Comprehensive Physiology Wiley Online Library

Uriniferous Tubule: Structural and Functional Organization

Full Article on Wiley Online Library



Abstract

The uriniferous tubule is divided into the proximal tubule, the intermediate (thin) tubule, the distal tubule and the collecting duct. The present chapter is based on the chapters by Maunsbach and Christensen on the proximal tubule, and by Kaissling and Kriz on the distal tubule and collecting duct in the 1992 edition of the Handbook of Physiology, Renal Physiology. It describes the fine structure (light and electron microscopy) of the entire mammalian uriniferous tubule, mainly in rats, mice, and rabbits. The structural data are complemented by recent data on the location of the major transport‐ and transport‐regulating proteins, revealed by morphological means (immunohistochemistry, immunofluorescence, and/or mRNA in situ hybridization). The structural differences along the uriniferous tubule strictly coincide with the distribution of the major luminal and basolateral transport proteins and receptors and both together provide the basis for the subdivision of the uriniferous tubule into functional subunits. Data on structural adaptation to defined functional changes in vivo and to genetical alterations of specified proteins involved in transepithelial transport importantly deepen our comprehension of the correlation of structure and function in the kidney, of the role of each segment or cell type in the overall renal function, and our understanding of renal pathophysiology. © 2012 American Physiological Society. Compr Physiol 2:805‐861, 2012.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1.

Schematic of nephrons and collecting duct. This scheme depicts a short‐looped and a long‐looped nephron together with the collecting system. Not drawn to scale. Within the cortex a medullary ray is delineated by a dashed line. 1 = renal corpuscle including Bowman's capsule and the glomerulus (glomerular tuft); 2 = proximal convoluted tubule; 3 = proximal straight tubule; 4 = descending thin limb; 5 = ascending thin limb; 6 = distal straight tubule (thick ascending limb); 7 = macula densa located within the final portion of the thick ascending limb; 8 = distal convoluted tubule; 9 = connecting tubule (CNT); 9* = CNT of the juxtamedullary nephron that forms an arcade; 10 = cortical collecting duct; 11 = outer medullary collecting duct; and 12 = inner medullary collecting duct. From Kriz and Bankir, with permission, 339.

Figure 2. Figure 2.

(A) Schematic drawing, demonstrating the essential structural features of renal transporting epithelia; 1 paracellular route through the tight junction and the lateral intercellular spaces; 2 transcellular route, across the apical plasma membrane, the cytoplasm and the basolateral plasma membrane; the apical membrane area may be augmented by short microvilli, microfolds (not shown) or “ brush border” (long microvilli of uniform dimensions), the basolateral membrane may be augmented by infoldings of the basal plasma membrane or by lateral folding, giving rise to basolateral processes, which narrowly interdigitate with those of neighbouring cells, the lateral folds narrowly enclose large mitochondria. [(B)‐(F)] Salient features of interdigitated epithelia. (B) Freeze fracture electron micrograph of a tight junction (TJ) (e.g., thick ascending limb) consisting of several densely arranged parallel strands. Rabbit (Cooperation with A. Schiller and R. Taugner). (C) TEM of interdigitated lateral folds, enclosing large mitochondria (M), and narrow intercellular spaces (arrows) of regular width (e.g., thick ascending limb epithelium); cell adhesion molecules within the intercellular spaces are contrasted by tannic acid staining; BL, basal lamina; Rat; (cooperation with T. Sakai). (D) Freeze‐fracture electron micrograph of the luminal aspect of a tubule demonstrating the mode of cellular interdigitation (e.g., ascending thin limb and Psammomys obesus) [from Kriz et al., with permission 345]. (E) Freeze fracture electron micrograph of a TJ in noninterdigitated epithelia (e.g., collecting duct cell); the prominent tight junction consists of several anastomosing tight junctional strands. (F) Infolded basal membranes confining extracellular spaces (arrows) which may vary in width with function; BL, basal lamina. (G) Freeze fracture electron micrograph of the basal aspect of a noninterdigitated cell (e.g., cortical collecting duct); the polygonal shape of this cell is recognized by contours of basal slits through which lateral intercellular spaces (white arrows) open into the interstitium; basal infoldings of extracellular spaces (black arrows) are separated from lateral intercellular spaces. Bars ∼1 μm (D, F, and G) and ∼0.1 μm (B, C, and E). Adapted, with permission, from Kaissling and Kriz 291.

Figure 3. Figure 3.

Schematic three‐dimensional appearance of a proximal tubule cell from the convoluted part of the rat proximal tubule, illustrating the appearance of the lateral and basal ridges and processes. Adapted, with permission, from Maunsbach 426.

Figure 4. Figure 4.

Schematic representation of the typical mouse nephron and collecting duct (CD) organization: Tortuous descending thin limbs (DTL) of long‐loop nephrons (LLN; large arrow); winding course of thick ascending limbs (TAL) of short loop nephrons (SLN) and CD (arrow heads); a piece of TAL inserted in the DTL of LLN (LLNt, small arrow), which forms its bend just beneath the “transitional zone” within the inner medulla (IM); and three different types of SLN bends (SLN1, SLN2, and SLN3). Based on the distribution of the nephron segments, the renal zones are defined, including cortex, outer stripe of outer medulla (OSOM), inner stripe of outer medulla (ISOM), and IM. The postmacula densa segments and the directions of the proximal tubules (PT) arising from their glomeruli are also illustrated. Adapted, with permission, from Zhai et al. 762.

Figure 5. Figure 5.

Electron micrograph of rat kidney cortex showing several proximal tubules with open lumens and evenly arranged brush borders. The micrograph illustrates some of the differences between the two segments of the convoluted part of the proximal tubule. In the first segment (S1) the cells are slightly taller, microvilli of the brush border longer, and apical endocytic vacuoles more numerous than in the second segment (S2). The lysosomes in the first segment have light contents whereas the lysosomes in the second segment show densely stained contents. DCT, distal convoluted tubule. The tissue was fixed by vascular perfusion with glutaraldehyde. X 2,100. Adapted, with permission, from Maunsbach 424.

Figure 6. Figure 6.

Electron micrographs and schematic drawings of proximal tubule epithelium from rat and rabbit kidney illustrating the three segments of the proximal tubule. (A) Rat, first segment (S1), (B) rat second segment (S2), (C) rat, third segment (S3), (D) rabbit, third segment (S3), and (E) schematic drawings of the three segments in rat. In segments S1 and S2 the mitochondria (M) are oriented perpendicular to the basement membrane and endocytic vacuoles (E) are numerous. The brush border (BB) is taller in S1 than in S2. The matrix of the lysosomes (L) in S1 is lightly stained but rather homogenous and densely stained in S2. The mitochondria in S3 are more randomly oriented than in S1 and S2 and they are less numerous in the rabbit than in the rat cells. In both species the lysosomes in S3 cells appear smaller and fewer as compared to S1 and S2 of the rat. The brush border of this segment is significantly higher in rat than in rabbit. Part of a cilium (C) is present in the brush border in C and peroxisomes (P) are seen in the cytoplasm. Capillary (CAP). The diagram in E illustrates the differences in cell shapes and the different distributions of mitochondria (M), lysosomes (L), endocytic vacuoles (E), and peroxisomes (microbodies) (P). S1, first segment. S2, second segment. S3, third segment. Adapted, with permission, from Maunsbach 426 and Maunsbach and Christensen 429. [(A) and (B)] X 8,900 and [(C) and (D)] X 9,800.

Figure 7. Figure 7.

(A) Tight junction (arrowheads) between two rat proximal tubule cells close to the tubule lumen (LU). The triple‐layered plasma membranes are completely apposed. The tight junction has a depth in the luminal‐basal direction of only 100 to 200 Å and is followed on its peritubular side by an intermediate junction. X 167,000. Adapted, with permission, from Maunsbach 424. (B) Tangential section through luminal part of proximal tubule cells showing an intermediate junction, which extends between the two pairs of arrowheads. When the junction is sectioned at right angle it can be noted that the cytoplasm next to the triple‐layered plasma membranes has an increased density and that a dense material fills the gap between the membranes (compare with normal lateral intercellular space at LIS). Lumen (LU) is seen in the upper right corner. X 145,000. Adapted, with permission, from Maunsbach 426. (C) Higher magnification of gap junction between proximal tubule cells from Necturus maculosus. The 20‐Å wide space between the two plasma membranes is clearly observable (arrowheads). X 204,000. Adapted, with permission, from Maunsbach 426.

Figure 8. Figure 8.

Parts of proximal tubule cells from rat and rabbit kidney illustrating smooth surfaced endoplasmic reticulum and peroxisomes. (A) Peroxisomes (P) (microbodies) from rat proximal tubule cell limited by a single membrane and showing a uniformly stained matrix. Smooth‐surfaced endoplasmic reticulum closely surrounds the peroxisomes (arrowheads) and is seen in continuity with the rough endoplasmic reticulum at arrow. X 44,000. Adapted, with permission, from Maunsbach and Christensen 429. (B) Smooth‐surfaced endoplasmic reticulum adjacent to the lateral plasma membrane. This so‐called paramembraneous endoplasmic reticulum (PER) has fenestrated cisternae (arrows) that closely follow the lateral plasma membrane. However, it is never in contact with this membrane, nor is it in continuity with the plasma membrane facing the tubule lumen (LU), endocytic vacuoles (E), or lysosomes. (A) X 95,000. Adapted, with permission, from Maunsbach 428. (C) Peroxisomes (P) in cells of isolated perfused proximal tubule of rabbit. In this species, part of the limiting membrane is straight, thickened, and electron dense. The limiting membrane is closely related to the smooth endoplasmic reticulum. Since this tubule was incubated with horseradish peroxidase (HRP) in the bath, the intercellular spaces, except in gap junction (GJ), are filled with reaction product for HRP. Reaction product is also seen in a small endocytic vesicle (V). X 45,000. Adapted, with permission, from Nielsen and Christensen 470. (D) Demonstration of dextran in rat proximal tubule cells 1 h after termination of intravenous infusion of dextran T‐40. The kidney was fixed in vivo by dripping a lead‐containing fixative on the kidney surface. Small dense particles representing dextran are present in lysosomes (arrows), where they are located adjacent to the dense matrix material. A peroxisome (P) with a fingerlike projection is well stained by the lead‐containing fixative. X 15,800. Adapted, with permission, from Christensen and Maunsbach 118.

Figure 9. Figure 9.

Electron micrographs illustrating the vacuolar apparatus in rat proximal tubule cells. (A) A round lysosome (L) with an electron‐dense matrix devoid of inclusions and located close to a mitochondrion (M). The lysosome is limited by a triple‐layered membrane. X 100,000. (B) Coated pit connected to the tubular lumen (LU). The coated pit has a thick cell coat or glycocalyx (arrow) and a cytoplasmic clathrin coat (arrowheads). X 100,000. Adapted, with permission, by A.B. Maunsbach. (C) Dense apical tubules connected to endosomes (S) in apical cytoplasm. X 70,000. Adapted, with permission, from Cui and Christensen 138.

Figure 10. Figure 10.

Isolated perfused proximal tubule of rabbit incubated with horseradish peroxidase in the bath for 30 min. The reaction product is seen in the lateral intercellular spaces (LIS) but not in the lumen (LU). Note the reaction product in a multivesicular body (MVB) and in lysosomes (L). Unstained section. X 15,000. Adapted, with permission, from Nielsen and Christensen 470.

Figure 11. Figure 11.

Immunofluorescence (mouse kidney) and immunogold (rat proximal tubule) localization of megalin and cubilin. The immunofluorescence reveal strong apical labeling in proximal tubules but no obvious labeling in glomeruli (G). Distal tubules (D) and vessels (A) are unlabeled. Bar, 100 μm. Immunogold labeling, megalin, 10 nm gold and cubilin, 5 nm gold, reveals colocalization of the two receptors in coated pits (Inv), coated vesicles (C), endosomes (E), and in recycling dense apical tubules (arrows). Bar, 0.5 μm.

Figure 12. Figure 12.

Schematic drawing of megalin, cubilin, and amnionless (AMN) presenting known domains and motifs. The three receptors colocalize in the renal proximal convoluted tubule (PCT) where they cooperate in clearance of the ultrafiltrate. Megalin binds a variety of filtered molecules (more than 50 ligands have been identified) through its complement type repeats and is able to mediate endocytosis via NPXY motifs in the cytoplasmic tail. Cubilin on the other hand includes multiple binding domains (CUB domains), but only around 15 ligands have been identified. Cubilin is a peripheral membrane protein and is thereby dependent on megalin and/or AMN to assure internalization of its ligands. AMN contains a NPXY motif and is probably assisting cubilin in endocytosis as well as in transport during synthesis. Adapted, with permission, from Christensen et al. 123.

Figure 13. Figure 13.

The megalin‐ and cubilin‐mediated uptake of three vitamin carrier protein complexes: DBP‐vitamin D3, TC‐vitamin B12, and RBP retinol in renal proximal tubule. Likewise the cubilin chaperone protein amnionless, amnionless (AMN) is indicated. Following receptor‐mediated endocytosis via apical coated pits, the complexes accumulate in lysosomes for degradation of the proteins, while the receptors recycle to the apical plasma membrane via dense apical tubules. As illustrated here and detailed in the text, megalin mediates the uptake of cubilin and its ligands. Whether the two receptors are constitutively associated in the plasma membrane and remain associated during recycling in dense apical tubules is not known. Whereas TC and RBP apparently bind exclusively to megalin, DBP binds with similar affinity to both megalin and cubilin. The intracellular processing of the vitamins may include modifications such as hydroxylation of 25‐OH‐D3 to 1,25‐(OH)2‐D3, and metaobolism of B12. The mechanisms for the cellular release of the vitamins remain to be clarified. Adapted, with permission, from Verroust et al. 700.

Figure 14. Figure 14.

Proximal tubular uptake of albumin A and B and insulin C, in rat A and C and in mouse B. (A) Double immunogold labeling of endogenous albumin (18 nm gold) and cathepsin B (6 nm gold) on ultrathin cryosection from rat kidney. The albumin labeling is highly concentrated in a lysosome (L). X 43,000 [adapted, with permission, from Christensen et al. 123]. (B) Immunofluorescence of paraffin section of cortex from kidney‐specific megalin mosaic KO mice by using sheep anti‐rat megalin (green), rabbit anti‐human albumin (red). Proximal tubule cells not expressing megalin (arrows) have no labeling for albumin. X 550 [adapted, with permission, from Christensen et al. 123]. (C) Electron microscope autoradiograph of 125I‐labelled insulin uptake in rat proximal tubule, fixed 25 min after start of microinfusion of the insulin. Grains are mainly located over lysosomes in the apical part of the cytoplasm (arrows). Endocytic vacuoles (E) and some lysosomes (L) located deep in the cytoplasm do not contain 125I‐insulin. X 16,000. Adapted, with permission, from Hellfritzsch et al. 248.

Figure 15. Figure 15.

Epon section from border between the outer and inner stripe of the outer zone of the medulla of rat labeled for AQP‐1 labeled by immunoperoxidase by method described, with permission, by Zhai et al. 761. Segment 3 proximal tubules show an intense brush border labeling and basolateral labeling (arrowheads). Two transitions into nonlabeled, thin‐walled DTL type 1 epithelium of short‐loop nephrons (arrows) are shown. X 400.

Figure 16. Figure 16.

Schematic presentation of the tubular‐vascular relationship in the outer part of the inner stripe of outer medulla (ISOM). Tl: thick ascending limb of long‐looped nephrons. Ts: thick ascending limb of short‐looped nephron. Dl: descending thin limb of long‐looped nephron. Ds: descending thin limb of short‐looped nephron. C: collecting duct. VB: vascular bundle. IB: interbundle region. Kindly provided, with permission, by Dr. X.Y. Zhai].

Figure 17. Figure 17.

(A) Schematic of thin limb ultrastructure. Type 1 is found in descending thin limb (TL) of short loop nephrons (SLNs). Type 2 is found in upper part of descending TLs of long loop nephrons (LLNs); in most species (e.g., rat, mouse, and Psammomys), it is complexely organized, in others (e.g., rabbit and guinea pig), it is more simple. Type 3 occurs in lower parts of descending TL of LLNs. Type 4 is found at the loop bend and in ascending TL of LLNs. Modified, with permission, from Kaissling and Kriz 291. (B) Four distinct epithelia along the TL of SLN and LLN in mouse. The figure comprises two columns of images representing lower and higher magnifications (right). Type 1 is a cross section of the DTL of an SLN (440 μm from the transition of the PT to the DTL); higher magnification of type 1 shows a simple and thin epithelium with few microprojections, abundant ribosomes, and sparse mitochondria throughout the cytoplasm. Type 2 is a cross section of the DTL of an LLN (370 μm from the transition of the PT to the DTL); higher magnifications of type 2 show a highly specialized epithelium: abundant short and plump microvilli, basolateral infoldings, and highly interdigitated cellular processes with shallow tight junctions (small arrows) and radially oriented mitochondria. Type 3 is a cross section of a DTL of LLN around the boundary between the inner stripe of outer medulla (ISOM) and the inner medulla (IM) (2790 μm from the transition of PT to DTL); higher magnifications of type 3 show the highly differentiated membranes: Numerous relatively long microvilli, abundant finger‐like basolateral infoldings throughout the cytoplasm (large arrows), and the less shallow tight junctions (small arrow) and few mitochondria. Type 4 is a cross section of the ascending thin limb (ATL) of an LLN (700 μm from the transition of the ATL to the TAL); higher magnifications of type 4 show the bold and dense apical membrane, basolateral infoldings, cellular processes with tight junctions (small arrows), and various organelles with sparse ribosomes throughout the cytoplasm. Bars, 1 μm. Adapted, with permission, from Zhai et al. 762.

Figure 18. Figure 18.

Peroxidase immunohistochemistry of an Epon section showing the transition (arrows) from UT‐A2‐labeled DTL into the nonlabeled, thick ascending epithelium of a short loop nephron (SLN) of mouse kidney. X 200. Adapted, with permission, from Zhai et al. 762.

Figure 19. Figure 19.

Survey on location (left panel) and ultrastructure of the thick ascending limb of Henle's loop [TAL = distal straight tubule, including macula densa (MD)]; C, cortex; IS, inner stripe; OS, outer stripe; IZ, inner zone; the direction of the urinary flow is indicated by white arrows, (A) medullary part, (B) cortical part, and (C) MD. adapted, with permission, from 288,291.

Figure 20. Figure 20.

Overview on the histotopography of the thick ascending limb (T); (A) cross section of the inner stripe; the upper portion shows part of a vascular bundle with descending (a) and ascending vasa recta (v); in the lower half thick ascending limbs are intermingled with descending thin limbs (DL), collecting ducts (CDs), and capillaries from the interbundle capillary plexus. (B) Longitudinal section of the deep cortex in rat; the thick ascending limb (TAL) profiles reveal a much thinner epithelium than in the inner stripe; they are grouped around the CDs and surrounded by profiles of proximal tubules [adapted, with permission, from 291]. Inset: longitudinal section through the sharp transition (arrowheads) from the thin epithelium of the ascending limb (AL) to the TAL (T) in mouse. [(A) and inset] ∼10 μm and (B) ∼50 μm.

Figure 21. Figure 21.

Ultrastucture of thick ascending limb cells. (A) Deep level of the inner stripe; rat; (B) cortical part; rat; and (C) macula densa cells; rabbit. Mitochondria (arrow head) in A and B are confined in the interdigitated lateral folds and the intercellular spaces (long arrow) are narrow; in C the mitochondria are scattered in the perinuclear cytoplasm and the intercellular spaces are dilated; the macula densa (MD) cells are affixed to the extraglomerular mesangium (star); [(A) and (B) adapted, with permission, from 291 and (C) from 290]. Bars ∼ 1 μm.

Figure 22. Figure 22.

Macula densa (MD). (A) Preparation of a isolated human renal corpuscle (G) together with the cortical radial artery (A), glomerular arterioles (red), and tubules; proximal tubule (P; gray); the thick ascending limb (T, yellow) ascends along the cortical collecting duct (C; violet), bends toward the vascular pole of its glomerulus and passes between the glomerular arterioles; it continues as distal convoluted tubule (DCT, orange); the asterisk indicates the location of the MD. [Slightly modified, with permission, from Karl Peter (1909) 517]. (B) Scanning electron micrograph; the cortical thick ascending limb (CTAL) is open at the level of the MD, the luminal aspect of rabbit MD cell plaque is visible; single cilia are seen on MD cells as well as on CTAL cells; EGM, extraglomerular mesangium [adapted, with permission, from 291]. (C) Epon section of rat MD within the cortical thick ascending limb; the epithelium reveals conspicuously dilated intercellular spaces; those between thick ascending limb cells are narrow; shortly after the MD the epithelium transforms (arrowheads) to that of the DCT (D); arrow, indicates flow direction; (ea), efferent arteriole. (D) Epon section of mouse MD in contact with granulated rennin‐producing cells (GC) in the afferent glomerular arteriole; the MD cells display abundant microvilli on their luminal surface as well as single cilia (arrows); the intercellular spaces are narrow. Bars ∼ 10 μm.

Figure 23. Figure 23.

Organization of the transition from the DCT to the collecting duct in the cortex of rabbit. (A) Connecting tubule (CN) of a superficial nephron, opening into the beginning of a cortical collecting duct (CD) under the renal capsule; the sharp transition in the epithelial lining is indicated by a bar in the lumen. (B) Sharp transition (bar) of DCT (D) to CNT (CN); fusion of several CNTs establishes an arcade that ascends (arrow) in the cortex; note the progressive decrease in height of the CNT epithelium along the CNT. (C) Arcade (CN), ascending in the cortex in close proximity to the cortical radial artery (A), takes up several tributaries. (D) Opening of an arcade (CN) in the upper third of the cortex into a CCD within a medullary ray; the arrows indicate the flow directions; note the proximity of the arcade to an afferent arteriole (white arrow head). (E) Juxtaposition of an arcade (CN) to a lymphatic (L) in the periarterial connective tissue of the cortical radial artery (A). [(A) and (B)] Epon sections of rabbit kidney, (E) of mouse kidney; [(C) and (D)] paraffin sections of rabbit kidney. Bars ∼ 50 μm. [(A) and (B)] [adapted, with permission, from 292]; [(C) and (D)] [adapted, with permission, from 288], (E) [adapted, with permission, from 286].

Figure 24. Figure 24.

Survey on organization of the cortical distal segments and collecting ducts (CDs; left panel) and on ultrastructure of the segment‐specific cells (right panel); C ‐ cortex, OS, outer stripe, IS, inner stripe of the outer medulla; IM, inner medulla; dashed line, delimits the medullary ray; (a) distal convoluted tubule (DCT) and (A) DCT‐cell; (b) connecting tubule (CNT) and (B) CNT‐cell; (c) CCD and (C) CCD cell; (d) inner medulla (IM), and (D) IMCD cell; the black semicircles indicate the occurrence of intercalated (IC) cells in the segment; adapted, with permission, from 291.

Figure 25. Figure 25.

(A) The thick ascending limb (T) transforms shortly after the macula densa (MD), affixed to the granulated portion of the afferent glomerular arteriole (aa), to the distal convoluted tubule 1 (D); the abrupt change in the epithelial structure (arrowheads) coincides with the marked increase in contrast; the appearance of an intercalated cell (asterisk) in the DCT epithelium indicates the transition (arrowheads) to the transitional segment (D2), which shows a slightly lower density of mitochondria; mouse; epon section; Bar ∼ 10 μm. (B) DCT1 cell in rat; characteristic are the tall mitochondria enclosed in lateral interdigitating cell processes, the apical position and apical flattening of the nucleus and the narrow mitochondria‐free cytoplasmic rim on top of the nucleus, containing many small vesicles; arrowhead, single cilium. Epon section; bar ∼ 1 μm. Adapted, with permission, from 341.

Figure 26. Figure 26.

Comparison of salient features of DCT, CNT, and collecting duct segments and cells in the renal cortex. (A) Mouse renal cortex showing side by side a profile of the distal convoluted tubule 2 (D2), of a connecting tubule (CN), and a collecting duct (CCD); the decrease of density in mitochondria, their change in cellular position from basal infoldings to the apical cytoplasm, the decrease in cell height, and the increase in the frequency of intercalated cells (asterisks) in the epithelial lining is evident. Epon section; bar ∼ 10 μm. [(B)‐(D)] Higher magnification of a DCT cell (B), a CNT cell (C), and a CCD cell (D); compare with (A); the arrows point to interdigitating cell processes, narrowly enclosing large mitochondria (B), abundant deep infolding of the basal plasma membrane reaching between tall mitochondria (C) and to infoldings of the basal plasma membrane restricted to a narrow rim in the basal cell portion in the noninterdigitating CCD cell (D); all mitochondria and cell organelles are located above this basal rim. [(B) and (C)] From Psammomys obesus; (D) rat; fixation by reduced osmium. TEM, bar ∼ 1 μm. [(B)‐(D)] Modified, with permission, from 341.

Figure 27. Figure 27.

Connecting tubule (CNT) in rat. (A) Longitudinal section through renal cortex, showing profiles of CNT, forming arcades, ascending (arrows) in the cortex in close juxtaposition to the cortical radial vessels (CRV) and glomerular afferent arterioles (aa); bar ∼ 50 μm. (B) The CNT epithelium is composed of CNT cells and intercalated cells (arrows); bar ∼ 10 μm. (C) Characteristic CNT cell with abundant infoldings of the basal plasmalemm extending deeply into the cytoplasm; mitochondria are found between the infolded membranes and in the apical cytoplasm on top of the nucleus; bar ∼ 10 μm. (D) The infolded plasma membranes reveal numerous caveolae; bars ∼ 0.01 μm. Adapted, with permission, from 291.

Figure 28. Figure 28.

Cortical [(A) and (B)] and outer medullary [(C) and (D)] collecting duct. (A) Collecting duct in the upper cortex of rat; the epithelium is composed by collecting duct (CD) cells and intercalated cells (IC cells; asterisks); T, thick ascending limb: P, proximal tubule. (B) Cortical collecting duct (CCD) cell; infoldings of the basal plasma membrane are restricted to the basal cell portion; all mitochondria and cell organelles are located above the infolded membranes. (C) Collecting duct in the inner stripe of mouse; the epithelial organization is similar as in the cortex; note the different aspect of IC cells (asterisks); (T) thick ascending limb. (D) Outer medullary conduction duct (OMCD) cell in the inner stripe; the organization is similar as in the CCD; the infoldings of the basal plasmalemm and the amount of mitochondria decrease toward the medulla. Bars: [(A) and (C)] ∼ 10 μm; [(B) and (D)] ∼ 1 μm. [(B) adapted, with permission, from 291 and (D) adapted, with permission, from 341].

Figure 29. Figure 29.

Inner medullary collecting duct. (A) Tubular profile of a medullary collecting duct in the transitional zone from the inner stripe of the outer medulla to the inner zone in rabbit; the inner medullary conducting duct (IMCD) cells in the homogenous epithelium possess a dense apical cytoskelettal web under the luminal plasmalemma, indicated by a star in two cells, under the luminal plasmalemm; the cytoplasm reveals a paucity of cell organelles; the big round nucleus has a rather homogeneous chromatin structure. (B) Tip of the renal papilla in rat with openings of papillary CDs (paraffin section) into the renal pelvis; the urothelium of the pelvic wall is seen on the right. (C) Epithelium of the middle portion of an IMCD in rat. Within the epithelium three zones are seen: a basal zone with basal infoldings, a middle zone containing Golgi fields, mitochondria and lysosomal elements, and a thin apical zone with abundant tubular and vesicular profiles. Note the deep tight junction. (D) IMCD cells in the deep inner medulla; the tall cells have a large nucleus and relative to their size few cell organelles are encountered; lateral intercellular spaces (arrow) are filled with microvilli and microfolds; the cytoplasm immediately beneath the apical plasmalemm is devoid of cell organelles. Bars: (A) ∼ 10 μm; (B) ∼ 100 μm; (C) ∼ 1 μm; and (D) ∼ 10 μm. Adapted, with permission, from 341.

Figure 30. Figure 30.

Schematic distribution of the major apical transport proteins (NKCC2, black; NCC, green; TRPM6, turquoise; TRPV5, orange; ENaC, red; and AQP2, blue), along the cortical distal segments in rabbit 1, and in rat, mouse, and human 2; MR, medullary ray; TAL, thick ascending limb; G, renal corpuscle; DCT, distal convoluted tubule; CNT, connecting tubule; CCD, cortical collecting duct; restriction of a transporter to a tubular portion is indicated by a thick vertical bars, the continuation along the collecting duct (CD) by arrows. Modified, with permission, from 341.

Figure 31. Figure 31.

Transition from the thick ascending limb to the distal convoluted tubule [(A)‐(C)]: rabbit; [(D)‐(F)]: mouse; [(A) and (D)] 1 μm epon sections, all others cryostat sections; [(B) and (E)] immunofluorescence for NKCC2; (C) in situ hybridization for mRNA of SLC12a3 (coding for NCC), (F) immunofluorescence for NCC, same section as E (double fluorescence); in both species the sharp change in morphology is congruent with the abrupt change from NKCC2 to NCC (arrowheads). Bars: ∼ 50 μm. Adapted, with permission, from 385.

Figure 32. Figure 32.

