Comprehensive Physiology Wiley Online Library

Renal Medullary Circulation

Full Article on Wiley Online Library



Abstract

The renal medullary microcirculation is a distinctive arrangement of blood vessels that serve multiple functions in the renal medulla. This article begins with a description of the unique anatomy of this vascular bed and the role it plays in transport and countercurrent exchange in the medulla. A segment of the review is then devoted to the important role mathematical modeling has played in the understanding of this vascular bed's function. Succeeding sections focus upon the hematocrit in the vasa recta capillaries and methods utilized to assess blood flow in the renal medulla. An extensive portion of the article is then devoted to the regulation of the medullary circulation, from ion channel architecture to neurohormonal signaling. Finally, we discuss the importance of the renal medullary circulation in the regulation of fluid and electrolyte homeostasis and arterial blood pressure regulation. © 2012 American Physiological Society. Compr Physiol 2:97‐140, 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.

Microcirculation of the renal cortex and medulla. Within the renal cortex, interlobular arteries, derived from the arcuate artery, ascend toward the cortical surface. Superficial and midcortical glomeruli arise at obtuse and right angles, while juxtamedullary glomeruli arise at an acute, recurrent angle from the interlobular artery. The majority of blood flow to the medulla arises from juxtamedullary efferent arterioles. A minor fraction might also be derived from periglomerular shunt pathways. In the outer stripe of the outer medulla (OM), juxtamedullary efferent arterioles give rise to descending vasa recta (DVR) that coalesce with ascending vasa recta (AVR), and sometimes, thin descending limbs of Henle, to form vascular bundles. Vascular bundles are the prominent feature of the inner stripe of the OM. DVR on the periphery of vascular bundles perfuse the interbundle capillary plexus that supplies nephrons (thick‐ascending limbs, collecting ducts (CD), long looped thin descending limbs, not shown). DVR in the center of the bundles continue across the inner‐outer medullary junction to perfuse the inner medulla (IM). In some species, thin descending limbs of short‐looped nephrons migrate toward or become associated with vascular bundles. In the inner medulla, vascular bundles disappear and vasa recta become dispersed with thin loops of Henle and CDs. Blood from the interbundle capillary plexus is returned without rejoining vascular bundles. DVR have a continuous endothelium (inset) and are surrounded by contractile pericytes. Like cortical peritubular capillaries, the AVR endothelium is highly fenestrated. As blood flows toward the papillary tip, NaCl and urea diffuse into DVR and out of AVR. Transmural gradients of NaCl and urea drive water efflux across the DVR wall via aquaporin‐1 water channels.

Reproduced, with permission, from Pallone et al. 514.
Figure 2. Figure 2.

The intra‐arterial cushion. A longitudinal section through an afferent arteriole shows an intra‐arterial cushion at its origin from the interlobular artery (inset, × 160). The cushion protrudes into the lumen of the parent vessel. Smooth muscle cells of the cushion are embedded in a copious matrix (× 4000). Intra‐arterial cushions might affect the relative volume fraction of RBCs vs plasma (hematocrit) that is directed from intralobular arterioles to juxtamedullary glomeruli and renal medulla. It is also conceivable that they regulate the relative distribution of blood flow between the superficial and juxtamedullary cortex (see text).

Reproduced, with permission, from Moffat and Creasey 433.
Figure 3. Figure 3.

Structure and transition of cortical and medullary vessels. The proximal afferent arteriole is composed of at least two layers of smooth muscle cells. The muscularity and size of cortical efferent arterioles differ with location. Note the difference between the superficial and juxtamedullary efferent arterioles. The juxtamedullary efferent arteriole is larger, has a thicker, multilayered media, and its endothelium is composed of more numerous endothelial cells. In the illustration, a descending vas rectum (DVR) in a vascular bundle is adjacent to three fenestrated ascending vasa recta (AVR). The DVR wall is surrounded by a contractile pericyte. At the bottom right, DVR and AVR from the inner medulla are shown. Inner medullary DVR have a continuous endothelium through most of their length as pericytes become scarcer with medullary depth. Terminal DVR and the entire AVR wall are fenestrated.

