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: 365334|回复: 219
go

INVITED REVIEWSodium and calcium transport pathways along the mammaliandistal n [复制链接]

Rank: 1

积分
威望
0  
包包
0  
楼主
发表于 2009-4-21 13:35 |只看该作者 |倒序浏览 |打印
作者:JohannesLoffing and BrigitteKaissling作者单位:Institute of Anatomy, University of Zurich, CH-8057 Zurich,Switzerland
, t7 Q# e' g% e% ~6 `! o                  ) K, B4 x8 M9 q1 E2 H+ w! P6 i
                  / A) r1 V# c- l/ u  `5 E
          + p% c, ?3 t! k9 c9 {+ L2 M
                        
! v3 a) c+ `1 i              }% F- V# z, G/ u* |
            
: T% Y, U" ~9 x9 ~            $ a7 L' [2 _9 ~8 ^6 v( w
            & ~, L3 p/ P% s7 w8 r
                     
% m- F- g( ~* v) Y! Y" O        + |9 `" j* q6 E7 _( Z. M# Z+ h/ }1 s
        
  C5 g. ?! w* f. ~& O0 S, D) m% ?8 k        
3 v- o2 A/ X. c          【摘要】
( `! q3 t( O8 ]      The final adjustment of renalsodium and calcium excretion is achieved by the distal nephron, inwhich transepithelial ion transport is under control of varioushormones, tubular fluid composition, and flow rate. Acquired orinherited diseases leading to deranged renal sodium and calcium balancehave been linked to dysfunction of the distal nephron. Diuretic drugselicit their effects on sodium balance by specifically inhibitingsodium transport proteins in the apical plasma membrane of distalnephron segments. The identification of the major apical sodiumtransport proteins allows study of their precise distribution patternalong the distal nephron and helps address their cellular and molecularregulation under various physiological and pathophysiological settings.This review focuses on the topological arrangement of sodium andcalcium transport proteins along the cortical distal nephron and onsome aspects of their functional regulation. The availability of data on the distribution of transporters in various species points to thestrengths, as well as to the limitations, of animal models for theextrapolation to humans.   m  g" K2 O. d% R
          【关键词】 thiazidesensitive sodium chloride cotransporter amiloridesensitive epithelial sodium channel epithelial calciumchannel rat mouse* ?$ |1 }! U. e9 E, I. H  p
                  INTRODUCTION
% X! }) Y) ~% L9 Q2 @- p; W  v  ]$ A# D+ |) s' [
IN THE LAST DECADE, THE MAJOR salt and water transport proteins in the renal distal nephron have beenidentified, and their precise intrarenal localizations were explored byimmunomethods or by in situ hybridization. The resulting data confirmedthat the inventory of the transport proteins in the distal nephron identified so far is the same in all investigated mammalian species (rabbit, rat, mouse, and human). However, the transporter topology along the distal convolution shows subtle species differences, theunawareness of which might have been the cause for occasional discrepancies in the interpretation of experimental data. Furthermore, under altered functional conditions the abundance and extension alongthe distal nephron and intracellular localization of transporter proteins change. The studies also unveiled the strikingly consistent link between the given inventory of apical transport proteins along thedistal nephron and the fine structure of the respective cells. Thispoints to the potential of structural approaches in investigatingfunctional mechanisms in distal nephron electrolyte transport in vivo.
3 ^5 X  {; L) S
7 P1 j# I4 _' D5 D; xThe detailed physiology of the transporters and their involvement inhuman pathophysiology has been discussed in depth in excellent recentreviews ( 33, 44, 53, 95, 107, 110, 114, 138 ). Here, wewill review the distributions in the distal nephron of the majorsodium, calcium, and water transport proteins and regulation of theirrelated functions, in correlation with the structural organization ofthe distal nephron, with the main emphasis on the distal convolution.
) Z$ B6 d- H1 E9 D; H# a3 N' }2 `" K) E4 p" \$ S
CORTICAL DISTAL NEPHRON/ r7 n* S% Z* J  M9 \. D. F

+ ]) W9 V3 g& ^5 `; ?' PMicroanatomic Organization of the Cortical Distal Nephron; W, a) p9 p) }" x
- r) o7 A1 M0 d4 I/ @
The anatomic definition of the distal nephron takes into accountexclusively those tubular portions that originate from the metanephricblastema: the distal straight part [thick ascending limb of Henle'sloop (TAL)], located in the medullary rays, and the convolutedportion, the "distal convolution," located in the corticallabyrinth. The functionally defined "distal nephron" also includes,besides the TAL and the distal convolution, the cortical collectingduct (CCD), which embryologically originates from the most peripheralbranchings of the ureteral bud.
+ X! m; K6 y  g0 \3 N
/ u; d% a5 F6 S) N2 d+ E; _Microanatomically, the distal convolution of superficial nephrons is asimple tube, opening into the most peripheral extensions of a CCD( 70 ). In contrast, distal tubules of deeper nephron generations merge to so-called "arcades" ( 103 ). Theextent of nephron fusion and the ratio between nephrons drainingindividually or through an arcade into a CCD vary among and withinspecies ( 35, 103 ). The arcades ascend within the corticallabyrinth, proximate to the cortical radial vessels, before they openinto a CCD within the medullary rays ( 35, 57 ). The numberof nephrons drained by each CCD averages 11 in the human kidney( 98 ), 6 in rabbits ( 58 ) and rats( 69 ), and 5 in mice ( 71 ).1 K6 C8 k3 M4 U+ @3 a/ Q0 J

( q7 q4 I( C" W: q/ LThe present conventionally used subdivision of the distal convolutioninto the so-called "distal convoluted tubule" (DCT) and the"connecting tubule" (CNT; including arcades) is based on more orless quite obvious structural differences along the distal convolution.They were initially observed by Schweigger-Seidel in 1865 ( 118 ) and disclosed in microdissected preparations of kidneys from rabbits, humans, mice, sheep, cats, pigs, cattle, anddolphins in astounding detail by Peter and Inouye in 1909 ( 103 ). Some 70 years later, Peter's light microscopicobservations were confirmed by detailed electron microscopic studies inrabbits ( 58 ) and extended to rats ( 29 ).
( |% y7 M3 M0 u& b. Z9 _+ a% l
9 X3 f8 c7 {9 q0 p+ A) eIn all species, the TAL epithelium changes at various distancesdownstream of the macula densa abruptly to the DCT, which is theinitial segment of the distal convolution. In rabbits, the entirelength of the DCT epithelium is composed by one cell type, the DCTcells. An exceedingly high density of mitochondria, encased in narrowpalisade-like-arranged, interdigitated lateral cell processes,characterizes them. The DCT cells are abruptly replaced by the"CNT" cells, which by light and electron microscopy appear"lighter" and display among other irregularly arranged basolateralplasma membrane infoldings and fewer mitochondria than do DCT cells( 58 ). Intercalated cells do not show up before thetransition to the CNT and continue all along the CNT and CCD ( 58 ). In deep and intermediate nephrons, the change fromthe DCT to the CNT epithelium regularly occurs a few cells before fusion of two tubules; hence the arcades are entirely made up of CNTepithelium ( 58 ). The transition from the CNT to the CCD isgiven by the abrupt substitution of CNT by CCD cells (principal cells).Basolateral membrane infoldings in CCD cells are restricted to the mostbasal cell portion, and the few mitochondria are normally situated inthe cytoplasm above the infoldings ( 58 ).+ x- u# `7 M3 X- Z  Q/ ~& q( d

- l0 w/ O0 m! R- U. mIn rats and mice, the situation is different. On the basis of serial1-µm sections, Crayen and Thoenes ( 29 ) reconstructed thedistal convolution of a superficial rat nephron, from its beginningshortly downstream of the macula densa to the first confluence withanother tubule. They distinguished by light microscopy andultrastructure a total of four cell types; types 1-3 correspond toDCT, CNT, and CCD (principal) cells and type 4 to intercalated cells.Type 1 (DCT) cells exclusively comprised the first part of the tubularportion. In the direction of the flow, these cells became intermingledwith intercalated cells. Then, type 2 cells progressively replaced type1 cells, and type 3 cells progressively replaced type 2 cells.Furthermore, cell height, basolateral cell membranes, and mitochondrialdensity of cell types 1-3 gradually decreased in flow direction.Because of the gradual structural changes, Crayen and Thoenes concludedthat in rats segmentation of the distal convolution can be made onlyarbitrarily. A decade later, the lack of sharp segment borders in therat distal convolution was confirmed by Dorup ( 35 ), andexcellent ultrastructural descriptions of each of the four cell typeswere given by Madsen and Tisher ( 83 ). Dorup( 35 ) also reported, in addition to the gradual segmenttransitions, ultrastructural distinctions between the DCT cells in theearly and late part of the DCT. These were corroborated by theobservation of the abundant presence of caveolin in late DCT cells( 17 ), but not in early DCT cells, consistent with thestructural abundance of caveolae on the basolateral plasma membranes ofcells in this tubular region ( 57 ).4 [" `$ S) X8 |1 ^7 ^
* R) X+ `0 L$ H: f; u
In mice (Fig. 1 ), the distal convolutionseems to be similarly organized as it is in rats; however, systematicultrastructural studies are not available. Ultrastuctural descriptionsof the transitional regions in the human distal convolution are lacking as well.* S0 @8 E/ s* @9 S

9 k* @) |+ |: [Fig. 1. Mouse cortical distal nephron portions. a, d,and g : 1-µm Epon sections. b, c, e, f, h, and i : Cryostatsections. a - c : Arrows, transition from thethick ascending limb (T) to the early distal convoluted tubule (D1); P,proximal tubule. b and c : Doubleimmunofluorescence for bumetanide-sensitive sodium-2 chloride potassiumcotransporter (NKCC2) and thiazide-sensitive sodium-chloridecotransporter (NCC). d - f : Approximatetransition from the late distal convoluted tubule (DCT; D2) to theconnecting tubule (CN). e and f : Double labelingfor H   ATPase, prominent in intercalated cells, andaquaporin-2 (AQP2), confined to connecting tubule cells. g - i : Transition from CN to the corticalcollecting duct (CD) in the medullary rays. h and i : Consecutive cryostat sections stained for epithelialsodium channel (ENaC) and AQP2. Bars = ~ 50 µm.
3 A& H* M" Q3 _) R/ K
& O& v7 ?8 ^0 {! j3 }* v1 cDistribution of Transport Proteins Along the Distal Nephron
7 Z. C3 E, V5 j1 t6 L* C, }# m9 i, s; y
+ y8 {9 d! T) A0 x7 ?The following apical transport systems have been shown byimmunomethods and/or in situ hybridization to be confined in the kidneyexclusively to the distal nephron: the bumetanide-sensitive sodium-2chloride potassium cotransporter (NKCC2) ( 62, 82, 97 ), thethiazide-sensitive sodium-chloride cotransporter (NCC) ( 3, 96, 105 ), the amiloride-sensitive epithelial sodium channel (ENaC) ( 37 ), the recently discovered epithelialcalcium channel (ECaC1; CaT2; TRPV5) ( 54, 101 ), and thevasopressin-sensitive water channel [aquaporin-2 (AQP2)] ( 66, 94 ). The basolateral sodium/calcium exchanger (NCX) ( 96, 108 ) and the plasma membrane Ca-ATPase (PMCA) ( 13, 14 ), as well as the cytoplasmic calcium-binding proteincalbindin D 28k ( 10, 13, 112 ) are, in contrastto other nephron portions, particularly abundant in some parts of thedistal nephron.# T* {4 T  q. n) Z, i

+ j7 ~+ T& Y) b" j) kMapping the apical transport systems along the distal nephron ofrabbits ( 77 ), rats ( 26, 55, 116 ), mice( 19, 65, 78 ), and humans ( 11 ) revealed theirserial arrangement. NKCC2 is confined to the TAL, including the maculadensa ( 62, 82, 97 ), and distinguishes this segment fromall others and in all species. Salt subtraction from the tubular fluidvia NKCC2 in the (water impermeable) TAL is the precondition forurinary concentration. Solute reabsorption by the subsequent distalconvolution is the premise for fine-tuning of renal electrolyteexcretion. It proceeds by the concerted action of the apicaltransporters NCC, ENaC, ECaC1, and AQP2, as well as others notmentioned in this context. Of these transporters, NCC is withoutexception the most upstream transporter in the distal convolution andreplaces the NKCC2 exactly at the structural transition from the TAL tothe DCT ( 3, 96, 97, 105 ) (Fig. 1, a - c )., `$ l: ^/ ?* S4 A7 }

$ j$ h- d4 |* |6 P; b, zWhat differs markedly among the species is the site of start-off alongthe distal convolution of ENaC, ECaC1, and AQP2 expression. In therabbit kidney (Fig. 2, top; Fig. 4, a and b ),immunostaining for ENaC starts where in situ hybridization for NCC mRNAstops ( 77 ). NCC immunostaining, made with antibodiesdirected against a synthetic peptide, is replaced abruptly by ECaC1immunostaining (Fig. 4, a and b; LoffingJ and Loffing D, unpublished observations). Immunostaining witha monoclonal antibody against a metolazone-binding protein continuesbeyond the stop of the in situ signal for some distance into the CNT( 3 ), where it colocalizes with ECaC1 ( 52 ).The abrupt start of coexpression with ENaC of the vasopressin-sensitive water channel AQP2 (Fig. 3, a and b ) sharplymarks the beginning of the CCD ( 77 ), which structurally isdiscernible by abrupt replacement of CNT cells by CCD cells (principalcells). The basolateral NCX is immunohistochemically detectableexclusively in the CNT ( 3, 108 ), and calbindinD 28k is intermediate in the DCT, rises sharply with thebeginning of the CNT, and continues somewhat more weakly along the CCD( 77 ).5 v2 Q7 q; p% I1 n% a( S
7 N% K3 ]  L; P2 T
Fig. 2. Transitional regions from the DCT to the connectingtubule in a rabbit, mouse, and human. All sections are cryostatsections. Top : rabbit consecutive sections (modified fromRef. 77 ). Left, in situ hybridization for NCCmRNA, confined to the DCT (D); right, immunostaining forENaC in CN; arrowheads, definite stop of NCC mRNA and the abruptbeginning of apical ENaC staining. Middle : mouse sections(modified from Ref. 78 ). Shown are double immunostainingfor NCC and ENaC, exclusive presence of NCC immunostaining in D1, andcoexpression of NCC and ENaC by cells in D2 (arrows). Bottom : human consecutive sections (modified from Ref. 11 ). Shown are immunostaining for NCC and ENaC, exclusiveNCC immunostaining in early profiles of distal convoluted tubule (D1),and weak NCC staining colocalized with ENaC in late profiles (D2).Bars = ~50 µm.8 B5 y, e4 \5 R
5 x+ P5 r7 b1 v1 W9 g$ W
Fig. 3. Farthest upstream sites of AQP2 in the distal nephronof a rabbit, mouse, and human. All sections are cryostat sectionsshowing immunofluorescence. a and b : Rabbitsections (modified from Ref. 77 ). Shown are doublelabeling for calbindin D 28k (CB) and AQP2, resepctively,and abrupt start of AQP2 immunostaining, coincident with sharp decreaseof CB immunostaining at the sharp transition (arrowheads) from CN toCD. D, DCT. c and d : Mouse (unpublishedobservations). Shown are double labeling for NCC and AQP2,respectively, intermingling of NCC- and AQP2-positive cells(arrowheads) at the end of DCT (D2), and beginning of the CN, where NCCdisappears. e and f : Human consecutive cryostatsections (modified from Ref. 11 ) labeled for ENaC andAQP2, respectively. Shown is the lack of AQP2 immunostaining in mostupstream ENaC-labeled CN profiles (CN*). Bars = ~ 50 µm.  d+ i" `9 w6 A9 i. s: E