Transitional regions from the distal convoluted tubule (D) to the connecting tubule (CN) and the cortical collecting duct (CCD) in rabbit. [(A) and (E)] 1 μm epon sections, all others cryostat sections. (B) In situ hybridization for mRNA of SLC12a3 (coding for NCC). (C) Same section as (B), immunostaining for the β subunit of epithelial sodium channel (ENaC); no overlap of NCC and ENaC, arrow heads point to the definite stop of NCC mRNA and the abrupt beginning of apical ENaC staining; no overlap of NCC and ENaC in the same cell. (D) Immunostaining of a consecutive section for Calbindin k28D (CB); the end of NCC expression and the beginning of apical ENaC coincides with a marked increase of CB staining in the cytoplasm of tubular cells and the appearance of intercalated cells in the tubular epithelium (asterisks). (E) The transition of the CNT to the CCD is marked by the obvious change in the epithelial structure at the opening of the CNT (CN) to the CCD (arrowheads) indicates the transition of the CNT to the CCD. (F) The structural change coincides with a shift of luminal ENaC immunostaining to the cytoplasm (arrowheads); (G) with a sharp reduction of CB immunostaining (arrowheads); and (H) and the clear‐cut onset of luminal AQP2 immunostaining. Bars: ∼ 50 μm. Adapted, with permission, from 380.

Figure 33. Figure 33.

Transitional regions from the distal convoluted tubule to the connecting tubule and the cortical collecting duct (CCD); mouse. [(A) and (E)] 1 μm epon sections, all others, cryostat sections. (A) The most upstream occurrence of intercalated cells (IC cells; asterisks) indicates the transition from the DCT1 (D1) to the transitional segment of the DCT, the DCT2 (D2); the frequency of IC cells increases toward and along the CNT (CN), note the marked decrease of epithelial lining along the CNT segment. [(B) and (C)] Double immunostaining for NCC and the β subunit of epithelial sodium channel (ENaC); NCC, and ENaC overlap in the cells of DCT2 (D2); (D) consecutive section, immunostained for the vasopressine‐sensitive water channel aquaporin 2 (AQP2); its onset coincides with the disappearance of NCC expression and defines the beginning of the CNT. (E) Transition from the CNT (CN) to the CCD. [(F) and (G)] Consecutive sections, immunostained for β subunit of ENaC and AQP2; both are coexpressed by the CNT cells as well as by the collecting duct cells (CD). Bars: ∼ 50 μm. Adapted, with permission, from 385 and 380.

Figure 34. Figure 34.

Distribution of NCC, the epithelial calcium channel TRPV5 and the sodium calcium exchanger (NCX) along the distal nephron. Cryostat sections; [(A) and (B)] rabbit; [(C)‐(G)] mouse. [(A)‐(D)] Double immunostaining for NCC and TRPV5; [(A) and (B)] in rabbit NCC in the distal convoluted tubule (D) and TRPV5 in the connecting tubule (CN) are exclusive; [(C) and (D)] In mouse, NCC and TRPV5 are coexpressed in the DCT2 (D2), TRPV5 continues in the CNT (CN), whereas NCC is absent in the CNT. [(E) and (F)] Immunofluorescence for TRPV5 is the highest in the apical cytoplasm in DCT2 and decreases progressively and shifts to the cytoplasm along the CNT (CN); the arrow indicates the urinary flow direction; the segmental distribution of the sodium calcium exchanger (NCX) in the basolateral membrane parallels that of TRPV5; weak NCX staining is present in DCT1 (D). [(G) and (H)] TRPV5 and NCX expressions sharply stop at the transition from the CNT to the cortical collecting duct (CCD). Bars ∼ 50 μm. Adapted, with permission, from Loffing et al. 385 and Loffing and Kaissling 380.

Figure 35. Figure 35.

Rat cortical collecting duct with two distinct types of intercalated cells (IC cells). Type A IC cells marked with an asterix, type B IC cells with #. Original magnification ∼2200×. Adapted, with permission, from 291.

Figure 36. Figure 36.

Scheme of the typical appearance of the different types of intercalated cells (IC cells). (A) Type A IC cell. (B) Type B IC cell. (C) Medullary IC cell. Adapted, with permission, from 291.

Figure 37. Figure 37.

Rat intercalated cells (IC cells). (A) Type A IC cell with abundant apical microfolds and many mitochondria. (B) Type B IC cells cell with a rather narrow apical cell pole, a narrow rim of dense cytoplasm exempt of vesicles under the apical plasma membrane, abundant smooth surfaced vesicles in the apical cytoplasm, a huge Golgi complex, and abundant mitochondria along the basolateral plasma membrane. Transmission electron microscopy ∼7000× magnification.

Figure 38. Figure 38.

Expression of H+‐ATPase subunits in intercalated cells (IC cells): (A) mouse connecting tubule stained with antibodies against the B1 H+‐ATPase subunit (red) and the segment cell specific water channel aquaporin 2 (AQP2) (green). B1 subunits are detected in luminal (cells with arrows) and/or basolateral membranes (cell with asterix). Adapted, with permission, from 189. (B) The a4 H+‐ATPase subunit is expressed in various nephron segments including the proximal tubule and also in IC cells. Mouse CNT stained with antibodies against the a4 H+‐ATPase (red) and the AQP2 water channel labeling segment specific cells (green).

Figure 39. Figure 39.

Expression of intercalated cell (IC cell) specific anion exchangers AE1 and pendrin. (A) Mouse outer medullary collecting duct stained against AE1 (red) and aquaporin 2 (AQP2) water channels (green). AE1 shows a basolateral localization in cells negative for AQP2. (B) Colocalization of the E subunit of apical H+‐ATPase (green) and basolateral AE1 (red) in rat outer medullary collecting duct IC cells. (C) The anion exchanger pendrin (red) is localized at the luminal pole of non‐type A (i.e., type B and non‐A/non‐B IC cells) IC cells in mouse CNT. Segment specific cells are identified by the presence of the AQP2 water channel. Some cells are negative for pendrin and AQP2 (asterix), presumably type A IC cells.



Figure 1.

Schematic of nephrons and collecting duct. This scheme depicts a short‐looped and a long‐looped nephron together with the collecting system. Not drawn to scale. Within the cortex a medullary ray is delineated by a dashed line. 1 = renal corpuscle including Bowman's capsule and the glomerulus (glomerular tuft); 2 = proximal convoluted tubule; 3 = proximal straight tubule; 4 = descending thin limb; 5 = ascending thin limb; 6 = distal straight tubule (thick ascending limb); 7 = macula densa located within the final portion of the thick ascending limb; 8 = distal convoluted tubule; 9 = connecting tubule (CNT); 9* = CNT of the juxtamedullary nephron that forms an arcade; 10 = cortical collecting duct; 11 = outer medullary collecting duct; and 12 = inner medullary collecting duct. From Kriz and Bankir, with permission, 339.



Figure 2.

(A) Schematic drawing, demonstrating the essential structural features of renal transporting epithelia; 1 paracellular route through the tight junction and the lateral intercellular spaces; 2 transcellular route, across the apical plasma membrane, the cytoplasm and the basolateral plasma membrane; the apical membrane area may be augmented by short microvilli, microfolds (not shown) or “ brush border” (long microvilli of uniform dimensions), the basolateral membrane may be augmented by infoldings of the basal plasma membrane or by lateral folding, giving rise to basolateral processes, which narrowly interdigitate with those of neighbouring cells, the lateral folds narrowly enclose large mitochondria. [(B)‐(F)] Salient features of interdigitated epithelia. (B) Freeze fracture electron micrograph of a tight junction (TJ) (e.g., thick ascending limb) consisting of several densely arranged parallel strands. Rabbit (Cooperation with A. Schiller and R. Taugner). (C) TEM of interdigitated lateral folds, enclosing large mitochondria (M), and narrow intercellular spaces (arrows) of regular width (e.g., thick ascending limb epithelium); cell adhesion molecules within the intercellular spaces are contrasted by tannic acid staining; BL, basal lamina; Rat; (cooperation with T. Sakai). (D) Freeze‐fracture electron micrograph of the luminal aspect of a tubule demonstrating the mode of cellular interdigitation (e.g., ascending thin limb and Psammomys obesus) [from Kriz et al., with permission 345]. (E) Freeze fracture electron micrograph of a TJ in noninterdigitated epithelia (e.g., collecting duct cell); the prominent tight junction consists of several anastomosing tight junctional strands. (F) Infolded basal membranes confining extracellular spaces (arrows) which may vary in width with function; BL, basal lamina. (G) Freeze fracture electron micrograph of the basal aspect of a noninterdigitated cell (e.g., cortical collecting duct); the polygonal shape of this cell is recognized by contours of basal slits through which lateral intercellular spaces (white arrows) open into the interstitium; basal infoldings of extracellular spaces (black arrows) are separated from lateral intercellular spaces. Bars ∼1 μm (D, F, and G) and ∼0.1 μm (B, C, and E). Adapted, with permission, from Kaissling and Kriz 291.



Figure 3.

Schematic three‐dimensional appearance of a proximal tubule cell from the convoluted part of the rat proximal tubule, illustrating the appearance of the lateral and basal ridges and processes. Adapted, with permission, from Maunsbach 426.



Figure 4.

Schematic representation of the typical mouse nephron and collecting duct (CD) organization: Tortuous descending thin limbs (DTL) of long‐loop nephrons (LLN; large arrow); winding course of thick ascending limbs (TAL) of short loop nephrons (SLN) and CD (arrow heads); a piece of TAL inserted in the DTL of LLN (LLNt, small arrow), which forms its bend just beneath the “transitional zone” within the inner medulla (IM); and three different types of SLN bends (SLN1, SLN2, and SLN3). Based on the distribution of the nephron segments, the renal zones are defined, including cortex, outer stripe of outer medulla (OSOM), inner stripe of outer medulla (ISOM), and IM. The postmacula densa segments and the directions of the proximal tubules (PT) arising from their glomeruli are also illustrated. Adapted, with permission, from Zhai et al. 762.



Figure 5.

Electron micrograph of rat kidney cortex showing several proximal tubules with open lumens and evenly arranged brush borders. The micrograph illustrates some of the differences between the two segments of the convoluted part of the proximal tubule. In the first segment (S1) the cells are slightly taller, microvilli of the brush border longer, and apical endocytic vacuoles more numerous than in the second segment (S2). The lysosomes in the first segment have light contents whereas the lysosomes in the second segment show densely stained contents. DCT, distal convoluted tubule. The tissue was fixed by vascular perfusion with glutaraldehyde. X 2,100. Adapted, with permission, from Maunsbach 424.



Figure 6.

Electron micrographs and schematic drawings of proximal tubule epithelium from rat and rabbit kidney illustrating the three segments of the proximal tubule. (A) Rat, first segment (S1), (B) rat second segment (S2), (C) rat, third segment (S3), (D) rabbit, third segment (S3), and (E) schematic drawings of the three segments in rat. In segments S1 and S2 the mitochondria (M) are oriented perpendicular to the basement membrane and endocytic vacuoles (E) are numerous. The brush border (BB) is taller in S1 than in S2. The matrix of the lysosomes (L) in S1 is lightly stained but rather homogenous and densely stained in S2. The mitochondria in S3 are more randomly oriented than in S1 and S2 and they are less numerous in the rabbit than in the rat cells. In both species the lysosomes in S3 cells appear smaller and fewer as compared to S1 and S2 of the rat. The brush border of this segment is significantly higher in rat than in rabbit. Part of a cilium (C) is present in the brush border in C and peroxisomes (P) are seen in the cytoplasm. Capillary (CAP). The diagram in E illustrates the differences in cell shapes and the different distributions of mitochondria (M), lysosomes (L), endocytic vacuoles (E), and peroxisomes (microbodies) (P). S1, first segment. S2, second segment. S3, third segment. Adapted, with permission, from Maunsbach 426 and Maunsbach and Christensen 429. [(A) and (B)] X 8,900 and [(C) and (D)] X 9,800.



Figure 7.

(A) Tight junction (arrowheads) between two rat proximal tubule cells close to the tubule lumen (LU). The triple‐layered plasma membranes are completely apposed. The tight junction has a depth in the luminal‐basal direction of only 100 to 200 Å and is followed on its peritubular side by an intermediate junction. X 167,000. Adapted, with permission, from Maunsbach 424. (B) Tangential section through luminal part of proximal tubule cells showing an intermediate junction, which extends between the two pairs of arrowheads. When the junction is sectioned at right angle it can be noted that the cytoplasm next to the triple‐layered plasma membranes has an increased density and that a dense material fills the gap between the membranes (compare with normal lateral intercellular space at LIS). Lumen (LU) is seen in the upper right corner. X 145,000. Adapted, with permission, from Maunsbach 426. (C) Higher magnification of gap junction between proximal tubule cells from Necturus maculosus. The 20‐Å wide space between the two plasma membranes is clearly observable (arrowheads). X 204,000. Adapted, with permission, from Maunsbach 426.



Figure 8.

Parts of proximal tubule cells from rat and rabbit kidney illustrating smooth surfaced endoplasmic reticulum and peroxisomes. (A) Peroxisomes (P) (microbodies) from rat proximal tubule cell limited by a single membrane and showing a uniformly stained matrix. Smooth‐surfaced endoplasmic reticulum closely surrounds the peroxisomes (arrowheads) and is seen in continuity with the rough endoplasmic reticulum at arrow. X 44,000. Adapted, with permission, from Maunsbach and Christensen 429. (B) Smooth‐surfaced endoplasmic reticulum adjacent to the lateral plasma membrane. This so‐called paramembraneous endoplasmic reticulum (PER) has fenestrated cisternae (arrows) that closely follow the lateral plasma membrane. However, it is never in contact with this membrane, nor is it in continuity with the plasma membrane facing the tubule lumen (LU), endocytic vacuoles (E), or lysosomes. (A) X 95,000. Adapted, with permission, from Maunsbach 428. (C) Peroxisomes (P) in cells of isolated perfused proximal tubule of rabbit. In this species, part of the limiting membrane is straight, thickened, and electron dense. The limiting membrane is closely related to the smooth endoplasmic reticulum. Since this tubule was incubated with horseradish peroxidase (HRP) in the bath, the intercellular spaces, except in gap junction (GJ), are filled with reaction product for HRP. Reaction product is also seen in a small endocytic vesicle (V). X 45,000. Adapted, with permission, from Nielsen and Christensen 470. (D) Demonstration of dextran in rat proximal tubule cells 1 h after termination of intravenous infusion of dextran T‐40. The kidney was fixed in vivo by dripping a lead‐containing fixative on the kidney surface. Small dense particles representing dextran are present in lysosomes (arrows), where they are located adjacent to the dense matrix material. A peroxisome (P) with a fingerlike projection is well stained by the lead‐containing fixative. X 15,800. Adapted, with permission, from Christensen and Maunsbach 118.



Figure 9.

Electron micrographs illustrating the vacuolar apparatus in rat proximal tubule cells. (A) A round lysosome (L) with an electron‐dense matrix devoid of inclusions and located close to a mitochondrion (M). The lysosome is limited by a triple‐layered membrane. X 100,000. (B) Coated pit connected to the tubular lumen (LU). The coated pit has a thick cell coat or glycocalyx (arrow) and a cytoplasmic clathrin coat (arrowheads). X 100,000. Adapted, with permission, by A.B. Maunsbach. (C) Dense apical tubules connected to endosomes (S) in apical cytoplasm. X 70,000. Adapted, with permission, from Cui and Christensen 138.



Figure 10.

Isolated perfused proximal tubule of rabbit incubated with horseradish peroxidase in the bath for 30 min. The reaction product is seen in the lateral intercellular spaces (LIS) but not in the lumen (LU). Note the reaction product in a multivesicular body (MVB) and in lysosomes (L). Unstained section. X 15,000. Adapted, with permission, from Nielsen and Christensen 470.



Figure 11.

Immunofluorescence (mouse kidney) and immunogold (rat proximal tubule) localization of megalin and cubilin. The immunofluorescence reveal strong apical labeling in proximal tubules but no obvious labeling in glomeruli (G). Distal tubules (D) and vessels (A) are unlabeled. Bar, 100 μm. Immunogold labeling, megalin, 10 nm gold and cubilin, 5 nm gold, reveals colocalization of the two receptors in coated pits (Inv), coated vesicles (C), endosomes (E), and in recycling dense apical tubules (arrows). Bar, 0.5 μm.



Figure 12.

Schematic drawing of megalin, cubilin, and amnionless (AMN) presenting known domains and motifs. The three receptors colocalize in the renal proximal convoluted tubule (PCT) where they cooperate in clearance of the ultrafiltrate. Megalin binds a variety of filtered molecules (more than 50 ligands have been identified) through its complement type repeats and is able to mediate endocytosis via NPXY motifs in the cytoplasmic tail. Cubilin on the other hand includes multiple binding domains (CUB domains), but only around 15 ligands have been identified. Cubilin is a peripheral membrane protein and is thereby dependent on megalin and/or AMN to assure internalization of its ligands. AMN contains a NPXY motif and is probably assisting cubilin in endocytosis as well as in transport during synthesis. Adapted, with permission, from Christensen et al. 123.



Figure 13.

The megalin‐ and cubilin‐mediated uptake of three vitamin carrier protein complexes: DBP‐vitamin D3, TC‐vitamin B12, and RBP retinol in renal proximal tubule. Likewise the cubilin chaperone protein amnionless, amnionless (AMN) is indicated. Following receptor‐mediated endocytosis via apical coated pits, the complexes accumulate in lysosomes for degradation of the proteins, while the receptors recycle to the apical plasma membrane via dense apical tubules. As illustrated here and detailed in the text, megalin mediates the uptake of cubilin and its ligands. Whether the two receptors are constitutively associated in the plasma membrane and remain associated during recycling in dense apical tubules is not known. Whereas TC and RBP apparently bind exclusively to megalin, DBP binds with similar affinity to both megalin and cubilin. The intracellular processing of the vitamins may include modifications such as hydroxylation of 25‐OH‐D3 to 1,25‐(OH)2‐D3, and metaobolism of B12. The mechanisms for the cellular release of the vitamins remain to be clarified. Adapted, with permission, from Verroust et al. 700.



Figure 14.

Proximal tubular uptake of albumin A and B and insulin C, in rat A and C and in mouse B. (A) Double immunogold labeling of endogenous albumin (18 nm gold) and cathepsin B (6 nm gold) on ultrathin cryosection from rat kidney. The albumin labeling is highly concentrated in a lysosome (L). X 43,000 [adapted, with permission, from Christensen et al. 123]. (B) Immunofluorescence of paraffin section of cortex from kidney‐specific megalin mosaic KO mice by using sheep anti‐rat megalin (green), rabbit anti‐human albumin (red). Proximal tubule cells not expressing megalin (arrows) have no labeling for albumin. X 550 [adapted, with permission, from Christensen et al. 123]. (C) Electron microscope autoradiograph of 125I‐labelled insulin uptake in rat proximal tubule, fixed 25 min after start of microinfusion of the insulin. Grains are mainly located over lysosomes in the apical part of the cytoplasm (arrows). Endocytic vacuoles (E) and some lysosomes (L) located deep in the cytoplasm do not contain 125I‐insulin. X 16,000. Adapted, with permission, from Hellfritzsch et al. 248.



Figure 15.

Epon section from border between the outer and inner stripe of the outer zone of the medulla of rat labeled for AQP‐1 labeled by immunoperoxidase by method described, with permission, by Zhai et al. 761. Segment 3 proximal tubules show an intense brush border labeling and basolateral labeling (arrowheads). Two transitions into nonlabeled, thin‐walled DTL type 1 epithelium of short‐loop nephrons (arrows) are shown. X 400.



Figure 16.

Schematic presentation of the tubular‐vascular relationship in the outer part of the inner stripe of outer medulla (ISOM). Tl: thick ascending limb of long‐looped nephrons. Ts: thick ascending limb of short‐looped nephron. Dl: descending thin limb of long‐looped nephron. Ds: descending thin limb of short‐looped nephron. C: collecting duct. VB: vascular bundle. IB: interbundle region. Kindly provided, with permission, by Dr. X.Y. Zhai].



Figure 17.

(A) Schematic of thin limb ultrastructure. Type 1 is found in descending thin limb (TL) of short loop nephrons (SLNs). Type 2 is found in upper part of descending TLs of long loop nephrons (LLNs); in most species (e.g., rat, mouse, and Psammomys), it is complexely organized, in others (e.g., rabbit and guinea pig), it is more simple. Type 3 occurs in lower parts of descending TL of LLNs. Type 4 is found at the loop bend and in ascending TL of LLNs. Modified, with permission, from Kaissling and Kriz 291. (B) Four distinct epithelia along the TL of SLN and LLN in mouse. The figure comprises two columns of images representing lower and higher magnifications (right). Type 1 is a cross section of the DTL of an SLN (440 μm from the transition of the PT to the DTL); higher magnification of type 1 shows a simple and thin epithelium with few microprojections, abundant ribosomes, and sparse mitochondria throughout the cytoplasm. Type 2 is a cross section of the DTL of an LLN (370 μm from the transition of the PT to the DTL); higher magnifications of type 2 show a highly specialized epithelium: abundant short and plump microvilli, basolateral infoldings, and highly interdigitated cellular processes with shallow tight junctions (small arrows) and radially oriented mitochondria. Type 3 is a cross section of a DTL of LLN around the boundary between the inner stripe of outer medulla (ISOM) and the inner medulla (IM) (2790 μm from the transition of PT to DTL); higher magnifications of type 3 show the highly differentiated membranes: Numerous relatively long microvilli, abundant finger‐like basolateral infoldings throughout the cytoplasm (large arrows), and the less shallow tight junctions (small arrow) and few mitochondria. Type 4 is a cross section of the ascending thin limb (ATL) of an LLN (700 μm from the transition of the ATL to the TAL); higher magnifications of type 4 show the bold and dense apical membrane, basolateral infoldings, cellular processes with tight junctions (small arrows), and various organelles with sparse ribosomes throughout the cytoplasm. Bars, 1 μm. Adapted, with permission, from Zhai et al. 762.



Figure 18.

Peroxidase immunohistochemistry of an Epon section showing the transition (arrows) from UT‐A2‐labeled DTL into the nonlabeled, thick ascending epithelium of a short loop nephron (SLN) of mouse kidney. X 200. Adapted, with permission, from Zhai et al. 762.



Figure 19.

Survey on location (left panel) and ultrastructure of the thick ascending limb of Henle's loop [TAL = distal straight tubule, including macula densa (MD)]; C, cortex; IS, inner stripe; OS, outer stripe; IZ, inner zone; the direction of the urinary flow is indicated by white arrows, (A) medullary part, (B) cortical part, and (C) MD. adapted, with permission, from 288,291.



Figure 20.

Overview on the histotopography of the thick ascending limb (T); (A) cross section of the inner stripe; the upper portion shows part of a vascular bundle with descending (a) and ascending vasa recta (v); in the lower half thick ascending limbs are intermingled with descending thin limbs (DL), collecting ducts (CDs), and capillaries from the interbundle capillary plexus. (B) Longitudinal section of the deep cortex in rat; the thick ascending limb (TAL) profiles reveal a much thinner epithelium than in the inner stripe; they are grouped around the CDs and surrounded by profiles of proximal tubules [adapted, with permission, from 291]. Inset: longitudinal section through the sharp transition (arrowheads) from the thin epithelium of the ascending limb (AL) to the TAL (T) in mouse. [(A) and inset] ∼10 μm and (B) ∼50 μm.



Figure 21.

Ultrastucture of thick ascending limb cells. (A) Deep level of the inner stripe; rat; (B) cortical part; rat; and (C) macula densa cells; rabbit. Mitochondria (arrow head) in A and B are confined in the interdigitated lateral folds and the intercellular spaces (long arrow) are narrow; in C the mitochondria are scattered in the perinuclear cytoplasm and the intercellular spaces are dilated; the macula densa (MD) cells are affixed to the extraglomerular mesangium (star); [(A) and (B) adapted, with permission, from 291 and (C) from 290]. Bars ∼ 1 μm.



Figure 22.

Macula densa (MD). (A) Preparation of a isolated human renal corpuscle (G) together with the cortical radial artery (A), glomerular arterioles (red), and tubules; proximal tubule (P; gray); the thick ascending limb (T, yellow) ascends along the cortical collecting duct (C; violet), bends toward the vascular pole of its glomerulus and passes between the glomerular arterioles; it continues as distal convoluted tubule (DCT, orange); the asterisk indicates the location of the MD. [Slightly modified, with permission, from Karl Peter (1909) 517]. (B) Scanning electron micrograph; the cortical thick ascending limb (CTAL) is open at the level of the MD, the luminal aspect of rabbit MD cell plaque is visible; single cilia are seen on MD cells as well as on CTAL cells; EGM, extraglomerular mesangium [adapted, with permission, from 291]. (C) Epon section of rat MD within the cortical thick ascending limb; the epithelium reveals conspicuously dilated intercellular spaces; those between thick ascending limb cells are narrow; shortly after the MD the epithelium transforms (arrowheads) to that of the DCT (D); arrow, indicates flow direction; (ea), efferent arteriole. (D) Epon section of mouse MD in contact with granulated rennin‐producing cells (GC) in the afferent glomerular arteriole; the MD cells display abundant microvilli on their luminal surface as well as single cilia (arrows); the intercellular spaces are narrow. Bars ∼ 10 μm.



Figure 23.

Organization of the transition from the DCT to the collecting duct in the cortex of rabbit. (A) Connecting tubule (CN) of a superficial nephron, opening into the beginning of a cortical collecting duct (CD) under the renal capsule; the sharp transition in the epithelial lining is indicated by a bar in the lumen. (B) Sharp transition (bar) of DCT (D) to CNT (CN); fusion of several CNTs establishes an arcade that ascends (arrow) in the cortex; note the progressive decrease in height of the CNT epithelium along the CNT. (C) Arcade (CN), ascending in the cortex in close proximity to the cortical radial artery (A), takes up several tributaries. (D) Opening of an arcade (CN) in the upper third of the cortex into a CCD within a medullary ray; the arrows indicate the flow directions; note the proximity of the arcade to an afferent arteriole (white arrow head). (E) Juxtaposition of an arcade (CN) to a lymphatic (L) in the periarterial connective tissue of the cortical radial artery (A). [(A) and (B)] Epon sections of rabbit kidney, (E) of mouse kidney; [(C) and (D)] paraffin sections of rabbit kidney. Bars ∼ 50 μm. [(A) and (B)] [adapted, with permission, from 292]; [(C) and (D)] [adapted, with permission, from 288], (E) [adapted, with permission, from 286].



Figure 24.

Survey on organization of the cortical distal segments and collecting ducts (CDs; left panel) and on ultrastructure of the segment‐specific cells (right panel); C ‐ cortex, OS, outer stripe, IS, inner stripe of the outer medulla; IM, inner medulla; dashed line, delimits the medullary ray; (a) distal convoluted tubule (DCT) and (A) DCT‐cell; (b) connecting tubule (CNT) and (B) CNT‐cell; (c) CCD and (C) CCD cell; (d) inner medulla (IM), and (D) IMCD cell; the black semicircles indicate the occurrence of intercalated (IC) cells in the segment; adapted, with permission, from 291.



Figure 25.

(A) The thick ascending limb (T) transforms shortly after the macula densa (MD), affixed to the granulated portion of the afferent glomerular arteriole (aa), to the distal convoluted tubule 1 (D); the abrupt change in the epithelial structure (arrowheads) coincides with the marked increase in contrast; the appearance of an intercalated cell (asterisk) in the DCT epithelium indicates the transition (arrowheads) to the transitional segment (D2), which shows a slightly lower density of mitochondria; mouse; epon section; Bar ∼ 10 μm. (B) DCT1 cell in rat; characteristic are the tall mitochondria enclosed in lateral interdigitating cell processes, the apical position and apical flattening of the nucleus and the narrow mitochondria‐free cytoplasmic rim on top of the nucleus, containing many small vesicles; arrowhead, single cilium. Epon section; bar ∼ 1 μm. Adapted, with permission, from 341.



Figure 26.

Comparison of salient features of DCT, CNT, and collecting duct segments and cells in the renal cortex. (A) Mouse renal cortex showing side by side a profile of the distal convoluted tubule 2 (D2), of a connecting tubule (CN), and a collecting duct (CCD); the decrease of density in mitochondria, their change in cellular position from basal infoldings to the apical cytoplasm, the decrease in cell height, and the increase in the frequency of intercalated cells (asterisks) in the epithelial lining is evident. Epon section; bar ∼ 10 μm. [(B)‐(D)] Higher magnification of a DCT cell (B), a CNT cell (C), and a CCD cell (D); compare with (A); the arrows point to interdigitating cell processes, narrowly enclosing large mitochondria (B), abundant deep infolding of the basal plasma membrane reaching between tall mitochondria (C) and to infoldings of the basal plasma membrane restricted to a narrow rim in the basal cell portion in the noninterdigitating CCD cell (D); all mitochondria and cell organelles are located above this basal rim. [(B) and (C)] From Psammomys obesus; (D) rat; fixation by reduced osmium. TEM, bar ∼ 1 μm. [(B)‐(D)] Modified, with permission, from 341.



Figure 27.

Connecting tubule (CNT) in rat. (A) Longitudinal section through renal cortex, showing profiles of CNT, forming arcades, ascending (arrows) in the cortex in close juxtaposition to the cortical radial vessels (CRV) and glomerular afferent arterioles (aa); bar ∼ 50 μm. (B) The CNT epithelium is composed of CNT cells and intercalated cells (arrows); bar ∼ 10 μm. (C) Characteristic CNT cell with abundant infoldings of the basal plasmalemm extending deeply into the cytoplasm; mitochondria are found between the infolded membranes and in the apical cytoplasm on top of the nucleus; bar ∼ 10 μm. (D) The infolded plasma membranes reveal numerous caveolae; bars ∼ 0.01 μm. Adapted, with permission, from 291.



Figure 28.