Reproduced, with permission, from Jamison and Kriz 291.
Figure 4. Figure 4.

Distribution of descending vasa recta (DVR) pericytes. (A and B) Immunofluorescent staining of DVR pericytes using anti α‐smooth muscle actin as primary antibody. The pericytes are present on DVR from outer medullary vascular bundles (Panel A) and those from the inner medulla (Panel B) (1000 ×). (C) Low power image of immunofluorescent staining of DVR pericytes using anti α‐smooth muscle actin antibody. Some vessels show pericytes throughout their length to the papillary tip. Black vessels are filled with India ink (× 100).

Reproduced, with permission, from Park et al. 529.
Figure 5. Figure 5.

Electron micrograph of descending vasa recta (DVR) and ascending vasa recta (AVR). Electron micrograph of DVR and AVR in rat vascular bundles. DVR have a continuous endothelium and AVR are fenestrated. Note the minimal interstitium that exists between vessels in this region (× 12,400).

Reproduced, with permission, from Pallone et al. 516.
Figure 6. Figure 6.

Arterial injection of Psammomys obesus. (A) Photograph of the microvasculature of the desert rodent Psammomys obesus obtained by injecting the arteries of the kidney with Microfil and digesting the tissue. The distinct arteriolar patterns of the cortex, outer and inner medulla are apparent. In Psammomys, the separation of the outer medulla into vascular bundles and the dense capillary plexus of the interbundle region (*) is striking because vasa recta coalesce into giant vascular bundles. OM, outer medulla; IM, inner medulla. Designations on the original figure are C = cortex, TR = transitional region (outer stripe of the outer medulla), IS = inner stripe of the outer medulla, IZ = inner zone (inner medulla).

Reproduced, with permission, from Bankir et al.. (B and C) India ink injection study of vascular bundles in the outer medulla of the rat. In contrast to Psammomys, individual vascular bundles do not coalesce into giant bundles. The bundles are more evenly dispersed throughout the inner stripe of the outer medulla. This pattern is typical of many mammalian species including the rat, mouse and human. Reproduced, with permission, from Bankir et al., Moffat and Fourman) (29, 33).
Figure 7. Figure 7.

Tubular‐vascular relationships in the outer medulla (OM). Organization of the inner stripe of the OM. Top and bottom panels show longitudinal and cross‐sectional views, respectively. The extent to which the thin descending limbs of Henle (tDLH) of short looped nephrons associate with vascular bundles varies between species. In the rabbit, no association exists, whereas in the rat and mouse, the tDLH migrates to the periphery or becomes incorporated within vascular bundles, respectively. Abbreviations: VB, vascular bundle; IB, interbundle region; CD, collecting duct; 1 and 2, thin descending and thick ascending limbs of long‐looped nephrons; 3 and 4, thin descending and thick ascending limbs of short‐looped nephrons.

Reproduced, with permission, from Bankir and de Rouffignac 29.
Figure 8. Figure 8.

Renal medullary interstitial cells (RMICs). RMICs appear to be tethered between thin limbs and vasa recta in the inner medulla. Interstitial spaces lie between the cells and the cells are stacked like rungs of a ladder. RMICs are contractile and secrete vasoactive paracrine agents (see text). The stacked arrangement of RMICs in some species has been suggested to help retard axial diffusion that would otherwise tend to dissipate corticomedullary gradients of NaCl and urea. AVR, venous or ascending vasa recta. Arrows point to lipid droplets within RMIC.

Reproduced, with permission, from Lemley and Kriz 355.
Figure 9. Figure 9.

Tubular vascular relationships and collecting duct clusters in the inner medulla. (A) A CD cluster (blue) is surrounded by descending vasa recta (DVR) (red) and thin DLH (green). Neither DVR nor thin DLH are incorporated into the center of the cluster. (B) A CD cluster (blue) has both ascending vasa recta (AVR) (red) and thin ALH (green) in its surroundings and within the cluster. (C) Idealized cross‐sectional depiction of a CD cluster (blue) in relationship to AVR (red) and thin ALH (green). AVR and thin ALH are diffusely distributed around and within the CD cluster. (D) Idealized cross‐sectional depiction of thin DLH (purple) and DVR (aqua) surrounding open regions in which CD clusters reside.