) `$ U8 I) B$ \' ~; {- p5 GThe congruency of structural segmentation and distribution pattern ofapical transporters along the rabbit distal nephron are explicit. Thedistribution pattern of apical transporters along the rat distalconvolution mirrors the above-described lack of sharp structuraltransitions in this species. DCT cells in this late portion differ insome structural aspects from those in the early portion (see above) andcoexpress, in addition to NCC, the apical channels ENaC( 116 ) and ECaC1 ( 55 ). Furthermore, theydisplay a very high abundance of cytoplasmic calbindin D 28k ( 76, 105 ) and basolateral NCX ( 96 ). In maleSprague-Dawley rats, the fractional length of the late NCC-displayingtubular portion, defined by its high abundance of basolateral NCX, has been estimated to amount to ~20% of the total NCC-positive portion ( 26 ).' d6 j1 i, e$ a3 K  F5 X- h

, w2 b' U: ]5 L* BA further significant difference between rats and rabbits concerns thedistribution of AQP2 in the distal nephron. In rats, immunostaining forAQP2 has been detected to start with the break-off of NCC-staining( 77 ). Occasionally, AQP2-positive cells may beintermingled even with the last few NCC-positive cells. This observation agrees with the former structural data on intermingling ofcell types along the distal convolution ( 29, 35 ).Quantitative estimation by Western blot analysis of AQP2 inpreparations of isolated rat CNT and CCD revealed ~60% AQP2abundance in the CNT from that in the CCD ( 66 ), consistentwith weaker AQP2 immunostaining along the CNT than in the CCD of rats( 26, 66, 77 ). The CCD cells in the successive segmentcoexpress ENaC and AQP2, but not ECaC1. In the cortex of maleSprague-Dawley rats, the fractional volumes of DCT, CNT, and CCDcorrespond to ~1:1:1 ( 26 ).
! j1 z( c% l3 A& D0 C& Y1 g1 b& ~: A( v1 O5 D
In mice, the distribution of NCC, ENaC, ECaC1, and AQP2 (Fig. 1, c and f; Fig 2, middle; Fig 3, c and d; Fig. 4, c and d ) resembles that in rats. Inthe late part of the DCT, the apical transporters NCC, ENaC, and ECaC1are coexpressed ( 78 ) and, additionally, the mostdownstream NCC-positive cells might even coexpress AQP2 (Loffing J,unpublished observations). A significant abundance ofcytoplasmic calbindin D 28k, basolateral NCX ( 19, 78 ), and PMCA ( 78 ) has also been demonstrated byimmunomethods in the early portion of the DCT. Nevertheless, a markedjump in abundance of these latter proteins was consistently observed at the sites where, in addition to NCC, coexpression of ENaC and ECaC1began (Fig. 5 ) ( 78 ). Thefunctional link between apical ECaC1 and NCX and PMCA is furtherevidenced by the parallel decreases in apical ECaC1 and basolateral NCXand PMCA immunostaining along the CNT and their simultaneous cessationat the histotopographically recognized CCD( 78 ). Single cells, coexpressing ECaC1, NCX, PMCA,and calbindin D 28k, are occasionally interspersed inthe CCD epithelium ( 78 ). In female C57/BL6 mice, thefractional volumes of early DCT, late DCT, and CNT correspond to~2:1:2 ( 75 ).
, V4 g$ K3 U* _- V% \5 ]* Z3 g6 t2 X7 K) d- }) S2 Y
Fig. 4. NCC and ECaC1 in the distal nephron of rabbit and mouse(unpublished observations). All sections are cryostat sections showingdouble immunostaining with a rabbit-anti-NCC antibody (characterized inreference Ref. 11 ) and a guinea pig anti-ECaC1 antibody(characterized in Ref. 54 ). a and b :Rabbit sections. Arrowheads, abrupt replacement of apical NCC by apicalECaC1 labeling. Basement membrane staining is due to binding of rabbitimmunoglobulins to the secondary anti-rabbit antibody. c and d: Mousesections. Shown are strong apical coexpression of NCC and ECaC1 in lateD2 profiles and cytoplasmic labeling for ECaC1 in NCC-negative CNprofiles. Bars = ~ 50 µm.% P, E2 w3 X1 x4 J0 W. d! ~/ _
% X9 _* C. e+ c0 B
Fig. 5. ECaC1 and basolateral sodium-calcium exchanger (NCX)double immunolabeling in mouse sections (modified from Ref. 78 ). All sections are cryostat sections. Shown are theabrupt onset of apical ECaC1 staining in D2, coincident with abruptrise in NCX immunostaining (arrowheads) and gradual translocation ofECaC1 staining from apical toward cytoplasmic sites in the CN,paralleled by a decrease in basolateral NCX staining. Bars = ~50µm.
. a. f/ _4 p- _" Y5 U; w5 d$ H2 I4 Y( ]9 @8 n/ S+ B# U% J
In humans, the tubular portion with overlap of NCC and ENaC is rathershort. NCC characterizes ~30-35% of profiles of the distalconvolution and ENaC ~70-75% ( 11 ). AQP2 does notdirectly supersede the NCC, but the initial ~15% of the CNT [atleast the portions before the fusion with arcades display ENaC alone(Fig. 3, e and f )] ( 11 ). Thedistribution of ECaC1 along the human nephron has not been assessed byimmunostaining so far. Similarly to mice, NCX, PMCA, and calbindinD 28k are traceable to varying extents even in the initialportion of the DCT, and, in pronounced difference from all otherspecies, continue in significant abundance along the corticalcollecting duct ( 11 ).
+ o* L& R5 B, N4 P' [' [+ t% Y( S  Y" j& E9 ~" s" x# @# Y" ?4 r/ r
Definition of the Distal Nephron Based on Distribution ofTransport Proteins) o! E7 V/ k6 @4 H: c: s; t/ U

! u8 ~; o" q3 u$ W( RThe clear-cut structural and functional organization of the rabbitdistal nephron constitutes a simple model, which aids in anunderstanding of the more complex distal nephron organization in rats,mice, and humans (Fig. 6 ). In all fourmammalian species analyzed so far, of the proteins discussed in thisarticle, NKCC2 is the most upstream apical salt transporter andunequivocally defines the TAL. The succeeding salt transporter is theNCC, the onset and end of which in the distal convolution ofall species are clearly discernible, and which defines the DCT. Thenext apical ion transporters in the series are ENaC and ECaC1. Inrabbits, they abruptly replace NCC in the apical membrane. In rats,mice, and humans, ENaC and ECaC1 seem to be "pushed" more or lessupstream along the distal convolution into the NCC-displaying lateportion of the DCT, giving rise to a portion with apical coexpression of NCC, ENaC, and ECaC1. Accordingly, in these latter species the DCTcan be further subdivided into an early and a late portion. Besides thecoexpression with NCC of the apical transporters ENaC ( 11, 79, 116 ) and ECaC1 ( 55, 78 ), the second portion differsfrom the first, e.g., by the presence of intercalated cells ( 35, 83 ), by very prominent cytoplasmic calbindinD 28k ( 76, 105 ), by basolateral NCX( 96 ) and PMCA ( 78 ), as well as by discretestructural differences ( 35 ). The conspicuous onset ofbasolateral NCX immunostaining in the NCC-positive segment of the rat( 96 ) prompted the groups of Bachmann ( 3 ) andEllison ( 40 ) to propose the subdivision of the DCT intoDCT1 (NCX negative) and DCT2 (NCX positive). According to thiscriterion, neither in mice ( 19 ) nor in humans could suchsubdivision of the DCT be made. The onset of traceability of ENaC andECaC1, as far as has been investigated to date, coincides approximatelywith the marked start (rat) ( 96 ) or rise (mouse)( 78 ) in NCX, PMCA, and calbindin D 28k immunostaining. The coexpression of NCC and ENaC has been used as wellto distinguish the late (~DCT2) from the early (only NCC expressing)DCT (~DCT1) ( 78 ). That the late part of the DCT variesin length among species, probably also among species strains, possiblyalong with age, sex, and other factors, has been emphasized in aneditorial comment by Wade ( 134 ).
3 y* ~7 I. |) M3 G. [
6 _. r. Z+ A) O$ X. JFig. 6. Distribution patterns of apical and basolateral transport proteinsalong distal nephron in rabbit, rat, mouse, and human (modified fromRef. 11 ). G, glomerulus; TAL, thick ascending limb; CNT,connecting tubule; CCD, cortical collecting duct; MR, medullary ray.Colored bars, schematic representation of transport protein locations;arrows, continuation of given transporter along the CCD; light shading,consistent weak immmunostaining (for axial gradients in proteinabundance and changes in intracellular locations, see text). Apicaltransport proteins: light green, NKCC2; dark green, NCC; red, ENaC;orange, ECaC1 (not yet traced in humans); blue, AQP2. Basolateraltransport proteins: orange, coexpression of plasma membrane Ca-ATPase(PMCA) and NCX. Site of onset of immunostaining varies among and withinspecies.! c" ?/ }5 c( v4 }% y* r

. O6 n, r3 S6 |* E) N" BThe end of NCC immunostaining can be used in all species to mark thebeginning of the CNT. In rabbits, it comes along with the onset of ENaCand ECaC1 and basolateral NCX immunostaining and in rats and mice withonset of additional AQP2 coexpression (see Fig. 6 ). In all species, thepresence of AQP2 and the histotopographical location of the tubule inthe medullary ray (or in superficial nephrons the proximity to therenal capsule) defines a CCD. The overall length of the distalconvolution (DCT and CNT) can be conveniently estimated by doubleimmunostaining for NCC and NCX ( 26, 96 ) or NCC andcalbindin D 28k ( 11 ).! g* l, A5 T+ H0 B
. V1 A! R/ j9 d! u" L6 v) h1 w: E
Relevance of Specific Transporter Topology
1 A" _6 ^' E3 p, w/ R
/ M  i. K! F4 v) NThe specific sequence of electrolyte transporters along the distalnephron probably guarantees sodium recovery under a large range ofphysiopathological situations. It seems to imply the possibility ofpartially compensating for inadequate salt reabsorption by adaptingsalt reabsorption via other and differentially regulated salt transportsystems in downstream tubular portions. For instance, impairment ofNaCl reabsorption in the TAL, by whatever means, drastically increasesthe NaCl load in the downstream segments of the TAL, which respond withhigher salt reabsorption rates, and over longer periods, with increasedtransport capacity and associated epithelial hypertrophy( 70 ). Such compensatory mechanisms along the nephron arethought to occur also under impaired sodium uptake due to mutations ofgenes for some transport proteins (e.g., NKCC2 in Barrter syndrome, NCCin Gitelman syndrome) ( 49, 107 ).& T7 @. u# v: f8 x

. X5 P$ d1 w; E+ rAlthough at first glance the species differences in the distributionpattern of transporters appear to be rather trivial, their functionalrelevance might be substantial. The definite (rabbit) or gradual (rat,mouse, human) changes in the distribution of specific transportpathways along the distal nephron are also reflected in respectivedistribution patterns of sensitivities to various peptide hormones( 87 ). For instance, the sites of vasopressin sensitivityalong the distal nephron, assessed in preparations of isolated distaltubules from rabbit, rat, mouse, and human kidneys ( 87 ),match precisely in all cases the respective sites of AQP2 occurrence.The upstream shifting of vasopressin-sensitive water retrieval into theENaC-displaying CNT may be functionally relevant for at least tworeasons. It might favor ENaC-mediated sodium entry into the cells, bycreating a favorable gradient for sodium entry by water subtractionfrom the (salt diluted) tubular fluid in the CNT. The differentialhormonal control of AQP2 and ENaC activity opens subtle and complexpossibilities for regulatory interactions. Furthermore, the AQP2upstream shifting of vasopressin-regulated water subtraction in theCNTs might go along with a considerable gain in the overall waterreabsorption ( 66 )., {' Y8 n0 N2 y