Cortical [(A) and (B)] and outer medullary [(C) and (D)] collecting duct. (A) Collecting duct in the upper cortex of rat; the epithelium is composed by collecting duct (CD) cells and intercalated cells (IC cells; asterisks); T, thick ascending limb: P, proximal tubule. (B) Cortical collecting duct (CCD) cell; infoldings of the basal plasma membrane are restricted to the basal cell portion; all mitochondria and cell organelles are located above the infolded membranes. (C) Collecting duct in the inner stripe of mouse; the epithelial organization is similar as in the cortex; note the different aspect of IC cells (asterisks); (T) thick ascending limb. (D) Outer medullary conduction duct (OMCD) cell in the inner stripe; the organization is similar as in the CCD; the infoldings of the basal plasmalemm and the amount of mitochondria decrease toward the medulla. Bars: [(A) and (C)] ∼ 10 μm; [(B) and (D)] ∼ 1 μm. [(B) adapted, with permission, from 291 and (D) adapted, with permission, from 341].



Figure 29.

Inner medullary collecting duct. (A) Tubular profile of a medullary collecting duct in the transitional zone from the inner stripe of the outer medulla to the inner zone in rabbit; the inner medullary conducting duct (IMCD) cells in the homogenous epithelium possess a dense apical cytoskelettal web under the luminal plasmalemma, indicated by a star in two cells, under the luminal plasmalemm; the cytoplasm reveals a paucity of cell organelles; the big round nucleus has a rather homogeneous chromatin structure. (B) Tip of the renal papilla in rat with openings of papillary CDs (paraffin section) into the renal pelvis; the urothelium of the pelvic wall is seen on the right. (C) Epithelium of the middle portion of an IMCD in rat. Within the epithelium three zones are seen: a basal zone with basal infoldings, a middle zone containing Golgi fields, mitochondria and lysosomal elements, and a thin apical zone with abundant tubular and vesicular profiles. Note the deep tight junction. (D) IMCD cells in the deep inner medulla; the tall cells have a large nucleus and relative to their size few cell organelles are encountered; lateral intercellular spaces (arrow) are filled with microvilli and microfolds; the cytoplasm immediately beneath the apical plasmalemm is devoid of cell organelles. Bars: (A) ∼ 10 μm; (B) ∼ 100 μm; (C) ∼ 1 μm; and (D) ∼ 10 μm. Adapted, with permission, from 341.



Figure 30.

Schematic distribution of the major apical transport proteins (NKCC2, black; NCC, green; TRPM6, turquoise; TRPV5, orange; ENaC, red; and AQP2, blue), along the cortical distal segments in rabbit 1, and in rat, mouse, and human 2; MR, medullary ray; TAL, thick ascending limb; G, renal corpuscle; DCT, distal convoluted tubule; CNT, connecting tubule; CCD, cortical collecting duct; restriction of a transporter to a tubular portion is indicated by a thick vertical bars, the continuation along the collecting duct (CD) by arrows. Modified, with permission, from 341.



Figure 31.

Transition from the thick ascending limb to the distal convoluted tubule [(A)‐(C)]: rabbit; [(D)‐(F)]: mouse; [(A) and (D)] 1 μm epon sections, all others cryostat sections; [(B) and (E)] immunofluorescence for NKCC2; (C) in situ hybridization for mRNA of SLC12a3 (coding for NCC), (F) immunofluorescence for NCC, same section as E (double fluorescence); in both species the sharp change in morphology is congruent with the abrupt change from NKCC2 to NCC (arrowheads). Bars: ∼ 50 μm. Adapted, with permission, from 385.



Figure 32.

Transitional regions from the distal convoluted tubule (D) to the connecting tubule (CN) and the cortical collecting duct (CCD) in rabbit. [(A) and (E)] 1 μm epon sections, all others cryostat sections. (B) In situ hybridization for mRNA of SLC12a3 (coding for NCC). (C) Same section as (B), immunostaining for the β subunit of epithelial sodium channel (ENaC); no overlap of NCC and ENaC, arrow heads point to the definite stop of NCC mRNA and the abrupt beginning of apical ENaC staining; no overlap of NCC and ENaC in the same cell. (D) Immunostaining of a consecutive section for Calbindin k28D (CB); the end of NCC expression and the beginning of apical ENaC coincides with a marked increase of CB staining in the cytoplasm of tubular cells and the appearance of intercalated cells in the tubular epithelium (asterisks). (E) The transition of the CNT to the CCD is marked by the obvious change in the epithelial structure at the opening of the CNT (CN) to the CCD (arrowheads) indicates the transition of the CNT to the CCD. (F) The structural change coincides with a shift of luminal ENaC immunostaining to the cytoplasm (arrowheads); (G) with a sharp reduction of CB immunostaining (arrowheads); and (H) and the clear‐cut onset of luminal AQP2 immunostaining. Bars: ∼ 50 μm. Adapted, with permission, from 380.



Figure 33.

Transitional regions from the distal convoluted tubule to the connecting tubule and the cortical collecting duct (CCD); mouse. [(A) and (E)] 1 μm epon sections, all others, cryostat sections. (A) The most upstream occurrence of intercalated cells (IC cells; asterisks) indicates the transition from the DCT1 (D1) to the transitional segment of the DCT, the DCT2 (D2); the frequency of IC cells increases toward and along the CNT (CN), note the marked decrease of epithelial lining along the CNT segment. [(B) and (C)] Double immunostaining for NCC and the β subunit of epithelial sodium channel (ENaC); NCC, and ENaC overlap in the cells of DCT2 (D2); (D) consecutive section, immunostained for the vasopressine‐sensitive water channel aquaporin 2 (AQP2); its onset coincides with the disappearance of NCC expression and defines the beginning of the CNT. (E) Transition from the CNT (CN) to the CCD. [(F) and (G)] Consecutive sections, immunostained for β subunit of ENaC and AQP2; both are coexpressed by the CNT cells as well as by the collecting duct cells (CD). Bars: ∼ 50 μm. Adapted, with permission, from 385 and 380.



Figure 34.

Distribution of NCC, the epithelial calcium channel TRPV5 and the sodium calcium exchanger (NCX) along the distal nephron. Cryostat sections; [(A) and (B)] rabbit; [(C)‐(G)] mouse. [(A)‐(D)] Double immunostaining for NCC and TRPV5; [(A) and (B)] in rabbit NCC in the distal convoluted tubule (D) and TRPV5 in the connecting tubule (CN) are exclusive; [(C) and (D)] In mouse, NCC and TRPV5 are coexpressed in the DCT2 (D2), TRPV5 continues in the CNT (CN), whereas NCC is absent in the CNT. [(E) and (F)] Immunofluorescence for TRPV5 is the highest in the apical cytoplasm in DCT2 and decreases progressively and shifts to the cytoplasm along the CNT (CN); the arrow indicates the urinary flow direction; the segmental distribution of the sodium calcium exchanger (NCX) in the basolateral membrane parallels that of TRPV5; weak NCX staining is present in DCT1 (D). [(G) and (H)] TRPV5 and NCX expressions sharply stop at the transition from the CNT to the cortical collecting duct (CCD). Bars ∼ 50 μm. Adapted, with permission, from Loffing et al. 385 and Loffing and Kaissling 380.



Figure 35.

Rat cortical collecting duct with two distinct types of intercalated cells (IC cells). Type A IC cells marked with an asterix, type B IC cells with #. Original magnification ∼2200×. Adapted, with permission, from 291.



Figure 36.

Scheme of the typical appearance of the different types of intercalated cells (IC cells). (A) Type A IC cell. (B) Type B IC cell. (C) Medullary IC cell. Adapted, with permission, from 291.



Figure 37.

Rat intercalated cells (IC cells). (A) Type A IC cell with abundant apical microfolds and many mitochondria. (B) Type B IC cells cell with a rather narrow apical cell pole, a narrow rim of dense cytoplasm exempt of vesicles under the apical plasma membrane, abundant smooth surfaced vesicles in the apical cytoplasm, a huge Golgi complex, and abundant mitochondria along the basolateral plasma membrane. Transmission electron microscopy ∼7000× magnification.



Figure 38.

Expression of H+‐ATPase subunits in intercalated cells (IC cells): (A) mouse connecting tubule stained with antibodies against the B1 H+‐ATPase subunit (red) and the segment cell specific water channel aquaporin 2 (AQP2) (green). B1 subunits are detected in luminal (cells with arrows) and/or basolateral membranes (cell with asterix). Adapted, with permission, from 189. (B) The a4 H+‐ATPase subunit is expressed in various nephron segments including the proximal tubule and also in IC cells. Mouse CNT stained with antibodies against the a4 H+‐ATPase (red) and the AQP2 water channel labeling segment specific cells (green).



Figure 39.

Expression of intercalated cell (IC cell) specific anion exchangers AE1 and pendrin. (A) Mouse outer medullary collecting duct stained against AE1 (red) and aquaporin 2 (AQP2) water channels (green). AE1 shows a basolateral localization in cells negative for AQP2. (B) Colocalization of the E subunit of apical H+‐ATPase (green) and basolateral AE1 (red) in rat outer medullary collecting duct IC cells. (C) The anion exchanger pendrin (red) is localized at the luminal pole of non‐type A (i.e., type B and non‐A/non‐B IC cells) IC cells in mouse CNT. Segment specific cells are identified by the presence of the AQP2 water channel. Some cells are negative for pendrin and AQP2 (asterix), presumably type A IC cells.