Reproduced, with permission, from Pannabecker and Dantzler 522.
Figure 10. Figure 10.

Relationships of vasa recta and loops of Henle to collecting duct clusters. (A) Three‐dimensional reconstruction of single CD segment (blue) with multiple ascending vasa recta (AVR) (red). Top: 90° axial rotation of segments shown in adjacent panels. Scale bar, 100 μm. (B) Electron micrographs showing transverse sections of CDs and AVR from 1.5 mm (i, ii, and iv) and 4 mm (iii) below the base of the IM. i: CD surrounded by 4 AVR (*). Other tubular structures surrounding the CD are ATLs. Scale bar, 10 μm. ii: AVR abuts CD with minimal direct contact. Scale bar, 1 μm. iii: AVR abuts CD with microvillus (arrow). IS, interstitium. Scale bar, 1 μm. iv: AVR abuts CD with microvilli (arrows). Scale bar, 1 μm.

Reproduced, with permission, from Pannabecker and Dantzler 522.
Figure 11. Figure 11.

Hemodynamic effects on intrarenal oxygenation. Intrarenal oxygen tension (pO2) was measured in the cortex and medulla with a microelectrode. Cortical pO2 falls and medullary pO2 increases during an episode of hypotension induced by either hemorrhage, aortic ligation, or nitroprusside infusion (Panel A). Inhibition of transport in the thick ascending limb of Henle with a loop diuretic (Panel B) increases basal pO2 in the medulla (compare to Panel A) and eliminates the effect of hypotension to raise medullary pO2. Inhibition of vasodilatory prostaglandins and nitric oxide, or blockade of adenosine receptors reduces basal pO2 in the medulla and accentuates the increase in pO2 caused by hypotension (Panel C). Intrarenal tissue pO2 decreases with medullary depth (Panel D). * indicates significance from baseline (P < .001).

Reproduced, with permission, from Brezis et al. and Brezis et al. 55,56.
Figure 12. Figure 12.

Vasa recta solute permeabilities. [14C]urea permeability (PU, ordinate) vs 22Na permeability (PNa, abscissa) is shown for outer medullary descending vasa recta (DVR) (OMDVR) isolated from Sprague‐Dawley rats and perfused in vitro. Results are also shown for inner medullary DVR and ascending vasa recta (AVR) (IMDVR, IMAVR) perfused on the surface of the exposed papilla of Munich‐Wistar rats in vivo. The dashed line is identity. PU and PNa are highly correlated and nearly equal in inner medullary vasa recta. In contrast, PU of outer medullary DVR is always very high and is not correlated with PNa. The dissociation of PNa and PU in OMDVR results (at least in part) from the expression of the UTB‐facilitated urea carrier. In separate experiments (not shown), PU of OMDVR was inhibited by exposure to urea analogues or phloretin.

Data reproduced, with permission, from Pallone et al. 516.
Figure 13. Figure 13.

Osmotic water permeability (Pf) of outer medullary descending vasa recta (DVR). (A) Pf was measured in glutaraldehyde fixed rat DVR by measuring the rate of transmural water flux generated by a bath > lumen NaCl gradient. Sequential measurements in controls were stable. In contrast, exposure to p‐chloromercuribenzene sulfonate (pCMBS, 2 mM), an agent that covalently binds to cysteine residues on aquaporin‐1, reduced Pf to nearly zero. In these experiments, glutaraldehyde fixation was necessary to prevent deterioration of the vessel caused by pCMBS and other harsh conditions of the experiment. (B) Pf was measured in AQP1 null (−/−) or replete (+/+) murine DVR by transmural gradients of NaCl, urea, glucose or raffinose. When NaCl was the solute used to drive water flux, deletion of AQP1 reduced Pf from ∼1100 μm/s to nearly zero. Water flux driven by raffinose (MW 564) was markedly reduced by AQP1 deletion (compare AQP1 −/− to +/+), but remained unexpectedly high. Similarly, glucose (MW 180) and urea (MW 60) gradients drove measurable water flux across AQP1 (−/−) DVR.

Reproduced, with permission, from Pallone et al. 501,504.
Figure 14. Figure 14.