4 n2 h! p  W0 R0 X2 kThus the final result of solute excretion depends not only on thepresence and abundance of given transport proteins in the kidney butalso on their species-specific topological arrangement.
0 S, ]6 L% p  B. M, H' }
8 R' a* c% J5 c& _; Y# l; W7 m7 ?CONTROL OF TRANSPORT ACROSS APICAL PROTEINS
6 ?8 }/ C2 Y3 G, M1 `8 G
6 @' m# `+ x' U2 V" {The differences in the specific distribution patterns of transportproteins are probably evolutionary responses to species-specific livingconditions. Evidently, within this given frame there is room forindividual adaptation to the actual needs. Immunomethods and/ormorphology can disclose the nephron sites in which, under specificfunctional conditions, in vivo adaptive changes took place.
+ G, x/ F1 U! H* J9 T$ A, h7 X
/ U" F% V2 q4 x0 E$ E, t/ G9 c, U" r6 BRegulation of NCC-Mediated Salt Reabsorption
8 d6 J# i1 B% v8 e+ _* f3 g% x* X$ n/ @  t
NCC mediates electroneutral NaCl uptake into the DCT cells.Because renal NCC is confined to the DCT, data on NCC derived fromwhole-organ homogenates truly reflect corresponding changes in NCCabundance in this segment. As long as specific anti-NCC antibodies werenot available, abundance of NCC was often assessed by its specificbinding to metolazone, a thiazide-like diuretic ( 5 ).Changes in metolazone binding protein, NCC abundance, or NCC mRNA havebeen observed under many different pathophysiological or clinicalsituations (for a review, see Refs. 47, 67,and 107 ). What might be the common denominator under thedifferential conditions, controlling specifically apical NCC abundanceand NCC-mediated cellular salt uptake?
  @# K3 c/ y( }, h% G3 S
4 b. E4 z* h6 _4 kTubular sodium load and flow rates. Some data from the time when neither[ 3 H]metolazone-binding studies nor NCC antibodies wereavailable suggested that tubular salt load and/or flow rate in the DCTdirectly controls sodium reabsorption in the DCT. Rabbits under chronichigh-sodium, reduced-potassium diets (with respective chronic very lowendogenous plasma levels of aldosterone) revealed marked hypertrophy ofthe DCT epithelium, indicating chronically upregulated transportcapacity in the DCT epithelium ( 59 ). Micropuncture studiesin rats under furosemide-induced impairment of NaCl reabsorption (withsimultaneous replacement of salt and fluid loss) confirmed that themarked increases in surface basolateral plasma membrane and inmitochondrial volume in the distal convolution cells mirrorcorrespondingly increased sodium reabsorption rates ( 40, 60, 122 ). In situ hybridization ( 96 ) as well as RNAseprotection assays ( 140 ) disclosed upregulation of mRNA forNCC under the given conditions. Others were unable to detect anyincrease for NCC mRNA ( 1, 88 ). Indications for NCCmRNA-independent regulation of NCC protein abundance, e.g., bymechanisms that control protein translation and/or stability, have beenobtained in some studies ( 1, 137 ), but so farintracellular processing and trafficking of NCC in vivo have not beenaddressed. Studies in rats under the above experimental conditionsdemonstrated the involvement of IGF-1 and IGF binding protein 3 in thehypertrophy of the DCT epithelium ( 68 ).
  v9 H: F9 j- V( Y6 Z  h
1 k+ A" h! \# g; V0 \Steroid hormones. mineralocorticoids and glucocorticoids. In rabbits, drastically increased endogenous aldosterone levelsassociated with chronic low-sodium, high-potassium intake did notincrease the transport machinery in the DCT but did markedly in the CNT( 59 ). Adaptive structural and functional hypertrophy inthe DCT epithelium after furosemide-induced inhibition of salt reabsorption in the TAL occurred in adrenalectomized rats with clampedplasma levels of mineralocorticoid and glucocorticoid hormones tosimilar extents as in intact rats ( 60, 122 ). These collected observations suggested that changes in salt delivery and flowrate by themselves might be sufficient to initiate modulation of salttransport rates in the DCT. Nevertheless, several studies advocated theimplication of aldosterone in the control of apical NCC abundanceand/or NCC-mediated salt reabsorption. In rat kidneys, low dietarysodium intake, which promotes rises of endogenous aldosterone plasmalevels, as well as exogeneous aldosterone application increased theabundance of NCC protein, as assessed by Western blotting andimmunohistochemistry ( 64 ). Increased binding of [ 3 H]metolazone to kidney homogenates was observed afterexogenously applied aldosterone ( 22 ) but not with dietarysodium restriction ( 21 ). Velazquez et al.( 128 ) observed that the low electroneutral sodiumtransport activity, measured in vivo in the early distal convolution ofadrenalectomized rats, can be restored to normal and further increasedby replacement of aldosterone and/or high doses of glucocorticoids( 128 ). The increase in NCC-mediated sodium transport andNCC protein after furosemide treatment or dietary sodium restrictionwas significantly lower when aldosterone receptors were blocked byspironolactone ( 1, 93 ). This experimental setting providedindirect evidence for the role of aldosterone in the observed changes;however, the possible contribution of systemic or upstream renalparameters to the given observations has not been experimentally excluded.( H0 R; S8 j+ ]: Y/ |+ b" y- V
  x- g' n/ Z: M1 a0 O; V* i
MINERALOCORTICOID RECEPTORS AND11 - HYDROXYSTEROID DEHYDROGENSASE TYPE 2. Canonically, a direct role of mineralocorticoid hormones in the controlof apical NCC abundance and of NCC-mediated NaCl transport requires thepresence in the DCT of mineralocorticoid receptors (MR) and of theenzyme 11 -hydroxysteroid dehydrogensase type 2 (11 -HSD2), whichconfers mineralocorticoid specificity to the MR by the rapidmetabolization of glucocorticoids. MR and 11 -HSD2 have been revealedby various methods in the distal nephron of rats, rabbits, and humans(for a review, see Refs. 42 and 107 ) and11 -HSD2 also in mice ( 19 ). Whereas MR seem to bepresent all along the rat DCT and CNT ( 15 ), 11 -HSD2 isnot detectable in the early (ENaC-lacking) portion (DCT1) but is welldetectable in the second DCT portion (DCT2), in the CNT, and CD( 15, 116 ). These observations indicate that the effects ofaldosterone on NCC-mediated sodium transport might differ along theaxis of the DCT, and they corroborate the suggestion that the DCT mayexhibit some promiscuity with respect to the selectivity formineralocorticoids and glucocorticoids ( 107 ). Reilly andEllison ( 107 ) proposed that the latter may stimulateNCC-mediated sodium transport primarily in the early DCT (DCT1),whereas aldosterone may stimulate sodium transport predominantly in thelate DCT (displaying NCC and ENaC ), where MR and 11 -HSD2 are bothhighly expressed. Putatively, control of NCC abundance by aldosteronevia MR-independent noncanonical pathways is conceivable.
+ h% @1 L  S: u& h. X, s
+ }6 l: |6 E  z" xSEX HORMONES. Sex hormones seem to affect directly or indirectly NCC-mediated salttransport. RT-PCR ( 74 ) and autoradiographic ( 30, 124 ) experiments pointed to the expression of estrogen receptors in rat kidneys. Chen et al. ( 23 ) and Verlander et al.( 130 ) demonstrated that estrogens given to ovarectomizedrats increase [ 3 H]metolazone binding sites and NCCabundance, respectively, in the renal cortex. These observations wereused to explain why female rats respond with a more pronounced diuresisto thiazides than do male rats ( 23 ). Whether increasedNCC-mediated sodium transport contributes to the sodium retention seenunder estrogen therapy (e.g., the contraceptive pill) is not known." q  Q; M+ E) w8 I1 ^8 k) A: E
3 X, ]5 t" a% f4 l- b
Peptide hormones. The presence of receptors for peptide hormones such as calcitonin, PTH,and isoproterenol had been shown indirectly by the increases in cAMPactivity after application of these hormones to morphologicallydefined, microdissected distal tubule segments ( 87 ).Blakely et al. ( 12 ) reported increased[ 3 H]metolazone binding in kidney homogenates ofcalcitonin-treated rats./ k0 U/ _0 v+ n4 ]" g

! ]0 C" `5 q$ E$ i0 o) MAngiotensin II is involved in the control of arterial blood pressureand whole body sodium homeostasis. Independently from its effect onadrenal aldosterone secretion, angiotensin II stimulates sodiumreabsorption in the distal tubule ( 135 ). It has beendiscussed that the hypertrophy in the DCT under increased salt load ofthe DCT (after furosemide treatment, combined with high salt and water load) might be ascribed, in part, to an effect of angiotensin II( 6 ). The role of angiotensin II in NCC regulation can be deduced from recent experiments in mice with targeted disruption of theangiotensin II type 1 (AT 1 ) receptor ( 18 ).Unlike wild-type mice, these AT 1 knockout mice lackincreases in NCC protein abundance in response to a low dietary sodiumintake. Thus in the rodent DCT part of the adaptation to low dietarysodium intake is possibly mediated by increased angiotensin II levels.) ~; m9 Y6 G8 F+ @1 C& D4 R

0 o" c! ^; z2 z3 VInhibition of NCC-mediated transport. Thiazide diuretics are frequently used in the treatment ofhypertension. Thiazides inhibit the uptake of NaCl into the DCT cell bybinding to NCC. Prolonged reduction of sodium entry into the cellsresults in chronically lower transport rates, structurally reflected bylowering of the epithelium with a reduction of the active-salttransporting machinery ( 70 ). In the DCT of mice, treatmentwith thiazides is indeed associated with lowering of DCT epithelium(Valderrabano V and Loffing J, unpublished observations), andNCC knockout mice have a marked hypotrophy of the DCT epithelium ( 117 ). However, treatment of rats with metolazone for 3 days or with hydrochlorothiazide induced massive apoptosisexclusively in the early DCT ( 76 ). In rats treatedcontinuously for 2 and 4 wk with metolazone, the epithelium of theearly DCT was markedly simplified, displaying fewer basolateralmembranes and mitochondria, and de- and regenerating cells. Theseobservations suggest chronically lower transport rates and corroborateformer data by Morsing et al. ( 89 ), who measured thatpost-chronic in vivo blockade of NaCl transport by thiazides reducedtransport capacity of rat distal tubules, despite a significantlyincreased number of thiazide binding sites. The authors pointed outthat increases in the number of thiazide receptors are not necessarilysynonymous with increases in transport activity. The different effectsof thiazides on the DCT in rats (apoptosis) and mice(hypotrophy) once again emphasize species differences, even betweenclosely related species.
: `0 ~  f$ l9 O6 }" I( l& {8 P* w
* o% [' E" \+ D, ?0 v- SRegulation of ENaC-Mediated Sodium Transport
6 }+ N+ o1 Y) C! O4 Y" A
- r! Q1 A& T3 b) s% i- o$ _# iThe second major sodium transporter in the distal convolution isENaC. ENaC is the major player in aldosterone-regulated sodium reabsorption, not only in the kidneys but also in other organs (e.g.,distal colon, salivary glands) ( 131 ). In the kidney, all ENaC-positive tubule portions have been subsumed under the term "aldosterone-sensitive distal nephron" (ASDN) ( 81 ),which comprises the CNT and CD in all species and includes, in somespecies at least, the late part of the DCT (see above). The essentialrole of ENaC in control of salt and volume homeostasis is highlighted by the fact that some hereditary forms of severe salt-sensitive arterial hypertension (Liddle's syndrome) and severe renal salt wasting (pseudohypoaldosteronism type I) are related to gain- andloss-of-function mutations, respectively, of ENaC genes (for a review,see Ref. 110 ).& ]& _' i4 @" X' r' n2 O1 I
8 W& G% o& r( r6 E
Sodium uptake into the cell via ENaC is electrogenic and can bemeasured by corresponding sodium currents that are inhibited byamiloride. ENaC-related sodium transport favors potassium secretion, which proceeds most probably via the apical renal outer medulla potassium channel ROMK, coexisting with ENaC in the same cells ( 136 ). Putative potassium reabsorption by intercalatedcells, which are regularly interspersed among the ENaC-expressingcells, might modify the rigid link between sodium reabsorption via ENaC and K secretion via ROMK.
- g0 f' i, I7 h" ?4 P% Q
$ N( u$ q1 K8 [! ]# _& bENaC is composed of three subunits (,, ) ( 110 ).All three subunits have been located along the ASDN ( 37 ),although in differential imunnohistochemical abundance andintracellular sites ( 50 ). Interestingly, -ENaC, but not - and -ENaC, has been evidenced by RT-PCR in microdissectedrabbit DCTs; however, amiloride-sensitive sodium currents could not bedetected in this segment ( 129 ). Expression of -ENaCmRNA has been also revealed by in situ RT-PCR in the mouse TAL and DCT( 25 ).5 f. \) C% K' B- K" Q. X

5 k' a# D" z1 U9 A0 ^9 ]Regulation of renal sodium transport via ENaC could intervene 1 ) directly at the level of single channels in the membrane, by changing the channel gating kinetics; in vitro studies in amphibian cells and heterologous expressions systems suggested this possibility (for a review, see Refs. 38 and 48 ), but toour knowledge, such effects have not been described so far for the ASDNin vivo; or 2 ) at the level of density of functional apicalchannels. This can result either from altered synthesis rates of ENaCsubunits and/or from altered exo- and endocytosis rates of alreadypresent ENaC subunits.
( W8 L  L: j' m9 {% Z6 `
, b" i3 j. a3 S0 v$ \Role of synthesis of ENaC subunits. Although the exact stoichiometry of the subunits in the channel isstill debated, there seems to be consensus that the -subunit plays apivotal role in the assembling of functionally active channels( 120 ). In the kidney, primarily the abundance of -ENaC appears to be regulated by aldosterone. Exogenous aldosterone application increases the abundance of -ENaC at the mRNA and proteinlevel ( 84, 123 ). Dietary sodium restriction, which increases endogenous aldosterone production, has been shown to induce -ENaC in some ( 84, 85, 140 ) but not all studies( 109, 123 ). Based on previous studies in Xenopuslaevis A6 cells, it has been proposed that the induction of -ENaC might be a prerequiste for the apical translocation of ENaC( 86 ). Sufficient availability of -ENaC may allow fullassembly of ENaC channels and their subsequent release from theendoplasmic reticulum and delivery to the cell surface. The inductionof -ENaC (at least in nonadrenalectomized rats) in response toaldosterone, however, is rather small, and it is conceivable that invivo the induction of -ENaC alone does not account for the apicaltargeting of all three ENaC subunits. Interestingly, Nielsen andco-workers ( 93 ) recently reported that MR inhibition byspironolactone blunts the induction of -ENaC but does not preventthe apical redistribution of all three ENaC subunits in response to alow-sodium diet. Short-term adaptation of the kidneys to 24 h ofdietary sodium restriction apparently does not involve significantupregulation of -ENaC ( 85 ). However, amiloride-sensitive renal sodium reabsorption was shown to be significantly increased after 4 h of sodium restriction( 46 ).+ B2 z" F; L: L& B9 ?: B, w

# q# M2 N& S5 K( G- C6 {! yOther hormones besides aldosterone affect the synthesis rate of ENaCsubunits as well, and thereby they might also intervene in the controlby aldosterone of ENaC-mediated sodium reabsorption. For example, inthe rat kidney in vivo chronic increases in vasopressin levels induce - and -ENaC at the mRNA level ( 92 ), in agreement with studies in rat CCD cells in vitro ( 34 ). Similarly,water restriction in normal rats and chronic infusion of dDAVP (astable analog of vasopressin) into Brattleboro rats (a rat strain that lacks endogenous vasopressin) increase - and -ENaC proteinabundance ( 39 ).- i' U# v+ i" o  K$ O5 t
$ L' B5 F7 Z0 J. J# n0 k+ D6 z7 J' J
Angiotensin II rapidly stimulates ENaC activity in isolated distalnephron segments ( 135 ), an effect most likely mediated byAT 1 receptors ( 104 ). Such acute effects ofangiotensin II are unlikely to be mediated by an increased ENaCsynthesis rate, but recent experiments in AT 1 receptorknockout mice suggest that angiotensin II might have a direct impact onENaC abundance as well. Despite elevated plasma aldosterone levels, theknockout mice express less -ENaC than wild-type mice( 18 ).% e3 s% P0 W  d* ~0 k