References
 1. Abrahamson DR, Irwin MH, St John PL, Perry EW, Accavitti MA, Heck LW, Couchman JR. Selective immunoreactivities of kidney basement membranes to monoclonal antibodies against laminin: Localization of the end of the long arm and the short arms to discrete microdomains. J Cell Biol 109: 3477‐3491, 1989.
 2. Ackermann D, Gresko N, Carrel M, Loffing‐Cueni D, Habermehl D, Gomez‐Sanchez CE, Rossier BC, Loffing J. In vivo nuclear translocation of mineralocorticoid and glucocorticoid receptors in rat kidney: Differential effect of corticosteroids along the distal tubule. Am J Physiol Renal Physiol 299: F1473‐F1485, 2010.
 3. Agre P, Saboori AM, Asimos A, Smith BL. Purification and partial characterization of the Mr 30,000 integral membrane protein associated with the erythrocyte Rh(D) antigen. J Biol Chem 262: 17497‐17503, 1987.
 4. Ahn KY, Kone BC. Expression and cellular localization of mRNA encoding the “gastric” isoform of H(+)‐K(+)‐ATPase alpha‐subunit in rat kidney. Am J Physiol 268: F99‐F109, 1995.
 5. Ahn KY, Park KY, Kim KK, Kone BC. Chronic hypokalemia enhances expression of the H(+)‐K(+)‐ATPase alpha 2‐subunit gene in renal medulla. Am J Physiol 271: F314‐F321, 1996.
 6. Alcorn D, Anderson WP, Ryan GB. Morphological changes in the renal macula densa during natriuresis and diuresis. Ren Physiol 9: 335‐347, 1986.
 7. Allen F, Tisher CC. Morphology of the ascending thick limb of Henle. Kidney Int 9: 8‐22, 1976.
 8. Alper SL, Natale J, Gluck S, Lodish HF, Brown D. Subtypes of intercalated cells in rat kidney collecting duct defined by antibodies against erythroid band 3 and renal vacuolar H+‐ATPase. Proc Natl Acad Sci U S A 86: 5429‐5433, 1989.
 9. Amann K, Plank C, Dotsch J. Low nephron number–a new cardiovascular risk factor in children? Pediatr Nephrol 19: 1319‐1323, 2004.
 10. Ambuhl PM, Amemiya M, Danczkay M, Lotscher M, Kaissling B, Moe OW, Preisig PA, Alpern RJ. Chronic metabolic acidosis increases NHE3 protein abundance in rat kidney. Am J Physiol 271: F917‐F925, 1996.
 11. Amemiya M, Loffing J, Lotscher M, Kaissling B, Alpern RJ, Moe OW. Expression of NHE‐3 in the apical membrane of rat renal proximal tubule and thick ascending limb. Kidney Int 48: 1206‐1215, 1995.
 12. Amsellem S, Gburek J, Hamard G, Nielsen R, Willnow TE, Devuyst O, Nexo E, Verroust PJ, Christensen EI, Kozyraki R. Cubilin is essential for albumin reabsorption in the renal proximal tubule. J Am Soc Nephrol 21: 1859‐1867, 2010.
 13. Anderson JM, Van Itallie CM. Tight junctions and the molecular basis for regulation of paracellular permeability. Am J Physiol 269: G467‐G475, 1995.
 14. Anderson JM, Van Itallie CM. Physiology and function of the tight junction. Cold Spring Harb Perspect Biol 1: a002584, 2009.
 15. Andrews PM. Morphological alterations of the glomerular (visceral) epithelium in response to pathological and experimental situations. J Electron Microsc Tech 9: 115‐144, 1988.
 16. Andrews PM, Porter KR. A scanning electron microscopic study of the nephron. Am J Anat 140: 81‐115, 1974.
 17. Angelow S, Ahlstrom R, Yu AS. Biology of claudins. Am J Physiol Renal Physiol 295: F867‐F876, 2008.
 18. Ardiles LG, Loyola F, Ehrenfeld P, Burgos ME, Flores CA, Valderrama G, Caorsi I, Egido J, Mezzano SA, Figueroa CD. Modulation of renal kallikrein by a high potassium diet in rats with intense proteinuria. Kidney Int 69: 53‐59, 2006.
 19. Arpin‐Bott MP, Kaissling B, Waltisperger E, Rabhi M, Saussine P, Freund‐Mercier MJ, Stoeckel ME. Historadioautographic localization of oxytocin and V1a vasopressin binding sites in the kidney of developing and adult rabbit, mouse and merione and of adult human. Exp Nephrol 10: 196‐208, 2002.
 20. Bachmann S, Dawnay AB, Bouby N, Bankir L. Tamm‐Horsfall protein excretion during chronic alterations in urinary concentration and protein intake in the rat. Ren Physiol Biochem 14: 236‐245, 1991.
 21. Bachmann S, Koeppen‐Hagemann I, Kriz W. Ultrastructural localization of Tamm‐Horsfall glycoprotein (THP) in rat kidney as revealed by protein A‐gold immunocytochemistry. Histochemistry 83: 531‐538, 1985.
 22. Bachmann S, Kriz W. Histotopography and ultrastructure of the thin limps of the loop of Henle in the hamster. Cell Tissue Reseach 225: 111‐127, 1982.
 23. Bachmann S, Kriz W, Kuhn C, Franke WW. Differentiation of cell types in the mammalian kidney by immunofluorescence microscopy using antibodies to intermediate filament proteins and desmoplakins. Histochemistry 77: 365‐394, 1983.
 24. Bachmann S, Mutig K, Bates J, Welker P, Geist B, Gross V, Luft FC, Alenina N, Bader M, Thiele BJ, Prasadan K, Raffi HS, Kumar S. Renal effects of Tamm‐Horsfall protein (uromodulin) deficiency in mice. Am J Physiol Renal Physiol 288: F559‐F567, 2005.
 25. Bachmann S, Velazquez H, Obermuller N, Reilly RF, Moser D, Ellison DH. Expression of the thiazide‐sensitive Na‐Cl cotransporter by rabbit distal convoluted tubule cells. J Clin Invest 96: 2510‐2514, 1995.
 26. Bacic D, Capuano P, Gisler SM, Pribanic S, Christensen EI, Biber J, Loffing J, Kaissling B, Wagner CA, Murer H. Impaired PTH‐induced endocytotic down‐regulation of the renal type IIa Na+/Pi‐cotransporter in RAP‐deficient mice with reduced megalin expression. Pflugers Arch 446: 475‐484, 2003.
 27. Bacic D, LeHir M, Biber J, Kaissling B, Murer H, Wagner CA. The renal Na+/phosphate cotransporter NaPi‐IIa is internalized via the receptor‐mediated endocytic route in response to parathyroid hormone. Kidney Int 69: 495‐503, 2006.
 28. Bacic D, Wagner CA, Hernando N, Kaissling B, Biber J, Murer H. Novel aspects in regulated expression of the renal type IIa Na/Pi‐cotransporter. Kidney Int Suppl 91: S5‐S12, 2004.
 29. Bagherie‐Lachidan M, McNeill H. What drives cyst formation in PKD? J Am Soc Nephrol 21: 200‐202, 2010.
 30. Baines AD, de RC. Functional heterogeneity of nephrons. II. Filtration rates, intraluminal flow velocities and fractional water reabsorption. Pflugers Arch 308: 260‐276, 1969.
 31. Bakris GL, Fonseca VA, Sharma K, Wright EM. Renal sodium‐glucose transport: Role in diabetes mellitus and potential clinical implications. Kidney Int 75: 1272‐1277, 2009.
 32. Balkovetz DF. Claudins at the gate: Determinants of renal epithelial tight junction paracellular permeability. Am J Physiol Renal Physiol 290: F572‐F579, 2006.
 33. Balkovetz DF. Tight junction claudins and the kidney in sickness and in health. Biochim Biophys Acta 1788: 858‐863, 2009.
 34. Bankir L, Bouby N, Trinh‐Trang‐Tan MM, Kaissling B. Thick ascending limb–anatomy and function: Role in urine concentrating mechanisms. Adv Nephrol Necker Hosp 16: 69‐102, 1987.
 35. Bankir L, Fischer C, Fischer S, Jukkala K, Specht HC, Kriz W. Adaptation of the rat kidney to altered water intake and urine concentration. Pflugers Arch 412: 42‐53, 1988.
 36. Bankir L, Kaissling B, Rouffignac CD, Kriz W. Vascular organization of the kidney of Psammomys obesus. Anat Embryol (Berl) 155: 149‐160, 1979.
 37. Barajas L. The ultrastructure of the juxtaglomerular apparatus as disclosed by three‐dimensional reconstructions from serial sections. The anatomical relationship between the tubular and vascular components. J Ultrastruct Res 33: 116‐147, 1970.
 38. Bargmann W, Krisch B, Leonhardt H. Lipids in the proximal convoluted tubule of the cat kidney and the reabsorption of cholesterol. Cell Tissue Res 177: 523‐538, 1977.
 39. Barrett JM, Kriz W, Kaissling B, de RC. The ultrastructure of the nephrons of the desert rodent (Psammomys obesus) kidney. II. Thin limbs of Henle of long‐looped nephrons. Am J Anat 151: 499‐514, 1978.
 40. Beard ME, Novikoff AB. Distribution of peroxisomes (microbodies) in the nephron of the rat: A cytochemical study. J Cell Biol 42: 501‐518, 1969.
 41. Beeuwkes R III. The vascular organization of the kidney. Annu Rev Physiol 42: 531‐542, 1980.
 42. Belge H, Gailly P, Schwaller B, Loffing J, Debaix H, Riveira‐Munoz E, Beauwens R, Devogelaer JP, Hoenderop JG, Bindels RJ, Devuyst O. Renal expression of parvalbumin is critical for NaCl handling and response to diuretics. Proc Natl Acad Sci U S A 104: 14849‐14854, 2007.
 43. Bell PD, Komlosi P, Zhang ZR. ATP as a mediator of macula densa cell signalling. Purinergic Signal 5: 461‐471, 2009.
 44. Bentzel CJ, Tourville DR, Parsa B, Tomasi TB. Bidirectional transport of horseradish peroxidase in proximal tubule of Necturus kidney. J Cell Biol 48: 197‐202, 1971.
 45. Bergeron M, Gaffiero P, Thiéry G. Segmental variations in the organization of the endoplasmic reticulum of the rat nephron. Cell Tissue Reseach 247: 215‐225, 1987.
 46. Bergeron M, Guerette D, Forget J, Thiery G. Three‐dimensional characteristics of the mitochondria of the rat nephron. Kidney Int 17: 175‐185, 1980.
 47. Berry CA. Heterogeneity of tubular transport processes in the nephron. Annu Rev Physiol 44: 181‐201, 1982.
 48. Biber J, Gisler SM, Hernando N, Murer H. Protein/protein interactions (PDZ) in proximal tubules. J Membr Biol 203: 111‐118, 2005.
 49. Biber J, Gisler SM, Hernando N, Wagner CA, Murer H. PDZ interactions and proximal tubular phosphate reabsorption. Am J Physiol Renal Physiol 287: F871‐F875, 2004.
 50. Biemesderfer D, DeGray B, Aronson PS. Active (9.6 s) and inactive (21 s) oligomers of NHE3 in microdomains of the renal brush border. J Biol Chem 276: 10161‐10167, 2001.
 51. Biemesderfer D, Pizzonia J, Abu‐Alfa A, Exner M, Reilly R, Igarashi P, Aronson PS. NHE3: A Na+/H +exchanger isoform of renal brush border. Am J Physiol 265: F736‐F742, 1993.
 52. Biemesderfer D, Rutherford PA, Nagy T, Pizzonia JH, Abu‐Alfa AK, Aronson PS. Monoclonal antibodies for high‐resolution localization of NHE3 in adult and neonatal rat kidney. Am J Physiol 273: F289‐F299, 1997.
 53. Bindels RJ. A molecular switch controlling renal sodium and potassium excretion. Nat Genet 35: 302‐303, 2003.
 54. Biner HL, Arpin‐Bott MP, Loffing J, Wang X, Knepper M, Hebert SC, Kaissling B. Human cortical distal nephron: Distribution of electrolyte and water transport pathways. J Am Soc Nephrol 13: 836‐847, 2002.
 55. Birn H, Christensen EI, Nielsen S. Kinetics of endocytosis in renal proximal tubule studied with ruthenium red as membrane marker. Am J Physiol 264: F239‐F250, 1993.
 56. Birn H, Vorum H, Verroust PJ, Moestrup SK, Christensen EI. Receptor‐associated protein is important for normal processing of megalin in kidney proximal tubules. J Am Soc Nephrol 11: 191‐202, 2000.
 57. Bishop JM, Verlander JW, Lee HW, Nelson RD, Weiner AJ, Handlogten ME, Weiner ID. Role of the rhesus glycoprotein, Rh B glycoprotein, in renal ammonia excretion. Am J Physiol Renal Physiol 299: F1065‐F1077, 2010.
 58. Biver S, Belge H, Bourgeois S, Van VP, Nowik M, Scohy S, Houillier P, Szpirer J, Szpirer C, Wagner CA, Devuyst O, Marini AM. A role for Rhesus factor Rhcg in renal ammonium excretion and male fertility. Nature 456: 339‐343, 2008.
 59. Bjerke T, Christensen EI, Boye N. Tubular handling of neurotensin in the rat kidney as studied by micropuncture and HPLC. Am J Physiol 256: F100‐F106, 1989.
 60. Blackman B, Russell T, Nordeen SK, Medina D, Neville MC. Claudin 7 expression and localization in the normal murine mammary gland and murine mammary tumors. Breast Cancer Res 7: R248‐R255, 2005.
 61. Bobulescu IA, Moe OW. Luminal Na(+)/H (+) exchange in the proximal tubule. Pflugers Arch 458: 5‐21, 2009.
 62. Boettger T, Hubner CA, Maier H, Rust MB, Beck FX, Jentsch TJ. Deafness and renal tubular acidosis in mice lacking the K‐Cl co‐transporter Kcc4. Nature 416: 874‐878, 2002.
 63. Boim MA, Ho K, Shuck ME, Bienkowski MJ, Block JH, Slightom JL, Yang Y, Brenner BM, Hebert SC. ROMK inwardly rectifying ATP‐sensitive K+ channel. II. Cloning and distribution of alternative forms. Am J Physiol 268: F1132‐F1140, 1995.
 64. Bonnici B, Wagner CA. Postnatal expression of transport proteins involved in acid‐base transport in mouse kidney. Pflugers Arch 448: 16‐28, 2004.
 65. Borke JL, Caride A, Verma AK, Penniston JT, Kumar R. Plasma membrane calcium pump and 28‐kDa calcium binding protein in cells of rat kidney distal tubules. Am J Physiol 257: F842‐F849, 1989.
 66. Boros S, Bindels RJ, Hoenderop JG. Active Ca(2+) reabsorption in the connecting tubule. Pflugers Arch 458: 99‐109, 2009.
 67. Bostanjoglo M, Reeves WB, Reilly RF, Velazquez H, Robertson N, Litwack G, Morsing P, Dorup J, Bachmann S, Ellison DH. 11Beta‐hydroxysteroid dehydrogenase, mineralocorticoid receptor, and thiazide‐sensitive Na‐Cl cotransporter expression by distal tubules. J Am Soc Nephrol 9: 1347‐1358, 1998.
 68. Bouby N, Bankir L. Effect of high protein intake on sodium, potassium‐dependent adenosine triphosphatase activity in the thick ascending limb of Henle's loop in the rat. Clin Sci (Lond) 74: 319‐329, 1988.
 69. Bouby N, Trinh‐Trang‐Tan MM, Kriz W, Bankir L. Possible role of the thick ascending limb and of the urine concentrating mechanism in the protein‐induced increase in GFR and kidney mass. Kidney Int Suppl 22: S57‐S61, 1987.
 70. Bourdeau JE, Chen ERY, Carone FA. Insulin uptake in the renal proximal tubule. Am J Physiol 225: 1399‐1404, 1973.
 71. Bourgeois S, Meer LV, Wootla B, Bloch‐Faure M, Chambrey R, Shull GE, Gawenis LR, Houillier P. NHE4 is critical for the renal handling of ammonia in rodents. J Clin Invest 120: 1895‐1904, 2010.
 72. Breton S, Lisanti MP, Tyszkowski R, McLaughlin M, Brown 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.
 73. Breton S, Wiederhold T, Marshansky V, Nsumu NN, Ramesh V, Brown D. The B1 subunit of the H+ATPase is a PDZ domain‐binding protein. Colocalization with NHE‐RF in renal B‐intercalated cells. J Biol Chem 275: 18219‐18224, 2000.
 74. Breyer MD, Kokko JP, Jacobson HR. Regulation of net bicarbonate transport in rabbit cortical collecting tubule by peritubular pH, carbon dioxide tension, and bicarbonate concentration. J Clin Invest 77: 1650‐1660, 1986.
 75. Briggs JP, Schnermann JB. Whys and wherefores of juxtaglomerular apparatus function. Kidney Int 49: 1724‐1726, 1996.
 76. Brooks HL, Sorensen AM, Terris J, Schultheis PJ, Lorenz JN, Shull GE, Knepper MA. Profiling of renal tubule Na+ transporter abundances in NHE3 and NCC null mice using targeted proteomics. J Physiol 530: 359‐366, 2001.
 77. Brown AC, Hallouane D, Mawby WJ, Karet FE, Saleem MA, Howie AJ, Toye AM. RhCG is the major putative ammonia transporter expressed in the human kidney, and RhBG is not expressed at detectable levels. Am J Physiol Renal Physiol 296: F1279‐F1290, 2009.
 78. Brown D. The ins and outs of aquaporin‐2 trafficking. Am J Physiol Renal Physiol 284: F893‐F901, 2003.
 79. Brown D, Gluck S, Hartwig J. Structure of the novel membrane‐coating material in proton‐secreting epithelial cells and identification as an H+ATPase. J Cell Biol 105: 1637‐1648, 1987.
 80. Brown D, Hirsch S, Bluck S. An H+‐ATPase in opposite plasma membrane domains in kidney epithelial cell subpopulations. Nature 331: 622‐624, 1988.
 81. Brown D, Hirsch S, Gluck S. Localization of a proton‐pumping ATPase in rat kidney. J Clin Invest 82: 2114‐2126, 1988.
 82. Brown D, Kumpulainen T. Immunocytochemical localization of carbonic anhydrase on ultrathin frozen sections with protein A‐gold. Histochemistry 83: 153‐158, 1985.
 83. Brown D, Kumpulainen T, Roth J, Orci L. Immunohistochemical localization of carbonic anhydrase in postnatal and adult rat kidney. Am J Physiol 245: F110‐F118, 1983.
 84. Brown D, Orci L. Junctional complexes and cell polarity in the urinary tubule. J Electron Microsc Tech 9: 145‐170, 1988.
 85. Brown D, Roth J, Kumpulainen T, Orci L. Ultrastructural immunocytochemical localization of carbonic anhydrase. Presence in intercalated cells of the rat collecting tubule. Histochemistry 75: 209‐213, 1982.
 86. Brown D, Roth J, Orci L. Lectin‐gold cytochemistry reveals intercalated cell heterogeneity along rat kidney collecting ducts. Am J Physiol 248: C348‐C356, 1985.
 87. Brown D, Weyer P, Orci L. Nonclathrin‐coated vesicles are involved in endocytosis in kidney collecting duct intercalated cells. Anat Rec 218: 237‐242, 1987.
 88. Brown RW, Diaz R, Robson AC, Kotelevtsev YV, Mullins JJ, Kaufman MH, Seckl JR. The ontogeny of 11 beta‐hydroxysteroid dehydrogenase type 2 and mineralocorticoid receptor gene expression reveal intricate control of glucocorticoid action in development. Endocrinology 137: 794‐797, 1996.
 89. Bruce LJ, Cope DL, Jones GK, Schofield AE, Burley M, Povey S, Unwin RJ, Wrong O, Tanner MJ. Familial distal renal tubular acidosis is associated with mutations in the red cell anion exchanger (Band 3, AE1) gene. J Clin Invest 100: 1693‐1707, 1997.
 90. Bulger RE, Cronin RE, Dobyan DC. Survey of the morphology of the dog kidney. Anat Rec 194: 41‐65, 1979.
 91. Bulger RE, Dobyan DC. Recent advances in renal morphology. Annu Rev Physiol 44: 147‐179, 1982.
 92. Bulger RE, Dobyan DC. Recent structure‐function relationships in normal and injured mammalian kidneys. Anat Rec 205: 1‐ll, 1983.
 93. Bulger RE, Siegel FL, Pendergrass R. Scanning and transmission electron microscopy of the rat kidney. Am J Anat 139: 483‐501, 1974.
 94. Bulger RE, Tisher CC, Myers CH, Trump BF. Human renal ultrastructure. II. The thin limb of Henle's loop and the interstitium in healthy individuals. Lab Invest 16: 124‐141, 1967.
 95. Burckhardt G, Wolff NA, Bahn A. Molecular characterization of the renal organic anion transporter 1. Cell Biochem Biophys 36: 169‐174, 2002.
 96. Butlen D, Vadrot S, Roseau S, Morel F. Insulin receptors along the rat nephron: 125I insulin binding in microdissected glomeruli and tubules. Pflügers Arch 412: 604‐612, 1988.
 97. Butterworth MB, Edinger RS, Frizzell RA, Johnson JP. Regulation of the epithelial sodium channel by membrane trafficking. Am J Physiol Renal Physiol 296: F10‐F24, 2009.
 98. Calvet JP. New insights into ciliary function: Kidney cysts and photoreceptors. Proc Natl Acad Sci U S A 100: 5583‐5585, 2003.
 99. Campean V, Kricke J, Ellison D, Luft FC, Bachmann S. Localization of thiazide‐sensitive Na(+)‐Cl(‐) cotransport and associated gene products in mouse DCT. Am J Physiol Renal Physiol 281: F1028‐F1035, 2001.
 100. Capasso G, Rizzo M, Pica A, Di Maio FS, Moe OW, Alpern RJ, De Santo NG. Bicarbonate reabsorption and NHE‐3 expression: Abundance and activity are increased in Henle's loop of remnant rats. Kidney Int 62: 2126‐2135, 2002.
 101. Capasso G, Unwin R, Rizzo M, Pica A, Giebisch G. Bicarbonate transport along the loop of Henle: Molecular mechanisms and regulation. J Nephrol 15 Suppl 5: S88‐S96, 2002.
 102. Carone FA, Christensen EI, Flouret G. Degradation and transport of AVP by proximal tubule. Am J Physiol 253: F1120‐F1128, 1987.
 103. Carone FA, Peterson DR. Hydrolysis and transport of small peptides by the proximal tubule. Am J Physiol 238: F151‐F158, 1980.
 104. Carone FA, Pullmann TN, Oparil S, Nakamura S. Micropuncture evidence of rapid hydrolysis of bradykinin by rat proximal tubule. Am J Physiol 230: 1420‐1424, 1976.
 105. Cha SK, Huang CL. WNK4 kinase stimulates caveola‐mediated endocytosis of TRPV5 amplifying the dynamic range of regulation of the channel by protein kinase C. J Biol Chem 285: 6604‐6611, 2010.
 106. Chambrey R, Goossens D, Bourgeois S, Picard N, Bloch‐Faure M, Leviel F, Geoffroy V, Cambillau M, Colin Y, Paillard M, Houillier P, Cartron JP, Eladari D. Genetic ablation of Rhbg in the mouse does not impair renal ammonium excretion. Am J Physiol Renal Physiol 289: F1281‐F1290, 2005.
 107. Cheval L, Barlet‐Bas C, Khadouri C, Feraille E, Marsy S, Doucet A. K(+)‐ATPase‐mediated Rb +transport in rat collecting tubule: Modulation during K+ deprivation. Am J Physiol 260: F800‐F805, 1991.
 108. Choate KA, Kahle KT, Wilson FH, Nelson‐Williams C, Lifton RP. WNK1, a kinase mutated in inherited hypertension with hyperkalemia, localizes to diverse Cl‐ ‐transporting epithelia. Proc Natl Acad Sci U S A 100: 663‐668, 2003.
 109. Christensen EI. Rapid protein uptake and digestion in proximal tubule lysosomes. Kidney Int 10: 301‐310, 1976.
 110. Christensen EI. Rapid membrane recycling in renal proximal tubule cells. Eur J Cell Biol 29: 43‐49, 1982.
 111. Christensen EI, Birn H. Megalin and cubilin: Multifunctional endocytic receptors. Nat Rev Mol Cell Biol 3: 256‐266, 2002.
 112. Christensen EI, Birn H, Rippe B, Maunsbach AB. Controversies in nephrology: Renal albumin handling, facts, and artifacts! Kidney Int 72: 1192‐1194, 2007.
 113. Christensen EI, Birn H, Verroust P, Moestrup SK. Membrane receptors for endocytosis in the renal proximal tubule. Int Rev Cytol 180: 237‐284, 1998.
 114. Christensen EI, Carone FA, Rennke HG. Effect of molecular charge on endocytic uptake of ferritin in renal proximal tubule cells. Lab Invest 44: 351‐358, 1981.
 115. Christensen EI, Devuyst O, Dom G, Nielsen R, Van Der Smissen P, Verroust P, Leruth M, Guggino WB, Courtoy PJ. Loss of chloride channel ClC‐5 impairs endocytosis by defective trafficking of megalin and cubilin in kidney proximal tubules. Proc Natl Acad Sci U S A 100: 8472‐8477, 2003.
 116. Christensen EI, Madsen KM. Renal age changes: Observations on the rat kidney cortex with special reference to structure and function of the lysosomal system in the proximal tubule. Lab Invest 39: 289‐297, 1978.
 117. Christensen EI, Maunsbach AB. Intralysosomal digestion of lysozyme in renal proximal tubule cells. Kidney Int 6: 396‐407, 1974.
 118. Christensen EI, Maunsbach AB. Effects of dextran on lysosomal ultrastructure and protein digestion in renal proximal tubule. Kidney Int 16: 301‐311, 1979.
 119. Christensen EI, Maunsbach AB. Proteinuria induced by sodium maleate in rats. Effects on ultrastructure and protein handling in renal proximal tubule. Kidney Int 17: 771‐787, 1980.
 120. Christensen EI, Maunsbach AB. Dextran is resistent to lysosomal digestion in kidney tubules. Virchows Arch B Cell Pathol Incl Mol Pathol 37: 49‐59, 1981.
 121. Christensen EI, Maunsbach AB, Lüllmann‐Rauch R. Renal lysosomal protein digestion in experimental lipidosis. Virchows Arch B Cell Pathol Incl Mol Pathol 43: 309‐316, 1983.
 122. Christensen EI, Moskaug JO, Vorum H, Jacobsen C, Gundersen TE, Nykjær A, Blomhoff R, Willnow TE, Moestrup SK. Evidence for an essential role of megalin in transepithelial transport of retinol. J Am Soc Nephrol 10: 685‐695, 1999.
 123. Christensen EI, Verroust PJ, Nielsen R. Receptor‐mediated endocytosis in renal proximal tubule. Pflugers Arch 458: 1039‐1048, 2009.
 124. Clapp WL, Madsen KM, Verlander JW, Tisher CC. Intercalated cells of the rat inner medullary collecting duct. Kidney Int 31: 1080‐1087, 1987.
 125. Clapp WL, Park CH, Madsen KM, Tisher CC. Axial heterogeneity in the handling of albumin by the rabbit proximal tubule. Lab Invest 58: 549‐558, 1988.
 126. Clarke H, Marano CW, Peralta SA, Mullin JM. Modification of tight junction function by protein kinase C isoforms. Adv Drug Deliv Rev 41: 283‐301, 2000.
 127. Claude P, Goodenough DA. Fracture faces of zonulae occludentes from “tight” and “leaky” epithelia. J Cell Biol 58: 390‐400, 1973.
 128. Codina J, Delmas‐Mata JT, DuBose TD Jr. Expression of HKalpha2 protein is increased selectively in renal medulla by chronic hypokalemia. Am J Physiol 275: F433‐F440, 1998.
 129. Colegio OR, Van Itallie CM, McCrea HJ, Rahner C, Anderson JM. Claudins create charge‐selective channels in the paracellular pathway between epithelial cells. Am J Physiol Cell Physiol 283: C142‐C147, 2002.
 130. Colegio OR, Van IC, Rahner C, Anderson JM. Claudin extracellular domains determine paracellular charge selectivity and resistance but not tight junction fibril architecture. Am J Physiol Cell Physiol 284: C1346‐C1354, 2003.
 131. Coleman RA, Wu DC, Liu J, Wade JB. Expression of aquaporins in the renal connecting tubule. Am J Physiol Renal Physiol 279: F874‐F883, 2000.
 132. Comper WD, Haraldsson B, Deen WM. Resolved: Normal glomeruli filter nephrotic levels of albumin. J Am Soc Nephrol 19: 427‐432, 2008.
 133. Comper WD, Hilliard LM, Nikolic‐Paterson DJ, Russo LM. Disease‐dependent mechanisms of albuminuria. Am J Physiol Renal Physiol 295: F1589‐F1600, 2008.
 134. Costanzo LS, Windhager EE. Calcium and sodium transport by the distal convoluted tubule of the rat. Am J Physiol 235: F492‐F506, 1978.
 135. Coudroy G, Gburek J, Kozyraki R, Madsen M, Trugnan G, Moestrup SK, Verroust PJ, Maurice M. Contribution of cubilin and amnionless to processing and membrane targeting of cubilin‐amnionless complex. J Am Soc Nephrol 16: 2330‐2337, 2005.
 136. Crayen ML, Thoenes W. Architecture and cell structures in the distal nephron of the rat kidney. Cytobiologie 17: 197‐211, 1978.
 137. Cruz DN, Simon DB, Nelson‐Williams C, Farhi A, Finberg K, Burleson L, Gill JR, Lifton RP. Mutations in the Na‐Cl cotransporter reduce blood pressure in humans. Hypertension 37: 1458‐1464, 2001.
 138. Cui S, Christensen EI. Three‐dimensional organization of the vacuolar apparatus involved in endocytosis and membrane recycling of rat kidney proximal tubule cells. An electron‐microscopic study of serial sections. Exp Nephrol 1: 175‐184, 1993.
 139. Daniel H, Rubio‐Aliaga I. An update on renal peptide transporters. Am J Physiol Renal Physiol 284: F885‐F892, 2003.
 140. Davenport JR, Yoder BK. An incredible decade for the primary cilium: A look at a once‐forgotten organelle. Am J Physiol Renal Physiol 289: F1159‐F1169, 2005.
 141. Davidson SJ. Protein absorption by renal cells. II. Very rapid lysosomal digestion of exogenous ribonuclease in vitro. J Cell Biol 59: 213‐222, 1973.
 142. Dawson TP, Gandhi R, LeHir M, Kaissling B. Ecto‐5’‐nucleotidase: Localization in rat kidney by light microscopic histochemical and immunohistochemical methods. J Histochem Cytochem 37: 39‐47, 1989.
 143. de Borst MH. On the origin of albuminuria. Kidney Int 72: 1409‐1410, 2007.
 144. de Duve C, Wattiaux R. Functions of lysosomes. Annu Rev Physiol 28: 435‐492, 1966.
 145. de Groot T, Bindels RJ, Hoenderop JG. TRPV5: An ingeniously controlled calcium channel. Kidney Int 74: 1241‐1246, 2008.
 146. De Jong JC, Van Der Vliet WA, van den Heuvel LP, Willems PH, Knoers NV, Bindels RJ. Functional expression of mutations in the human NaCl cotransporter: Evidence for impaired routing mechanisms in Gitelman's syndrome. J Am Soc Nephrol 13: 1442‐1448, 2002.
 147. Desjardins M, Bendayan M. Heterogenous distribution of type IV collagen, entactin, heparan sulfate proteoglycan, and laminin among renal basement membranes as demonstrated by quantitative immunocytochemistry. J Histochem Cytochem 37: 885‐897, 1989.
 148. Devuyst O, Christie PT, Courtoy PJ, Beauwens R, Thakker RV. Intra‐renal and subcellular distribution of the human chloride channel, CLC‐5, reveals a pathophysiological basis for Dent's disease. Hum Mol Genet 8: 247‐257, 1999.
 149. Di Sole F, Cerull R, Babich V, Quinones H, Gisler SM, Biber J, Murer H, Burckhardt G, Helmle‐Kolb C, Moe OW. Acute regulation of Na/H exchanger NHE3 by adenosine A(1) receptors is mediated by calcineurin homologous protein. J Biol Chem 279: 2962‐2974, 2004.
 150. Dieterich HJ, Barrett JM, Kriz W, Bulhoff JP. The ultrastructure of the thin loop limbs of the mouse kidney. Anat Embryol (Berl) 147: 1‐18, 1975.
 151. Dietert SC. The columnar cells occurring in the parietal layer of Bowman's capsule. Cellular fine structure and protein transport. J Cell Biol 35: 435‐444, 1967.
 152. Dijkink L, Hartog A, Deen PM, van Os CH, Bindels RJ. Time‐dependent regulation by aldosterone of the amiloride‐sensitive Na+ channel in rabbit kidney. Pflugers Arch 438: 354‐360, 1999.
 153. Dobyan DC, Bulger RE. Renal carbonic anhydrase. Am J Physiol 243: F311‐F324, 1982.
 154. Dobyan DC, Bulger RE. Morphology of the minipig kidney. J Electron microscopy technique 9: 213‐234, 1988.
 155. Dobyan DC, Magill LS, Friedman PA, Hebert SC, Bulger RE. Carbonic anhydrase histochemistry in rabbit and mouse kidneys. Anat Rec 204: 185‐197, 1982.
 156. Docherty NG, Calvo IF, Quinlan MR, Perez‐Barriocanal F, McGuire BB, Fitzpatrick JM, Watson RW. Increased E‐cadherin expression in the ligated kidney following unilateral ureteric obstruction. Kidney Int 75: 205‐213, 2009.