UTB and urea recycling in the medulla. Schematic of vascular and tubular urea recycling in the kidney. Short and long loops of Henle and vasa recta are shown. The UTA2 urea transporter is expressed in the thin descending limbs of Henle. The UTB urea transporter is expressed in descending vasa recta (DVR) endothelium and red blood cells (not shown). Thin descending limbs of short‐looped nephrons become associated with vascular bundles (see Fig. 9) so that urea recycling from thin limbs to DVR via UTA2 and UTB is accommodated. UTB is not expressed by the ascending vasa recta (AVR) endothelium but AVR are fenestrated and urea permeability is high. Thus urea in AVR plasma and RBCs can readily recycle back to DVR in vascular bundles using UTB in the RBC membrane and DVR endothelium. The UTA1, A3 and A4 collecting duct urea transporters conduct urea from the lumen to the inner medullary interstitium. C, cortex; OS, outer stripe of outer medulla; IS, inner stripe of outer medulla; IM, inner medulla.

Reproduced, with permission, from Yang and Bankir 706.
Figure 15. Figure 15.

Effect of AQP1 deletion on predictions of renal medullary interstitial osmolality. A mathematical simulation of the renal medulla was solved to predict interstitial osmolality. Interstitial osmolality is shown as a function of corticomedullary axis (x/liter = 0 is the corticomedullary junction, x/liter = 1 is the papillary tip). Various curves denote predictions for different values of Pf [descending vasa recta (DVR) osmotic water permeability]. Pf was varied between 0 (equivalent to AQP1 deletion) and 2000 μm/s. AQP1 expression in DVR is predicted to enhance concentrating ability by conducting water flux from DVR to interstitium where it is then taken up by ascending vasa recta. The net result is a secondary reduction of blood flow in the deepest regions of the inner medulla (papillary tip).

Reproduced, with permission, from Pallone et al. 501.
Figure 16. Figure 16.

Predicted oxygen tension (PO2) profiles in the rat outer medullary interstitium. The structural organization of the OM is represented by means of four concentric regions centered on a vascular bundle: an innermost region containing the central vascular bundle (R1), where long descending vasa recta are sequestered; a peripheral region of the vascular bundle (R2); a region neighboring the vascular bundle (R3), where most thick ascending limbs reside; and the region most distant from the vascular bundle (R4), where collecting ducts are located. Vertical dotted lines mark the boundary between the outer stripe (OS) and the inner stripe (IS), and x/liter denotes the ratio of the axial coordinate to total length of outer medulla. Model suggests that the OM anatomy has a significant impact on the radial distribution of oxygen. It preserves O2 delivery to the inner medulla, but limits active Na+ reabsorption across medullary thick ascending limbs and thereby decreases the OM urinary concentrating capacity.

Reproduced, with permission, from Chen et al. 89.
Figure 17. Figure 17.

Distribution of hematocrit in the kidney. 51Cr‐RBCs and 131I‐IgM (plasma volume marker) were simultaneously infused into the kidney. An equilibration period of either 1 or 10 min followed before ligation of the renal artery and vein. The distribution of RBCs and plasma was inferred by measuring activity of the isotopes in tissue and dividing their ratio by the systemic ratio. Results show that the hematocrit of inner medullary blood is lower than that of either whole kidney, cortex, or outer medulla.

Data redrawn, with permission, from Rasmussen 561.
Figure 18. Figure 18.

Connexin staining in descending vasa recta. Immunostaining of Cx40. Immunostaining is shown with antibody directed against α‐smooth muscle actin (SMA red) or Cx40 (green). A merged image is shown along with a corresponding white light micrograph. Cx40 showed linear staining confined to the endothelium with very little SMA colocalization. Bar = 10 μm.

Reproduced, with permission, from Zhang et al. 725.
Figure 19. Figure 19.

Effect of arginine vasopressin (AVP) on inner medullary blood flow and urine osmolality. To control plasma vasopressin concentrations, decerebrate rats were infused with AVP. Increasing AVP concentration within the physiological range caused a reduction of inner medullary blood flow and an improvement in urinary concentration. * indicates significance from control period (P < .05).

