidylinositol 4,5- bis phosphate. Curr Opin Neurobiol 15: 370-378, 2005./ C: e- Y, B# p3 t2 s; w( z
' Y% v4 l7 q3 b( ?$ W& g
5 n% B3 R% v/ }; C2 }1 w+ X
  z/ L6 c, ~; Z% V
Tong Q, Booth RE, Worrell RT, and Stockand JD. Regulation of Na   transport by aldosterone: signaling convergence and cross talk between the PI3-K and MAPK1/2 cascades. Am J Physiol Renal Physiol 286: F1232-F1238, 2004.0 a* v6 M4 J% ]. `
% e& \: J# k, i3 g- k$ E; N  w& F7 R  e

& |, q; ^( g: v
$ ~0 F8 C4 u  s7 T/ H5 ?# [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- bis phosphate produced by phosphoinositide 3-OH kinase. J Biol Chem 279: 22654-22663, 2004.: q' \( H) B! Q7 e; m& {

' f- L8 E# \: @; T' U- N: d: R" ]* k4 ?, Y! B7 D
7 B4 ]* P1 @' ~. j1 M5 i9 s0 m
Tong Q and Stockand JD. Receptor tyrosine kinases mediate epithelial Na   channel inhibition by epidermal growth factor. Am J Physiol Renal Physiol 288: F150-F161, 2005.+ k& s  e7 F+ E% y6 t, {0 }8 K7 q, q

9 U6 v3 i9 M2 j$ z7 ^* E/ y8 @

& S" `0 T: o, J1 yVerrey F. Transcriptional control of sodium transport in tight epithelial by adrenal steroids. J Membr Biol 144: 93-110, 1995.
- e- `2 K9 D. O! A0 o1 N* D
0 v, d% ^0 h' g' K5 R7 f1 B
2 r* p7 o; P" p  M3 {3 f7 e
3 m7 E8 H/ F6 o: M9 i! j4 C; L- m/ AWang 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.$ X& t8 y5 o. S* f1 z$ B

/ N! c9 `& N! i# y  p
) Y% V+ j; P( S2 a2 A$ @! F% T" K) v- `3 ]: I
Weernink PA, Meletiadis K, Hommeltenberg S, Hinz M, Ishihara H, Schmidt M, and Jakobs KH. Activation of type I phosphatidylinositol 4-phosphate 5-kinase isoforms by the Rho GTPases, RhoA, Rac1, and Cdc42. J Biol Chem 279: 7840-7849, 2004.
' L9 z- v3 d# x1 P* T- c- _1 g. M2 T5 m) S& z. N0 z! N

1 M$ Y# j1 T/ m; H5 ^1 `6 e: J, j8 s3 Z
Wu L, Bauer CS, Zhen XG, Xie C, and Yang J. Dual regulation of voltage-gated calcium channels by PtdIns(4,5)P 2. Nature 419: 947-952, 2002.
+ i9 b$ w( H* I* @* j" y2 |5 n4 b  v% I) ^

( f+ t* p6 w; J6 x- O' [; _4 L! b4 R, q5 P7 X
Xiao J, Zhen XG, and Yang J. Localization of PIP 2 activation gate in inward rectifier K   channels. Nat Neurosci 6: 811-818, 2003., M& |1 z- L0 J  ?& L- Y
/ Y# w- ?+ t6 W

9 ]3 }2 f) {. e
6 g2 @' R  c$ w  YYang SA, Carpenter CL, and Abrams CS. Rho and Rho-kinase mediate thrombin-induced phosphatidylinositol 4-phosphate 5-kinase trafficking in platelets. J Biol Chem 279: 42331-42336, 2004.) h2 I$ t( c, R" D" `4 E+ W. `

' f6 N2 k: U( z* P1 R8 g8 h
( B0 @4 A3 e/ c$ R# H2 ~" s4 P8 _4 H
Yue G, Malik B, Yue G, and Eaton DC. Phosphatidylinositol 4,5- bis phosphate (PIP 2 ) stimulates epithelial sodium channel activity in A6 cells. J Biol Chem 277: 11965-11969, 2002.
3 Z1 }! ~* V9 t, ?' d" }
' a# }, h1 Y+ Z: ^7 x1 @+ x( q' h6 ?& H8 k+ R) x

% o' r: t2 L$ q- d# |9 m0 zZhainazarov AB, Spehr M, Wetzel CH, Hatt H, and Ache BW. Modulation of the olfactory CNG channel by Ptdlns(3,4,5)P 3. J Membr Biol 201: 51-57, 2004.& S6 _5 b& s. x- h' W. H' n! n$ d3 n

" e: w7 w; D. P8 k
# e3 b& K' [% H
6 z  u5 U/ y9 Q' d* GZhang H, He C, Yan X, Mirshahj T, and Logothetis D. Activation of inwardly rectifying K channels by distinct PtdIns(4,5)P 2 interactions. Nat Cell Biol 1: 193-188, 1999.! A" i7 H6 N* l. j% p+ i

# h- J3 r, a7 g9 E/ d& S4 u3 x. _5 ?3 d6 f* a- R
4 k$ R9 k3 _" t; K" V
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
75 
威望
75  
包包
2118  
沙发
发表于 2015-6-12 22:43 |只看该作者
细胞治疗行业  

Rank: 2

积分
66 
威望
66  
包包
1790  
藤椅
发表于 2015-7-7 07:25 |只看该作者
自己知道了  

Rank: 2

积分
72 
威望
72  
包包
1859  
板凳
发表于 2015-7-18 08:53 |只看该作者
干细胞之家微信公众号
神经干细胞

Rank: 2

积分
129 
威望
129  
包包
1788  
报纸
发表于 2015-7-21 21:38 |只看该作者
羊水干细胞

Rank: 2

积分
69 
威望
69  
包包
1788  
地板
发表于 2015-8-8 09:16 |只看该作者
不是吧  

Rank: 2

积分
73 
威望
73  
包包
1833  
7
发表于 2015-9-12 14:26 |只看该作者
你还想说什么啊....  

Rank: 2

积分
162 
威望
162  
包包
1724  
8
发表于 2015-9-22 14:43 |只看该作者
加油啊!!!!顶哦!!!!!支持楼主,支持你~  

Rank: 2

积分
101 
威望
101  
包包
1951  
9
发表于 2015-9-29 23:16 |只看该作者
勤奋真能造就财富吗?  

Rank: 2

积分
163 
威望
163  
包包
1852  
10
发表于 2015-10-29 20:01 |只看该作者
一个人最大的破产是绝望,最大的资产是希望。  
‹ 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, 2024-4-30 10:46

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.