 157. Dorup J. Ultrastructure of distal nephron cells in rat renal cortex. J Ultrastruct Res 92: 101‐118, 1985.
 158. 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.
 159. Dorup J, Maunsbach AB. Three‐dimensional organization and segmental ultrastructure of rat proximal tubules. Exp Nephrol 5: 305‐317, 1997.
 160. Doucet A. H+, K(+)‐ATPASE in the kidney: Localization and function in the nephron. Exp Nephrol 5: 271‐276, 1997.
 161. Doucet A, Katz AI, Morel F. Determination of Na‐K‐ATPase activity in single segments of the mammalian nephron. Am J Physiol 237: F105‐F113, 1979.
 162. Doucet A, Marsy S. Characterization of K‐ATPase activity in distal nephron: Stimulation by potassium depletion. Am J Physiol 253: F418‐F423, 1987.
 163. Drenckhahn D, Schluter K, Allen DP, Bennett V. Colocalization of band 3 with ankyrin and spectrin at the basal membrane of intercalated cells in the rat kidney. Science 230: 1287‐1289, 1985.
 164. DuBose TD Jr, Codina J, Burges A, Pressley TA. Regulation of H(+)‐K(+)‐ATPase expression in kidney. Am J Physiol 269: F500‐F507, 1995.
 165. Duong Van Huyen JP, Cheval L, Bloch‐Faure M, Belair MF, Heudes D, Bruneval P, Doucet A. GDF15 triggers homeostatic proliferation of acid‐secreting collecting duct cells. J Am Soc Nephrol 19: 1965‐1974, 2008.
 166. Ecelbarger CA, Kim GH, Wade JB, Knepper MA. Regulation of the abundance of renal sodium transporters and channels by vasopressin. Exp Neurol 171: 227‐234, 2001.
 167. Ecelbarger CA, Knepper MA, Verbalis JG. Increased abundance of distal sodium transporters in rat kidney during vasopressin escape. J Am Soc Nephrol 12: 207‐217, 2001.
 168. Ecelbarger CA, Terris J, Hoyer JR, Nielsen S, Wade JB, Knepper MA. Localization and regulation of the rat renal Na(+)‐K(+)‐2Cl‐ cotransporter, BSC‐1. Am J Physiol 271: F619‐F628, 1996.
 169. Eladari D, Cheval L, Quentin F, Bertrand O, Mouro I, Cherif‐Zahar B, Cartron JP, Paillard M, Doucet A, Chambrey R. Expression of RhCG, a new putative NH(3)/NH(4)(+) transporter, along the rat nephron. J Am Soc Nephrol 13: 1999‐2008, 2002.
 170. Elling F, Hasselager E, Friis C. Perfusion fixation of kidneys in adult pigs for electron microscopy. Acta Anat (Basel) 98: 340‐342, 1977.
 171. Ellison DH. Divalent cation transport by the distal nephron: Insights from Bartter's and Gitelman's syndromes. Am J Physiol Renal Physiol 279: F616‐F625, 2000.
 172. Ellison DH. The thiazide‐sensitive na‐cl cotransporter and human disease: Reemergence of an old player. J Am Soc Nephrol 14: 538‐540, 2003.
 173. Ellison DH, Velazquez H, Wright FS. Thiazide‐sensitive sodium chloride cotransport in early distal tubule. Am J Physiol 253: F546‐F554, 1987.
 174. ElMoghrabi S, Houillier P, Picard N, Sohet F, Wootla B, Bloch‐Faure M, Leviel F, Cheval L, Frische S, Meneton P, Eladari D, Chambrey R. Tissue kallikrein permits early renal adaptation to potassium load. Proc Natl Acad Sci U S A 107: 13526‐13531, 2010.
 175. Enck AH, Berger UV, Yu AS. Claudin‐2 is selectively expressed in proximal nephron in mouse kidney. Am J Physiol Renal Physiol 281: F966‐F974, 2001.
 176. Ericsson JL, Trump BF. Electron microscopic studies of the epithelium of the proximal tubule of the rat kidney. I. The intracellular localization of acid phosphatase. Lab Invest 13: 1427‐1456, 1964.
 177. Ericsson JLE. Absorption and decomposition of homologous hemoglobin in renal proximal tubular cells. An experimental light and electron microscopic study. Acta Pathologica et Microbiologica Scandinavica 168: 1‐120, 1964.
 178. Ericsson JLE, Bergstrand A, Andres G, Bucht H, Cinotti G. Morphology of the renal tubular epithelium in young, healthy humans. Acta Pathologica et Microbiologica Scandinavica 63: 361‐384, 1965.
 179. Ericsson JLE, Trump BF, Weibel J. Electron microscopic studies of the proximal tubule of the rat kidney. II. Cytosegresomes and cytosomes: Their relationship to each other and to the lysosome concept. Lab Invest 14: 1341‐1365, 1965.
 180. Ernst SA, Schreiber JH. Ultrastructural localization of Na+,K+‐ATPase in rat and rabbit kidney medulla. J Cell Biol 91: 803‐813, 1981.
 181. Estevez R, Boettger T, Stein V, Birkenhager R, Otto E, Hildebrandt F, Jentsch TJ. Barttin is a Cl‐ channel beta‐subunit crucial for renal Cl‐ reabsorption and inner ear K+ secretion. Nature 414: 558‐561, 2001.
 182. Evan AP, Gattone VH II, Schwartz GJ. Development of solute transport in rabbit proximal tubule. II. Morphologic segmentation. Am J Physiol 245: F391‐F407, 1983.
 183. Evan AP, Gattone VH, Blomgren PM. Application of scanning electron microscopy to kidney development and nephron maturation. Scan Electron Microsc Pt 1: 455‐473, 1984.
 184. Farquhar MG, Palade GE. Junctional complexes in various epithelia. J Cell Biol 17: 375‐412, 1963.
 185. Fejes‐Toth G, Naray‐Fejes‐Toth A. Immunohistochemical localization of colonic H‐K‐ATPase to the apical membrane of connecting tubule cells. Am J Physiol Renal Physiol 281: F318‐F325, 2001.
 186. Fenton RA, Knepper MA. Mouse models and the urinary concentrating mechanism in the new millennium. Physiol Rev 87: 1083‐1112, 2007.
 187. Fenton RA, Stewart GS, Carpenter B, Howorth A, Potter EA, Cooper GJ, Smith CP. Characterization of mouse urea transporters UT‐A1 and UT‐A2. Am J Physiol Renal Physiol 283: F817‐F825, 2002.
 188. Finberg KE, Wagner CA, Stehberger PA, Geibel JP, Lifton RP. Molecular cloning and characterization of Atp6v1b1, the murine vacuolar H+ ‐ATPase B1‐subunit. Gene 318: 25‐34, 2003.
 189. Findley MK, Koval M. Regulation and roles for claudin‐family tight junction proteins. IUBMB Life 61: 431‐437, 2009.
 190. Fogelgren B, Yang S, Sharp IC, Huckstep OJ, Ma W, Somponpun SJ, Carlson EC, Uyehara CF, Lozanoff S. Deficiency in Six2 during prenatal development is associated with reduced nephron number, chronic renal failure, and hypertension in Br/+ adult mice. Am J Physiol Renal Physiol 296: F1166‐F1178, 2009.
 191. Forgac M. Vacuolar ATPases: Totary proton pumps in physiology and pathophysiology. Nat Rev Mol Cell Biol 8: 917‐929, 2007.
 192. Forssmann WG. A method for in vivo diffusion tracer studies combining perfusion fixation with intravenous tracer injection. Histochemie 20: 277‐286, 1969.
 193. Forster IC, Hernando N, Biber J, Murer H. Proximal tubular handling of phosphate: A molecular perspective. Kidney Int 70: 1548‐1559, 2006.
 194. Friedman PA. Mechanisms of renal calcium transport. Exp Nephrol 8: 343‐350, 2000.
 195. Frigeri A, Gropper MA, Turck CW, Verkman AS. Immunolocalization of the mercurial‐insensitive water channel and glycerol intrinsic protein in epithelial cell plasma membranes. Proc Natl Acad Sci U S A 92: 4328‐4331, 1995.
 196. Frindt G, Palmer LG. Surface expression of sodium channels and transporters in rat kidney: Effects of dietary sodium. Am J Physiol Renal Physiol 297: F1249‐F1255, 2009.
 197. Fyfe JC, Madsen M, Hojrup P, Christensen EI, Tanner SM, de la Chapelle A, He Q, Moestrup SK. The functional cobalamin (vitamin B12)‐intrinsic factor receptor is a novel complex of cubilin and amnionless. Blood 103: 1573‐1579, 2004.
 198. Gamba G. Role of WNK kinases in regulating tubular salt and potassium transport and in the development of hypertension. Am J Physiol Renal Physiol 288: F245‐F252, 2005.
 199. Gamba G. The thiazide‐sensitive Na+‐Cl‐ cotransporter: Molecular biology, functional properties, and regulation by WNKs. Am J Physiol Renal Physiol 297: F838‐F848, 2009.
 200. Ganote CE, Grantham JJ, Moses HL, Burg MB, Orloff J. Ultrastructural studies of vasopressin effect on isolated perfused renal collecting tubules of the rabbit. J Cell Biol 36: 355‐367, 1968.
 201. Garg LC, Knepper MA, Burg MB. Mineralocorticoid effects on Na‐K‐ATPase in individual nephron segments. Am J Physiol 240: F536‐F544, 1981.
 202. Garg LC, Narang N. Suppression of ouabain‐insensitive K‐ATPase activity in rabbit nephron segments during chronic hyperkalemia. Ren Physiol Biochem 12: 295‐301, 1989.
 203. Ge Y, Ahn D, Stricklett PK, Hughes AK, Yanagisawa M, Verbalis JG, Kohan DE. Collecting duct‐specific knockout of endothelin‐1 alters vasopressin regulation of urine osmolality. Am J Physiol Renal Physiol 288: F912‐F920, 2005.
 204. Geering K. FXYD proteins: New regulators of Na‐K‐ATPase. Am J Physiol Renal Physiol 290: F241‐F250, 2006.
 205. Gekle M. Renal albumin handling: A look at the dark side of the filter. Kidney Int 71: 479‐481, 2007.
 206. Gekle M, Drumm K, Mildenberger S, Freudinger R, Gassner B, Silbernagl S. Inhibition of Na+‐H+ exchange impairs receptor‐mediated albumin endocytosis in renal proximal tubule‐derived epithelial cells from opossum. J Physiol 520: 709‐721, 1999.
 207. Gekle M, Serrano OK, Drumm K, Mildenberger S, Freudinger R, Gassner B, Jansen HW, Christensen EI. NHE3 serves as a molecular tool for cAMP‐mediated regulation of receptor‐mediated endocytosis. Am J Physiol Renal Physiol 283: F549‐F558, 2002.
 208. Gekle M, Volker K, Mildenberger S, Freudinger R, Shull GE, Wiemann M. NHE3 Na+/H+ exchanger supports proximal tubular protein reabsorption in vivo. Am J Physiol Renal Physiol 287: F469‐F473, 2004.
 209. Giebisch G. Renal potassium channels: Function, regulation, and structure. Kidney Int 60: 436‐445, 2001.
 210. Giebisch G, Aronson PS. The proximal nephron. In: Andreoli TE, Hoffmann JF, Fanestil DD, Schultz SG, editors. Physiology of Membrane Disorders. New York: Plenum, 1986, p. 669‐700.
 211. Gisler SM, Pribanic S, Bacic D, Forrer P, Gantenbein A, Sabourin LA, Tsuji A, Zhao ZS, Manser E, Biber J, Murer H. PDZK1: I. A major scaffolder in brush borders of proximal tubular cells. Kidney Int 64: 1733‐1745, 2003.
 212. Giurgea‐Marion L, Toubeau G, Laurent G, Heuson‐Stiennon JA, Tulkens PM. Impairment of lysosome‐pinocytic vesicle fusion in rat kidney proximal tubules after treatment with gentamicin at low doses. Toxicol Appl Pharmacol 86: 271‐285, 1986.
 213. Gkika D, Topala CN, Chang Q, Picard N, Thebault S, Houillier P, Hoenderop JG, Bindels RJ. Tissue kallikrein stimulates Ca(2+) reabsorption via PKC‐dependent plasma membrane accumulation of TRPV5. EMBO J 25: 4707‐4716, 2006.
 214. Glaudemans B, Knoers NV, Hoenderop JG, Bindels RJ. New molecular players facilitating Mg(2+) reabsorption in the distal convoluted tubule. Kidney Int 77: 17‐22, 2010.
 215. Gluck S, Cannon C, Al‐Awqati Q. Exocytosis regulates urinary acidification in turtle bladder by rapid insertion of H+ pumps into the luminal membrane. Proc Natl Acad Sci U S A 79: 4327‐4331, 1982.
 216. Golbang AP, Cope G, Hamad A, Murthy M, Liu CH, Cuthbert AW, O'shaughnessy KM. Regulation of the expression of the Na/Cl cotransporter by WNK4 and WNK1: Evidence that accelerated dynamin‐dependent endocytosis is not involved. Am J Physiol Renal Physiol 291: F1369‐F1376, 2006.
 217. Goligorsky M, Hruska KA. Transcytosis in cultured proximal tubular cells. J Membr Biol 93: 237‐247, 1986.
 218. Gonzalez CB, Figueroa CD, Reyes CE, Caorsi CE, Troncoso S, Menzel D. Immunolocalization of V1 vasopressin receptors in the rat kidney using anti‐receptor antibodies. Kidney Int 52: 1206‐1215, 1997.
 219. Gottschalk CW, MYLLE M. Micropuncture study of the mammalian urinary concentrating mechanism: Evidence for the countercurrent hypothesis. Am J Physiol 196: 927‐936, 1959.
 220. Grantham JJ. Mode of water transport in mammalian renal collecting tubules. Fed Proc 30: 14‐21, 1971.
 221. Greger R, Schlatter E, Lang F. Evidence for electroneutral sodium chloride cotransport in the cortical thick ascending limb of Henle's loop of rabbit kidney. Pflugers Arch 396: 308‐314, 1983.
 222. Grimm PR, Foutz RM, Brenner R, Sansom SC. Identification and localization of BK‐beta subunits in the distal nephron of the mouse kidney. Am J Physiol Renal Physiol 293: F350‐F359, 2007.
 223. Guder WG, Hallbach J, Fink E, Kaissling B, Wirthensohn G. Kallikrein (kininogenase) in the mouse nephron: Effect of dietary potassium. Biol Chem Hoppe Seyler 368: 637‐645, 1987.
 224. Guder WG, Wagner S, Wirthensohn G. Metabolic fuels along the nephron: Pathways and intracellular mechanisms of interaction. Kidney Int 29: 41‐45, 1986.
 225. Gunther W, Luchow A, Cluzeaud F, Vandewalle A, Jentsch TJ. ClC‐5, the chloride channel mutated in Dent's disease, colocalizes with the proton pump in endocytotically active kidney cells. Proc Natl Acad Sci U S A 95: 8075‐8080, 1998.
 226. Gunzel D, Yu AS. Function and regulation of claudins in the thick ascending limb of Henle. Pflugers Arch 458: 77‐88, 2009.
 227. Guo R, Liu L, Barajas L. RT‐PCR study of the distribution of connexin 43 mRNA in the glomerulus and renal tubular segments. Am J Physiol 275: R439‐R447, 1998.
 228. Gurich RW, Warnock DG. Electrically neutral Na+‐H+ exchange in endosomes obtained from rabbit renal cortex. Am J Physiol 251: F702‐F709, 1986.
 229. Gustafson CE, Katsura T, McKee M, Bouley R, Casanova JE, Brown D. Recycling of AQP2 occurs through a temperature‐ and bafilomycin‐sensitive trans‐Golgi‐associated compartment. Am J Physiol Renal Physiol 278: F317‐F326, 2000.
 230. Haas M, Forbush B III. The Na‐K‐Cl cotransporters. J Bioenerg Biomembr 30: 161‐172, 1998.
 231. Hafner P, Grimaldi R, Capuano P, Capasso G, Wagner CA. Pendrin in the mouse kidney is primarily regulated by Cl‐ excretion but also by systemic metabolic acidosis. Am J Physiol Cell Physiol 295: C1658‐C1667, 2008.
 232. Haga HJ, Andersen KJ, Rygh T, Iversen BM, Matre R. Changes in lysosome populations in the rat kidney cortex induced by experimental proteinuria. Int J Biochem 20: 793‐800, 1988.
 233. Hagege J, Richet G. Dark cells of the distal convoluted tubules and collecting ducts. I. Morphological data. Fortschr Zool 23: 288‐298, 1975.
 234. Halperin ML, Kamel KS, Oh MS. Mechanisms to concentrate the urine: An opinion. Curr Opin Nephrol Hypertens 17: 416‐422, 2008.
 235. Hammerman MR. Interaction of insulin with the renal proximal tubular cell. Am J Physiol 249: F1‐F11, 1985.
 236. Hammerman MR, Gavin JR III. Insulin‐stimulated phosphorylation and insulin binding in canine renal basolateral membranes. Am J Physiol 247: F408‐F417, 1984.
 237. Han KH, Woo SK, Kim WY, Park SH, Cha JH, Kim J, Kwon HM. Maturation of TonEBP expression in developing rat kidney. Am J Physiol Renal Physiol 287: F878‐F885, 2004.
 238. Harris RC, McKanna JA, Akai Y, Jacobson HR, Dubois RN, Breyer MD. Cyclooxygenase‐2 is associated with the macula densa of rat kidney and increases with salt restriction. J Clin Invest 94: 2504‐2510, 1994.
 239. Harrison‐Bernard LM, Navar LG, Ho MM, Vinson GP, el‐Dahr SS. Immunohistochemical localization of ANG II AT1 receptor in adult rat kidney using a monoclonal antibody. Am J Physiol 273: F170‐F177, 1997.
 240. Hay DA, Evan AP. Maturation of the proximal tubule in the puppy kidney: A comparison to the adult. Anat Rec 195: 273‐299, 1979.
 241. Hays SR. Mineralocorticoid modulation of apical and basolateral membrane H+/OH‐/HCO3‐ transport processes in the rabbit inner stripe of outer medullary collecting duct. J Clin Invest 90: 180‐187, 1992.
 242. Hebert SC. Bartter syndrome. Curr Opin Nephrol Hypertens 12: 527‐532, 2003.
 243. Hebert SC, Andreoli TE. Control of NaCl transport in the thick ascending limb. Am J Physiol 246: F745‐F756, 1984.
 244. Hebert SC, Culpepper RM, Andreoli TE. NaCl transport in mouse medullary thick ascending limbs. I. Functional nephron heterogeneity and ADH‐stimulated NaCl cotransport. Am J Physiol 241: F412‐F431, 1981.
 245. Hediger MA, Rhoads DB. Molecular physiology of sodium‐glucose cotransporters. Physiol Rev 74: 993‐1026, 1994.
 246. Heiskala M, Peterson PA, Yang Y. The roles of claudin superfamily proteins in paracellular transport. Traffic 2: 93‐98, 2001.
 247. Hellfritzsch M, Christensen EI, Sonne O. Luminal uptake and intracellular transport of insulin in renal proximal tubules. Kidney Int 29: 983‐988, 1986.
 248. Hellfritzsch M, Nielsen S, Christensen EI, Nielsen JT. Basolateral tubular handling of insulin in the kidney. In: Bianchi C, Bocci V, Carone FA, Rabkin R, editors. Kidney and Proteins in Health and Disease. Basel: Karger, 1988, p. 86‐91.
 249. Hernando N, Wagner CA, Gisler SM, Biber J, Murer H. PDZ proteins and proximal ion transport. Curr Opin Nephrol Hypertens 13: 569‐574, 2004.
 250. Hjälm G, Murray E, Crumley G, Harazim W, Lundgren S, Onyango I, Ek B, Larsson M, Juhlin C, Hellman P, Davis H, Åkerström G, Rask L, Morse B. Cloning and sequencing of human gp330, a Ca(2+)‐binding receptor with potential intracellular signaling properties. Eur J Biochem 239: 132‐137, 1996.
 251. Hjelle JT, Oparil S, Peterson DR. Subcellular sites of insulin hydrolysis in renal proximal tubules. Am J Physiol 246: F409‐F416, 1984.
 252. Holthofer H, Schulte BA, Pasternack G, Siegel G, Spicer SS. Immunocytochemical characterization of carbonic anhydrase‐rich cells in the rat kidney collecting duct. Lab Invest 57: 150‐156, 1987.
 253. Holthofer H, Schulte BA, Pasternack G, Siegel GJ, Spicer SS. Three distinct cell populations in rat kidney collecting duct. Am J Physiol 253: C323‐C328, 1987.
 254. Holthofer H, Schulte BA, Spicer SS. Expression of binding sites for Dolichos biflorus agglutinin at the apical aspect of collecting duct cells in rat kidney. Cell Tissue Reseach 249: 481‐485, 1987.
 255. Hopkins AM, Li D, Mrsny RJ, Walsh SV, Nusrat A. Modulation of tight junction function by G protein‐coupled events. Adv Drug Deliv Rev 41: 329‐340, 2000.
 256. Hou J, Renigunta A, Gomes AS, Hou M, Paul DL, Waldegger S, Goodenough DA. Claudin‐16 and claudin‐19 interaction is required for their assembly into tight junctions and for renal reabsorption of magnesium. Proc Natl Acad Sci U S A 106: 15350‐15355, 2009.
 257. Hou J, Renigunta A, Konrad M, Gomes AS, Schneeberger EE, Paul DL, Waldegger S, Goodenough DA. Claudin‐16 and claudin‐19 interact and form a cation‐selective tight junction complex. J Clin Invest 118: 619‐628, 2008.
 258. Hou X, Mrug M, Yoder BK, Lefkowitz EJ, Kremmidiotis G, D'Eustachio P, Beier DR, Guay‐Woodford LM. Cystin, a novel cilia‐associated protein, is disrupted in the cpk mouse model of polycystic kidney disease. J Clin Invest 109: 533‐540, 2002.
 259. Hruban Z, Rechcigl M Jr. Microbodies and related particles. Morphology, biochemistry, and physiology. Int Rev Cytol Suppl 1: 1‐296, 1969.
 260. Huber S, Asan E, Jons T, Kerscher C, Puschel B, Drenckhahn D. Expression of rat kidney anion exchanger 1 in type A intercalated cells in metabolic acidosis and alkalosis. Am J Physiol 277: F841‐F849, 1999.
 261. Igarashi P, Somlo S. Genetics and pathogenesis of polycystic kidney disease. J Am Soc Nephrol 13: 2384‐2398, 2002.
 262. Imai M, Hayashi M, Araki M. Functional heterogeneity of the descending limbs of Henle's loop. I. Internephron heterogeneity in the hamster kidney. Pflugers Arch 402: 385‐392, 1984.
 263. Imbert‐Teboul M, Charbardes D, Morel F. Vasopressin and catecholamines sites of action along rabbit, mouse and rat nephron. Contrib Nephrol 21: 41‐47, 1980.
 264. Ishibashi K, Imai M, Sasaki S. Cellular localization of aquaporin 7 in the rat kidney. Exp Nephrol 8: 252‐257, 2000.
 265. Ishibashi K, Sasaki S, Fushimi K, Yamamoto T, Kuwahara M, Marumo F. Immunolocalization and effect of dehydration on AQP3, a basolateral water channel of kidney collecting ducts. Am J Physiol 272: F235‐F241, 1997.
 266. Jacobsen NO, Jørgensen F. Ultrastructural observations on the pars descendens of the proximal tubule in the kidney of the male rat. Z Zellforsch 136: 479‐499, 1973.
 267. Jacobson HR. Functional segmentation of the mammalian nephron. Am J Physiol 241: F203‐F218, 1981.
 268. Jaisser F, Escoubet B, Coutry N, Eugene E, Bonvalet JP, Farman N. Differential regulation of putative K(+)‐ATPase by low‐K+ diet and corticosteroids in rat distal colon and kidney. Am J Physiol 270: C679‐C687, 1996.
 269. Jamison RL, Kriz W. Proximal tubule. In: Jamison RL, Kriz W, editors. Urinary Concentrating Mechanism: Structure and Function. New York: Oxford University Press, 1982, p. 113‐132.
 270. Jeck N, Schlingmann KP, Reinalter SC, Komhoff M, Peters M, Waldegger S, Seyberth HW. Salt handling in the distal nephron: Lessons learned from inherited human disorders. Am J Physiol Regul Integr Comp Physiol 288: R782‐R795, 2005.
 271. Jedrzejewski K, Kugler P. Peptidases in the kidney and urine of rats after castration. Histochemistry 74: 63‐84, 1982.
 272. Jentsch TJ, Hubner CA, Fuhrmann JC. Ion channels: Function unravelled by dysfunction. Nat Cell Biol 6: 1039‐1047, 2004.
 273. Jones DB. Scanning electron microscopy of isolated dog renal tubules. Scan Electron Microsc Pt 2: 805‐813, 1982.
 274. Jouret F, Auzanneau C, Debaix H, Wada GH, Pretto C, Marbaix E, Karet FE, Courtoy PJ, Devuyst O. Ubiquitous and kidney‐specific subunits of vacuolar H+‐ATPase are differentially expressed during nephrogenesis. J Am Soc Nephrol 16: 3235‐3246, 2005.
 275. Jung JY, Song JH, Li C, Yang CW, Kang TC, Won MH, Jeong YG, Han KH, Choi KB, Lee SH, Kim J. Expression of epidermal growth factor in the developing rat kidney. Am J Physiol Renal Physiol 288: F227‐F235, 2005.
 276. Kahle KT, Gimenez I, Hassan H, Wilson FH, Wong RD, Forbush B, Aronson PS, Lifton RP. WNK4 regulates apical and basolateral Cl‐ flux in extrarenal epithelia. Proc Natl Acad Sci U S A 101: 2064‐2069, 2004.
 277. Kahle KT, Macgregor GG, Wilson FH, Van Hoek AN, Brown D, Ardito T, Kashgarian M, Giebisch G, Hebert SC, Boulpaep EL, Lifton RP. Paracellular Cl‐ permeability is regulated by WNK4 kinase: Insight into normal physiology and hypertension. Proc Natl Acad Sci U S A 101: 14877‐14882, 2004.
 278. Kahle KT, Wilson FH, Lalioti M, Toka H, Qin H, Lifton RP. WNK kinases: Molecular regulators of integrated epithelial ion transport. Curr Opin Nephrol Hypertens 13: 557‐562, 2004.
 279. Kaissling B. Ultrastructural organization of the transition from the distal nephron to the collecting duct in the desert rodent Psammomys obesus. Cell Tissue Res 212: 475‐495, 1980.
 280. Kaissling B. Structural aspects of adaptive changes in renal electrolyte excretion. Am J Physiol 243: F211‐F226, 1982.
 281. Kaissling B. Cellular heterogeneity of the distal nephron and its relation to function. Klin Wochenschr 63: 868‐876, 1985a.
 282. Kaissling B. Structural adaptation to altered electrolyte metabolism by cortical distal segments. Fed Proc 44: 2710‐2716, 1985b.
 283. Kaissling B, Bachmann S, Kriz W. Structural adaption of the distal convoluted tubule to prolonged furosemide treatment. Am J Physiol 248: F374‐F381, 1985.
 284. Kaissling B, de RC, Barrett JM, Kriz W. The structural organization of the kidney of the desert rodent Psammomys obesus. Anat Embryol (Berl) 148: 121‐143, 1975.
 285. Kaissling B, Hegyi I, Loffing J, LeHir M. Morphology of interstitial cells in the healthy kidney. Anat Embryol (Berl) 193: 303‐318, 1996.
 286. Kaissling B, Koeppen B, LeHir M, Wade JB. Effect of mineralocorticoids on the structure of intercalated cells in renal collecting ducts. Acta Anat 111: 72, 1981.
 287. Kaissling B, Kriz W. Structural analysis of the rabbit kidney. Adv Anat Embryol Cell Biol 56: 1‐123, 1979.
 288. Kaissling B, Kriz W. Axial heterogeneity of the ‘distal tubule’. Contrib Nephrol 33: 29‐47, 1982a.
 289. Kaissling B, Kriz W. Variability of intercellular spaces between macula densa cells: A transmission electron microscopic study in rabbits and rats. Kidney Int Suppl 12: S9‐S17, 1982b.
 290. Kaissling B, Kriz W. Morphology of the loop of Henle, distal tubule, and collecting duct. In: Windhager EE, editor. Handbook of Physiology. Renal Physiology. Am. Physiol. Soc. New York: Oxford University Press, 1992, sect. 8, p. 109‐167.
 291. Kaissling B, LeHir M. Distal tubular segments of the rabbit kidney after adaptation to altered Na‐ and K‐intake. I. Structural changes. Cell Tissue Res 224: 469‐492, 1982.
 292. Kaissling B, LeHir M. The renal cortical interstitium: Morphological and functional aspects. Histochem Cell Biol 130: 247‐262, 2008.
 293. Kaissling B, Loffing J. Cell growth and cell death in renal distal tubules, associated with diuretic treatment. Nephrol Dial Transplant 13: 1341‐1343, 1998.
 294. Kaissling B, Peter S, Kriz W. The transition of the thick ascending limb of Henle's loop into the distal convoluted tubule in the nephron of the rat kidney. Cell Tissue Res 182: 111‐118, 1977.
 295. Kaissling B, Stanton BA. Adaptation of distal tubule and collecting duct to increased sodium delivery. I. Ultrastructure. Am J Physiol 255: F1256‐F1268, 1988a.
 296. Kaissling B, Stanton BA. Chronic furosemide treatment alters the ultrastructure of intercalated cells in renal collecting ducts. Clin Res 36: 521, 1988b..
 297. Karet FE, Gainza FJ, Gyory AZ, Unwin RJ, Wrong O, Tanner MJ, Nayir A, Alpay H, Santos F, Hulton SA, Bakkaloglu A, Ozen S, Cunningham MJ, di PA, Walker WG, Lifton RP. Mutations in the chloride‐bicarbonate exchanger gene AE1 cause autosomal dominant but not autosomal recessive distal renal tubular acidosis. Proc Natl Acad Sci U S A 95: 6337‐6342, 1998.
 298. Kashgarian M, Biemesderfer D, Caplan M, Forbush B. Monoclonal antibody to Na,K‐ATPase: Immunocytochemical localization along nephron segments. Kidney Int 28: 899‐913, 1985.
 299. Kato Y, Sai Y, Yoshida K, Watanabe C, Hirata T, Tsuji A. PDZK1 directly regulates the function of organic cation/carnitine transporter OCTN2. Mol Pharmacol 67: 734‐743, 2005.
 300. Katz AI, Doucet A, Morel F. Na‐K‐ATPase activity along the rabbit, rat, and mouse nephron. Am J Physiol 237: F114‐F120, 1979.
 301. Kausalya PJ, Amasheh S, Gunzel D, Wurps H, Muller D, Fromm M, Hunziker W. Disease‐associated mutations affect intracellular traffic and paracellular Mg2+ transport function of Claudin‐16. J Clin Invest 116: 878‐891, 2006.
 302. Kempson SA, Lotscher M, Kaissling B, Biber J, Murer H, Levi M. Parathyroid hormone action on phosphate transporter mRNA and protein in rat renal proximal tubules. Am J Physiol 268: F784‐F791, 1995.
 303. Kerjaschki D, Farquhar MG. The pathogenic antigen of Heymann nephritis is a membrane glycoprotein of the renal proximal tubule brush border. Proc Natl Acad Sci U S A 79: 5557‐5561, 1982.
 304. Kerjaschki D, Noronha‐Blob L, Sacktor B, Farquhar MG. Microdomains of distinctive glycoprotein composition in the kidney proximal tubule brush border. J Cell Biol 98: 1505‐1513, 1984.
 305. Kerjaschki D, Sacktor B, Reiss U, Farquhar MG. Microdomains of distinctive glycoprotein composition in the kidney proximal tubule brush border. J Cell Biol 98: 178a, 1984.
 306. Keusch I, Traebert M, Lotscher M, Kaissling B, Murer H, Biber J. Parathyroid hormone and dietary phosphate provoke a lysosomal routing of the proximal tubular Na/Pi‐cotransporter type II. Kidney Int 54: 1224‐1232, 1998.
 307. Kim GH, Ecelbarger C, Knepper MA, Packer RK. Regulation of thick ascending limb ion transporter abundance in response to altered acid/base intake. J Am Soc Nephrol 10: 935‐942, 1999.
 308. Kim GH, Ecelbarger CA, Mitchell C, Packer RK, Wade JB, Knepper MA. Vasopressin increases Na‐K‐2Cl cotransporter expression in thick ascending limb of Henle's loop. Am J Physiol 276: F96‐F103, 1999.
 309. Kim HY, Verlander JW, Bishop JM, Cain BD, Han KH, Igarashi P, Lee HW, Handlogten ME, Weiner ID. Basolateral expression of the ammonia transporter family member Rh C glycoprotein in the mouse kidney. Am J Physiol Renal Physiol 296: F543‐F555, 2009.
 310. Kim J, Kim YH, Cha JH, Tisher CC, Madsen KM. Intercalated cell subtypes in connecting tubule and cortical collecting duct of rat and mouse. J Am Soc Nephrol 10: 1‐12, 1999.
 311. Kim YH, Kwon TH, Frische S, Kim J, Tisher CC, Madsen KM, Nielsen S. Immunocytochemical localization of pendrin in intercalated cell subtypes in rat and mouse kidney. Am J Physiol Renal Physiol 283: F744‐F754, 2002.
 312. Kirk A, Campbell S, Bass P, Mason J, Collins J. Differential expression of claudin tight junction proteins in the human cortical nephron. Nephrol Dial Transplant 25: 2107‐2119, 2010.