Reproduced, with permission, from Franchini and Cowley 189.
Figure 20. Figure 20.

In vitro microperfusion. (A) An afferent arteriole is cannulated with concentric pipettes and perfused toward the glomerulus.

Reproduced, with permission, from Hansen et al. (B and C) Perfusion of an isolated descending vasa recta (DVR). Images obtained before (B) and after (C) exposure to angiotensin II (AngII). Compare the thick smooth muscle layer of the afferent arteriole to the sporadically distributed pericyte cell bodies of DVR. Reproduced, with permission, from Hansen et al. and Pallone et al. 233,511.
Figure 21. Figure 21.

Arachidonic acid (AA) metabolites. (A) Abbreviated schematic showing signaling molecules generated from arachadonic acid by cyclo‐oxygenase (COX). Prostaglandins (PG) and thromboxanes (Tx) are vasoactive end products. B. Abbreviated schematic showing signaling molecules generated from the cytochrome P450 metabolism of AA. Hydrolases generate the potent constrictor 20‐hydroxyeicosatetraenoic acid (20‐HETE) and epoxygenases generate the epoxyeicosatrienoic acids that most often function as vasodilators. The lipoxygenase pathway of arachidonic acid metabolism is not shown.

Reproduced, with permission, from Pallone and Cao 499.
Figure 22. Figure 22.

Effect of intrarenal infusion of adenosine receptor A1 or A2 subtype agonists. Top and bottom panels show the effect of adenosine agonist infusions on intrarenal blood flow. Cortical and medullary measurements were obtained using laser‐Doppler flowmetry with optical fibers placed on the kidney surface or inserted into the renal parenchyma, respectively. Left and right panels show the respective effects of either A1 or A2 receptor stimulation with subtype‐specific agonists. At time = 0, the A1 agonist N6‐cyclopentyladenosine (left panels) or the A2 agonist CGS‐21680C (right panels) was transiently infused (1 min) into the renal parenchyma. The A1 agonist transiently reduced both cortical and medullary blood flow, while the A2 agonist caused a preferential increase in blood flow to the medulla.

Reproduced, with permission, from Agmon et al. 5.
Figure 23. Figure 23.

Autoregulation in different regions of the kidney. An electromagnetic flow device on the renal artery was used to measure total renal blood flow (RBF). Laser‐Doppler flow probes were inserted into the renal parenchyma at various depths to measure regional blood flow in the outer and inner medulla (IM). Total RBF and cortical tissue blood flow shows intact autoregulation (stability of blood flow over a range of perfusion pressure). In contrast, in these volume‐expanded rats (see text), the small fraction of blood flow that reaches the outer or IM is not autoregulated.

Reproduced, with permission, from Mattson et al. 410.
Figure 24. Figure 24.

Pressure natriuresis. (A) Medullary blood flow is autoregulated in hydropenic but not volume expanded rats. (B) Renal interstitial hydrostatic pressure (RIHP) is higher and increases to a greater degree with renal perfusion pressure in volume expanded animals. (C) When renal perfusion pressure is increased, urinary sodium excretion (UNaV) increases much more markedly in volume expanded than in hydropenic animals.

Reproduced, with permission, from Cowley 108.
Figure 25. Figure 25.

Acute NOS inhibition in the renal medulla. Line graph (left) showing changes in renal cortical and medullary blood flow (measured by laser‐Doppler flowmetry) in response to renal medullary interstitial infusion of the nitric oxide synthase inhibitor NG‐nitro‐l‐arginine methyl ester (L‐NAME, 120 μg/h). Bar graph (right) shows changes in urine volume (UVol) and sodium excretion (UNaV) accompanying medullary interstitial infusion of L‐NAME in the infused kidney and the noninfused contralateral kidney. * indicates significance from control (P < 0.05).

Reproduced, with permission, from Mattson et al. 415.
Figure 26. Figure 26.

Chronic influence of renal medullary interstitial infusion of the nitric oxide synthase inhibitor NG‐nitro‐l‐arginine methyl ester (L‐NAME, 8.6 mg/kg/day) on renal medullary blood flow (top), daily sodium balance (middle), and mean arterial blood pressure (bottom) in conscious Sprague Dawley rats. Vertical‐hashed marks indicate the L‐NAME infusion period. * indicates significance from control (P < 0.05).