9 F  f2 y: `3 _$ X4 f6 F4 O* j8 f# JRegulation of ENaC activity by redistribution of ENaC subunits. In rodent kidneys, dietary sodium restriction causes animmunohistochemically traceable redistribution of ENaC subunits from intracellular compartments toward the apical cell surface ( 79, 84 ). These observations correlate well with previous patch-clamp studies in isolated rat collecting ducts by Pacha and co-workers ( 99 ), who recorded increases in the number of open sodiumchannels in the apical plasma membrane after 1 wk of dietary sodiumrestriction ( 99 ). The increases in active channels were inparallel to increases in endogenous plasma aldosterone levels( 99 ) and were observable even within 15 h aftersodium restriction ( 45 ). Immunohistochemistry revealedthat in adrenalectomized rats within less than 4 h after onesingle aldosterone injection, the apical ENaC density increased ( 81 ). The rapid upregulation of apical ENaC activity andabundance indicates that such changes might be relevant not only forthe long-term adaptation of renal sodium excretion but also in the adaptation to circadian variations of dietary sodium intake( 45 ).
8 q3 F' ~& Y7 v( w# Y7 E. _( v0 x- S, M
The molecular mechanisms underlying the rapid accumulation of ENaC inthe apical cell surface are not well understood. Data suggestinvolvement of the serum and glucocorticoid-regulated kinase SGK1,which is an aldosterone-induced protein ( 20, 90 ). SGK1induction by aldosterone is clearly dose dependent ( 119 ) and occurs even under small variations in endogenous plasma aldosterone levels provoked by alterations in dietary sodium intake( 56 ). Coexpression of ENaC with SGK1 in X. laevis oocytes manifestly stimulates ENaC activity ( 20, 90 ) and cell surface abundance ( 32 ). The latter isthought to be related to SGK1-dependent phosphorylation andinactivation of the ubiquitinligase Nedd4-2 that downregulatesENaC ( 31, 121 ). The importance and regulation of SGK1 andNedd4-2 under various experimental conditions are summarized inseveral recent reviews ( 61, 73, 80, 100, 120 ).
: q$ i5 \6 j$ Y* z% w# {  t
2 ?7 W8 r( x& D5 ^The induction of SGK1 by aldosterone, detectable by immunomethodsexclusively in the ENaC-positive DCT2, CNT, and CD cells ( 81 ), precedes the apical targeting ( 81 ) andfunctional activation of ENaC ( 9 ). An in vivo role of SGK1in ENaC regulation can be deduced from recent findings made in SGK1knockout mice ( 141 ). However, the salt-loosing phenotypeof the SGK1 knockout mice is mild compared with that of MR knockoutmice ( 7 ) or that of the -, -, or -ENaC knockoutmice ( 110 ). These observations in SGK1 knockout micesuggest that SGK1 might be an important, but not the sole, playernecessary for apical ENaC activity.6 i- j; d( o9 M& b9 |" E
: N8 `, C3 h) f2 D" q7 v
Axial differences in ENaC density. Under standard conditions of sodium intake, all three subunits aretraceable in the apical membranes only in the initial parts of the ASDN( 77, 79 ). Farther downstream in particular, - and -subunits seem to vanish from the apical membrane and become increasingly prominent in intracellular compartments ( 50, 77, 79, 84 ).
0 a, Y  y$ i! Z8 p) u  a3 \/ c( i6 l7 l+ Y
Former studies on net sodium reabsorption in microperfused rat tubules( 27, 106, 125 ) and in isolated rabbit tubules( 2 ) demonstrated that in upstream portions of the ASDN(late DCT and CNT; formerly designated together as "late distaltubule"), sodium transport was consistently present and several timeshigher than in CCDs located downstream. The axial decrease in thebasolateral Na-K-ATPase ( 63 ) and the morphologicalcorrelate of the sodium extrusion apparatus ( 59 ), i.e.,the basolateral membrane infoldings and the mitochondrial volumes, alsomirrors progressive decreases in the sodium transport rates along theASDN. The functional and morphological data match the progressiveimmunohistochemical decrease along the ASDN of the apical expression ofENaC. Under chronic high-salt intake, associated with low aldosteronelevels, apical ENaC is barely detectable, even at the very beginning ofthe ASDN ( 79 ). Under moderate standard salt intake byanimals in European laboratories, apical -, -, and -ENaC arefound to extend at best along the DCT2 and the early CNT ( 77, 79 ). Under salt restriction, inducing rises in endogeneousaldosterone plasma levels, apical ENaC is detectable also in the CCDand outer medullary CD ( 79 )./ D2 o' |& r% J6 Y0 m. `. q
; o5 D2 _- p0 t/ H
What causes the axial differences in apical ENaC and, consequently, insodium transport along the ASDN? Theoretically, an axially decreasinggradient of apical ENaC localization might be connected withcorrespondingly distributed MR density along the ASDN. However, formerdata by Farman et al. ( 43 ) as well as Doucet and Katz( 36 ) showed similar densities or even rather increasing MRdensities along the ASDN in rats. Equal sensitivity for aldosteronealong the collecting system is also suggested by thealdosterone-dependent induction of SGK1 and -ENaC, which occurreduniformly all along the collecting system ( 81 ). This implies that other factors superimpose the systemic changes in aldosterone levels and contribute to the differential regulation ofapical ENaC abundance along the distal nephron. Whetherdifferential sensitivities along the ASDN for other hormones, known toaffect ENaC function (e.g., insulin, vasopressin, angiotensin II),exist and whether these hormones influence ENaC surface expression are not known. Extracellular proteases [e.g., kallikrein,channel-activating protease-1 (CAP1)] ( 24, 126, 133 )secreted into the tubular fluid might contribute to axiallydifferential regulation of ENaC-mediated salt transport. Kallikrein issynthezized in the most upstream portions of the ASDN( 132 ). Prostasin, the human homologue of X. laevis CAP1, has been recently shown to be increased in urine inhumans under conditions of increased plasma aldosterone levels ( 91 ). However, the extracellular proteases seem to affectchannel activity ( 24, 126, 133 ) rather than ENaCcell-surface expression.
5 G) W, h8 o) a9 ~" {
  A+ D2 ~8 G/ ?What necessarily changes in tubular flow direction is the tubular fluidcomposition. Osmolarity changes and intra- and extracellular ionconcentrations (e.g., for sodium, calcium) have been discussed to playa role in ENaC regulation ( 48 ). Also, effects of tubular flow rate on ENaC-mediated sodium transport in isolated rabbit collecting tubules have been reported ( 113 ).% Y! Q9 P# x$ u6 n* `9 g
" I, I* y0 ?3 n' D7 R" Q3 h
Taken together, the marked gradient in apical ENaC density, assessed byin vivo experiments, agrees with functional data on sodium transport indifferent ASDN portions. The causes for this gradient along the ASDNare still unclear. The findings suggest that local factors along thetubule, presumably via the tubular fluid composition, importantlymodify the effects of systemic factors on ENaC-mediated transport.
6 }& i& F8 j5 ~
2 {% F0 Z% i" R: h+ qSites of Calcium Transport in the Distal Nephron7 m3 K% N2 [  e8 |7 d, w

2 [  e0 b; F( ~Former micropuncture experiments and studies in isolated tubulesof rabbits attributed active, transcellular movement of calcium to thedistal segments downstream of the macula densa (reviewed in Refs. 44, 53, and 107 ). Theobservation that, in humans, the tubular portion with a high abundanceof the calcium-extruding proteins PMCA and NCX, andcalbindinD 28k, is much longer than in rabbits, rats, andmice (see Fig. 6 ) suggests that, in humans, relevant transcellularcalcium transport may occur over a much longer tubular portion thanin laboratory animals, i.e., all along the distal convolution and alsoin the cortical collecting duct.
) f# [# ?0 ~8 M4 I' n* m  {$ h' B% C& u; ]$ v
The specific apical calcium channels in the distal tubules remainedelusive for a long time. Verapamil (dihydropyridine)-sensitive (cardiacL-type) calcium channels have been implicated in transcellular calciummovements ( 4, 72 ). In an immortalized mouse DCT cell line,antisense oligonucleotides directed against the 1c - or the 3 -subunits of verapamil-sensitive calcium channelsinhibited the rise in intracellular calcium concentrations in responseto PTH or chlorothiazide ( 4 ). Targeted disruption of the 3 -subunit in gene-modified mice blunted (in vivo) thehypocalciuric action of thiazides ( 8 ). These data couldsupport the hypothesis that verapamil-sensitive calcium channels indeedmay be implicated in transcellular calcium transport in the distalnephron. However, the predominantly cytoplasmic and basolateraldistribution pattern of 1c -subunits of the cardiacL-type calcium channel is more compatible with a role of these channelsin intracellular and membrane signaling processes rather than intranscellular calcium movement ( 142 ). The epithelialcalcium channel ECaC1 exhibits highly selective calcium permeability,is activated by hyperpolarization, and yet is insensitive to verapamil( 53 ). A homologous channel was identified from rat kidneyby Peng et al. ( 101 ) and called "calcium transporter2" (CaT2) ( 101 ), to distinguish it from the apicalcalcium channel CaT1 in intestine described earlier ( 102 ).
1 [6 j. ?+ w, [+ o% ]- T
" ^3 T+ i5 k1 ZInterestingly, immunohistochemical ECaC1 traceability in the kidney ofrabbits ( 52 ), rats ( 55 ), and mice( 78 ) goes precisely in parallel with immunohistochemicalabundance of PMCA and NCX in distal nephron portions. Particularly wellevident is the axial decrease in ECaC1 localization in the apicalmembrane from the most upstream ECaC1-positive portions towardcytoplasmic domains in portions further downstream in mice( 78 ) and the parallel decreases in basolateral NCX andPMCA (Fig. 5 ). These observations suggest a parallel reduction ofcalcium transport rates. Whether the intracellulary localized ECaC1molecules are recruited to the apical plasma membrane, and in responseto which stimulus, is unknown as yet. Like NCC (see above), ECaC1 alsoappears to be upregulated by estrogens ( 127 ). Consistentwith a role of ECaC1 in regulated transcellular calcium transport isthe response in vivo at the mRNA and protein level to vitaminD 3 ( 55 ) and the reduced ECaC1 expression inkidneys of 25-hydroxyvitamin D 3 -1 -hydroxylase knockoutmice ( 51 ). These observations, taken together, speak infavor of ECaC1 as the key apical calcium channel in renal transcellular calcium transport in vivo ( 53 ). However, the notion thatNCX and other proteins involved in transcellular calcium movement aredetectable, although in comparably lower abundance, in distal siteslacking ECaC1 suggests that these tubular portions and other apicaltransporters may contribute to some calcium homeostasis as well.
+ Q+ ]8 `/ q) B: B" u+ L
8 J* j- R- h/ j8 Q* j; kInteraction of sodium and calcium transport along the distalconvolution. Costanzo and Windhager ( 27, 28 ) demonstrated in a seriesof microperfusion experiments that the acute application of thiazides or amiloride has a direct stimulatory effect on transcellular calciumreabsorption in the renal distal convolution. The obvious inverserelationship of sodium and calcium transport in the distal convolutionhas been explained by two rationales. 1 ) Ion transport inhibition by thiazide and amiloride hyperpolarizes the cells, andthereby activates calcium channels within the apical plasma membrane( 44 ). The hyperpolarization of the cells is thought to beeither indirectely related to the lowered chloride entry via NCC,causing an enhanced chloride influx across chloride channels, or isdirectly related to impaired sodium entry via ENaC, respectively. 2 ) The reduced apical sodium entry lowers intracellularsodium concentration and thus may increase the driving force forbasolateral Na/Ca exchange ( 44 ). These mechanisms couldexplain the hypocalciuric effect of thiazide diuretics and amiloride,as well as the reduced urinary calcium excretion found in patients withloss-of-function mutations for NCC (Gitelman syndrome) ( 44, 115 ). However, alternative explanations for the hypocalciuriaseen under these conditions are conceivable. For example, reducedurinary calcium excretion may be secondary to extracellular volumecontraction, contributing to enhanced sodium and calcium reabsorptionin the proximal tubules ( 16, 139 ).. s; R2 L5 a+ J0 K8 @6 O, a
/ |! H* ^8 o/ e! C. A* P
CONCLUDING REMARKS
' w/ M9 `1 S6 V' A0 ~" G3 O# h1 p3 h. l7 p' I2 [
The cortical distal nephron is the site of fine regulation ofrenal electrolyte excretion. Along this nephron portion, three different salt transporters are arranged in series: the twocotransporters NKCC2 and NCC, and ENaC. In addition, within the distalconvolution ENaC is consistently coexpressed (as far as is known) withECaC1, and in some species also with the vasopressin-sensitive water channel AQP2. Because the transport activity of each of these transporters is differentially controlled, the serial arrangement mightguarantee, teleologically speaking, optimal electrolyte recovery undera vast range of environmental conditions.4 k* p! L* W* m1 E/ F6 E

; u5 `6 D& k" e# JAlthough the inventory and the basic sequence of transporters along thedistal cortical nephron are the same in kidneys of the investigatedmammalian species, there exist, however, subtle differences that mightbe functionally relevant. The distinctions pertain to the extent ofcoexpression of NCC with ENaC and ECaC1, ranging from not existent inthe rabbit to considerable in the mouse, and to the lengths of overlapof ENaC and AQP2. The species-specific distributions of transportproteins along the distal nephron coincide with the respectivestructural organization, either with sharp (rabbit) or with more orless progressive (mouse, rat human) structural segment transitions inthe distal convolution.
2 F" w% a) y/ f
1 _5 s, r1 f0 q! J0 g, H! gThese species differences present some problems. One is a semantic oneregarding criteria for subdivision of the distal convolution. Thesegments DCT, CNT, and CCD are clearly demarcated by structure andcongruent distribution of transport proteins only in rabbits. The moreor less gradual structural changes and the partial coexpressions oftransporters in the other species make segmentation a matter ofdefinition. Thus interpretation of functional data from the distalnephron needs precise criteria defining the investigated nephron portion.
' b# c% {5 T0 N( x% T7 h
' m5 k6 G1 r& q$ EA presumably more challenging problem is implicated in the functionalconsequences of the different species-specific topological arrangementsof the transporters. Any maneuver affecting a given transporteractivity in the distal nephron will result in slightly differentsecondary changes along the nephron and in the final urinaryelectrolyte excretion pattern in each species. These differences mightbe amplified by the exceedingly functional plasticity of the distalconvolution, in particular of the DCT, triggered by the manifoldfactors mentioned in this article and probably by many others notdiscussed here.7 u5 i* X0 }3 N# e: J
" v; Q/ f9 \5 z1 G. s
Thus despite the large congruence among the species with respect to theoccurrence of transport proteins in the distal convolution, thedistinct differences in the topology of transporters along the distalconvolution limit direct extrapolation of data from one species to theother. Discrepancies between the urinary excretion pattern of humanssuffering from a defined gene defect for a given transport protein andgenetically engineered mouse models with the respective defect mightlie, in part, in the different species-specific organizations of thedistal convolution and of the kidney.
7 ~$ R; h4 R6 D; t5 C
9 U$ w6 ^/ L/ Q# W9 t+ JACKNOWLEDGEMENTS. h- ^4 Q3 D5 H/ _; e$ U. K  F( @. Z, b
' y4 _+ {. d5 C/ |
The authors thank Dr. D. Loffing-Cueni for contributing Fig. 4.# C8 W; ~/ J) Q" i
          【参考文献】  d* @$ O5 ]5 r2 C( Z
1. Abdallah, JG,Schrier RW,Edelstein C,Jennings SD,Wyse B,andEllison DH. Loop diuretic infusion increases thiazide-sensitive Na   /Cl -cotransporter abundance: role of aldosterone. J Am Soc Nephrol 12:1335-1341,2001 .
1 u+ L7 F4 p, b6 C- L
" Y5 K- N9 ~- ]# I4 H+ ]+ p6 y0 g# h  {. o* D

+ W) F; p: U9 j- t6 p' [4 [- E2. Almeida, AJ,andBurg MB. Sodium transport in the rabbit connecting tubule. Am J Physiol Renal Fluid Electrolyte Physiol 243:F330-F334,1982 .8 v+ H- e3 O0 f3 `" H# x' t