 313. Kirk KL, Bell PD, Barfuss DW, Ribadeneira M. Direct visualization of the isolated and perfused macula densa. Am J Physiol 248: F890‐F894, 1985.
 314. Kishore BK, Mandon B, Oza NB, DiGiovanni SR, Coleman RA, Ostrowski NL, Wade JB, Knepper MA. Rat renal arcade segment expresses vasopressin‐regulated water channel and vasopressin V2 receptor. J Clin Invest 97: 2763‐2771, 1996.
 315. Kiuchi‐Saishin Y, Gotoh S, Furuse M, Takasuga A, Tano Y, Tsukita S. Differential expression patterns of claudins, tight junction membrane proteins, in mouse nephron segments. J Am Soc Nephrol 13: 875‐886, 2002.
 316. Kleyman TR, Myerburg MM, Hughey RP. Regulation of ENaCs by proteases: An increasingly complex story. Kidney Int 70: 1391‐1392, 2006.
 317. Knepper MA, Kim GH, Fernandez‐Llama P, Ecelbarger CA. Regulation of thick ascending limb transport by vasopressin. J Am Soc Nephrol 10: 628‐634, 1999.
 318. Knepper MA, Roch‐Ramel F. Pathways of urea transport in the mammalian kidney. Kidney Int 31: 629‐633, 1987.
 319. Knepper MA, Wade JB, Terris J, Ecelbarger CA, Marples D, Mandon B, Chou CL, Kishore BK, Nielsen S. Renal aquaporins. Kidney Int 49: 1712‐1717, 1996.
 320. Knoers NV, De Jong JC, Meij IC, van den Heuvel LP, Bindels RJ. Genetic renal disorders with hypomagnesemia and hypocalciuria. J Nephrol 16: 293‐296, 2003.
 321. Knohl SJ, Scheinman SJ. Inherited hypercalciuric syndromes: Dent's disease (CLC‐5) and familial hypomagnesemia with hypercalciuria (paracellin‐1). Semin Nephrol 24: 55‐60, 2004.
 322. Ko B, Kamsteeg EJ, Cooke LL, Moddes LN, Deen PM, Hoover RS. RasGRP1 stimulation enhances ubiquitination and endocytosis of the sodium‐chloride cotransporter. Am J Physiol Renal Physiol 299: F300‐F309, 2010.
 323. Ko SB, Luo X, Hager H, Rojek A, Choi JY, Licht C, Suzuki M, Muallem S, Nielsen S, Ishibashi K. AE4 is a DIDS‐sensitive Cl(‐)/HCO(‐)(3) exchanger in the basolateral membrane of the renal CCD and the SMG duct. Am J Physiol Cell Physiol 283: C1206‐C1218, 2002.
 324. Koechlin N, Elalouf JM, Kaissling B, Roinel N, de RC. A structural study of the rat proximal and distal nephron: Effect of peptide and thyroid hormones. Am J Physiol 256: 814‐822, 1989.
 325. Komlosi P, Fintha A, Bell PD. Unraveling the relationship between macula densa cell volume and luminal solute concentration/osmolality. Kidney Int 70: 865‐871, 2006.
 326. Kone BC, Madsen KM, Tisher CC. Ultrastructure of the thick ascending limp of Henle in the rat kidney. Am J Anat 171: 217‐226, 1984.
 327. Konrad M, Schaller A, Seelow D, Pandey AV, Waldegger S, Lesslauer A, Vitzthum H, Suzuki Y, Luk JM, Becker C, Schlingmann KP, Schmid M, Rodriguez‐Soriano J, Ariceta G, Cano F, Enriquez R, Juppner H, Bakkaloglu SA, Hediger MA, Gallati S, Neuhauss SC, Nurnberg P, Weber S. Mutations in the tight‐junction gene claudin 19 (CLDN19) are associated with renal magnesium wasting, renal failure, and severe ocular involvement. Am J Hum Genet 79: 949‐957, 2006.
 328. Konrad M, Schlingmann KP, Gudermann T. Insights into the molecular nature of magnesium homeostasis. Am J Physiol Renal Physiol 286: F599‐F605, 2004.
 329. Konrad M, Weber S. Recent advances in molecular genetics of hereditary magnesium‐losing disorders. J Am Soc Nephrol 14: 249‐260, 2003.
 330. Koob R, Zimmermann M, Schoner W, Drenckhahn D. Colocalization and coprecipitation of ankyrin and Na+,K+‐ATPase in kidney epithelial cells. Eur J Cell Biol 45: 230‐237, 1988.
 331. Koval M. Claudins–key pieces in the tight junction puzzle. Cell Commun Adhes 13: 127‐138, 2006.
 332. Kozyraki R, Kristiansen M, Silahtaroglu A, Hansen C, Jacobsen C, Tommerup N, Verroust PJ, Moestrup SK. The human intrinsic factor‐vitamin B12 receptor, cubilin: Molecular characterization and chromosomal mapping of the gene to 10p within the autosomal recessive megaloblastic anemia (MGA1) region. Blood 91: 3593‐3600, 1998.
 333. Kraut JA, Helander KG, Helander HF, Iroezi ND, Marcus EA, Sachs G. Detection and localization of H+‐K+‐ATPase isoforms in human kidney. Am J Physiol Renal Physiol 281: F763‐F768, 2001.
 334. Kriz W. The architectonic and functional structure of the rat kidney. Z Zellforsch Mikrosk Anat 82: 495‐535, 1967.
 335. Kriz W. Structural organization of the renal medulla: Comparative and functional aspects. Am J Physiol 241: R3‐R16, 1981.
 336. Kriz W. A periarterial pathway for intrarenal distribution of renin. Kidney Int Suppl 20: S51‐S56, 1987.
 337. Kriz W. Ontogenetic development of the filtration barrier. Nephron Exp Nephrol 106: e44‐e50, 2007.
 338. Kriz W, Bankir L. A standard nomenclature for structures of the kidney. The Renal Commission of the International Union of Physiological Sciences (IUPS). Kidney Int 33: 1‐7, 1988.
 339. Kriz W, Barrett JM, Peter S. The renal vasculature: Anatomical‐functional aspects. Int Rev Physiol 11: 1‐21, 1976.
 340. Kriz W, Kaissling B. Structural organization of the mammalian kidney. In: Alpern RJ, Hebert SC, editors. Seldin and Giebisch's The kidney. Amsterdam: Elsevier Inc., 2008, p. 479‐563.
 341. Kriz W, Kaissling B, Schiller A, Taugner R. Morphological characteristics of transport epithelia. Klin Wochenschr 57: 967‐975, 1979.
 342. Kriz W, Koepsell H. The structural organization of the mouse kidney. Z Anat Entwicklungsgesch 144: 137‐163, 1974.
 343. Kriz W, Schiller A, Kaissling B, Taugner R. Comparative and functional aspects of thin loop limb ultrastructure. In: Maunsbach AB, Olsen TS, Christensen EI, editors. Functional Ultrastructure of the Kidney. London: Academic Press, 1980, p. 239‐250.
 344. Kriz W, Schiller A, Taugner R. Freeze‐fracture studies on the thin limps of Henle's loop in psammomys obesus. Am J Anat 162: 23‐33, 1981.
 345. Kriz W, Schnermann J, Koepsell H. The position of short and long loops of Henle in the rat kidney. Z Anat Entwicklungsgesch 138: 301‐319, 1972.
 346. Kugler P. Quantitative histochemistry of the lysosomal dipeptidyl aminopeptidase II in the proximal tubule of the rat kidney. Histochemistry 76: 557‐566, 1982.
 347. Kugler P, Vornberger G. Renal cathepsin‐B activities in rats after castration and treatment with sex hormones. Histochemistry 85: 157‐161, 1986.
 348. Kugler P, Wolf G, Scherberich J. Histochemical demonstration of peptidases in the human kidney. Histochemistry 83: 337‐341, 1985.
 349. Kuhn K, Reale E. Junctional complexes of the tubular cells in the human kidney as revealed with freeze‐fracture. Cell Tissue Res 160: 193‐205, 1975.
 350. Kuhn W, Ramel A. Aktiver salztransport als moeglicher (und wahrscheinlicher) einzeleffekt bei der harnkonzentrierung in der niere. Helv Chim Acta 42: 628‐660, 1959.
 351. Kultz D. Hypertonicity and TonEBP promote development of the renal concentrating system. Am J Physiol Renal Physiol 287: F876‐F877, 2004.
 352. Kyossev Z, Walker PD, Reeves WB. Immunolocalization of NAD‐dependent 11 beta‐hydroxysteroid dehydrogenase in human kidney and colon. Kidney Int 49: 271‐281, 1996.
 353. Lal‐Nag M, Morin PJ. The claudins. Genome Biol 10: 235, 2009.
 354. Lambert P. Sur les potentialités de résorption du tube contourné chez les urodèles. Soc Biol 110: 114‐116, 1932.
 355. Langer KH, Thoenes W. Zur genese “optisch leerer vakuolen” im epithel des energetisch insuffizienten proximalen nierentubulus. Untersuchungen an der rattenniere nach kaliumcyanidvergiftung und hämorrhagischen schock. Verh Dtsch Ges Pathol 394‐401, 1939.
 356. Larsson L, Clapp WL III, Park CH, Cannon JK, Tisher CC. Ultrastructural localization of acidic compartments in cells of isolated rabbit PCT. Am J Physiol 253: F95‐F103, 1987.
 357. Laurie GW, Leblond CP, Inoue S, Martin GR, Chung A. Fine structure of the glomerular basement membrane and immunolocalization of five basement membrane components to the lamina densa (basal lamina) and its extensions in both glomeruli and tubules of the rat kidney. Am J Anat 169: 463‐481, 1984.
 358. Leak LV. Ultrastructure of proximal tubule cells in mouse kidney as revealed by freeze etching. J Ultrastruct Res 25: 253‐270, 1968.
 359. Lee DB, Huang E, Ward HJ. Tight junction biology and kidney dysfunction. Am J Physiol Renal Physiol 290: F20‐F34, 2006.
 360. Lee DH, Riquier AD, Yang LE, Leong PK, Maunsbach AB, McDonough AA. Acute hypertension provokes acute trafficking of distal tubule Na‐Cl cotransporter (NCC) to subapical cytoplasmic vesicles. Am J Physiol Renal Physiol 296: F810‐F818, 2009.
 361. Lee HW, Verlander JW, Bishop JM, Igarashi P, Handlogten ME, Weiner ID. Collecting duct‐specific Rh C glycoprotein deletion alters basal and acidosis‐stimulated renal ammonia excretion. Am J Physiol Renal Physiol 296: F1364‐F1375, 2009.
 362. Lee HW, Verlander JW, Bishop JM, Nelson RD, Handlogten ME, Weiner ID. Effect of intercalated cell‐specific Rh C glycoprotein deletion on basal and metabolic acidosis‐stimulated renal ammonia excretion. Am J Physiol Renal Physiol 299: F369‐F379, 2010.
 363. LeFurgey A, Tisher CC. Morphology of rabbit collecting duct. Am J Anat 155: 111‐124, 1979.
 364. LeHir M, Dubach UC. The cellular specificity of lectin binding in the kidney. I. A light microscopical study in the rat. Histochemistry 74: 521‐530, 1982a.
 365. LeHir M, Dubach UC. The cellular specificity of lectin binding in the kidney. II. A light microscopical study in the rabbit. Histochemistry 74: 531‐540, 1982b.
 366. LeHir M, Dubach UC, Guder WG. Distribution of acid hydrolases in the nephron of normal and diabetic rats. Int J Biochem 12: 41‐45, 1980.
 367. LeHir M, Dubach UC, Schmidt U. Quantitative distribution of lysosomal hydrolases in the rat nephron. Histochemistry 63: 245‐251, 1979.
 368. LeHir M, Kaissling B, Dubach UC. Distal tubular segments of the rabbit kidney after adaptation to altered Na‐ and K‐intake. II. Changes in Na‐K‐ATPase activity. Cell Tissue Res 224: 493‐504, 1982.
 369. LeHir M, Kaissling B, Koeppen BM, Wade JB. Binding of peanut lectin to specific epithelial cell types in kidney. Am J Physiol 242: C117‐C120, 1982.
 370. Levi M, Lotscher M, Sorribas V, Custer M, Arar M, Kaissling B, Murer H, Biber J. Cellular mechanisms of acute and chronic adaptation of rat renal P(i) transporter to alterations in dietary P(i). Am J Physiol 267: F900‐F908, 1994.
 371. Leviel F, Hubner CA, Houillier P, Morla L, El MS, Brideau G, Hatim H, Parker MD, Kurth I, Kougioumtzes A, Sinning A, Pech V, Riemondy KA, Miller RL, Hummler E, Shull GE, Aronson PS, Doucet A, Wall SM, Chambrey R, Eladari D. The Na+‐dependent chloride‐bicarbonate exchanger SLC4A8 mediates an electroneutral Na +reabsorption process in the renal cortical collecting ducts of mice. J Clin Invest 120: 1627‐1635, 2010.
 372. Levine DZ, Jacobson HR. The regulation of renal acid secretion: New observations from studies of distal nephron segments. Kidney Int 29: 1099‐1109, 1986.
 373. Levine DZ, Roinel N, de RC. Flow‐correlated influx of K, Ca, P, and Mg during continuous microperfusion of the loop of Henle in the rat. Kidney Int 22: 634‐639, 1982.
 374. Li WY, Huey CL, Yu AS. Expression of claudin‐7 and ‐8 along the mouse nephron. Am J Physiol Renal Physiol 286: F1063‐F1071, 2004.
 375. Lima WR, Parreira KS, Devuyst O, Caplanusi A, N'kuli F, Marien B, Van Der Smissen P, Alves PM, Verroust P, Christensen EI, Terzi F, Matter K, Balda MS, Pierreux CE, Courtoy PJ: ZONAB promotes proliferation and represses differentiation of proximal tubule epithelial cells. J Am Soc Nephrol 21: 478‐488, 2010.
 376. Lin DH, Sterling H, Wang WH. The protein tyrosine kinase‐dependent pathway mediates the effect of K intake on renal K secretion. Physiology (Bethesda) 20: 140‐146, 2005.
 377. Liu W, Morimoto T, Woda C, Kleyman TR, Satlin LM. Ca2+ dependence of flow‐stimulated K secretion in the mammalian cortical collecting duct. Am J Physiol Renal Physiol 293: F227‐F235, 2007.
 378. Liu W, Xu S, Woda C, Kim P, Weinbaum S, Satlin LM. Effect of flow and stretch on the [Ca2+]i response of principal and intercalated cells in cortical collecting duct. Am J Physiol Renal Physiol 285: F998‐F1012, 2003.
 379. Loffing J, Kaissling B. Sodium and calcium transport pathways along the mammalian distal nephron: From rabbit to human. Am J Physiol Renal Physiol 284: F628‐F643, 2003.
 380. Loffing J, Korbmacher C. Regulated sodium transport in the renal connecting tubule (CNT) via the epithelial sodium channel (ENaC). Pflugers Arch 458: 111‐135, 2009.
 381. Loffing J, LeHir M, Kaissling B. Modulation of salt transport rate affects DNA synthesis in vivo in rat renal tubules. Kidney Int 47: 1615‐1623, 1995.
 382. Loffing J, Loffing‐Cueni D, Hegyi I, Kaplan MR, Hebert SC, LeHir M, Kaissling B. Thiazide treatment of rats provokes apoptosis in distal tubule cells. Kidney Int 50: 1180‐1190, 1996.
 383. Loffing J, Loffing‐Cueni D, Macher A, Hebert SC, Olson B, Knepper MA, Rossier BC, Kaissling B. Localization of epithelial sodium channel and aquaporin‐2 in rabbit kidney cortex. Am J Physiol Renal Physiol 278: F530‐F539, 2000.
 384. Loffing J, Loffing‐Cueni D, Valderrabano V, Klausli L, Hebert SC, Rossier BC, Hoenderop JG, Bindels RJ, Kaissling B. Distribution of transcellular calcium and sodium transport pathways along mouse distal nephron. Am J Physiol Renal Physiol 281: F1021‐F1027, 2001.
 385. Loffing J, Lotscher M, Kaissling B, Biber J, Murer H, Seikaly M, Alpern RJ, Levi M, Baum M, Moe OW. Renal Na/H exchanger NHE‐3 and Na‐PO4 cotransporter NaPi‐2 protein expression in glucocorticoid excess and deficient states. J Am Soc Nephrol 9: 1560‐1567, 1998.
 386. Loffing J, Pietri L, Aregger F, Bloch‐Faure M, Ziegler U, Meneton P, Rossier BC, Kaissling 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.
 387. Loffing J, Summa V, Zecevic M, Verrey F. Mediators of aldosterone action in the renal tubule. Curr Opin Nephrol Hypertens 10: 667‐675, 2001.
 388. Loffing J, Vallon V, Loffing‐Cueni D, Aregger F, Richter K, Pietri L, Bloch‐Faure M, Hoenderop JG, Shull GE, Meneton P, Kaissling B. Altered renal distal tubule structure and renal Na(+) and Ca(2+) handling in a mouse model for Gitelman's syndrome. J Am Soc Nephrol 15: 2276‐2288, 2004.
 389. Loffing J, Zecevic M, Feraille E, Kaissling B, Asher C, Rossier BC, Firestone GL, Pearce D, Verrey F. Aldosterone induces rapid apical translocation of ENaC in early portion of renal collecting system: Possible role of SGK. Am J Physiol Renal Physiol 280: F675‐F682, 2001.
 390. Lonnerholm G, Ridderstrale Y. Intracellular distribution of carbonic anhydrase in the rat kidney. Kidney Int 17: 162‐174, 1980.
 391. Lonnerholm G, Wistrand PJ. Carbonic anhydrase in the human kidney: A histochemical and immunocytochemical study. Kidney Int 25: 886‐898, 1984.
 392. Lorenz JN, Baird NR, Judd LM, Noonan WT, Andringa A, Doetschman T, Manning PA, Liu LH, Miller ML, Shull GE. Impaired renal NaCl absorption in mice lacking the ROMK potassium channel, a model for type II Bartter's syndrome. J Biol Chem 277: 37871‐37880, 2002.
 393. Lotscher M, Kaissling B, Biber J, Murer H, Levi M. Role of microtubules in the rapid regulation of renal phosphate transport in response to acute alterations in dietary phosphate content. J Clin Invest 99: 1302‐1312, 1997.
 394. Lotscher M, Scarpetta Y, Levi M, Halaihel N, Wang H, Zajicek HK, Biber J, Murer H, Kaissling B. Rapid downregulation of rat renal Na/P(i) cotransporter in response to parathyroid hormone involves microtubule rearrangement. J Clin Invest 104: 483‐494, 1999.
 395. Lu M, Wang T, Yan Q, Wang W, Giebisch G, Hebert SC. ROMK is required for expression of the 70‐pS K channel in the thick ascending limb. Am J Physiol Renal Physiol 286: F490‐F495, 2004.
 396. Luyckx VA, Goda FO, Mount DB, Nishio T, Hall A, Hebert SC, Hammond TG, Yu AS. Intrarenal and subcellular localization of rat CLC5. Am J Physiol 275: F761‐F769, 1998.
 397. Maack T. Changes in the activity of acid hydrolases during renal reabsorption of lysozyme. J Cell Biol 35: 268‐273, 1967.
 398. Madara JL. Regulation of the movement of solutes across tight junctions. Annu Rev Physiol 60: 143‐159, 1998.
 399. Madjdpour C, Bacic D, Kaissling B, Murer H, Biber J. Segment‐specific expression of sodium‐phosphate cotransporters NaPi‐IIa and ‐IIc and interacting proteins in mouse renal proximal tubules. Pflugers Arch 448: 402‐410, 2004.
 400. Madri JA, Roll FJ, Furthmayr H, Foidart JM. Ultrastructural localization of fibronectin and laminin in the basement membranes of the murine kidney. J Cell Biol 86: 682‐687, 1980.
 401. Madsen KM, Christensen EI. Effects of mercury on lysosomal protein digestion in the kidney proximal tubule. Lab Invest 38: 165‐174, 1978.
 402. Madsen KM, Clapp WL, Verlander JW. Structure and function of the inner medullary collecting duct. Kidney Int 34: 441‐454, 1988.
 403. Madsen KM, Harris RH, Tisher CC. Uptake and intracellular distribution of ferritin in the rat distal convoluted tubule. Kidney Int 21: 354‐361, 1982.
 404. Madsen KM, Hyung Park C. Lysosome distribution and cathepsin B and L activity along the rabbit proximal tubule. Am J Physiol 253: F1290‐F1301, 1987.
 405. Madsen KM, Maunsbach AB. Effects of chronic mercury exposure on the rat kidney cortex as studied morphometrically by light and electron microscopy. Virchows Arch B Cell Pathol Incl Mol Pathol 37: 137‐152, 1981.
 406. Madsen KM, Tisher CC. Structural‐functional relationships along the distal nephron. Am J Physiol 250: F1‐F15, 1986.
 407. Madsen KM, Verlander JW, Tisher CC. Relationship between structure and function in distal tubule and collecting duct. J Electron Microsc Tech 9: 187‐208, 1988.
 408. Marchetti J, Imbert‐Teboul M, Alhenc‐Gelas F, Allegrini J, Menard J, Morel F. Kallikrein along the rabbit microdissected nephron: A micromethod for its measurement. Effect of adrenalectomy and DOCA treatment. Pflugers Arch 401: 27‐33, 1984.
 409. Marino M, Andrews D, Brown D, McCluskey RT. Transcytosis of retinol‐binding protein across renal proximal tubule cells after megalin (gp 330)‐mediated endocytosis. J Am Soc Nephrol 12: 637‐648, 2001.
 410. Marples D, Schroer TA, Ahrens N, Taylor A, Knepper MA, Nielsen S. Dynein and dynactin colocalize with AQP2 water channels in intracellular vesicles from kidney collecting duct. Am J Physiol 274: F384‐F394, 1998.
 411. Marshansky V, Ausiello DA, Brown D. Physiological importance of endosomal acidification: Potential role in proximal tubulopathies. Curr Opin Nephrol Hypertens 11: 527‐537, 2002.
 412. Marsy S, Elalouf JM, Doucet A. Quantitative RT‐PCR analysis of mRNAs encoding a colonic putative H, K‐ATPase alpha subunit along the rat nephron: Effect of K+ depletion. Pflugers Arch 432: 494‐500, 1996.
 413. Martinez‐Hernandez A, Chung AE. The ultrastructural localization of two basement membrane components: Entactin and laminin in rat tissues. J Histochem Cytochem 32: 289‐298, 1984.
 414. Masilamani S, Kim GH, Mitchell C, Wade JB, Knepper MA. Aldosterone‐mediated regulation of ENaC alpha, beta, and gamma subunit proteins in rat kidney. J Clin Invest 104: R19‐R23, 1999.
 415. Matlin K, Bainton DF, Pesonen M, Louvard D, Genty N, Simons K. Transepithelial transport of a viral membrane glycoprotein implanted into the apical plasma membrane of Madin‐Darby canine kidney cells. I. Morphological evidence. J Cell Biol 97: 627‐637, 1983.
 416. Matlin KS. Clues to occludin. Focus on “Knockdown of occludin expression leads to diverse phenotypic alterations in epithelial cells”. Am J Physiol Cell Physiol 288: C1191‐C1192, 2005.
 417. Maude DL, Handelsman DG, Babu M, Gordon EE. Handling of insulin by the isolated perfused rat kidney. Am J Physiol 240: F288‐F294, 1981.
 418. Maunsbach AB. Absorption of ferritin by rat kidney proximal tubule cells. Electron microscopic observations of the initial uptake phase in cells. J Ultrastruct Res 16: 1‐12, 1966a.
 419. Maunsbach AB. Absorption of I125‐labeled homologous albumin by rat kidney proximal tubule cells. A study of microperfused single proximal tubules by electron microscopic autoradiography and histochemistry. J Ultrastruct Res 15: 197‐241, 1966b.
 420. Maunsbach AB. Electron microscopic observations of cytoplasmatic bodies with crystalline patterns in rat kidney proximal tubule cells. J Ultrastruct Res 14: 167‐189, 1966c.
 421. Maunsbach AB. Observations on the segmentation of the proximal tubule in the rat kidney. J Ultrastruct Res 16: 239‐258, 1966d.
 422. Maunsbach AB. Observations on the ultrastructure and acid phosphatase activity of the cytoplasmic bodies in rat kidney proximal tubule cells. With a comment on their classification. J Ultrastruct Res 16: 197‐238, 1966e.
 423. Maunsbach AB. The influence of different fixatives and fixation methods on the ultrastructure of rat kidney proximal tubule cells. I. Comparison of different perfusion fixation methods and of glutaraldehyde, formaldehyde and osmium tetroxide fixatives. J Ultrastruct Res 15: 242‐282, 1966f.
 424. Maunsbach AB. Ultrastructure and digestive activity of lysosomes from proximal tubule cells. Proc 4th Int Congr Nephrol 1: 102‐115, 1969.
 425. Maunsbach AB. Ultrastructure of the proximal tubule. In: Orloff J, Berliner RW, editors. Handbook of Physiology. Washington D.C.: American Physiological Society, 1973, p. 31‐79.
 426. Maunsbach AB. Cellular mechanisms of tubular protein transport. In: Thurau K, editor. International Review of Physiology, Kidney and Urinary Tract Physiology II. Baltimore: University Park Press, 1976, p. 145‐167.
 427. Maunsbach AB. Ultrastructure of epithelia as related to models of iso‐osmotic transport. In: Salánki J, editor. Physiology of Non‐excitable Cells. Budapest: Akadémiai Kiadó, 1981, p. 81‐88.
 428. Maunsbach AB, Christensen EI. Functional ultrastructure of the proximal tubule. In: Windhager EE, editor. Handbook of Physiology. Renal Physiology. Am. Physiol. Soc. New York: Oxford University Press, 1992, sect. 8, p. 41‐107.
 429. Maunsbach AB, Giebisch GH, Stanton BA. Effects of flow rate on proximal tubule ultrastructure. Am J Physiol 253: F582‐F587, 1987.
 430. McCulloch F, Chambrey R, Eladari D, Peti‐Peterdi J. Localization of connexin 30 in the luminal membrane of cells in the distal nephron. Am J Physiol Renal Physiol 289: F1304‐F1312, 2005.
 431. McDonough AA, Biemesderfer D. Does membrane trafficking play a role in regulating the sodium/hydrogen exchanger isoform 3 in the proximal tubule? Curr Opin Nephrol Hypertens 12: 533‐541, 2003.
 432. McDonough AA, Leong PK, Yang LE. Mechanisms of pressure natriuresis: How blood pressure regulates renal sodium transport. Ann N Y Acad Sci 986: 669‐677, 2003.
 433. McKinney TD, Davidson KK. Bicarbonate transport in collecting tubules from outer stripe of outer medulla of rabbit kidneys. Am J Physiol 253: F816‐F822, 1987.
 434. McNeill H. Planar cell polarity and the kidney. J Am Soc Nephrol 20: 2104‐2111, 2009.
 435. Meade P, Hoover RS, Plata C, Vazquez N, Bobadilla NA, Gamba G, Hebert SC. cAMP‐dependent activation of the renal‐specific Na+‐K+‐2Cl‐ cotransporter is mediated by regulation of cotransporter trafficking. Am J Physiol Renal Physiol 284: F1145‐F1154, 2003.
 436. Meezan E, Pillion DJ, Elgavish A. Binding and degradation of 125I‐insulin by isolated rat renal brush border membranes: Evidence for low affinity, high capacity insulin recognition sites. J Membr Biol 105: 113‐129, 1988.
 437. Meij IC, Koenderink JB, De Jong JC, De Pont JJ, Monnens LA, van den Heuvel LP, Knoers NV. Dominant isolated renal magnesium loss is caused by misrouting of the Na+,K+‐ATPase gamma‐subunit. Ann N Y Acad Sci 986: 437‐443, 2003.
 438. Melander O, Orho‐Melander M, Bengtsson K, Lindblad U, Rastam L, Groop L, Hulthen UL. Genetic variants of thiazide‐sensitive NaCl‐cotransporter in Gitelman's syndrome and primary hypertension. Hypertension 36: 389‐394, 2000.
 439. Meneton P, Loffing J, Warnock DG. Sodium and potassium handling by the aldosterone‐sensitive distal nephron: The pivotal role of the distal and connecting tubule. Am J Physiol Renal Physiol 287: F593‐F601, 2004.
 440. Mennitt PA, Wade JB, Ecelbarger CA, Palmer LG, Frindt G. Localization of ROMK channels in the rat kidney. J Am Soc Nephrol 8: 1823‐1830, 1997.
 441. Miller F, Palade GE. Lytic activities in renal protein absorption droplets. An electron microscopic cytochemical study. J Cell Biol 23: 519‐552, 1964.
 442. Milton A, Hellfritzsch M, Christensen EI. Basolateral binding and uptake of 125‐I‐insulin in proximal tubular cells after peritubular extraction in the avian kidney. Diabetes Res 7: 189‐195, 1988.
 443. Mitic LL, Anderson JM. Molecular architecture of tight junctions. Annu Rev Physiol 60: 121‐142, 1998.
 444. Moestrup SK, Birn H, Fischer PB, Petersen CM, Verroust PJ, Sim RB, Christensen EI, Nexø E. Megalin‐mediated endocytosis of transcobalamin‐vitamin‐B12 complexes suggests a role of the receptor in vitamin‐B12 homeostasis. Proc Natl Acad Sci U S A 93: 8612‐8617, 1996.
 445. Moestrup SK, Cui S, Vorum H, Bregengård C, Bjørn SE, Norris K, Gliemann J, Christensen EI. Evidence that epithelial glycoprotein 330/megalin mediates uptake of polybasic drugs. J Clin Invest 96: 1404‐1413, 1995.
 446. Moestrup SK, Kozyraki R, Kristiansen M, Kaysen JH, Rasmussen HH, Brault D, Pontillon F, Goda FO, Christensen EI, Hammond TG, Verroust PJ. The intrinsic factor‐vitamin B12 receptor and target of teratogenic antibodies is a megalin‐binding peripheral membrane protein with homology to developmental proteins. J Biol Chem 273: 5235‐5242, 1998.
 447. Moller JC. Proximal tubules in long‐term compensatory renal growth. Quantitative light‐ and electron‐microscopic analyses. APMIS Suppl 4: 82‐86, 1988.
 448. Moller JC, Skriver E. Quantitative ultrastructure of human proximal tubules and cortical interstitium in chronic renal disease (hydronephrosis). Virchows Arch A Pathol Anat Histopathol 406: 389‐406, 1985.
 449. Molony DA, Reeves WB, Hebert SC, Andreoli TE. ADH increases apical Na+, K+, 2Cl‐ entry in mouse medullary thick ascending limbs of Henle. Am J Physiol 252: F177‐F187, 1987.
 450. Morel F, Chabardes D, Imbert M. Functional segmentation of the rabbit distal tubule by microdetermination of hormone‐dependent adenylate cyclase activity. Kidney Int 9: 264‐277, 1976.
 451. Morel F, Chabardès D, Imbert‐Teboul M. Heterogeneity of hormonal control in the distal nephron. Barcolo R, editor. Proceedings of the VII International Congress of Nephrology Basel: Karger, 1978, p. 209‐216.
 452. Mouro‐Chanteloup I, Cochet S, Chami M, Genetet S, Zidi‐Yahiaoui N, Engel A, Colin Y, Bertrand O, Ripoche P. Functional reconstitution into liposomes of purified human RhCG ammonia channel. PLoS One 5: e8921, 2010.
 453. Mujais SK, Kauffman S, Katz AI. Angiotensin II binding sites in individual segments of the rat nephron. J Clin Invest 77: 315‐318, 1986.
 454. Mundel P, Bachmann S, Bader M, Fischer A, Kummer W, Mayer B, Kriz W. Expression of nitric oxide synthase in kidney macula densa cells. Kidney Int 42: 1017‐1019, 1992.
 455. Murer H, Forster IC, Hernando N, Biber J. Proximal tubular handling of phosphate: Na/Pi‐cotransporters and their regulation. In: Alpern RJ, Hebert SC, editors. Seldin and Giebisch's The Kidney. New York: Academic Press, 2008, p. 1979‐1988.
 456. Mutig K, Saritas T, Uchida S, Kahl T, Borowski T, Paliege A, Bohlick A, Bleich M, Shan Q, Bachmann S. Short‐term stimulation of the thiazide‐sensitive Na+‐Cl‐ cotransporter by vasopressin involves phosphorylation and membrane translocation. Am J Physiol Renal Physiol 298: F502‐F509, 2010.
 457. Myers CE, Bulger RE, Tisher CC, Trump BF. Human ultrastructure. IV. Collecting duct of healthy individuals. Lab Invest 15: 1921‐1950, 1966.
 458. Nakamura R, Emmanouel DS, Katz AI. Insulin binding in various segments of the rabbit nephron. J Clin Invest 72: 388‐392, 1983.
 459. Nakamura S, Amlal H, Galla JH, Soleimani M. Colonic H+‐K+‐ATPase is induced and mediates increased HCO3‐ reabsorption in inner medullary collecting duct in potassium depletion. Kidney Int 54: 1233‐1239, 1998.