Reproduced, with permission, from Mattson et al. 409.
Figure 27. Figure 27.

Hypothetical mechanisms whereby changes in renal medullary perfusion may lead to an alteration in arterial blood pressure.

Reproduced, with permission, from Mattson 404.


Figure 1.

Microcirculation of the renal cortex and medulla. Within the renal cortex, interlobular arteries, derived from the arcuate artery, ascend toward the cortical surface. Superficial and midcortical glomeruli arise at obtuse and right angles, while juxtamedullary glomeruli arise at an acute, recurrent angle from the interlobular artery. The majority of blood flow to the medulla arises from juxtamedullary efferent arterioles. A minor fraction might also be derived from periglomerular shunt pathways. In the outer stripe of the outer medulla (OM), juxtamedullary efferent arterioles give rise to descending vasa recta (DVR) that coalesce with ascending vasa recta (AVR), and sometimes, thin descending limbs of Henle, to form vascular bundles. Vascular bundles are the prominent feature of the inner stripe of the OM. DVR on the periphery of vascular bundles perfuse the interbundle capillary plexus that supplies nephrons (thick‐ascending limbs, collecting ducts (CD), long looped thin descending limbs, not shown). DVR in the center of the bundles continue across the inner‐outer medullary junction to perfuse the inner medulla (IM). In some species, thin descending limbs of short‐looped nephrons migrate toward or become associated with vascular bundles. In the inner medulla, vascular bundles disappear and vasa recta become dispersed with thin loops of Henle and CDs. Blood from the interbundle capillary plexus is returned without rejoining vascular bundles. DVR have a continuous endothelium (inset) and are surrounded by contractile pericytes. Like cortical peritubular capillaries, the AVR endothelium is highly fenestrated. As blood flows toward the papillary tip, NaCl and urea diffuse into DVR and out of AVR. Transmural gradients of NaCl and urea drive water efflux across the DVR wall via aquaporin‐1 water channels.

Reproduced, with permission, from Pallone et al. 514.


Figure 2.

The intra‐arterial cushion. A longitudinal section through an afferent arteriole shows an intra‐arterial cushion at its origin from the interlobular artery (inset, × 160). The cushion protrudes into the lumen of the parent vessel. Smooth muscle cells of the cushion are embedded in a copious matrix (× 4000). Intra‐arterial cushions might affect the relative volume fraction of RBCs vs plasma (hematocrit) that is directed from intralobular arterioles to juxtamedullary glomeruli and renal medulla. It is also conceivable that they regulate the relative distribution of blood flow between the superficial and juxtamedullary cortex (see text).

Reproduced, with permission, from Moffat and Creasey 433.


Figure 3.

Structure and transition of cortical and medullary vessels. The proximal afferent arteriole is composed of at least two layers of smooth muscle cells. The muscularity and size of cortical efferent arterioles differ with location. Note the difference between the superficial and juxtamedullary efferent arterioles. The juxtamedullary efferent arteriole is larger, has a thicker, multilayered media, and its endothelium is composed of more numerous endothelial cells. In the illustration, a descending vas rectum (DVR) in a vascular bundle is adjacent to three fenestrated ascending vasa recta (AVR). The DVR wall is surrounded by a contractile pericyte. At the bottom right, DVR and AVR from the inner medulla are shown. Inner medullary DVR have a continuous endothelium through most of their length as pericytes become scarcer with medullary depth. Terminal DVR and the entire AVR wall are fenestrated.

Reproduced, with permission, from Jamison and Kriz 291.


Figure 4.

Distribution of descending vasa recta (DVR) pericytes. (A and B) Immunofluorescent staining of DVR pericytes using anti α‐smooth muscle actin as primary antibody. The pericytes are present on DVR from outer medullary vascular bundles (Panel A) and those from the inner medulla (Panel B) (1000 ×). (C) Low power image of immunofluorescent staining of DVR pericytes using anti α‐smooth muscle actin antibody. Some vessels show pericytes throughout their length to the papillary tip. Black vessels are filled with India ink (× 100).