2 L) R3 S- x# c* [# i1 Z% ~
% j4 p, I! m* T+ G# `! c- c9 ~: S" y9 Y; u
3. Bachmann, S,Velazquez H,Obermüller N,Reilly RF,Moser D,andEllison DH. Expression of the thiazide-sensitive Na-Cl cotransporter by rabbit distal convoluted tubule cells. J Clin Invest 96:2510-2514,1995  .
3 ], }, V  M* J; J) Z, I/ ?. t0 _
! ^4 @& W8 N2 D4 {
4 R. J) T( L5 I2 {+ L" E/ O' ^! R
4. Barry, ELR,Gesek FA,Yu ASL,Lytton J,andFriedman PA. Distinct calcium channel isoforms mediate parathyroid hormone and chlorothiazide-stimulated calcium entry in transporting epithelial cells. J Membr Biol 161:55-64,1998  .
+ I7 ^; a7 o& z$ d5 v+ z
" r9 z, u8 Q- {1 |5 S
8 V* W9 Z. ]9 m" l+ L  a  b' c% P3 K1 l4 U, b$ e5 t
5. Beaumont, K,Vaughn DA,andFanestil DD. Thiazide diuretic drug receptors in rat kidney: identification with ( 3 H)metolazone. Proc Natl Acad Sci USA 85:2311-2314,1988 .8 o' M! ?0 p7 P  G* W  @4 p4 I

' Z2 F8 D# m6 C* w3 \5 p: w1 v$ B( h% G9 \! q$ M6 b  C0 S& F% u
7 ?( o# X+ H% W, _# h9 X6 G2 H
6. Beck, FX,Ohno A,Müller E,Seppi T,andPfaller W. Inhibition of angiotensin-converting enzyme modulates structural and functional adaptation to loop diuretic-induced diuresis. Kidney Int 51:36-43,1997  .
9 ~! z' F/ F4 @/ e8 J) X
- h: c6 S" j# K: C( X/ z! v* ]
& H: g$ Q% e4 N: x
4 S/ m, I8 @1 E( G+ y7 V0 R7. Berger, S,Bleich M,Schmid W,Cole TJ,Peters J,Watanabe H,Kriz W,Warth R,Greger R,andSchutz G. Mineralocorticoid receptor knockout mice: pathophysiology of Na   metabolism. Proc Natl Acad Sci USA 95:9424-9429,1998 .
' g( c0 \* H- Z0 g; l/ M0 l
6 E% K/ j: N* J2 P8 Q% R9 X
  @- H/ a+ r  `% u+ o4 o& A& f& S- _" A# z+ D3 `0 Z2 z8 ^8 }
8. Bernardo, JF,Sneddon WB,andFriedman PA. Mice lacking the calcium channel 3 subunit are refractory to chlorothiazide (Abstract). J Am Soc Nephrol 12:27A,2001.
, s  Y  O  m, W% |7 r: h/ G1 o& v: u, z* K) k) X
0 Y9 _; w0 L2 o# `" m
8 c9 a. \9 Z9 E. ]# e
9. Bhargava, A,Fullerton MJ,Myles K,Purdy TM,Funder JW,Pearce D,andCole TJ. The serum- and glucocorticoid-induced kinase is a physiological mediator of aldosterone action. Endocrinology 142:1587-1594,2001 ./ ^0 n; f0 o' c( I, r5 d4 s% P

$ J. V- o6 u3 r6 {6 b# E3 C: F; T3 C* ?3 K, ]
' ^  a7 ^5 N% H) i' n0 H
10. Bindels, RJ,Hartog A,Timmermans A,andVan Os CH. Immunocytochemical localization of calbindin-D 28k, calbindin-D 9k and parvalbumin in rat kidney. Contrib Nephrol 91:7-13,1991 .0 _' c& {4 k1 |3 k, N8 q3 o- s

8 z% F! @- T$ \% l2 y  J$ @* n( X& B3 A& Z
, K5 t% C  m2 M: G( t* t& p
11. Biner, HL,Arpin-Bott MP,Loffing J,Wang X,Knepper M,Hebert SC,andKaissling B. Human cortical distal nephron: distribution of electrolyte and water transport pathways. J Am Soc Nephrol 13:836-847,2002 .$ E$ j8 j$ \4 _% ~" ?/ A! e, m5 h6 V

% e, Y6 M2 k' r# L! V. d/ y" |8 A

( G0 |) F. F( \+ }5 M12. Blakely, P,Vaughn DA,andFanestil DD. Amylin, calcitonin gene-related peptide, and adrenomedullin: effects on thiazide receptor and calcium. Am J Physiol Renal Fluid Electrolyte Physiol 272:F410-F415,1997 .
7 ~+ l9 w% K6 s. t4 x' p0 q! _! y' A! k8 B6 I& I" U$ O: B+ F9 q

' x, e# u% L9 i7 f# J+ X
# R6 U4 ]4 u6 M; Z6 X13. Borke, JL,Caride A,Verma AK,Penniston JT,andKumar R. Plasma membrane calcium pump and 28-kDa calcium binding protein in cells of rat kidney distal tubules. Am J Physiol Renal Fluid Electrolyte Physiol 257:F842-F849,1989 .4 d) p3 I* m8 {

; I( Y8 A/ Y& s+ S- @) t  |5 t% v$ B% f9 E

0 h% L% e5 t/ {4 b14. Borke, JL,Minami J,Verma A,Penniston JT,andKumar R. Monoclonal antibodies to human erythrocyte membrane Ca 2  -Mg 2  adenosine triphosphatase pump recognize an epitope in the basolateral membrane of human kidney distal tubule cells. J Clin Invest 80:1225-1231,1987  .
" L+ \# x8 g, w0 w; ~2 z( t
; e3 k/ E* s/ O+ O% E6 c5 R, @8 f9 A' @

9 k+ z( f2 m4 x' p1 b15. Bostonjoglo, M,Reeves WB,Reilly RF,Velazques H,Robertson N,Litwack G,Morsing P,Dorup J,Bachmann S,andEllison DH. 11 -Hydroxysteroid dehydrogenase, mineralocorticoid receptor, and thiazide-sensitive Na-Cl cotransporter expression by distal tubules. J Am Soc Nephrol 9:1347-1358,1998 .
. ]; q; J+ u) z
- k6 H; J' ?% ~9 N, l3 M) C1 N, ^

: z' C4 ^4 J# y6 `2 q& u16. Breslau, N,Moses AM,andWeiner IM. The role of volume contraction in the hypocalciuric action of chlorothiazide. Kidney Int 10:164-170,1976  .
$ g; t  b2 M) [  d% G2 T" a. b) s
- [) @; b6 E, f3 \5 U( V9 X: O% X, G
8 S/ C  n# X0 |4 j9 l( c9 j
17. Breton, S,Lisanti MP,Tyszkowski R,McLaughlin M,andBrown D. Basolateral distribution of caveolin-1 in the kidney: absence from H   -ATPase-coated endocytic vesicles in intercalated cells. J Histochem Cytochem 46:205-214,1998 .
$ |1 b9 c$ X* h+ s1 h
0 ~6 m( m7 m, s0 C/ I& j: {0 i2 ~* ~- g
' i# V: Q; x) k3 r1 l6 N5 s# v, G
18. Brooks, HL,Allred AJ,Beutler KT,Coffman TM,andKnepper MA. Targeted proteomic profiling of renal Na   transporter and channel abundances in angiotensin II type 1a receptor knockout mice. Hypertension 39:470-473,2002 .
% m# y. A! K6 Q1 a9 `2 U; a
' f4 P9 U6 c5 e2 ?6 f. [0 B4 _/ X' _+ t( A3 @2 `- P

& H# y$ t* ~# {+ h9 U* B19. Campean, V,Kricke J,Ellison D,Luft FC,andBachmann S. Localization of thiazide-sensitive Na   -Cl cotransport and associated gene products in mouse DCT. Am J Physiol Renal Physiol 281:F1028-F1035,2001 .
% m# R' q3 _4 Y/ r+ T& k, Y
" I) @0 \5 e. D, [7 I. L, i3 R& r4 b& A% _; O. @

0 y' Q" n! l6 s4 i( n20. Chen, SY,Bhargava A,Mastroberardino L,Meijer OC,Wang J,Buse P,Firestone GL,Verrey F,andPearce D. Epithelial sodium channel regulated by aldosterone-induced protein sgk. Proc Natl Acad Sci USA 96:2514-2519,1999 .; [) G/ @% K* \

. v, n5 U) ]$ `2 O' y& P: b' `6 `  a' X% Z

- S  z: s+ L; i5 P$ e21. Chen, Z,Vaughn DA,Beaumont K,andFanestil DD. Effects of diuretic treatment and of dietary sodium on renal binding of 3 H-metolazone. J Am Soc Nephrol 1:91-98,1990 .$ d5 P5 U' U: U4 f' u
' G, L9 T5 D- z9 u1 Q

' j# F& {: T+ D
& f9 |9 d% j! {( \7 ?5 b. `# }22. Chen, Z,Vaughn DA,Blakely P,andFanestil DD. Adrenocortical steroids increase renal thiazide diuretic receptor density and response. J Am Soc Nephrol 5:1361-1368,1994 .# Q8 W7 a" s) _! k  P1 B+ r7 {

- [4 [4 i$ z% g: f9 ^$ A! J, X4 f0 I% y* Q) Z) c

# `$ {( p8 N7 H" b23. Chen, Z,Vaughn DA,andFanestil DD. Influence of gender on renal thiazide diuretic receptor density and response. J Am Soc Nephrol 5:1112-1119,1994 .
# ?& U* b+ }* Z" T* d# Q1 L8 w/ T# N% _- T

5 E6 P8 F3 Q1 M1 b9 e/ K+ N: `0 f/ I  M4 F5 B  e
24. Chraibi, A,Vallet V,Firsov D,Hess SK,andHorisberger JD. Protease modulation of the activity of the epithelial sodium channel expressed in Xenopus oocytes. J Gen Physiol 111:127-138,1998 .
( F, c7 L  ^. r2 ]$ ^# c, M" j2 C/ u6 L: Q4 A7 ^% p8 y
1 G8 f+ X; y  e

% Y9 _: }5 p8 h. i9 C. N" a6 y25. Ciampolillo, F,McCoy DE,Green RB,Karlson KH,Dagenais A,Molday RS,andStanton BA. Cell-specific expression of amiloride-sensitive, Na   -conducting ion channels in the kidney. Am J Physiol Cell Physiol 271:C1303-C1315,1996 .
; g4 x: b. g& s  j  K! c* `# _/ x+ {% l7 }( Q2 q
: ?% N( K8 H/ I( `8 \4 A
# Y, w' o: w  f5 B# g) p9 ?
26. Coleman, RA,Wu DC,Liu J,andWade JB. Expression of aquaporins in the renal connecting tubule. Am J Physiol Renal Physiol 279:F874-F883,2000 .% ^$ D8 s; W; i. r) N! [: U* l& F& m& X
. e  E8 q, K) W, r$ H# N4 [; }! z

+ `% h% A7 r$ H' Z
8 _: P, C9 W7 _27. Costanzo, LS. Comparison of calcium and sodium transport in early and late rat distal tubules: effect of amiloride. Am J Physiol Renal Fluid Electrolyte Physiol 246:F937-F945,1984 .
$ T$ t/ E/ J% u) A& S) S  l2 U* k! W  i5 T# ^) R9 \0 `+ A% {
! x# \8 `$ L1 ^& R/ E# k/ `9 ?
9 z/ H; s: |% Q: A) V! U! r
28. Costanzo, LS,andWindhager EE. Calcium and sodium transport by the distal convoluted tubule of the rat. Am J Physiol Renal Fluid Electrolyte Physiol 235:F497-F506,1978.( m: \+ ?8 p8 [, }

; M( `, b2 g. z& R( D( [) ?$ s/ i4 G7 a( G8 H6 l9 a

- E. p6 s# w  \, h29. Crayen, ML,andThoenes W. Architecture and cell structures in the distal nephron of the rat kidney. Cytobiologie 17:197-211,1978  .
! M# m4 V, u# p
2 f! R- H, z* G3 m" c, L/ k  g
2 K8 L1 ~: C3 ^' P  a
( I7 |# ~9 V6 A0 l30. Davidoff, M,Caffier H,andSchiebler TH. Steroid hormone binding receptors in the rat kidney. Histochemistry 69:39-48,1980  .. W- s5 h. Q1 n2 q

0 j* P: }( ]2 U3 z0 s* h
  s$ ]0 X4 f% s- {6 x6 e# C$ G" }7 _3 m. Y9 |" h! B1 o& b
31. Debonneville, C,Flores SY,Kamynina E,Plant PJ,Tauxe C,Thomas MA,Munster C,Chraibi A,Pratt JH,Horisberger JD,Pearce D,Loffing J,andStaub O. Phosphorylation of Nedd4-2 by Sgk1 regulates epithelial Na   channel cell surface expression. Embo J 20:7052-7059,2001  .' ?; u1 P$ T; F! G  d# F

3 c6 g$ S! Q0 }. B
- o, [2 {( R3 N' n, s! i, R/ z
32. De la Rosa, AD,Zhang P,Naray-Fejes-Toth A,Fejes-Toth G,andCanessa CM. The serum of glucocorticoid kinase sgk increases the abundance of epithelial sodium channels in the plasma membrane of Xenopus oocytes. J Biol Chem 274:37834-37839,1999 .
; d1 i: D' [& I3 i+ D
0 }0 x+ N; g" a( @$ g' b* h& a4 {- y
) _2 b" W* Q* {  A4 X+ {  `
33. Delpire, E,andMount DB. Human and murine phenotypes associated with defects in cation-chloride cotransport. Annu Rev Physiol 64:803-843,2002  .
) U  }: u0 z, u9 H! B1 l9 ?, m$ S* ]( W

+ J6 ?( Q9 y/ Q8 V4 N
8 X- h* E* g8 u1 h5 T& y3 G34. Djelidi, S,Fay M,Cluzeaud F,Escoubet B,Eugene E,Capurro C,Bonvalet JP,Farman N,andBlot-Chabaud M. Transcriptional regulation of sodium transport by vasopressin in renal cells. J Biol Chem 272:32919-32924,1997 .
5 I8 y5 T8 y+ y5 w# a8 x. b, E9 P2 }0 I9 o1 n0 f

2 B. j0 z) E: U( H) p0 Q' F: z9 w$ m, q! S! {1 V  G$ W
35. Dorup, J. Ultrastructure of three-dimensionally localized distal nephron segments in superficial cortex of the rat kidney. J Ultrastruct Mol Struct Res 99:169-187,1988 .' N6 m$ o+ m" d4 T8 {
7 D. `% F3 ~0 ~) N' f

3 b) U9 J) ^  k" j% L3 I/ ^/ b* Z  |1 F; \* T2 z
36. Doucet, A,andKatz AI. Mineralocorticoid receptors along the nephron: [ 3 H]aldosterone binding in rabbit tubules. Am J Physiol Renal Fluid Electrolyte Physiol 241:F605-F611,1981 .8 d1 J# A7 J/ q
0 G% |0 i0 J; l