 460. Nakamura S, Amlal H, Soleimani M, Galla JH. Pathways for HCO3‐reabsorption in mouse medullary collecting duct segments. J Lab Clin Med 136: 218‐223, 2000.
 461. Nauli SM, Alenghat FJ, Luo Y, Williams E, Vassilev P, Li X, Elia AE, Lu W, Brown EM, Quinn SJ, Ingber DE, Zhou J. Polycystins 1 and 2 mediate mechanosensation in the primary cilium of kidney cells. Nat Genet 33: 129‐137, 2003.
 462. Nejsum LN, Elkjaer M, Hager H, Frokiaer J, Kwon TH, Nielsen S. Localization of aquaporin‐7 in rat and mouse kidney using RT‐PCR, immunoblotting, and immunocytochemistry. Biochem Biophys Res Commun 277: 164‐170, 2000.
 463. Nelson CM, Jean RP, Tan JL, Liu WF, Sniadecki NJ, Spector AA, Chen CS. Emergent patterns of growth controlled by multicellular form and mechanics. Proc Natl Acad Sci U S A 102: 11594‐11599, 2005.
 464. Nelson RD, Guo XL, Masood K, Brown D, Kalkbrenner M, Gluck S. Selectively amplified expression of an isoform of the vacuolar H(+)‐ATPase 56‐kilodalton subunit in renal intercalated cells. Proc Natl Acad Sci U S A 89: 3541‐3545, 1992.
 465. Nelson WJ, Nusse R. Convergence of Wnt, beta‐catenin, and cadherin pathways. Science 303: 1483‐1487, 2004.
 466. Nicco C, Bankir L, Bouby N. Effect of salt and water intake on epithelial sodium channel mRNA abundance in the kidney of salt‐sensitive Sabra rats. Clin Exp Pharmacol Physiol 30: 963‐965, 2003.
 467. Nicco C, Wittner M, DiStefano A, Jounier S, Bankir L, Bouby N. Chronic exposure to vasopressin upregulates ENaC and sodium transport in the rat renal collecting duct and lung. Hypertension 38: 1143‐1149, 2001.
 468. Nielsen JT, Christensen EI. Ultrastructure of isolated perfused proximal tubules from rabbit kidney. A comparison with proximal tubules fixed by perfusion in vivo. Lab Invest 49: 400‐411, 1983.
 469. Nielsen JT, Christensen EI. Basolateral endocytosis of protein in isolated perfused proximal tubules. Kidney Int 27: 39‐45, 1985.
 470. Nielsen JT, Nielsen S, Christensen EI. Transtubular transport of proteins in rabbit proximal tubules. J Ultrastruct Res 92: 133‐145, 1985.
 471. Nielsen JT, Nielsen S, Christensen EI. Handling of lysozyme in isolated, perfused proximal tubules. Am J Physiol 251: F822‐F830, 1986.
 472. Nielsen R, Courtoy PJ, Jacobsen C, Dom G, Lima WR, Jadot M, Willnow TE, Devuyst O, Christensen EI. Endocytosis provides a major alternative pathway for lysosomal biogenesis in kidney proximal tubular cells. Proc Natl Acad Sci U S A 104: 5407‐5412, 2007.
 473. Nielsen S, Chou CL, Marples D, Christensen EI, Kishore BK, Knepper MA. Vasopressin increases water permeability of kidney collecting duct by inducing translocation of aquaporin‐CD water channels to plasma membrane. Proc Natl Acad Sci U S A 92: 1013‐1017, 1995.
 474. Nielsen S, Christensen EI. Insulin absorption in renal proximal tubules: A quantitative immunocytochemical study. J Ultrastruct Mol Struct Res 102: 205‐220, 1989.
 475. Nielsen S, Frokiaer J, Marples D, Kwon TH, Agre P, Knepper MA. Aquaporins in the kidney: From molecules to medicine. Physiol Rev 82: 205‐244, 2002.
 476. Nielsen S, Knepper MA. Vasopressin activates collecting duct urea transporters and water channels by distinct physical processes. Am J Physiol 265: F204‐F213, 1993.
 477. Nielsen S, Maunsbach AB, Ecelbarger CA, Knepper MA. Ultrastructural localization of Na‐K‐2Cl cotransporter in thick ascending limb and macula densa of rat kidney. Am J Physiol 275: F885‐F893, 1998.
 478. Nielsen S, Nielsen JT. Influence of flow rate and perfused load on insulin absorption in isolated proximal tubules. Am J Physiol 254: F802‐F812, 1988.
 479. Nielsen S, Nielsen JT, Christensen EI. Luminal and basolateral uptake of insulin in isolated, perfused proximal tubules. Am J Physiol 253: F857‐F867, 1987.
 480. Nielsen S, Pallone T, Smith BL, Christensen EI, Agre P, Maunsbach AB. Aquaporin‐1 water channels in short and long loop descending thin limbs and in descending vasa recta in rat kidney. Am J Physiol 268: F1023‐F1037, 1995.
 481. Nielsen S, Smith BL, Christensen EI, Knepper MA, Agre P. CHIP28 water channels are localized in constitutively water‐permeable segments of the nephron. J Cell Biol 120: 371‐383, 1993.
 482. Nijenhuis T, Hoenderop JG, Loffing J, van der Kemp AW, van Os CH, Bindels RJ. Thiazide‐induced hypocalciuria is accompanied by a decreased expression of Ca2+ transport proteins in kidney. Kidney Int 64: 555‐564, 2003.
 483. Nishi T, Forgac M. The vacuolar (H+)‐ATPases–nature's most versatile proton pumps. Nat Rev Mol Cell Biol 3: 94‐103, 2002.
 484. Nishio S, Hatano M, Nagata M, Horie S, Koike T, Tokuhisa T, Mochizuki T. Pkd1 regulates immortalized proliferation of renal tubular epithelial cells through p53 induction and JNK activation. J Clin Invest 115: 910‐918, 2005.
 485. Novarino G, Weinert S, Rickheit G, Jentsch TJ. Endosomal chloride‐proton exchange rather than chloride conductance is crucial for renal endocytosis. Science 328: 1398‐1401, 2010.
 486. Novikoff AB. The rat kidney: Cytochemical and electron microscopic studies. In: Quinn EL, Kass EH, editors. Biology of Pyelonephritis. Boston: Little, Brown, 1960, p. 113‐144.
 487. Novikoff AB. Lysosomes in the physiology and pathology of cells: Contributions of staining methods. In: De Reuck AVS, Cameron MP, editors. Ciba Found. Symp., Lysosomes. Boston: Little, Brown, 1963, p. 36‐77.
 488. Nyengaard JR, Bendtsen TF. Glomerular number and size in relation to age, kidney weight, and body surface in normal man. Anat Rec 232: 194‐201, 1992.
 489. Nykjær A, Dragun D, Walther D, Vorum H, Jacobsen C, Herz J, Melsen F, Christensen EI, Willnow TE. An endocytic pathway essential for renal uptake and activation of the steroid 25‐(OH) vitamin D3. Cell 96: 507‐515, 1999.
 490. Nykjaer A, Fyfe JC, Kozyraki R, Leheste JR, Jacobsen C, Nielsen MS, Verroust PJ, Aminoff M, de la Chapelle A, Moestrup SK, Ray R, Gliemann J, Willnow TE, Christensen EI. Cubilin dysfunction causes abnormal metabolism of the steroid hormone 25(OH) vitamin D(3). Proc Natl Acad Sci U S A 98: 13895‐13900, 2001.
 491. O'Neil RG, Hayhurst RA. Functional differentiation of cell types of cortical collecting duct. Am J Physiol 248: F449‐F453, 1985.
 492. Obermuller N, Kunchaparty S, Ellison DH, Bachmann 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.
 493. Odgaard E, Jakobsen JK, Frische S, Praetorius J, Nielsen S, Aalkjaer C, Leipziger J. Basolateral Na+‐dependent HCO3‐ transporter NBCn1‐mediated HCO3‐ influx in rat medullary thick ascending limb. J Physiol 555: 205‐218, 2004.
 494. Oka T, Murata Y, Namba M, Yoshimizu T, Toyomura T, Yamamoto A, Sun‐Wada GH, Hamasaki N, Wada Y, Futai M. a4, a unique kidney‐specific isoform of mouse vacuolar H+‐ATPase subunit a. J Biol Chem 276: 40050‐40054, 2001.
 495. Olbricht CJ, Cannon JK, Tisher CC. Cathepsin B and L in nephron segments of rats with puromycin aminonucleoside nephrosis. Kidney Int 32: 354‐361, 1987.
 496. Oliver J. Nephrons and Kidneys. New York: Harper & Row, Hoeber Medical, 1968.
 497. Oliver J, MacDowell M, Lee YC. Cellular mechanisms of protein metabolism in the nephron. I. The structural aspects of proteinuria: Tubular absorption, droplet formation and the disposal of proteins. J Exp Med 99: 589‐604, 1954.
 498. Omata K, Carretero OA, Itoh S, Scicli AG. Active and inactive kallikrein in rabbit connecting tubules and urine during low and normal sodium intake. Kidney Int 24: 714‐718, 1983.
 499. Omata K, Carretero OA, Scicli AG, Jackson BA. Localization of active and inactive kallikrein (kininogenase activity) in the microdissected rabbit nephron. Kidney Int 22: 602‐607, 1982.
 500. Orlando RA, Rader K, Authier F, Yamazaki H, Posner BI, Bergeron JJ, Farquhar MG. Megalin is an endocytic receptor for insulin. J Am Soc Nephrol 9: 1759‐1766, 1998.
 501. Ortiz PA. cAMP increases surface expression of NKCC2 in rat thick ascending limbs: Role of VAMP. Am J Physiol Renal Physiol 290: F608‐F616, 2006.
 502. Osathanondh V, Potter EL. Development of Human Kidney as shown by microdissection. III. Formation and interrelationship of collecting tubules and nephrons. Arch Pathol 76: 290‐302, 1963.
 503. Ottosen PD. Reversible peritubular binding of a cationic protein (lysozyme) to flounder kidney tubules. Cell Tissue Res 194: 207‐218, 1978.
 504. Ottosen PD, Bode F, Madsen KM, Maunsbach AB. Renal handling of lysozyme in the rat. Kidney Int 15: 246‐254, 1979.
 505. Ottosen PD, Maunsbach AB. Transport of peroxidase in flounder kidney tubules studies by electron microscope histochemistry. Kidney Int 3: 315‐326, 1973.
 506. Palmer LG, Frindt G. High‐conductance K channels in intercalated cells of the rat distal nephron. Am J Physiol Renal Physiol 292: F966‐F973, 2007.
 507. Pannabecker TL, Abbott DE, Dantzler WH. Three‐dimensional functional reconstruction of inner medullary thin limbs of Henle's loop. Am J Physiol Renal Physiol 286: F38‐F45, 2004.
 508. Park CH. Time course and vectorial nature of albumin metabolism in isolated perfused rabbit PCT. Am J Physiol 255: F520‐F528, 1988.
 509. Park CH, Maack T. Albumin absorption and catabolism by isolated perfused proximal convoluted tubules of the rabbit. J Clin Invest 73: 767‐777, 1984.
 510. Patel V, Li L, Cobo‐Stark P, Shao X, Somlo S, Lin F, Igarashi P. Acute kidney injury and aberrant planar cell polarity induce cyst formation in mice lacking renal cilia. Hum Mol Genet 17: 1578‐1590, 2008.
 511. Paunescu TG, Ljubojevic M, Russo LM, Winter C, McLaughlin MM, Wagner CA, Breton S, Brown D. cAMP stimulates apical V‐ATPase accumulation, microvillar elongation, and proton extrusion in kidney collecting duct A‐intercalated cells. Am J Physiol Renal Physiol 298: F643‐F654, 2010.
 512. Pech V, Kim YH, Weinstein AM, Everett LA, Pham TD, Wall SM. Angiotensin II increases chloride absorption in the cortical collecting duct in mice through a pendrin‐dependent mechanism. Am J Physiol Renal Physiol 292: F914‐F920, 2007.
 513. Pech V, Zheng W, Pham TD, Verlander JW, Wall SM. Angiotensin II activates H+‐ATPase in type A intercalated cells. J Am Soc Nephrol 19: 84‐91, 2008.
 514. Pedersen NB, Hofmeister MV, Rosenbaek LL, Nielsen J, Fenton RA. Vasopressin induces phosphorylation of the thiazide‐sensitive sodium chloride cotransporter in the distal convoluted tubule. Kidney Int 78: 160‐169, 2010.
 515. Persson AE, Bachmann S. Constitutive nitric oxide synthesis in the kidney–functions at the juxtaglomerular apparatus. Acta Physiol Scand 169: 317‐324, 2000.
 516. Peter K. Untersuchungen über Bau und Entvicklung der Niere. Jena, Germany: Fisher, 1909.
 517. Peter Y, Goodenough D. Claudins. Curr Biol 14: R293‐R294, 2004.
 518. Peterson DR, Carone FA, Oparil S, Christensen EI. Differences between renal tubular processing of glucagon and insulin. Am J Physiol 242: F112‐F118, 1982.
 519. Peterson DR, Oparil S, Flouret G, Carone FA. Handling of angiotensin II and oxytocin by renal tubular segments perfused in vitro. Am J Physiol 232: F319‐F324, 1977.
 520. Peti‐Peterdi J. Independent two‐photon measurements of albumin GSC give low values. Am J Physiol Renal Physiol 296: F1255‐F1257, 2009.
 521. Peti‐Peterdi J, Sipos A. A high‐powered view of the filtration barrier. J Am Soc Nephrol 21: 1841, 2010.
 522. Peti‐Peterdi J, Warnock DG, Bell PD. Angiotensin II directly stimulates ENaC activity in the cortical collecting duct via AT(1) receptors. J Am Soc Nephrol 13: 1131‐1135, 2002.
 523. Pfaller W. Structure function correlation on rat kidney. Quantitative correlation of structure and function in the normal and injured rat kidney. Adv Anat Embryol Cell Biol 70: 1‐106, 1982.
 524. Pham PC, Devuyst O, Pham PT, Matsumoto N, Shih RN, Jo OD, Yanagawa N, Sun AM. Hypertonicity increases CLC‐5 expression in mouse medullary thick ascending limb cells. Am J Physiol Renal Physiol 287: F747‐F752, 2004.
 525. Picard N, Van AM, Campone C, Seiler M, Bloch‐Faure M, Hoenderop JG, Loffing J, Meneton P, Bindels RJ, Paillard M, Alhenc‐Gelas F, Houillier P. Tissue kallikrein‐deficient mice display a defect in renal tubular calcium absorption. J Am Soc Nephrol 16: 3602‐3610, 2005.
 526. Piepenhagen PA, Nelson WJ. Differential expression of cell‐cell and cell‐substratum adhesion proteins along the kidney nephron. Am J Physiol 269: C1433‐C1449, 1995.
 527. Piepenhagen PA, Peters LL, Lux SE, Nelson WJ. Differential expression of Na(+)‐K(+)‐ATPase, ankyrin, fodrin, and E‐cadherin along the kidney nephron. Am J Physiol 269: C1417‐C1432, 1995.
 528. Piwon N, Gunther W, Schwake M, Bosl MR, Jentsch TJ. CIC‐5 Cl‐ ‐channel disruption impairs endocytosis in a mouse model for Dent's disease. Nature 408: 369‐373, 2000.
 529. Planes C, Caughey GH. Regulation of the epithelial Na+ channel by peptidases. Curr Top Dev Biol 78: 23‐46, 2007.
 530. Plans V, Rickheit G, Jentsch TJ. Physiological roles of CLC Cl(‐)/H (+) exchangers in renal proximal tubules. Pflugers Arch 458: 23‐37, 2009.
 531. Pradervand S, Vandewalle A, Bens M, Gautschi I, Loffing J, Hummler E, Schild L, Rossier BC. Dysfunction of the epithelial sodium channel expressed in the kidney of a mouse model for Liddle syndrome. J Am Soc Nephrol 14: 2219‐2228, 2003.
 532. Praetorius HA, Spring KR. Bending the MDCK cell primary cilium increases intracellular calcium. J Membr Biol 184: 71‐79, 2001.
 533. Preston GM, Agre P. Isolation of the cDNA for erythrocyte integral membrane protein of 28 kilodaltons: Member of an ancient channel family. Proc Natl Acad Sci U S A 88: 11110‐11114, 1991.
 534. Preston GM, Carroll TP, Guggino WB, Agre P. Appearance of water channels in Xenopus oocytes expressing red cell CHIP28 protein. Science 256: 385‐387, 1992.
 535. Prozialeck WC, Lamar PC, Appelt DM. Differential expression of E‐cadherin, N‐cadherin and beta‐catenin in proximal and distal segments of the rat nephron. BMC Physiol 4: 10, 2004.
 536. Pullman TN, Oparil S, Carone FA. Fate of labeled angiotensin II microinfused into individual nephrons in the rat. Am J Physiol 228: 747‐751, 1975.
 537. Pushkin A, Kurtz I. SLC4 base (HCO3 ‐, CO3 2‐) transporters: Classification, function, structure, genetic diseases, and knockout models. Am J Physiol Renal Physiol 290: F580‐F599, 2006.
 538. Quamme GA, de RC. Epithelial magnesium transport and regulation by the kidney. Front Biosci 5: D694‐D711, 2000.
 539. Quentin F, Chambrey R, Trinh‐Trang‐Tan MM, Fysekidis M, Cambillau M, Paillard M, Aronson PS, Eladari D. The Cl‐/HCO3‐ exchanger pendrin in the rat kidney is regulated in response to chronic alterations in chloride balance. Am J Physiol Renal Physiol 287: F1179‐F1188, 2004.
 540. Quentin F, Eladari D, Cheval L, Lopez C, Goossens D, Colin Y, Cartron JP, Paillard M, Chambrey R. RhBG and RhCG, the putative ammonia transporters, are expressed in the same cells in the distal nephron. J Am Soc Nephrol 14: 545‐554, 2003.
 541. Quentin F, Eladari D, Frische S, Cambillau M, Nielsen S, Alper SL, Paillard M, Chambrey R. Regulation of the Cl‐/HCO3‐ exchanger AE2 in rat thick ascending limb of Henle's loop in response to changes in acid‐base and sodium balance. J Am Soc Nephrol 15: 2988‐2997, 2004.
 542. Rabkin R, Mahoney CA. Hormones and the kidney. In: Schrier RW, Gottschalk CW, editors. Diseases of the Kidney. Boston/Toronto: Little, Brown and Company, 1988, p. 309‐355.
 543. Rabkin R, Ross BD, Mako ME, Rubenstein AH. The handling of insulin, proinsulin, and C‐peptide by the isolated rat kidney. Diabetes 27 Suppl 1: 192‐196, 1978.
 544. Rahmoune H, Thompson PW, Ward JM, Smith CD, Hong G, Brown J. Glucose transporters in human renal proximal tubular cells isolated from the urine of patients with non‐insulin‐dependent diabetes. Diabetes 54: 3427‐3434, 2005.
 545. Raychowdhury R, Niles JL, McCluskey RT, Smith JA. Autoimmune target in Heymann nephritis is a glycoprotein with homology to the LDL receptor. Science 244: 1163‐1165, 1989.
 546. Reilly RF, Ellison DH. Mammalian distal tubule: Physiology, pathophysiology, and molecular anatomy. Physiol Rev 80: 277‐313, 2000.
 547. Reissinger A, Ludwig M, Utsch B, Promse A, Baulmann J, Weisser B, Vetter H, Kramer HJ, Bokemeyer D. Novel NCCT gene mutations as a cause of Gitelman's syndrome and a systematic review of mutant and polymorphic NCCT alleles. Kidney Blood Press Res 25: 354‐362, 2002.
 548. Remuzzi A, Sangalli F, Fassi A, Remuzzi G. Albumin concentration in the Bowman's capsule: Multiphoton microscopy vs micropuncture technique. Kidney Int 72: 1410‐1411, 2007.
 549. Renkema KY, Velic A, Dijkman HB, Verkaart S, van der Kemp AW, Nowik M, Timmermans K, Doucet A, Wagner CA, Bindels RJ, Hoenderop JG. The calcium‐sensing receptor promotes urinary acidification to prevent nephrolithiasis. J Am Soc Nephrol 20: 1705‐1713, 2009.
 550. Rhodin J. Anatomy of kidney tubules. Int Rev Cytol 7: 485‐534, 1948.
 551. Rhodin J. Correlations of Ultrastructural Organization and Function in Normal and Experimentally Changed Proximal Convoluted Tubule Cells of the Mouse Kidney. Stockholm: Aktiebolaget Godvil, 1954.
 552. Riccardi D, Brown EM. Physiology and pathophysiology of the calcium‐sensing receptor in the kidney. Am J Physiol Renal Physiol 298: F485‐F499, 2010.
 553. Riccardi D, Hall AE, Chattopadhyay N, Xu JZ, Brown EM, Hebert SC. Localization of the extracellular Ca2+/polyvalent cation‐sensing protein in rat kidney. Am J Physiol 274: F611‐F622, 1998.
 554. Ridderstrale Y, Kashgarian M, Koeppen B, Giebisch G, Stetson D, Ardito T, Stanton B. Morphological heterogeneity of the rabbit collecting duct. Kidney Int 34: 655‐670, 1988.
 555. Ritthaler T, Traebert M, Lotscher M, Biber J, Murer H, Kaissling B. Effects of phosphate intake on distribution of type II Na/Pi cotransporter mRNA in rat kidney. Kidney Int 55: 976‐983, 1999.
 556. Rodat‐Despoix L, Delmas P. Ciliar functions in the nephron. Pflugers Arch 458: 179‐187, 2009.
 557. Rodman JS, Kerjaschki D, Merisko E, Farquhar MG. Presence of an extensive clathrin coat on the apical plasmalemma of the rat kidney proximal tubule cell. J Cell Biol 98: 1630‐1636, 1984.
 558. Rodman JS, Mooseker M, Farquhar MG. Cytoskeletal proteins of the rat kidney proximal tubule brush border. Eur J Cell Biol 42: 319‐327, 1986.
 559. Roesinger B, Schiller A, Taugner R. A freeze‐fracture study of tight junctions in the pars convoluta and pars recta of the renal proximal tubule. Cell Tissue Res 186: 121‐133, 1978.
 560. Rogulski J, Pacanis A, Adamowicz W, Angielski S. On the mechanism of maleate action on rat kidney mitochondria. Effect on oxidative metabolism. Acta Biochim Pol 21: 403‐413, 1974.
 561. Rossier BC. Hormonal regulation of the epithelial sodium channel ENaC: N or P(o)? J Gen Physiol 120: 67‐70, 2002.
 562. Rossier BC, Pradervand S, Schild L, Hummler E. Epithelial sodium channel and the control of sodium balance: Interaction between genetic and environmental factors. Annu Rev Physiol 64: 877‐897, 2002.
 563. Rossier BC, Stutts MJ. Activation of the epithelial sodium channel (ENaC) by serine proteases. Annu Rev Physiol 71: 361‐379, 2009.
 564. Rostgaard J. Electron microscopy of filaments in the basal part of rat kidney tubule cells, and their in situ interaction with heavy meromyosin. Z Zellforsch Mikrosk Anat 132: 497‐521, 1972.
 565. Roth J, Taatjes DJ. Glycocalyx heterogeneity of rat kidney urinary tubule: Demonstration with a lectin‐gold technique specific for sialic acid. Eur J Cell Biol 39: 449‐457, 1985.
 566. Rothenberger F, Velic A, Stehberger PA, Kovacikova J, Wagner CA. Angiotensin II stimulates vacuolar H+ ‐ATPase activity in renal acid‐secretory intercalated cells from the outer medullary collecting duct. J Am Soc Nephrol 18: 2085‐2093, 2007.
 567. Royaux IE, Wall SM, Karniski LP, Everett LA, Suzuki K, Knepper MA, Green ED. Pendrin, encoded by the Pendred syndrome gene, resides in the apical region of renal intercalated cells and mediates bicarbonate secretion. Proc Natl Acad Sci U S A 98: 4221‐4226, 2001.
 568. Rubera I, Loffing J, Palmer LG, Frindt G, Fowler‐Jaeger N, Sauter D, Carroll T, McMahon A, Hummler E, Rossier BC. Collecting duct‐specific gene inactivation of alphaENaC in the mouse kidney does not impair sodium and potassium balance. J Clin Invest 112: 554‐565, 2003.
 569. Russo LM, Sandoval RM, McKee M, Osicka TM, Collins AB, Brown D, Molitoris BA, Comper WD. The normal kidney filters nephrotic levels of albumin retrieved by proximal tubule cells: Retrieval is disrupted in nephrotic states. Kidney Int 71: 504‐513, 2007.
 570. Rybak JN, Ettorre A, Kaissling B, Giavazzi R, Neri D, Elia G. In vivo protein biotinylation for identification of organ‐specific antigens accessible from the vasculature. Nat Methods 2: 291‐298, 2005.
 571. Sabolic I, Haase W, Burckhardt G. ATP‐dependent H+ pump in membrane vesicles from rat kidney cortex. Am J Physiol 248: F835‐F844, 1985.
 572. Sabolic I, Katsura T, Verbavatz JM, Brown D. The AQP2 water channel: Effect of vasopressin treatment, microtubule disruption, and distribution in neonatal rats. J Membr Biol 143: 165‐175, 1995.
 573. Sabolic I, Valenti G, Verbavatz J‐M, Van Hoek AN, Verkman AS, Ausiello DA, Brown D. Localization of the CHIP28 water channel in rat kidney. Am J Physiol Cell Physiol 263: C1225‐C1233, 1992.
 574. Sahali D, Mulliez N, Chatelet F, Dupuis R, Ronco P, Verroust P. Characterization of a 280‐kD protein restricted to the coated pits of the renal brush border and the epithelial cells of the yolk sac. Teratogenic effect of the specific monoclonal antibodies. J Exp Med 167: 213‐218, 1988.
 575. Saito A, Pietromonaco S, Loo AK, Farquhar MG. Complete cloning and sequencing of rat gp330/“megalin,” a distinctive member of the low density lipoprotein receptor gene family. Proc Natl Acad Sci U S A 91: 9725‐9729, 1994.
 576. Salido EC, Barajas L, Lechago J, Laborde MP, Fisher DA. Immunocytochemical localisation of epidermal growth factor in mouse kidney. J Histochem Cytochem 34: 1155‐1160, 1986.
 577. Salido EC, Lakshmanan J, Fisher DA, Shapiro LJ, Barajas L. Expression of epidermal growth factor in the rat kidney ‐ an immunocytochemical and in situ hybridization study. Histochemistry 96: 65‐72, 1991.
 578. San‐Cristobal P, Dimke H, Hoenderop JG, Bindels RJ. Novel molecular pathways in renal Mg2+ transport: A guided tour along the nephron. Curr Opin Nephrol Hypertens 19: 456‐462, 2010.
 579. Sandberg MB, Riquier AD, Pihakaski‐Maunsbach K, McDonough AA, Maunsbach AB. ANG II provokes acute trafficking of distal tubule Na+‐Cl(‐) cotransporter to apical membrane. Am J Physiol Renal Physiol 293: F662‐F669, 2007.
 580. Sands JM, Layton HE. The physiology of urinary concentration: An update. Semin Nephrol 29: 178‐195, 2009.
 581. Sangan P, Rajendran VM, Mann AS, Kashgarian M, Binder HJ. Regulation of colonic H‐K‐ATPase in large intestine and kidney by dietary Na depletion and dietary K depletion. Am J Physiol 272: C685‐C696, 1997.
 582. Satlin LM, Schwartz GJ. Cellular remodeling of HCO3(‐)‐secreting cells in rabbit renal collecting duct in response to an acidic environment. J Cell Biol 109: 1279‐1288, 1989.
 583. 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.
 584. Scheel O, Zdebik AA, Lourdel S, Jentsch TJ. Voltage‐dependent electrogenic chloride/proton exchange by endosomal CLC proteins. Nature 436: 424‐427, 2005.
 585. Schiebler TH, Danner KG. The effect of sex hormones on the proximal tubules in the rat kidney. Cell Tissue Res 192: 527‐549, 1978.
 586. Schild L. The epithelial sodium channel: From molecule to disease. Rev Physiol Biochem Pharmacol 151: 93‐107, 2004.
 587. Schiller A, Forssmann WG, Taugner R. The tight junctions of renal tubules in the cortex and outer medulla. Cell Tissue Res 212: 395‐413, 1980.
 588. Schiller A, Taugner R. Heterogeneity of tight junctions along the collecting duct in the renal medulla. A freeze‐fracture study in rat and rabbit. Cell Tissue Res 223: 603‐614, 1982.
 589. Schiller A, Taugner R, Kriz W. The thin limps of Henle's loop in the rabbit. Cell Tissue Res 207: 249‐265, 1980.
 590. Schiller A, Taugner R, Roesinger B. Comparative morphology of the zona occludens in the kidney tubules. Verh Anat Ges 229‐234, 1978.
 591. Schlatter E, Schurek HJ, Ziek R. Renal handling of homologous and heterologous insulin in the isolated perfused rat kidney. Pflügers Arch 393: 227‐231, 1982.
 592. Schlingmann KP, Gudermann T. A critical role of TRPM channel‐kinase for human magnesium transport. J Physiol 566: 301‐308, 2005.
 593. Schmitt R, Ellison DH, Farman N, Rossier BC, Reilly RF, Reeves WB, Oberbaumer I, Tapp R, Bachmann S. Developmental expression of sodium entry pathways in rat nephron. Am J Physiol 276: F367‐F381, 1999.
 594. Schmitt R, Kahl T, Mutig K, Bachmann S. Selectively reduced expression of thick ascending limb Tamm‐Horsfall protein in hypothyroid kidneys. Histochem Cell Biol 121: 319‐327, 2004.
 595. Schmitz C, Hilpert J, Jacobsen C, Boensch C, Christensen EI, Luft FC, Willnow TE. Megalin deficiency offers protection from renal aminoglycoside accumulation. J Biol Chem 277: 618‐622, 2002.
 596. Schneeberger EE, Lynch RD. The tight junction: A multifunctional complex. Am J Physiol Cell Physiol 286: C1213‐C1228, 2004.
 597. Schnermann J. Sodium transport deficiency and sodium balance in gene‐targeted mice. Acta Physiol Scand 173: 59‐66, 2001.
 598. Schnermann J, Chou CL, Ma T, Traynor T, Knepper MA, Verkman AS. Defective proximal tubular fluid reabsorption in transgenic aquaporin‐1 null mice. Proc Natl Acad Sci U S A 95: 9660‐9664, 1998.
 599. Schon DA, Backman KA, Hayslett JP. Role of the medullary collecting duct in potassium excretion in potassium‐adapted animals. Kidney Int 20: 655‐662, 1981.
 600. Schønheyder HC, Maunsbach AB. Ultrastructure of a specialized neck region in the rabbit nephron. Kidney Int 7: 145‐153, 1975.
 601. Schroter J, Timmermans G, Seyberth HW, Greven J, Bachmann S. Marked reduction of Tamm‐Horsfall protein synthesis in hyperprostaglandin E‐syndrome. Kidney Int 44: 401‐410, 1993.
 602. Schultheis PJ, Lorenz JN, Meneton P, Nieman ML, Riddle TM, Flagella M, Duffy JJ, Doetschman T, Miller ML, Shull GE. Phenotype resembling Gitelman's syndrome in mice lacking the apical Na+‐Cl‐ cotransporter of the distal convoluted tubule. J Biol Chem 273: 29150‐29155, 1998.
 603. Schurek HJ, Kriz W. Morphologic and functional evidence for oxygen deficiency in the isolated perfused rat kidney. Lab Invest 53: 145‐155, 1985.
 604. Schwartz GJ, Al‐Awqati Q. Carbon dioxide causes exocytosis of vesicles containing H+ pumps in isolated perfused proximal and collecting tubules. J Clin Invest 75: 1638‐1644, 1985.
 605. Schwartz GJ, Barasch J, Al Awqati Q. Plasticity of functional epithelial polarity. Nature 318: 368‐371, 1985.
 606. Schwartz GJ, Tsuruoka S, Vijayakumar S, Petrovic S, Mian A, Al‐Awqati Q. Acid incubation reverses the polarity of intercalated cell transporters, an effect mediated by hensin. J Clin Invest 109: 89‐99, 2002.
 607. Schwartz MM, Venkatachalam MA. Structural differences in thin limbs of Henle: Physiological implications. Kidney Int 6: 193‐208, 1974.
 608. Scott WN, Maude DL, Shehadeh I, Solomon AK. Transtubular movement of albumin in Necturus kidney. Am J Physiol 211: 1039‐1042, 1966.
 609. Seetharam B, Christensen EI, Moestrup SK, Hammond TG, Verroust PJ. Identification of rat yolk sac target protein of teratogenic antibodies, gp280, as intrinsic factor‐cobalamin receptor. J Clin Invest 99: 2317‐2322, 1997.
 610. Seetharam B, Levine JS, Ramasamy M, Alpers DH. Purification, properties, and immunochemical localization of a receptor for Intrinsic Factor‐Cobalamin complex in the rat kidney. J Biol Chem 263: 4443‐4449, 1988.
 611. Serafini‐Cessi F, Malagolini N, Cavallone D. Tamm‐Horsfall glycoprotein: Biology and clinical relevance. Am J Kidney Dis 42: 658‐676, 2003.
 612. Seshadri RM, Klein JD, Kozlowski S, Sands JM, Kim YH, Han KH, Handlogten ME, Verlander JW, Weiner ID. Renal expression of the ammonia transporters, Rhbg and Rhcg, in response to chronic metabolic acidosis. Am J Physiol Renal Physiol 290: F397‐F408, 2006.