Reproduced, with permission, from Park et al. 529.


Figure 5.

Electron micrograph of descending vasa recta (DVR) and ascending vasa recta (AVR). Electron micrograph of DVR and AVR in rat vascular bundles. DVR have a continuous endothelium and AVR are fenestrated. Note the minimal interstitium that exists between vessels in this region (× 12,400).

Reproduced, with permission, from Pallone et al. 516.


Figure 6.

Arterial injection of Psammomys obesus. (A) Photograph of the microvasculature of the desert rodent Psammomys obesus obtained by injecting the arteries of the kidney with Microfil and digesting the tissue. The distinct arteriolar patterns of the cortex, outer and inner medulla are apparent. In Psammomys, the separation of the outer medulla into vascular bundles and the dense capillary plexus of the interbundle region (*) is striking because vasa recta coalesce into giant vascular bundles. OM, outer medulla; IM, inner medulla. Designations on the original figure are C = cortex, TR = transitional region (outer stripe of the outer medulla), IS = inner stripe of the outer medulla, IZ = inner zone (inner medulla).

Reproduced, with permission, from Bankir et al.. (B and C) India ink injection study of vascular bundles in the outer medulla of the rat. In contrast to Psammomys, individual vascular bundles do not coalesce into giant bundles. The bundles are more evenly dispersed throughout the inner stripe of the outer medulla. This pattern is typical of many mammalian species including the rat, mouse and human. Reproduced, with permission, from Bankir et al., Moffat and Fourman) (29, 33).


Figure 7.

Tubular‐vascular relationships in the outer medulla (OM). Organization of the inner stripe of the OM. Top and bottom panels show longitudinal and cross‐sectional views, respectively. The extent to which the thin descending limbs of Henle (tDLH) of short looped nephrons associate with vascular bundles varies between species. In the rabbit, no association exists, whereas in the rat and mouse, the tDLH migrates to the periphery or becomes incorporated within vascular bundles, respectively. Abbreviations: VB, vascular bundle; IB, interbundle region; CD, collecting duct; 1 and 2, thin descending and thick ascending limbs of long‐looped nephrons; 3 and 4, thin descending and thick ascending limbs of short‐looped nephrons.

Reproduced, with permission, from Bankir and de Rouffignac 29.


Figure 8.

Renal medullary interstitial cells (RMICs). RMICs appear to be tethered between thin limbs and vasa recta in the inner medulla. Interstitial spaces lie between the cells and the cells are stacked like rungs of a ladder. RMICs are contractile and secrete vasoactive paracrine agents (see text). The stacked arrangement of RMICs in some species has been suggested to help retard axial diffusion that would otherwise tend to dissipate corticomedullary gradients of NaCl and urea. AVR, venous or ascending vasa recta. Arrows point to lipid droplets within RMIC.

Reproduced, with permission, from Lemley and Kriz 355.


Figure 9.

Tubular vascular relationships and collecting duct clusters in the inner medulla. (A) A CD cluster (blue) is surrounded by descending vasa recta (DVR) (red) and thin DLH (green). Neither DVR nor thin DLH are incorporated into the center of the cluster. (B) A CD cluster (blue) has both ascending vasa recta (AVR) (red) and thin ALH (green) in its surroundings and within the cluster. (C) Idealized cross‐sectional depiction of a CD cluster (blue) in relationship to AVR (red) and thin ALH (green). AVR and thin ALH are diffusely distributed around and within the CD cluster. (D) Idealized cross‐sectional depiction of thin DLH (purple) and DVR (aqua) surrounding open regions in which CD clusters reside.

Reproduced, with permission, from Pannabecker and Dantzler 522.


Figure 10.

Relationships of vasa recta and loops of Henle to collecting duct clusters. (A) Three‐dimensional reconstruction of single CD segment (blue) with multiple ascending vasa recta (AVR) (red). Top: 90° axial rotation of segments shown in adjacent panels. Scale bar, 100 μm. (B) Electron micrographs showing transverse sections of CDs and AVR from 1.5 mm (i, ii, and iv) and 4 mm (iii) below the base of the IM. i: CD surrounded by 4 AVR (*). Other tubular structures surrounding the CD are ATLs. Scale bar, 10 μm. ii: AVR abuts CD with minimal direct contact. Scale bar, 1 μm. iii: AVR abuts CD with microvillus (arrow). IS, interstitium. Scale bar, 1 μm. iv: AVR abuts CD with microvilli (arrows). Scale bar, 1 μm.