; d8 S8 z7 Y, |' `. _" a5 q# x; a1 r) r+ ?4 h& T' ~
37. Duc, C,Farman N,Canessa CM,Bonvalet JP,andRossier BC. Cell-specific expression of epithelial sodium channel,, and subunits in aldosterone-responsive epithelia from the rat: localization by in situ hybridization and immunocytochemistry. J Cell Biol 127:1907-1921,1994 .9 b$ Z. n) m* h
6 e) t# T& `. [- C7 a  G, |
  N  W. I& g- i4 K
  E0 ~- X& i6 h
38. Eaton, DC,Malik B,Saxena NC,Al-Khalili OK,andYue G. Mechanisms of aldosterone's action on epithelial Na   transport. J Membr Biol 184:313-319,2001  .
0 A( e5 x" I! N# u; u, ?: }
0 \" M0 z- E- i3 b- S2 b2 G6 n. T0 L, L' S% \
& \  N0 [. W* {" B& v/ H2 m8 g. ^
39. Ecelbarger, CA,Kim GH,Terris J,Masilamani S,Mitchell C,Reyes I,Verbalis JG,andKnepper MA. Vasopressin-mediated regulation of epithelial sodium channel abundance in rat kidney. Am J Physiol Renal Physiol 279:F46-F53,2000 .8 r  @( Y8 \* j; |
3 J  b. W* v; D5 l+ f% l; P1 s! Q
  E% ^+ H$ c# Q1 t/ H
  [8 |0 o& u: R& z, {- E- l& A
40. Ellison, DH,Velàzques H,andWright FS. Adaptation of distal convoluted tubule of the rat. J Clin Invest 83:113-126,1989  .# J) L3 z' F& l/ p0 E

+ b; v9 J; L6 O* V, _0 I9 q
7 C. P4 \' k2 {# c6 ~& B( R* [- _0 j/ F2 z$ }! L
42. Farman, N,andRafestin-Oblin ME. Multiple aspects of mineralocorticoid selectivity. Am J Physiol Renal Physiol 280:F181-F192,2001 .
! H3 K/ n; f5 W1 @
7 @$ E( A: J$ l* ]+ X& [, M! P- `0 c  H' B
( x/ `( R* R: H* }
43. Farman, N,Vandewalle A,andBonvalet JP. Aldosterone binding in isolated tubules. II. An autoradiographic study of concentration dependency in the rabbit nephron. Am J Physiol Renal Fluid Electrolyte Physiol 242:F69-F77,1982 .
2 x% e! b9 c( d; |+ I8 V3 o( a: O1 h# a0 h& n; ?5 ~

/ b. O5 ~0 q' E' t! t
+ z& ^1 K' A3 i- n7 s44. Friedman, PA. Mechanisms of renal calcium transport. Exp Nephrol 8:343-350,2000  .4 `2 {- |  x" t. i! I5 D
, w( F: F! W# U" \
+ J. S1 f2 k6 w

# G1 J2 |1 ]! I* H$ u45. Frindt, G,Masilamani S,Knepper MA,andPalmer LG. Activation of epithelial Na channels during short-term Na deprivation. Am J Physiol Renal Physiol 280:F112-F118,2001 .. @+ f5 ~1 \* E3 F% ]5 M9 Y

! l8 |0 }% A8 M4 }  a! B1 L% |3 z; ?. ]- g6 o9 [

& J& \  i3 O$ H/ y( @% F' X46. Frindt, G,McNair T,Dahlmann A,Jacobs-Palmer E,andPalmer LG. Epithelial Na channels and short-term renal response to salt deprivation. Am J Physiol Renal Physiol 283:F717-F726,2002 .2 |' ~* M" M# r9 c) c3 j
( V. u0 Y2 ]) h( V4 W+ b3 B

2 e# P6 k" m; x0 ^+ `) M3 U9 k0 v$ c) |7 K3 z5 ^: G
47. Gamba, G. Molecular biology of distal nephron sodium transport mechanisms. Kidney Int 56:1606-1622,1999  .
0 M4 a) a! C( @- u
9 U% e9 b2 v8 k/ c6 C+ r/ [: e( S' f3 Y: j" l7 E  _
" M# S- I0 b  m5 M, Y: l  }" f! X
48. Garty, H,andPalmer LG. Epithelial sodium channels: function, structure, and regulation. Physiol Rev 77:359-396,1997 .5 [* S; w' J, i0 S1 I

. q# ~% J, A1 G$ z) _. ~
; ^. Y( Q' F1 [6 u* W2 ]8 Y
/ l$ G% k, r" S1 {" \49. Greger, R. Physiology of renal sodium transport. Am J Med Sci 319:51-62,2000  .
/ e- T$ Z2 K" J$ {
9 H/ g0 Z' ~2 z0 C2 p7 p) V/ o8 }5 \2 j7 I& R; U* r

9 {; A+ {7 ~( y7 e7 h7 L50. Hager, H,Kwon TH,Vinnikova AK,Masilamani S,Brooks HL,Frøkiær J,Knepper MA,andNielsen S. Immunocytochemical and immunoelectron microscopic localization of -, -, and -ENaC in rat kidney. Am J Physiol Renal Physiol 280:F1093-F1106,2001 .
2 J. g3 w4 ^  k( W4 y, l6 C' S" j; Z3 ?* F+ j: F5 Y$ S

1 H; v" e! f. y7 h
/ n  P$ d( x- t1 }) E8 R+ r# b9 }+ ^51. Hoenderop, JG,Dardenne O,Van Abel M,Van Der Kemp AW,Van Os CH,St-Arnaud R,andBindels RJ. Modulation of renal Ca 2  transport protein genes by dietary Ca 2  and 1,25-dihydroxyvitamin D 3 in 25-hydroxyvitamin D 3 -1 -hydroxylase knockout mice. FASEB J 16:1398-1406,2002 .
6 e: `  T# Y5 X( w/ Q* S' D3 G

. S; H( \- B8 X) P+ t9 A+ N) |6 \  |& Z7 Z
52. Hoenderop, JG,Hartog A,Stuiver M,Doucet A,Willems PH,andBindels RJ. Localization of the epithelial Ca 2  channel in rabbit kidney and intestine. J Am Soc Nephrol 11:1171-1178,2000 .! z2 v' U8 J; [% e2 f' x
6 R$ x/ e. s$ D, e

+ F0 M, K9 X0 x4 D( W+ `* j2 c( t: c, e9 O9 ~
53. Hoenderop, JG,Nilius B,andBindels RJ. Molecular mechanism of active Ca 2  reabsorption in the distal nephron. Annu Rev Physiol 64:529-549,2002  .5 V5 O$ ^( F& I! `) Q; e8 d0 ^

- C, _! _. w; B$ m) f3 L4 A( [/ u# k6 s5 ^
6 P* D9 t2 m& e# J% W; x* L1 m
54. Hoenderop, JG,van der Kemp AW,Hartog A,van de Graaf SF,van Os CH,Willems PH,andBindels RJ. Molecular identification of the apical Ca 2  channel in 1,25-dihydroxyvitamin D 3 -responsive epithelia. J Biol Chem 274:8375-8378,1999 .7 O7 ^6 S7 ?* B6 N7 z

% J+ p& V' y4 @9 d8 `, P0 y; p$ O2 v7 M* }
6 Y; C  R% A/ h8 C$ V4 C
55. Hoenderop, JGJ,Müller D,van der Kemp AWCM,Hartog A,Suzuki M,Ishibashi K,Imai M,Sweep F,Willems PHGM,van Os CH,andBindels RJM Calcitriol controls the epithelial calcium channel in kidney. J Am Soc Nephrol 12:1342-1349,2001 .
5 e* q7 u: Z" ~' g4 O  Y% i, w
- @) b: d# n" O9 d. _0 K) D* l0 V& m( ^
' N' L% D0 }: ?. P
( P6 z4 L3 t5 d+ e) y) c56. Hou, J,Speirs HJ,Seckl JR,andBrown RW. Sgk1 gene expression in kidney and its regulation by aldosterone: spatio-temporal heterogeneity and quantitative analysis. J Am Soc Nephrol 13:1190-1198,2002 .
6 r  S& I# M3 y& }% I; m  p* k2 b% R; V
7 ^3 H' u: J5 w7 [, ^  F  Y
$ j! j) g, G3 P
57. Kaissling, B,andKriz W. Morphology of the loop of Henle, distal tubule, and collecting duct.In: Handbook of Physiology. Renal Physiology. Bethesda, MD: Am Physiol Soc, 1992, sect. 8, vol. I, chapt. 3, p. 109-168.3 L+ Q0 [2 E! w* I
7 z+ ^, ?( y1 p2 r' t

# @' x, N! T4 I6 g6 J4 T0 ^- R4 e5 e; j& R
58. Kaissling, B,andKriz W. Structural analysis of the rabbit kidney.In: Advances in Anatomy, Embryology and Cell Biology, edited by Brodal A,Hild W,van Limborgh J,Ortmann R,Schiebler TH,Töndury G,and Wolf E.. Berlin: Springer-Verlag, 1979, p. 1-123.
. h" j& e" ]5 O. W! G2 |: ~; Z
# g9 h: O' |- }6 Q; M/ p+ E# n4 M* f+ r

* y) l0 x: C8 V" f, P% p59. Kaissling, B,andLe Hir M. Distal tubular segments of the rabbit kidney after adaptation to altered Na- and K-intake. I. Structural changes. Cell Tiss Res 224:469-492,1982  .3 z: M8 J# _1 u* F; Q5 d) z
' g; |0 O+ |! ]/ e& k# R
$ ~9 v( {. |; q1 q! A4 o. g! g
' C8 j3 D4 `# L# p8 l
60. Kaissling, B,andStanton BA. Adaptation of distal tubule and collecting duct to increased sodium delivery. I. Ultrastructure. Am J Physiol Renal Fluid Electrolyte Physiol 255:F1256-F1268,1988 .! d) _( m: h! b/ P1 j
1 P+ q3 }6 k, C. X+ a$ G# D6 p
% Y4 v6 P) O% [
( h1 D+ \" {% v: @8 Z. u6 e
61. Kamynina, E,andStaub O. Concerted action of ENaC, Nedd4-2, and Sgk1 in transepithelial Na   transport. Am J Physiol Renal Physiol 283:F377-F387,2002 ." t( `( v6 t) Z& c' B( ^
" a7 P' `# K7 H" v/ B

% E. i2 K1 ~0 s! X1 X/ t# s2 g! s, b3 \, @8 ?) Q# ]8 R
62. Kaplan, MR,Plotkin MD,Lee WS,Xu ZC,Lytton J,andHebert SC. Apical localization of the Na-K-Cl cotransporter, rBSC1, on rat thick ascending limbs. Kidney Int 49:40-47,1996  .
1 y: o* s) n; |8 c2 C; j* F4 K/ v
# B, k; R1 N3 w8 C1 ?" X: g" p8 p# _! {$ ^9 ^" W
0 m/ G, `& j* J8 {
63. Katz, AI,Doucet A,andMorel F. Na-K-ATPase activity along the rabbit, rat and mouse nephron. Am J Physiol Renal Fluid Electrolyte Physiol 237:F114-F120,1979 .
) a; L4 T8 h7 V. _7 f0 o
; i6 x( R5 t* C  k% m
) P" a5 u8 Q9 m5 M/ }9 _8 Q' U/ [" I1 s! I& s
64. Kim, GH,Masilamani S,Turner R,Mitchell C,Wade JB,andKnepper MA. The thiazide-sensitive Na-Cl cotransporter is an aldosterone-induced protein. Proc Natl Acad Sci USA 95:14552-14557,1998 .
4 w. T+ w+ X, [0 i  v
( P( v' s# J2 N
0 [  C) v! @& [- x' Q, ~9 A( t% e* R& S8 A# m' T+ e* M8 A+ ~
65. Kim, J,Kim YH,Cha JH,Tisher CC,andMadsen KM. Intercalated cell subtypes in connecting tubule and cortical collecting duct of rat and mouse. J Am Soc Nephrol 10:1-12,1999 .
: M, A5 Z. S( j! ]" a* b5 y4 M% T( D9 {# J0 K% @/ H

  J7 ]( G/ W. j7 @6 f& K
, F9 @0 I' T# c2 M66. Kishore, BK,Mandon B,Oza NB,DiGiovanni SR,Coleman RA,Ostrowski NL,Wade JB,andKnepper MA. Rat renal arcade segment expresses vasopressin-regulated water channel and vasopressin V 2 receptor. J Clin Invest 97:2763-2771,1996  .
$ C- q% N+ c) k0 \4 m) O
1 _8 Q6 ^2 w, L1 Y, H  L" |5 w5 |- o$ J. D8 k; |2 J

- f9 V( U4 Q7 l' H- u67. Knepper, MA,andBrooks HL. Regulation of the sodium transporters NHE3, NKCC2 and NCC in the kidney. Curr Opin Nephrol Hypertens 10:655-659,2001  .
  h) d1 c, X9 q& z5 y6 }
0 b4 o' ^  z7 X4 U3 G' l& I& S0 w& R5 I7 d4 G; K; k3 x

+ h# b2 p; I: U- ^68. Kobayashi, S,Clemmons DR,Nogami H,Roy AK,andVenkatachalam MA. Tubular hypertrophy due to work load induced by furosemide is associated with increases of IGF-1 and IGFBP-1. Kidney Int 47:818-828,1995  .6 W% x  p& h) {5 s2 ~1 d

! M9 O$ L$ U5 t( m$ ?5 e7 @. ^4 u6 O) R+ l

& L( m! `1 n0 c2 A5 X. w69. Kriz, W. The architectonic and functional structure of the rat kidney. Z Zellforsch Mikrosk Anat 82:495-535,1967  .: r- O, n! X9 N/ e
8 ~1 I: D- a7 J7 @, i. ]

: x8 Q' d' i) G  I. G/ C
4 W! Z, w% N7 C. B8 n+ I7 {70. Kriz, W,andKaissling B. Structural organization of the distal nephron.In: The Kidney: Physiology and Pathophysiology, edited by Seldin DW,and Giebisch G.. Philadelphia, PA: Lippincot, 2000, p. 587-654.+ E+ D3 x7 h7 g9 c& T

  z2 {( ^4 J2 \
" @) y) q) O5 ^7 B- F
/ V* c; l9 Q# C# c71. Kriz, W,andKoepsell H. The structural organization of the mouse kidney. Z Anat Entwickl-Gesch 144:137-163,1974  .% [( _* S7 S" [) n
0 U1 A) o) K, ]+ U  {
/ @- ~3 L9 j, ~$ Q

, W6 ?% t: M& e( Z5 u) [& @: {3 E72. Lajeunesse, D,Bouhtiauy I,andBrunette MG. Parathyroid hormone and hydrochlorothiazide increase calcium transport by luminal membrane of rabbit distal nephron segments through different pathways. J Endocrinol 134:35-41,1994.& V# ~, T" L: b  b$ r
" Q* k7 ^  b0 |# L3 l

% d2 _/ X% t8 ^. J1 J! l1 U) I3 S8 ~
73. Lang, F,andCohen P. Regulation and physiological roles of serum- and glucocorticoid-induced protein kinase isoforms. Sci STKE 2001:RE17,2001 ./ c  x: R% i1 ^1 N

3 l, U! k6 n, K( Y. N+ L% \
1 j7 e7 L" k+ q, V7 V* |" B3 n1 ^7 w! N. J* U
74. Lim, SK,Won YJ,Lee HC,Huh KB,andPark YS. A PCR analysis of ERalpha and ERbeta mRNA abundance in rats and the effect of ovariectomy. J Bone Miner Res 14:1189-1196,1999  .
0 @/ a8 O! x& X3 I8 h. e. W
9 Y! K" M/ @# H  \8 O5 y; _9 U8 H; c7 b! ~( q3 r9 {
1 [6 |; }1 @: C+ G2 u8 \* @
75. Loffing, J,Aregger F,Pietri L,Bloch-Faure M,Schultheiss P,Shull G,Meneton P,andKaissling B. NCC( / ) mice lack the distal convoluted tubule 1 (Abstract). Kidney Blood Press Res 23:22A,2000.1 d- T/ w' o% m) s6 `4 n7 k+ V