 613. Seshadri RM, Klein JD, Smith T, Sands JM, Handlogten ME, Verlander JW, Weiner ID. Changes in subcellular distribution of the ammonia transporter, Rhcg, in response to chronic metabolic acidosis. Am J Physiol Renal Physiol 290: F1443‐F1452, 2006.
 614. Shen H, Smith DE, Yang T, Huang YG, Schnermann JB, Brosius FC III. Localization of PEPT1 and PEPT2 proton‐coupled oligopeptide transporter mRNA and protein in rat kidney. Am J Physiol 276: F658‐F665, 1999.
 615. Silbernagl S. The renal handling of amino acids and oligopeptides. Physiol Rev 68: 911‐1007, 1988.
 616. Simon DB, Lu Y, Choate KA, Velazquez H, Al‐Sabban E, Praga M, Casari G, Bettinelli A, Colussi G, Rodriguez‐Soriano J, McCredie D, Milford D, Sanjad S, Lifton RP. Paracellin‐1, a renal tight junction protein required for paracellular Mg2+ resorption. Science 285: 103‐106, 1999.
 617. Simons K. Membrane traffic in an epithelial cell line derived from the dog kidney. Kidney Int Suppl 23 32: S‐201‐S‐207, 1987.
 618. Skou JC. The influence of some cations on an adenosine triphosphatase from peripheral nerves. Biochim Biophys Acta 23: 394‐401, 1957.
 619. Skou JC. Preparation from mammallian brain and kidney of the enzyme system involved in active transport of Na ions and K ions. Biochim Biophys Acta 58: 314‐325, 1962.
 620. Smith AN, Jouret F, Bord S, Borthwick KJ, Al‐Lamki RS, Wagner CA, Ireland DC, Cormier‐Daire V, Frattini A, Villa A, Kornak U, Devuyst O, Karet FE. Vacuolar H+‐ATPase d2 subunit: Molecular characterization, developmental regulation, and localization to specialized proton pumps in kidney and bone. J Am Soc Nephrol 16: 1245‐1256, 2005.
 621. Smith HW. The Kidney: Structure and Function in Health and Disease. New York: Oxford University Press, 1951.
 622. Snyder PM. Minireview: Regulation of epithelial Na+ channel trafficking. Endocrinology 146: 5079‐5085, 2005.
 623. Sohara E, Uchida S, Sasaki S. Function of aquaporin‐7 in the kidney and the male reproductive system. Handb Exp Pharmacol 219‐231, 2009.
 624. Speller AM, Moffat DB. Tubulo‐vascular relationships in the developing kidney. J Anat 123: 487‐500, 1977.
 625. Stanton BA, Kaissling B. Adaptation of distal tubule and collecting duct to increased Na delivery. II. Na+ and K+ transport. Am J Physiol 255: F1269‐F1275, 1988.
 626. Stanton BA, Kaissling B. Regulation of renal ion transport and cell growth by sodium. Am J Physiol 257: F1‐10, 1989.
 627. Star RA, Burg MB, Knepper MA. Bicarbonate secretion and chloride absorption by rabbit cortical collecting ducts. Role of chloride/bicarbonate exchange. J Clin Invest 76: 1123‐1130, 1985.
 628. Stehberger PA, Schulz N, Finberg KE, Karet FE, Giebisch G, Lifton RP, Geibel JP, Wagner CA. Localization and regulation of the ATP6V0A4 (a4) vacuolar H+‐ATPase subunit defective in an inherited form of distal renal tubular acidosis. J Am Soc Nephrol 14: 3027‐3038, 2003.
 629. Stehberger PA, Shmukler BE, Stuart‐Tilley AK, Peters LL, Alper SL, Wagner CA. Distal renal tubular acidosis in mice lacking the AE1 (band3) Cl‐/HCO3‐ exchanger (slc4a1). J Am Soc Nephrol 18: 1408‐1418, 2007.
 630. Stetler‐Stevenson MA, Flouret G, Nakamura S, Gulczynski B, Carone FA. In vivo metabolism of tritiated LHRH by the whole kidney and individual tubules of rat. Am J Physiol 244: F628‐F632, 1983.
 631. Stetson DL, Wade JB, Giebisch G. Morphologic alterations in the rat medullary collecting duct following potassium depletion. Kidney Int 17: 45‐56, 1980.
 632. Stoessel A, Himmerkus N, Bleich M, Bachmann S, Theilig F. Connexin 37 is localized in renal epithelia and responds to changes in dietary salt intake. Am J Physiol Renal Physiol 298: F216‐F223, 2010.
 633. Storm T, Emma F, Verroust PJ, Hertz JM, Nielsen R, Christensen EI. Report of a cubilin deficient patient. N Engl J Med 346: 89‐91, 2011.
 634. Stoward PJ, Spicer SS, Miller RL. Histochemical reactivity of peanut lectin‐horseradish peroxidase conjugate. J Histochem Cytochem 28: 979‐990, 1980.
 635. Strange K, Spring KR. Methods for imaging renal tubule cells. Kidney Int 30: 192‐200, 1986.
 636. Straus W. Isolation and biochemical properties of droplets from the cells of rat kidney. J Biol Chem 207: 745‐755, 1954.
 637. Straus W. Concentration of acid phosphatase, ribonuclease, desoxyribonuclease, beta‐glucuronidase and cathepsin in “droplets” isolated from the kidney cells of normal rats. J Biophys Biochem Cytol 2: 513‐521, 1956.
 638. Straus W. Changes in “droplet” fractions from rat kidney cells after intraperitoneal injection of egg white. J Biophys Biochem Cytol 3: 933‐947, 1957.
 639. Straus W. Rapid cytochemical identification of phagosomes in various tissues of the rat and their differentiation from mitochondria by the peroxidase method. J Biophys Biochem Cytol 5: 193‐213, 1959.
 640. Straus W. Cytochemical investigation of phagosomes and related structures in cryostat sections of the kidney and liver of rats after intravenous administration of horseradish peroxidase. Exp Cell Res 27: 80‐94, 1962.
 641. Straus W. Cytochemical observations on the relationship between lysosomes and phagosomes in kidney and liver by combined staining for acid phosphatase and intravenously injected horseradish peroxidase. J Cell Biol 20: 497‐507, 1964.
 642. Straus W. Cytochemical observations on the relationship between lysosomes and phagosomes in kidney and liver by combined staining for acid phosphatase and intravenously injected horseradish peroxidase. 1964. J Am Soc Nephrol 12: 2528‐2537, 2001.
 643. Subramanya AR, Ellison DH. Sorting out lysosomal trafficking of the thiazide‐sensitive Na‐Cl Co‐transporter. J Am Soc Nephrol 21: 7‐9, 2010.
 644. Sun AM, Liu Y, Dworkin LD, Tse CM, Donowitz M, Yip KP. Na+/H+ exchanger isoform 2 (NHE2) is expressed in the apical membrane of the medullary thick ascending limb. J Membr Biol 160: 85‐90, 1997.
 645. Sun‐Wada GH, Murata Y, Namba M, Yamamoto A, Wada Y, Futai M. Mouse proton pump ATPase C subunit isoforms (C2‐a and C2‐b) specifically expressed in kidney and lung. J Biol Chem 278: 44843‐44851, 2003.
 646. Tago K, Schuster VL, Stokes JB. Stimulation of chloride transport by HCO3‐CO2 in rabbit cortical collecting tubule. Am J Physiol 251: F49‐F56, 1986.
 647. Tang VW, Goodenough DA. Paracellular ion channel at the tight junction. Biophys J 84: 1660‐1673, 2003.
 648. Tanner GA. Glomerular sieving coefficient of serum albumin in the rat: A two‐photon microscopy study. Am J Physiol Renal Physiol 296: F1258‐F1265, 2009.
 649. Tatum R, Zhang Y, Salleng K, Lu Z, Lin JJ, Lu Q, Jeansonne BG, Ding L, Chen YH. Renal salt wasting and chronic dehydration in claudin‐7‐deficient mice. Am J Physiol Renal Physiol 298: F24‐F34, 2010.
 650. Teng‐umnuay P, Verlander JW, Yuan W, Tisher CC, Madsen KM. Identification of distinct subpopulations of intercalated cells in the mouse collecting duct. J Am Soc Nephrol 7: 260‐274, 1996.
 651. Terris J, Ecelbarger CA, Marples D, Knepper MA, Nielsen S. Distribution of aquaporin‐4 water channel expression within rat kidney. Am J Physiol 269: F775‐F785, 1995.
 652. Thoenes W. Fine structure of lipid granules in proximal tubule cells of mouse kidney. J Cell Biol 12: 433‐437, 1962.
 653. Thoenes W, Langer KH. Die endocytose‐phase der eiweissresorption im proximalen nierebtubulus. Virchows Arch B Cell Pathol Incl Mol Pathol 2: 361‐379, 1969.
 654. Thomas CP, Itani OA. New insights into epithelial sodium channel function in the kidney: Site of action, regulation by ubiquitin ligases, serum‐ and glucocorticoid‐inducible kinase and proteolysis. Curr Opin Nephrol Hypertens 13: 541‐548, 2004.
 655. Thomson RB, Aronson PS. Immunolocalization of Ksp‐cadherin in the adult and developing rabbit kidney. Am J Physiol 277: F146‐F156, 1999.
 656. Tian W, Salanova M, Xu H, Lindsley JN, Oyama TT, Anderson S, Bachmann S, Cohen DM. Renal expression of osmotically responsive cation channel TRPV4 is restricted to water‐impermeant nephron segments. Am J Physiol Renal Physiol 287: F17‐F24, 2004.
 657. Tisher CC, Bulger RE, Trump BF. Human renal ultrastructure. I. Proximal tubule of healthy individuals. Lab Invest 15: 1357‐1394, 1966.
 658. Tisher CC, Bulger RE, Trump BF. Human renal ultrastructure. 3. The distal tubule in healthy individuals. Lab Invest 18: 655‐668, 1968.
 659. Tisher CC, Bulger RE, Valtin H. Morphology of renal medulla in water diuresis and vasopressin‐induced antidiuresis. Am J Physiol 220: 87‐94, 1971.
 660. Tisher CC, Kokko JP. Relationship between peritubular oncotic pressure gradients and morphology in isolated proximal tubules. Kidney Int 6: 146‐156, 1974.
 661. Tisher CC, Rosen S, Osborne GB. Ultrastructure of the proximal tubule of the rhesus monkey kidney. Am J Pathol 56: 469‐517, 1969.
 662. Topala CN, Bindels RJ, Hoenderop JG. Regulation of the epithelial calcium channel TRPV5 by extracellular factors. Curr Opin Nephrol Hypertens 16: 319‐324, 2007.
 663. Traebert M, Roth J, Biber J, Murer H, Kaissling B. Internalization of proximal tubular type II Na‐P(i) cotransporter by PTH: Immunogold electron microscopy. Am J Physiol Renal Physiol 278: F148‐F154, 2000.
 664. Traebert M, Volkl H, Biber J, Murer H, Kaissling B. Luminal and contraluminal action of 1‐34 and 3‐34 PTH peptides on renal type IIa Na‐P(i) cotransporter. Am J Physiol Renal Physiol 278: F792‐F798, 2000.
 665. Trenchev P, Dorling J, Webb J, Holborow EJ. Localization of smooth muscle‐like contractile proteins in kidney by immunoelectron microscopy. J Anat 121: 85‐95, 1976.
 666. Trinh‐Trang‐Tan MM, Bouby N, Kriz W, Bankir L. Functional adaptation of thick ascending limb and internephron heterogeneity to urine concentration. Kidney Int 31: 549‐555, 1987.
 667. Trump BF, Bulger RE. Morphology of the Kidney. In: Becker EL, editor. Structural Basis of Renal Disease. New York: Hoeber, 1968, p. 1‐92.
 668. Tsuganezawa H, Kobayashi K, Iyori M, Araki T, Koizumi A, Watanabe S, Kaneko A, Fukao T, Monkawa T, Yoshida T, Kim DK, Kanai Y, Endou H, Hayashi M, Saruta T. A new member of the HCO3(‐) transporter superfamily is an apical anion exchanger of beta‐intercalated cells in the kidney. J Biol Chem 276: 8180‐8189, 2001.
 669. Tsukita S, Furuse M. Claudin‐based barrier in simple and stratified cellular sheets. Curr Opin Cell Biol 14: 531‐536, 2002.
 670. Tsukita S, Furuse M, Itoh M. Multifunctional strands in tight junctions. Nat Rev Mol Cell Biol 2: 285‐293, 2001.
 671. Tsuruoka S, Schwartz GJ. Adaptation of the outer medullary collecting duct to metabolic acidosis in vitro. Am J Physiol 275: F982‐F990, 1998.
 672. Turksen K. Wasted salts and wasted bodies: New insight into the role of claudin 7 in the kidney. Am J Physiol Renal Physiol 298: F22‐F23, 2010.
 673. Uchida S, Sasaki S, Furukawa T, Hiraoka M, Imai T, Hirata Y, Marumo F. Molecular cloning of a chloride channel that is regulated by dehydration and expressed predominantly in kidney medulla. J Biol Chem 268: 3821‐3824, 1993.
 674. Uchida S, Sasaki S, Nitta K, Uchida K, Horita S, Nihei H, Marumo F. Localization and functional characterization of rat kidney‐specific chloride channel, ClC‐K1. J Clin Invest 95: 104‐113, 1995.
 675. Unwin RJ, Capasso G, Shirley DG. An overview of divalent cation and citrate handling by the kidney. Nephron Physiol 98: 15‐20, 2004.
 676. Usuda N, Usman MI, Reddy MK, Hashimoto T, Reddy JK, Rao MS. Immunocytochemical localization of urate oxidase, fatty acyl‐CoA oxidase and catalase in bovine kidney peroxisomes. J Histochem Cytochem 36: 253‐258, 1988.
 677. Vallet M, Picard N, Loffing‐Cueni D, Fysekidis M, Bloch‐Faure M, Deschenes G, Breton S, Meneton P, Loffing J, Aronson PS, Chambrey R, Eladari D. Pendrin regulation in mouse kidney primarily is chloride‐dependent. J Am Soc Nephrol 17: 2153‐2163, 2006.
 678. Vallon V, Schroth J, Lang F, Kuhl D, Uchida S. Expression and phosphorylation of the Na+‐Cl‐ cotransporter NCC in vivo is regulated by dietary salt, potassium, and SGK1. Am J Physiol Renal Physiol 297: F704‐F712, 2009.
 679. Valtin H. Physiological effects of vasopressin on the kidney. In: Gash DM, Boer GJ, editors. Vasopressin, Principles and Properties. New York: Plenum, 1987, p. 369‐387.
 680. van der Lubbe N, Lim CH, Fenton RA, Meima ME, Jan Danser AH, Zietse R, Hoorn EJ. Angiotensin II induces phosphorylation of the thiazide‐sensitive sodium chloride cotransporter independent of aldosterone. Kidney Int 79: 66‐76, 2011.
 681. van Deurs B, Christensen EI. Endocytosis in kidney proximal tubule cells and cultured fibroblasts: A review of the structural aspect of membrane recycling between the plasma membrane and endocytic vacuoles. Eur J Cell Biol 33: 163‐173, 1984.
 682. Van Itallie CM, Anderson JM. The molecular physiology of tight junction pores. Physiology (Bethesda) 19: 331‐338, 2004.
 683. Van Itallie CM, Anderson JM. Claudins and epithelial paracellular transport. Annu Rev Physiol 68: 403‐429, 2006.
 684. Van Itallie CM, Fanning AS, Bridges A, Anderson JM. ZO‐1 stabilizes the tight junction solute barrier through coupling to the perijunctional cytoskeleton. Mol Biol Cell 20: 3930‐3940, 2009.
 685. Van Itallie CM, Holmes J, Bridges A, Anderson JM. Claudin‐2‐dependent changes in noncharged solute flux are mediated by the extracellular domains and require attachment to the PDZ‐scaffold. Ann N Y Acad Sci 1165: 82‐87, 2009.
 686. Venkatachalam MA, Karnovsky MJ. Extravascular protein in the kidney. An ultrastructural study of its relation to renal peritubular capillary permeability using protein tracers. Lab Invest 27: 435‐444, 1972.
 687. Verkman AS. Lessons on renal physiology from transgenic mice lacking aquaporin water channels. J Am Soc Nephrol 10: 1126‐1135, 1999.
 688. Verkman AS, Shi LB, Frigeri A, Hasegawa H, Farinas J, Mitra A, Skach W, Brown D, Van Hoek AN, Ma T. Structure and function of kidney water channels. Kidney Int 48: 1069‐1081, 1995.
 689. Verlander JW, Hassell KA, Royaux IE, Glapion DM, Wang ME, Everett LA, Green ED, Wall SM. Deoxycorticosterone upregulates PDS (Slc26a4) in mouse kidney: Role of pendrin in mineralocorticoid‐induced hypertension. Hypertension 42: 356‐362, 2003.
 690. Verlander JW, Kim YH, Shin W, Pham TD, Hassell KA, Beierwaltes WH, Green ED, Everett L, Matthews SW, Wall SM. Dietary Cl(‐) restriction upregulates pendrin expression within the apical plasma membrane of type B intercalated cells. Am J Physiol Renal Physiol 291: F833‐F839, 2006.
 691. Verlander JW, Madsen KM, Larsson L, Cannon JK, Tisher CC. Immunocytochemical localization of intracellular acidic compartments: Rat proximal nephron. Am J Physiol 257: F454‐F462, 1989.
 692. Verlander JW, Madsen KM, Tisher CC. Effect of acute respiratory acidosis on two populations of intercalated cells in rat cortical collecting duct. Am J Physiol 253: F1142‐F1156, 1987.
 693. Verlander JW, Madsen KM, Tisher CC. Axial distribution of band 3‐positive intercalated cells in the collecting duct of control and ammonium chloride‐loaded rabbits. Kidney Int Suppl 57: S137‐S147.: S137‐S147, 1996.
 694. Verlander JW, Miller RT, Frank AE, Royaux IE, Kim YH, Weiner ID. Localization of the ammonium transporter proteins RhBG and RhCG in mouse kidney. Am J Physiol Renal Physiol 284: F323‐F337, 2003.
 695. Verlander JW, Moudy RM, Campbell WG, Cain BD, Wingo CS. Immunohistochemical localization of H‐K‐ATPase alpha(2c)‐subunit in rabbit kidney. Am J Physiol Renal Physiol 281: F357‐F365, 2001.
 696. Verlander JW, Tran TM, Zhang L, Kaplan MR, Hebert SC. Estradiol enhances thiazide‐sensitive NaCl cotransporter density in the apical plasma membrane of the distal convoluted tubule in ovariectomized rats. J Clin Invest 101: 1661‐1669, 1998.
 697. Verrey F, Loffing J, Zecevic M, Heitzmann D, Staub O. SGK1: Aldosterone‐induced relay of Na+ transport regulation in distal kidney nephron cells. Cell Physiol Biochem 13: 21‐28, 2003.
 698. Verrey F, Singer D, Ramadan T, Vuille‐dit‐Bille RN, Mariotta L, Camargo SM. Kidney amino acid transport. Pflugers Arch 458: 53‐60, 2009.
 699. Verroust PJ, Birn H, Nielsen R, Kozyraki R, Christensen EI. The tandem endocytic receptors megalin and cubilin are important proteins in renal pathology. Kidney Int 62: 745‐756, 2002.
 700. Vijayakumar S, Takito J, Gao X, Schwartz GJ, Al‐Awqati Q. Differentiation of columnar epithelia: The hensin pathway. J Cell Sci 119: 4797‐4801, 2006.
 701. Villa‐Bellosta R, Ravera S, Sorribas V, Stange G, Levi M, Murer H, Biber J, Forster IC. The Na+‐Pi cotransporter PiT‐2 (SLC20A2) is expressed in the apical membrane of rat renal proximal tubules and regulated by dietary Pi. Am J Physiol Renal Physiol 296: F691‐F699, 2009.
 702. Vio CP, Cespedes C, Gallardo P, Masferrer JL. Renal identification of cyclooxygenase‐2 in a subset of thick ascending limb cells. Hypertension 30: 687‐692, 1997.
 703. Vio CP, Figueroa CD. Evidence for a stimulatory effect of high potassium diet on renal kallikrein. Kidney Int 31: 1327‐1334, 1987.
 704. Voets T, Nilius B, Hoefs S, van der Kemp AW, Droogmans G, Bindels RJ, Hoenderop JG. TRPM6 forms the Mg2+ influx channel involved in intestinal and renal Mg2+ absorption. J Biol Chem 279: 19‐25, 2004.
 705. Voldstedlund M, Thuneberg L, Tranum‐Jensen J, Vinten J, Christensen EI. Caveolae, caveolin and cav‐p60 in smooth muscle and renin‐producing cells in the rat kidney. Acta Physiol Scand 179: 179‐188, 2003.
 706. von Möllendorf W. Der Exkretionsapparat. Handbuch der Mikroskopischen Anatomie des Menschen. In: von Möllendorf W, Bargmann W, editors. Berlin: Springer, 1930, p. 1.
 707. Wade JB, Lee AJ, Liu J, Ecelbarger CA, Mitchell C, Bradford AD, Terris J, Kim GH, Knepper MA. UT‐A2: A 55‐kDa urea transporter in thin descending limb whose abundance is regulated by vasopressin. Am J Physiol Renal Physiol 278: F52‐F62, 2000.
 708. Wade JB, O'Neil RG, Pryor JL, Boulpaep EL. Modulation of cell membrane area in renal collecting tubules by corticosteroid hormones. J Cell Biol 81: 439‐445, 1979.
 709. Wade JB, Stanton BA, Brown D. Structural correlates of transport in distal tubule and collecting duct segments. In: Windhager EE, editor. Handbook of Physiology. Renal Physiology. Am. Physiol. Soc. New York: Oxford University Press, 1992, sect. 8, p. 169‐226.
 710. Wade JB, Welling PA, Donowitz M, Shenolikar S, Weinman EJ. Differential renal distribution of NHERF isoforms and their colocalization with NHE3, ezrin, and ROMK. Am J Physiol Cell Physiol 280: C192‐C198, 2001.
 711. Wagner CA. Metabolic acidosis: New insights from mouse models. Curr Opin Nephrol Hypertens 16: 471‐476, 2007.
 712. Wagner CA, Devuyst O, Bourgeois S, Mohebbi N. Regulated acid‐base transport in the collecting duct. Pflugers Arch 458: 137‐156, 2009.
 713. Wagner CA, Finberg KE, Breton S, Marshansky V, Brown D, Geibel JP. Renal vacuolar H+‐ATPase. Physiol Rev 84: 1263‐1314, 2004.
 714. Wagner CA, Finberg KE, Stehberger PA, Lifton RP, Giebisch GH, Aronson PS, Geibel JP. Regulation of the expression of the Cl‐/anion exchanger pendrin in mouse kidney by acid‐base status. Kidney Int 62: 2109‐2117, 2002.
 715. Wall SM, Mehta P, DuBose TD Jr. Dietary K+ restriction upregulates total and Sch‐28080‐sensitive bicarbonate absorption in rat tIMCD. Am J Physiol 275: F543‐F549, 1998.
 716. Wang SS, Devuyst O, Courtoy PJ, Wang XT, Wang H, Wang Y, Thakker RV, Guggino S, Guggino WB. Mice lacking renal chloride channel, CLC‐5, are a model for Dent's disease, a nephrolithiasis disorder associated with defective receptor‐mediated endocytosis. Hum Mol Genet 9: 2937‐2945, 2000.
 717. Wang T, Giebisch G. Effects of angiotensin II on electrolyte transport in the early and late distal tubule in rat kidney. Am J Physiol 271: F143‐F149, 1996.
 718. Wang W. Renal potassium channels: Recent developments. Curr Opin Nephrol Hypertens 13: 549‐555, 2004.
 719. Wang WH. Regulation of ROMK (Kir1.1) channels: New mechanisms and aspects. Am J Physiol Renal Physiol 290: F14‐F19, 2006.
 720. Wang XY, Masilamani S, Nielsen J, Kwon TH, Brooks HL, Nielsen S, Knepper MA. The renal thiazide‐sensitive Na‐Cl cotransporter as mediator of the aldosterone‐escape phenomenon. J Clin Invest 108: 215‐222, 2001.
 721. Wang Y, Cai H, Cebotaru L, Hryciw DH, Weinman EJ, Donowitz M, Guggino SE, Guggino WB. ClC‐5: Role in endocytosis in the proximal tubule. Am J Physiol Renal Physiol 289: F850‐F862, 2005.
 722. Warnock DG. Liddle syndrome: Genetics and mechanisms of Na+ channel defects. Am J Med Sci 322: 302‐307, 2001.
 723. Warnock DG, Rossier BC. Renal sodium handling: The role of the epithelial sodium channel. J Am Soc Nephrol 16: 3151‐3153, 2005.
 724. Waugh D, Prentice RS, Yadav D. The structure of the proximal tubule: A morphological study of basement membrane cristae and their relationships in the renal tubule of the rat. Am J Anat 121: 775‐785, 1967.
 725. Wehrli P, Loffing‐Cueni D, Kaissling B, Loffing J. Replication of segment‐specific and intercalated cells in the mouse renal collecting system. Histochem Cell Biol 127: 389‐398, 2007.
 726. Welling D, Urani J, Welling L, Wagner E. Fractal analysis and imaging of the proximal nephron cell. Am J Physiol 270: C953‐C963, 1996.
 727. Welling LW, Evan AP, Welling DJ. Shape of cells and extracellular channels in rabbit cortical collecting ducts. Kidney Int 20: 211‐222, 1981.
 728. Welling LW, Evan AP, Welling DJ, Gattone VH III. Morphometric comparison of rabbit cortical connecting tubules and collecting ducts. Kidney Int 23: 358‐367, 1983.
 729. Welling LW, Grantham JJ. Physical properties of isolated perfused renal tubules and tubular basement membranes. J Clin Invest 51: 1063‐1075, 1972.
 730. Welling LW, Welling DJ. Surface areas of brush border and lateral cell walls in the rabbit proximal nephron. Kidney Int 8: 343‐348, 1975.
 731. Welling LW, Welling DJ. Shape of epithelial cells and intercellular channels in the rabbit proximal nephron. Kidney Int 9: 385‐394, 1976.
 732. Welling LW, Welling DJ, Hill JJ. Shape of cells and intercellular channels in rabbit thick ascending limp of Henle. Kidney Int 13: 144‐151, 1978.
 733. Welling LW, Welling DJ, Holsapple JW, Evan AP. Morphometric analysis of distinct microanatomy near the base of proximal tubule cells. Am J Physiol 253: F126‐F140, 1987.
 734. Wilcox CS, Welch WJ. Macula densa nitric oxide synthase: Expression, regulation, and function. Kidney Int Suppl 67: S53‐S57, 1998.
 735. Wilcox ER, Burton QL, Naz S, Riazuddin S, Smith TN, Ploplis B, Belyantseva I, Ben‐Yosef T, Liburd NA, Morell RJ, Kachar B, Wu DK, Griffith AJ, Riazuddin S, Friedman TB. Mutations in the gene encoding tight junction claudin‐14 cause autosomal recessive deafness DFNB29. Cell 104: 165‐172, 2001.
 736. Will C, Fromm M, Muller D. Claudin tight junction proteins: Novel aspects in paracellular transport. Perit Dial Int 28: 577‐584, 2008.
 737. Wilson FH, Disse‐Nicodeme S, Choate KA, Ishikawa K, Nelson‐Williams C, Desitter I, Gunel M, Milford DV, Lipkin GW, Achard JM, Feely MP, Dussol B, Berland Y, Unwin RJ, Mayan H, Simon DB, Farfel Z, Jeunemaitre X, Lifton RP. Human hypertension caused by mutations in WNK kinases. Science 293: 1107‐1112, 2001.
 738. Wilson FH, Kahle KT, Sabath E, Lalioti MD, Rapson AK, Hoover RS, Hebert SC, Gamba G, Lifton RP. Molecular pathogenesis of inherited hypertension with hyperkalemia: The Na‐Cl cotransporter is inhibited by wild‐type but not mutant WNK4. Proc Natl Acad Sci U S A 100: 680‐684, 2003.
 739. Wingo CS, Madsen KM, Smolka A, Tisher CC. H‐K‐ATPase immunoreactivity in cortical and outer medullary collecting duct. Kidney Int 38: 985‐990, 1990.
 740. Wingo CS, Smolka AJ. Function and structure of H‐K‐ATPase in the kidney. Am J Physiol 269: F1‐F16, 1995.
 741. Winter C, Schulz N, Giebisch G, Geibel JP, Wagner CA. Nongenomic stimulation of vacuolar H+‐ATPases in intercalated renal tubule cells by aldosterone. Proc Natl Acad Sci U S A 101: 2636‐2641, 2004.
 742. Wittner M, Di SA, Mandon B, Roinel N, de Rouffignac C. Stimulation of NaCl reabsorption by antidiuretic hormone in the cortical thick ascending limb of Henle's loop of the mouse. Pflugers Arch 419: 212‐214, 1991.
 743. Yang B, Bankir L. Urea and urine concentrating ability: New insights from studies in mice. Am J Physiol Renal Physiol 288: F881‐F896, 2005.
 744. Yang CL, Angell J, Mitchell R, Ellison DH. WNK kinases regulate thiazide‐sensitive Na‐Cl cotransport. J Clin Invest 111: 1039‐1045, 2003.
 745. Yang LE, Sandberg MB, Can AD, Pihakaski‐Maunsbach K, McDonough AA. Effects of dietary salt on renal Na+ transporter subcellular distribution, abundance, and phosphorylation status. Am J Physiol Renal Physiol 295: F1003‐F1016, 2008.
 746. Yang T, Hassan S, Huang YG, Smart AM, Briggs JP, Schnermann JB. Expression of PTHrP, PTH/PTHrP receptor, and Ca(2+)‐sensing receptor mRNAs along the rat nephron. Am J Physiol 272: F751‐F758, 1997.
 747. Yoder BK, Hou X, Guay‐Woodford LM. The polycystic kidney disease proteins, polycystin‐1, polycystin‐2, polaris, and cystin, are co‐localized in renal cilia. J Am Soc Nephrol 13: 2508‐2516, 2002.
 748. Yokota S, Kato K. The heterogeneity of rat kidney lysosomes revealed by immunoelectron microscopic staining for cathepsins B and H. Histochemistry 89: 499‐504, 1988.
 749. Yokota S, Nishimura Y, Kato K. Localization of cathepsin L in rat kidney revealed by immunoenzyme and immunogold techniques. Histochemistry 90: 277‐283, 1988.
 750. Yokota S, Tsuji H, Kato K. Immunocytochemical localization of cathepsin H in rat kidney. Histochemistry 85: 223‐230, 1986.
 751. Younes‐Ibrahim M, Barlet‐Bas C, Buffin‐Meyer B, Cheval L, Rajerison R, Doucet A. Ouabain‐sensitive and ‐insensitive K‐ATPases in rat nephron: Effect of K depletion. Am J Physiol 268: F1141‐F1147, 1995.
 752. Yu AS, Enck AH, Lencer WI, Schneeberger EE. Claudin‐8 expression in Madin‐Darby canine kidney cells augments the paracellular barrier to cation permeation. J Biol Chem 278: 17350‐17359, 2003.
 753. Yun J, Kenney RA. Preparation of cat kidney tissue for ultrastructural studies. J Electron Microsc (Tokyo) 25: 11‐23, 1976.
 754. Zaar K, Hartig F, Fahimi HD, Gorgas K. Peroxisomal aggregates forming large stacks in the lipid segment of the canine kidney. Acta Histochem Suppl 29: 165‐168, 1984.
 755. Zabel M, Schiebler TH. Histochemical, autoradiographic and electron microscopic investigations of the renal proximal tubule of male and female rats after castration. Histochemistry 69: 255‐276, 1980.
 756. Zampighi G, Kreman M. Intercellular fibrillar skeleton in the basal interdigitations of kidney tubular cells. J Membr Biol 88: 33‐43, 1985.
 757. Zeller J. On the cytochemistry of lysosomes in the rat kidney under normal and experimental conditions. Histochemie 35: 235‐262, 1973.
 758. Zhai XY, Birn H, Jensen KB, Thomsen JS, Andreasen A, Christensen EI. Digital three‐dimensional reconstruction and ultrastructure of the mouse proximal tubule. J Am Soc Nephrol 14: 611‐619, 2003.
 759. Zhai XY, Fenton RA, Andreasen A, Thomsen JS, Christensen EI. Aquaporin‐1 is not expressed in descending thin limbs of short‐loop nephrons. J Am Soc Nephrol 18: 2937‐2944, 2007.
 760. Zhai XY, Kristoffersen IB, Christensen EI. Immunocytochemistry of renal membrane proteins on epoxy sections. Kidney Int72: 731‐735, 2007.
 761. Zhai XY, Thomsen JS, Birn H, Kristoffersen IB, Andreasen A, Christensen EI. Three‐dimensional reconstruction of the mouse nephron. J Am Soc Nephrol 17: 77‐88, 2006.
 762. Zhang Q, Taulman PD, Yoder BK. Cystic kidney diseases: All roads lead to the cilium. Physiology (Bethesda) 19: 225‐230, 2004.
 763. Zhou B, Zhuang J, Gu D, Wang H, Cebotaru L, Guggino WB, Cai H. WNK4 enhances the degradation of NCC through a sortilin‐mediated lysosomal pathway. J Am Soc Nephrol 21: 82‐92, 2010.
 764. Zimmermann KW. Ueber den bau des glomerulus der saugerniere. Z Mikrosk Anat Forsch 32: 176‐278, 1933.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Erik Ilsø Christensen, Carsten A. Wagner, Brigitte Kaissling. Uriniferous Tubule: Structural and Functional Organization. Compr Physiol 2012, 2: 805-861. doi: 10.1002/cphy.c100073