. x) G: O. E$ v+ v9 U
' F" Y2 P. |+ G* H  |9 \4 a/ k+ L
76. Loffing, J,Loffing-Cueni D,Hegyi I,Kaplan MR,Hebert SC,Le Hir M,andKaissling B. Thiazide treatment of rats provokes apoptosis in distal tubule cells. Kidney Int 50:1180-1190,1996  .
6 o9 x8 ~, P5 t0 ^: o* v5 T  m; r& I- ^+ f, l1 f1 U' u* d" W
* [  r+ ~' i9 S; v* @& L, [

2 b  [+ V$ U! G77. Loffing, J,Loffing-Cueni D,Macher A,Hebert SC,Olson B,Knepper MA,Rossier BC,andKaissling B. Localization of epithelial sodium channel and aquaporin-2 in rabbit kidney cortex. Am J Physiol Renal Physiol 278:F530-F539,2000 .. M& H* L. m) {6 D

9 @0 W# a1 W% Q" Q+ P0 E/ t+ g# Y0 d8 ~' U, w

1 u8 X! v4 R! S6 I* @78. Loffing, J,Loffing-Cueni D,Valderrabano V,Klausli L,Hebert SC,Rossier BC,Hoenderop JG,Bindels RJ,andKaissling B. Distribution of transcellular calcium and sodium transport pathways along mouse distal nephron. Am J Physiol Renal Physiol 281:F1021-F1027,2001 .' {7 V+ g7 @! ^# b1 P% ]

7 o, u: x2 _9 Y& N3 ^* S) V: v
# t  n9 N/ C* C5 c1 [/ U! V: Z' M+ `4 }# K  ~( Z
79. Loffing, J,Pietri L,Aregger F,Bloch-Faure M,Ziegler U,Meneton P,Rossier BC,andKaissling B. Differential subcellular localization of ENaC subunits in mouse kidney in response to high- and low-Na diets. Am J Physiol Renal Physiol 279:F252-F258,2000 .
; t% Z3 g8 {+ ?0 @" E' Q& x! K2 g9 h) _

. ?9 q3 K+ w; V# P0 U6 m
" u9 t5 R- i& x9 C2 x1 Q) P80. Loffing, J,Summa V,Zecevic M,andVerrey F. Mediators of aldosterone action in the renal tubule. Curr Opin Nephrol Hypertens 10:667-675,2001  .4 g; c! \! P) N  A; f
1 i+ h! N5 C8 Z' ~/ l' n
% P7 ]( [5 n* H2 F! @
9 f! F$ U2 ?. `' _6 f0 C, |* D
81. Loffing, J,Zecevic M,Feraille E,Kaissling B,Asher C,Rossier BC,Firestone GL,Pearce D,andVerrey 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 .
# e+ g8 b* b; K8 o
: U- d7 T" v8 W! V# [" `. g" N
; p0 {. ?  g* z7 K& A) J2 V
, ]- P' Q" x8 v  L# Y82. Lytle, C,Xu JC,Biemesderfer D,andForbush B, III. Distribution and diversity of Na-K-Cl cotransport proteins: a study with monoclonal antibodies. Am J Physiol Cell Physiol 269:C1496-C1505,1995 .. o+ g9 A2 O3 C4 W  K# o, }
# V  _9 T. Q0 C1 r; d2 ^" G
# q, F$ J9 a7 h8 u8 d/ v
4 Y4 C9 _, l: H- @7 @" l
83. Madsen, K,andTisher CC. Structural-functional relationships along the distal nephrons. Am J Physiol Renal Fluid Electrolyte Physiol 250:F1-F15,1986 .
* N$ Q4 ~* y7 \/ y5 z, P1 x" J1 D* \2 _
- {. V; k- g: }9 m
" Q+ O" ]4 _6 y- d. }8 o
84. Masilamani, S,Kim GH,Mitchell C,Wade JB,andKnepper MA. Aldosterone-mediated regulation of ENaC,, and subunit proteins in rat kidney. J Clin Invest 104:R19-R23,1999 .6 h, M+ V2 Z, g) N0 y
* M4 n# G$ v' V  A

9 E, d9 b- m  R6 v' ?5 u" |, k+ P* a
85. Masilamani, S,Wang X,Kim GH,Brooks H,Nielsen J,Nielsen S,Nakamura K,Stokes JB,andKnepper MA. Time course of renal Na-K-ATPase, NHE3, NKCC2, NCC, and ENaC abundance changes with dietary NaCl restriction. Am J Physiol Renal Physiol 283:F648-F657,2002 .% b8 M/ p$ K1 X! i

0 \3 A( {- D# S
2 n% j) B1 t7 r  d+ P( j& D4 J/ A' u4 q
86. May, A,Puoti A,Gaeggeler HP,Horisberger JD,andRossier BC. Early effect of aldosterone on the rate of synthesis of the epithelial sodium channel alpha subunit in A6 renal cells. J Am Soc Nephrol 8:1813-1822,1997 .
! p1 O( h% n8 r7 \$ ^
# [4 d( |4 ]  Q  Q& ^$ }/ x- q
! n8 e; `9 e  |# O4 @/ d
5 N  }0 x& A) c# z0 H) ]& G1 g, p+ b87. Morel, F. Sites of hormone action in the mammalian nephron. Am J Physiol Renal Fluid Electrolyte Physiol 240:F159-F164,1981 .
* b  N* ^; ]: S7 ?; y& ~1 x/ B) @( G3 i# Q2 }

" e% M! o" a( H# K3 a6 B; y) `) n4 X
88. Moreno, G,Merino A,Mercado A,Herrera JP,Gonzàlez-Salazar J,Correa-Rotter R,Hebert SC,andGamba G. Electroneutral Na-coupled cotransporter expression in the kidney during variations of NaCl and water metabolism. Hypertension 31:1002-1006,1998 .
* j6 g* ]* P$ N7 j' M$ M  G  j  Z/ |& M; f) _. ?2 m0 C( [2 s
9 a; U0 \$ J% ]' p
8 V3 g0 ~, A8 U' A# j
89. Morsing, P,Velazquez H,Wright FS,andEllison DH. Adaptation of distal convoluted tubule of rats. II. Effects of chronic thiazide infusion. Am J Physiol Renal Fluid Electrolyte Physiol 261:F137-F143,1991 .
* ^. r, G" G9 @9 N1 F4 c% G/ b$ r! \8 O' I& l

0 k% }, J1 v" G* h; Q! t$ E
) C2 h" k+ L- I0 k$ }0 W6 i. h90. Naray-Fejes-Toth, A,Canessa C,Cleaveland ES,Aldrich G,andFejes-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 .
4 t* ?$ [6 y2 _( j: t4 J' S, d8 e. \' h% l
3 P3 @4 d' G4 Q% U( {
: d3 e( L# k% q: g8 K* H0 v& h
91. Narikiyo, T,Kitamura K,Adachi M,Miyoshi T,Iwashita K,Shiraishi N,Nonoguchi H,Chen LM,Chai KX,Chao J,andTomita K. Regulation of prostasin by aldosterone in the kidney. J Clin Invest 109:401-408,2002  .
5 I3 `1 x# J& H6 q8 b# G
& `2 w, Q$ w' I+ Z% ^) a" W  y! r6 H0 l$ p; p9 T

4 C) Q) ^5 H- @6 Z, i* Q: a# ^92. Nicco, C,Wittner M,DiStefano A,Jounier S,Bankir L,andBouby N. Chronic exposure to vasopressin upregulates ENaC and sodium transport in the rat renal collecting duct and lung. Hypertension 38:1143-1149,2001 .
& l5 Z. \! ]! g
8 X: P8 r+ E3 \
+ S* E( N, ^' {( y' O3 \2 P0 J+ i" ~2 H  b  H+ v3 K4 v) ?# o6 t
93. Nielsen, J,Kwon TH,Masilamani S,Beutler K,Hager H,Nielsen S,andKnepper MA. Sodium transporter abundance profiling in kidney: effect of spironolactone. Am J Physiol Renal Physiol 283:F923-F933,2002 .$ `( ]) G. W& C$ S

2 u( R% u7 x; M0 y' r% W9 r3 v; P2 r2 e# J7 x
- ~+ c% M4 z% Q% n6 _
94. Nielsen, S,DiGiovanni SR,Christensen EI,Knepper MA,andHarris HW. Cellular and subcellular immunolocalization of vasopressin-regulated water channel in rat kidney. Proc Natl Acad Sci USA 90:11663-11667,1993 ./ q# V) D; v1 ^; D
) N" m, `3 F  Y( N+ M

' N+ f1 h5 e% }2 X: d4 P' z' S4 N6 \6 A4 h0 {* ?
95. Nielsen, S,Frøkiær J,Marples D,Kwon TH,Agre P,andKnepper MA. Aquaporins in the kidney: from molecules to medicine. Physiol Rev 82:205-244,2002 .
7 K+ C3 s5 n, `% X' X( z/ U& e; u4 f. m/ B3 G% O) v/ O& C. v4 P
/ `3 q6 `1 _* i* @( g

0 l( n2 w, `; T# d96. Obermüller, N,Bernstein P,Velazquez H,Reilly R,Moser D,Ellison DH,andBachmann S. Expression of the thiazide-sensitive Na-Cl cotransporter in rat and human kidney. Am J Physiol Renal Fluid Electrolyte Physiol 269:F900-F910,1995 .
$ o$ K5 C( J: ~+ S
5 M& F; J8 R: ^8 z' e7 W& {
$ @8 q! _4 O$ F. d* W* \" a; e# l( u0 E' `/ K4 I5 c
97. Obermüller, N,Kunchaparty S,Ellison DH,andBachmann S. Expression of the Na-K-2Cl cotransporter by macula densa and thick ascending limb cells of rat and rabbit nephron. J Clin Invest 98:635-640,1996  .
. c/ r+ T+ |1 ^. c8 u% W8 B
. K" u; L5 d2 f9 B4 x6 W# W7 l5 H8 ?0 e, [! @, [
( I3 J! {& H. D
98. Oliver, J. Nephrons and Kidneys. New York: Harper & Row, 1968.
/ s: o: ^# Q1 d# V; [, Y2 S9 s( _% S  R: ~6 k
& i& L( s+ u0 J
7 {8 R& n& h" \
99. Pacha, J,Frindt G,Antonian L,Silver RB,andPalmer LG. Regulation of Na channels of the rat cortical collecting tubule by aldosterone. J Gen Physiol 102:25-42,1993 .: y$ _* g8 K; y6 T2 W% b

' v' S( h+ n( ?# k' `
( X8 |& ~' H! s: X. {7 @, H3 Q, G5 V8 R  B
100. Pearce, D. The role of SGK1 in hormone-regulated sodium transport. Trends Endocrinol Metab 12:341-347,2001  .
/ x; M) [$ h4 o, `# K  L! @
: g& \& P- X+ `! a
/ `5 g) C0 C4 E/ V
9 Y' Q+ m$ n, s% M9 {7 a101. Peng, JB,Chen XZ,Berger UV,Vassilev PM,Brown EM,andHediger MA. A rat kidney-specific calcium transporter in the distal nephron. J Biol Chem 275:28186-28194,2000 .
; Q5 f8 o. q( ~- r- A2 S0 a! U6 i7 G/ v
+ n/ {1 q3 L9 b2 I

! |; V% n7 H2 e102. Peng, JB,Chen XZ,Berger UV,Vassilev PM,Tsukaguchi H,Brown EM,andHediger MA. Molecular cloning and characterization of a channel-like transporter mediating intestinal calcium absorption. J Biol Chem 274:22739-22746,1999 .. N: z# S* X0 O( x
; K) m# B: ~* }6 T; o
8 j& j) l- T$ s' e  j7 }

, l, M1 N+ Y9 K. T$ _9 d; q103. Peter, K. Untersuchungen über Bau und Entwicklung der Niere. Jena, Germany: Fischer, 1909, vol. 1.' H% d: p4 ^) ^# B; Y+ Z3 r. @! ^  I
, f2 `; |; D: `& b& W/ W

; c* {  m+ ?3 L+ U8 a
2 T6 K3 h" H+ u5 @104. Peti-Peterdi, J,Warnock DG,andBell PD. Angiotensin II directly stimulates ENaC activity in the cortical collecting duct via AT 1 receptors. J Am Soc Nephrol 13:1131-1135,2002 ./ H; |2 j- f) K8 o( S8 N- ~6 f

& x6 w" O; J3 h0 v! F" d
% {" J& \0 z9 J( T5 D8 O4 Y  K0 [8 `% }, M$ S+ t5 D' `
105. Plotkin, MD,Kaplan MR,Verlander JW,Lee WS,Brown D,Poch E,Gullans SR,andHebert SC. Localization of the thiazide sensitive Na-Cl cotransporter, rTSC1, in the rat kidney. Kidney Int 50:174-183,1996  .
3 u2 f$ j; E/ u, H, K+ `  H
4 o' b3 L& h( i" L: A8 N
% u. N2 u/ n5 D0 K
3 Y( A# v9 _7 A2 o106. Reif, MC,Troutman SL,andSchafer JA. Sodium transport by rat cortical collecting tubule: effects of vasopressin and deoxycorticosterone. J Clin Invest 77:1291-1298,1986  .! v* B7 @+ E$ I; ~6 r- a

6 X! d; {1 |+ c' a+ F0 c. E
3 q0 @2 I" w7 {9 T8 b+ ]/ A. Q$ s' F+ U0 A5 S3 B' x6 R2 u% Q
107. Reilly, RF,andEllison DH. Mammalian distal tubule: physiology, pathophysiology, and molecular anatomy. Physiol Rev 80:277-313,2000 .
9 L3 M0 U& l" q5 q6 b# @3 l1 \/ R! Q% g
1 ]; k% `8 m6 L+ q9 Z, X9 a* G% s9 Y* }5 g

. p1 R& `' T2 Z$ O" ?1 f* R108. Reilly, RF,Shugrue CA,Lattanzi D,andBiemesderfer D. Immunolocalization of the Na   /Ca 2  exchanger in rabbit kidney. Am J Physiol Renal Fluid Electrolyte Physiol 265:F327-F332,1993 .
  B" y1 t! n4 ^2 Z. V7 {3 Z) W# i

+ }+ ~: J' Q! |# X/ _: E6 m3 R( A- m4 _/ N3 n; S: D' ], V
109. Renard, S,Voilley N,Bassilana F,Lazdunski M,andBarbry P. Localization and regulation by steroids of the, and subunits of the amiloride-sensitive Na   chanel in colon, lung and kidney. Pflügers Arch 430:299-307,1995  .
  |$ y4 f9 H; k1 [5 @2 d: w7 n9 w+ y1 f( T. o4 Q& K

' w! ]% \: u% V; @8 H0 w1 Q
* Z; ?; M  k: ^0 L# C( C110. Rossier, BC,Pradervand S,Schild L,andHummler E. Epithelial sodium channel and the control of sodium balance: interaction between genetic and environmental factors. Annu Rev Physiol 64:877-897,2002  .
$ z- E' J+ }" f/ P+ l, ]' z! p* @3 U$ F- |8 V9 l

% Z- K9 S2 ~1 c. C
0 W. B. S  \& W2 I/ X% R$ M112. Roth, J,Brown D,Norman AW,andOrci L. Localization of the vitamin D-dependent calcium-binding protein in mammalian kidney. Am J Physiol Renal Fluid Electrolyte Physiol 243:F243-F252,1982 .3 o! L, i) T7 t2 U0 {6 g

. _" A( P' _! p7 d+ ?- z  B3 I* G7 F6 C1 u* i) F
. q/ z& R9 y' E) D
113. Satlin, LM,Sheng S,Woda CB,andKleyman TR. Epithelial Na   channels are regulated by flow. Am J Physiol Renal Physiol 280:F1010-F1018,2001 .
5 u( d0 x# i% n. s5 c" i- {1 G( S$ ?' B( G# V5 r" k. h

7 F  S* G5 ~6 Q' R7 _- e6 P* n( G5 Y+ {$ n+ O1 [
114. Schafer, JA. Abnormal regulation of ENaC: syndromes of salt retention and salt wasting by the collecting duct. Am J Physiol Renal Physiol 283:F221-F235,2002 .
* x/ R/ c4 m% Q. o) r. ?: S2 N' ]. Y! |
4 \& y  D: X! R' u, P) |% _
: ?0 U. z  _6 {- x5 e2 s* w
115. Scheinman, SJ,Guay-Woodford LM,Thakker RV,andWarnock DG. Genetic disorders of renal electrolyte transport. N Engl J Med 340:1177-1187,1999 .
) {$ |9 a* g; p
0 P8 l' l4 J. M4 c
- T% R6 F, i! I' C
. P) |+ W/ E+ X( }' J) w116. Schmitt, R,Ellison DH,Farman N,Rossier BC,Reilly RF,Reeves WB,Oberbäumer I,Tapp R,andBachmann S. Developmental expression of sodium entry pathways in rat distal nephron. Am J Physiol Renal Physiol 276:F367-F381,1999 .6 ?, E+ K" m3 w! K8 }9 u
1 B$ a7 C2 j) a* A9 W# }; k  n
, ]' w, C! O/ l* U

0 M! K( m0 a0 i117. Schultheis, PJ,Lorenz JN,Meneton P,Niema ML,Riddle TM,Flagella M,Duffy JJ,Doetschman T,Miller ML,andShull GE. Phenotype resembling Gitelman's syndrome in mice lacking the apical Na   -Cl cotransporter of the distal convolute tubule. J Biol Chem 273:1-6,1998 .
9 q# V3 F! F9 s7 }) E) o- E( w6 x) J
1 O! [/ X4 ?/ P7 V

) u" D7 j% L) U5 A/ \9 U118. Schweigger-Seidel, F. Die Nieren der Menschen und der Säugetiere in ihrem feineren Baue.In: Handbuch der mikroskopischen Anatomie des Menschen. Harn- und Geschlechtsapparat, edited by von Möllendorff W.. Berlin: Springer, 1930, vol. vol. VII/1, p. 1-328.
- ^; g  K. U6 M6 \/ a" w
7 ]) @8 P! s: y9 [) R
" l1 s- S  @4 h( z+ C! W1 s; |! |7 [4 o) a+ R
119. Shigaev, A,Asher C,Latter H,Garty H,andReuveny E. Regulation of sgk by aldosterone and its effects on the epithelial Na   channel. Am J Physiol Renal Physiol 278:F613-F619,2000 .6 x  h1 Y: ]+ N+ f

2 D8 ~( w5 ^7 o+ x4 X: L5 J* V. x) ?  Y) K% E
/ g: c: R$ z0 S6 h+ R" Q
120. Snyder, PM. The epithelial Na   channel: cell surface insertion and retrieval in Na   homeostasis and hypertension. Endocr Rev 23:258-275,2002 .
2 P9 ~$ l7 F5 {2 k/ s' i6 I, j
* D  Q/ I$ L7 X2 e+ W, Y9 d6 o' t* b5 Q, B# E1 F6 Q+ Q. ~4 V

) [, D) y5 ]( q8 ]3 q6 X4 y( q121. Snyder, PM,Olson DR,andThomas BC. Serum and glucocorticoid-regulated kinase modulates Nedd4-2-mediated inhibition of the epithelial Na   channel. J Biol Chem 277:5-8,2002 ., @: \* J8 h# r0 V

. C9 r" U, O1 o$ d! c% e' c
) F9 M' E7 b2 r3 a6 s; Q: W, ]8 D- b0 R1 U3 p
122. Stanton, BA,andKaissling B. Adaptation of distal tubule and collecting duct to increased Na delivery. II. Na   and K   transport. Am J Physiol Renal Fluid Electrolyte Physiol 255:F1269-F1275,1988 .( O* u1 \1 |' p! M0 `, D

# R( D/ z" e# o5 {/ z! A, V, z! q5 G4 A

7 J" B! C* v1 k3 q2 {% s$ m123. Stokes, JB,andSigmund RD. Regulation of rENaC mRNA by dietary NaCl and steroids: organ, tissue, and steroid heterogeneity. Am J Physiol Cell Physiol 274:C1699-C1707,1998 .
2 @7 G1 [6 ]) b9 m% ]+ k1 P
" a" U5 j) X  @' b' i
" n' a6 r6 d; o% ~" f# v1 M8 H
, D' \" ?! q1 z. V124. Stumpf, WE,Sar M,Narbaitz R,Reid FA,DeLuca HF,andTanaka Y. Cellular and subcellular localization of 1,25-(OH)2-vitamin D 3 in rat kidney: comparison with localization of parathyroid hormone and estradiol. Proc Natl Acad Sci USA 77:1149-1153,1980 .
8 \8 d+ N# e* z4 a1 a
9 S/ {8 e: A  T  Q
- o1 ]: G* B2 w6 U  k9 t5 r4 N' ?4 a& J+ `. v
125. Tomita, K,Pisano JJ,andKnepper MA. Control of sodium and potassium transport in the cortical collecting duct of the rat. J Clin Invest 76:132-136,1985  .; o- u5 L/ @+ I$ d  ]# k$ S$ h8 H

0 T2 w5 Q: D1 o0 }
# {) V- \  C0 @# g7 `4 j" ?0 l6 w
2 q  Q" S+ Y' R( Y& n' N, V5 k* N5 W. P126. Vallet, V,Chraibi A,Gaeggeler HP,Horisberger JD,andRossier BC. An epithelial serine protease activates the amiloride-sensitive sodium channel. Nature 389:607-610,1997 .* {1 Y+ K: j5 n
$ S# B  a% m2 V
; G  m# N0 X% j6 g9 |: O6 B
, n; k5 c6 W% t+ |) s* h+ b: s
127. Van Abel, M,Hoenderop JG,Dardenne O,St Arnaud R,Van Os CH,Van Leeuwen HJ,andBindels RJ. 1,25-Dihydroxyvitamin D 3 -independent stimulatory effect of estrogen on the expression of ECaC1 in the kidney. J Am Soc Nephrol 13:2102-2109,2002 .
' ^6 a: A; O+ M! c
8 W' b+ U' a1 r9 V# L! y# `7 S# N4 T
$ S# {4 N' p: i& ]! L& Y
( E9 N9 C, F0 G: p+ \128. Velazquez, H,Bartiss A,Bernstein P,andEllison DH. Adrenal steroids stimulate thiazide-sensitive NaCl transport by rat renal distal tubules. Am J Physiol Renal Fluid Electrolyte Physiol 270:F211-F219,1996 .5 d; O3 B7 H, l; r8 t5 W( u! i

+ N# b1 X* K2 _0 J' c3 d8 q2 N) P2 o" }. e

) ]4 n0 U' l/ m( }1 O. I129. Velazquez, H,Silva T,Andujar E,Desir GV,Ellison DH,andGreger R. The distal convoluted tubule of rabbit kidney does not express a functional sodium channel. Am J Physiol Renal Physiol 280:F530-F539,2001 ." S9 k& x& k& y9 ^! p! x

- [) R3 y( Y5 `% U; w# O' c
& T2 u6 X1 z$ n. Q& d3 B; c& h4 A
130. Verlander, JW,Tran TM,Zhang L,Kaplan MR,andHebert SC. Estradiol enhances thiazide-sensitive NaCl cotransporter density in the apical plasma membrane of the distal convoluted tubule in ovarectomized rats. J Clin Invest 101:1661-1669,1998  .6 T( m0 z; b) o
/ [/ H/ @" Z7 d) q, \8 B2 X; i

' V" T$ d8 Q+ M+ u/ R( G# J  |! C* f3 v
131. Verrey, F,Hummler E,Schild L,andRossier BC. Control of Na   transport by aldosterone.In: The Kidney: Physiology and Pathophysiology (3rd ed.), edited by Seldin W,and Giebisch G.. Philadelphia, PA: Lippincott Williams & Wilkins, 2000.
+ m) ^' G' t( O- d1 `4 j
+ B2 N" _; w5 H7 j3 k9 g  D- h. A* {9 ^

0 }  M' `+ W; D6 S5 v% ^132. Vio, CP,andFigueroa CD. Subcellular localization of renal kallikrein by ultrastructural immunocytochemistry. Kidney Int 28:36-42,1985  .% N4 A' E, X& G! G  z  q
+ W. T( K& y% N- c- y# i) `

+ q: I' p4 \! r6 F/ H, H
2 n: O2 c& k% X# U; \) i3 n133. Vuagniaux, G,Vallet V,Jaeger NF,Pfister C,Bens M,Farman N,Courtois-Coutry N,Vandewalle A,Rossier BC,andHummler E. Activation of the amiloride-sensitive epithelial sodium channel by the serine protease mCAP1 expressed in a mouse cortical collecting duct cell line. J Am Soc Nephrol 11:828-834,2000 .
. M( r" [( j1 r+ o! X2 D! @# ~# c

8 o; K# ]* v8 K2 e9 z  i+ u5 y
+ o2 F& }# j1 h5 ^1 |8 d$ B134. Wade, JB. Distribution of transporters along the mouse distal nephron: something old, something borrowed, something new. Am J Physiol Renal Physiol 281:F1019-F1020,2001 .+ X0 E- z4 f9 e
: e. y5 p" E7 \  ]
& |: z9 E# c% z0 W) n5 g5 G# Q0 n$ H
! v+ Y0 ^+ }+ z
135. Wang, T,andGiebisch G. Effects of angiotensin II on electrolyte transport in the early and late distal tubule in rat kidney. Am J Physiol Renal Fluid Electrolyte Physiol 271:F143-F149,1996 .
# D8 e* F( M  O; g) ?' z! ~% E6 z: c
+ N! o$ ^. A% v& y. k
! T3 M# T- W  T+ s$ b. _6 b0 u( N6 U, E, m5 e' v6 B! m. @3 G0 l
136. Wang, W,Hebert SC,andGiebisch G. Renal K   channels: structure and function. Annu Rev Physiol 59:413-436,1997  .9 s! P6 E& {* \- h4 E7 I! x

1 O' O) G2 [/ r0 i3 d" K! g1 M! O8 o! ~0 q, K  m. X

9 }, x' ?$ z/ Q! ^137. Wang, XY,Masilamani S,Nielsen J,Kwon TH,Brooks HL,Nielsen S,andKnepper MA. The renal thiazide-sensitive Na-Cl cotransporter as mediator of the aldosterone-escape phenomenon. J Clin Invest 108:215-222,2001  .8 q6 q; U3 h; x+ X' ^; c4 {
+ m% c# L( v8 [
9 T* e7 h: E/ c. [; u  h8 _
0 n& x8 i' z+ N6 t5 H, f
138. Warnock, DG. Renal genetic disorders related to K   and Mg 2 . Annu Rev Physiol 64:845-876,2002  .
& D. r( @9 P# o2 s- A8 \* i5 F7 w$ E+ H" {' ]: b

% N( A/ q! @$ s  B
7 J. B( k6 o6 L* ?4 {139. Weinman, EJ,andEknoyan G. Chronic effects of chlorothiazide on reabsorption by the proximal tubule of the rat. Clin Sci Mol Med 49:107-113,1975  .) n( \7 r; z1 a6 B# }# V
4 B9 q( q" N. M* h7 x$ T

' h! J% p6 l9 N9 o! n' I! J* O# x$ q8 J; q* Z  S
140. Wolf, K,Castrop H,Riegger GA,Kurtz A,andKramer BK. Differential gene regulation of renal salt entry pathways by salt load in the distal nephron of the rat. Pflügers Arch 442:498-504,2001  .
- |, t& b! Y; w! d7 [. S" u% r% f* t1 p& \& C+ g7 H- _" }
3 j& n8 S& Y& c/ \+ n, P

% b- C, d  X7 s- u141. Wulff, P,Vallon V,Huang DY,Völkl H,Yu F,Richter K,Jansen M,Schlünz M,Klingel K,Loffing J,Kauselmann G,Bösl MR,Lang F,andKuhl D. Impaired renal Na   retention in the sgk1 -knockout mouse. J Clin Invest 110:1263-1268,2002  .
1 {! c. b4 x* w7 [/ F- T6 G$ C  n; @0 d

4 I% p1 }& O, Z; O* F
* n; u" j+ |: v6 z8 y1 E+ @142. Zhao, PL,Wang XT,Zhang XM,Cebotaru V,Cebotaru L,Guo G,Morales M,andGuggino SE. Tubular and cellular localization of the cardiac L-type calcium channel in rat kidney. Kidney Int 61:1393-1406,2002  .

Rank: 2

积分
162 
威望
162  
包包
1724  
沙发
发表于 2015-7-17 16:27 |只看该作者
任何的限制,都是从自己的内心开始的。  

Rank: 2

积分
73 
威望
73  
包包
1833  
藤椅
发表于 2015-7-23 22:16 |只看该作者
支持~~  

Rank: 2

积分
76 
威望
76  
包包
1772  
板凳
发表于 2015-7-26 09:43 |只看该作者
干细胞之家微信公众号
感觉好像在哪里看过了,汗~  

Rank: 2

积分
162 
威望
162  
包包
1746  
报纸
发表于 2015-7-30 19:18 |只看该作者
不知道说些什么  

Rank: 2

积分
80 
威望
80  
包包
1719  
地板
发表于 2015-8-3 10:01 |只看该作者
我毫不犹豫地把楼主的这个帖子收藏了  

Rank: 2

积分
75 
威望
75  
包包
2193  
7
发表于 2015-8-31 11:10 |只看该作者
祝干细胞之家 越办越好~~~~~~~~~`  

Rank: 2

积分
163 
威望
163  
包包
1852  
8
发表于 2015-9-2 06:10 |只看该作者
dddddddddddddd  

Rank: 2

积分
88 
威望
88  
包包
1897  
9
发表于 2015-9-28 11:44 |只看该作者
谢谢哦  

Rank: 2

积分
79 
威望
79  
包包
1769  
10
发表于 2015-9-30 14:00 |只看该作者
先看看怎么样!  
‹ 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, 2024-6-28 20:13

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.