Reproduced, with permission, from Pannabecker and Dantzler 522.


Figure 11.

Hemodynamic effects on intrarenal oxygenation. Intrarenal oxygen tension (pO2) was measured in the cortex and medulla with a microelectrode. Cortical pO2 falls and medullary pO2 increases during an episode of hypotension induced by either hemorrhage, aortic ligation, or nitroprusside infusion (Panel A). Inhibition of transport in the thick ascending limb of Henle with a loop diuretic (Panel B) increases basal pO2 in the medulla (compare to Panel A) and eliminates the effect of hypotension to raise medullary pO2. Inhibition of vasodilatory prostaglandins and nitric oxide, or blockade of adenosine receptors reduces basal pO2 in the medulla and accentuates the increase in pO2 caused by hypotension (Panel C). Intrarenal tissue pO2 decreases with medullary depth (Panel D). * indicates significance from baseline (P < .001).

Reproduced, with permission, from Brezis et al. and Brezis et al. 55,56.


Figure 12.

Vasa recta solute permeabilities. [14C]urea permeability (PU, ordinate) vs 22Na permeability (PNa, abscissa) is shown for outer medullary descending vasa recta (DVR) (OMDVR) isolated from Sprague‐Dawley rats and perfused in vitro. Results are also shown for inner medullary DVR and ascending vasa recta (AVR) (IMDVR, IMAVR) perfused on the surface of the exposed papilla of Munich‐Wistar rats in vivo. The dashed line is identity. PU and PNa are highly correlated and nearly equal in inner medullary vasa recta. In contrast, PU of outer medullary DVR is always very high and is not correlated with PNa. The dissociation of PNa and PU in OMDVR results (at least in part) from the expression of the UTB‐facilitated urea carrier. In separate experiments (not shown), PU of OMDVR was inhibited by exposure to urea analogues or phloretin.

Data reproduced, with permission, from Pallone et al. 516.


Figure 13.

Osmotic water permeability (Pf) of outer medullary descending vasa recta (DVR). (A) Pf was measured in glutaraldehyde fixed rat DVR by measuring the rate of transmural water flux generated by a bath > lumen NaCl gradient. Sequential measurements in controls were stable. In contrast, exposure to p‐chloromercuribenzene sulfonate (pCMBS, 2 mM), an agent that covalently binds to cysteine residues on aquaporin‐1, reduced Pf to nearly zero. In these experiments, glutaraldehyde fixation was necessary to prevent deterioration of the vessel caused by pCMBS and other harsh conditions of the experiment. (B) Pf was measured in AQP1 null (−/−) or replete (+/+) murine DVR by transmural gradients of NaCl, urea, glucose or raffinose. When NaCl was the solute used to drive water flux, deletion of AQP1 reduced Pf from ∼1100 μm/s to nearly zero. Water flux driven by raffinose (MW 564) was markedly reduced by AQP1 deletion (compare AQP1 −/− to +/+), but remained unexpectedly high. Similarly, glucose (MW 180) and urea (MW 60) gradients drove measurable water flux across AQP1 (−/−) DVR.

Reproduced, with permission, from Pallone et al. 501,504.


Figure 14.

UTB and urea recycling in the medulla. Schematic of vascular and tubular urea recycling in the kidney. Short and long loops of Henle and vasa recta are shown. The UTA2 urea transporter is expressed in the thin descending limbs of Henle. The UTB urea transporter is expressed in descending vasa recta (DVR) endothelium and red blood cells (not shown). Thin descending limbs of short‐looped nephrons become associated with vascular bundles (see Fig. 9) so that urea recycling from thin limbs to DVR via UTA2 and UTB is accommodated. UTB is not expressed by the ascending vasa recta (AVR) endothelium but AVR are fenestrated and urea permeability is high. Thus urea in AVR plasma and RBCs can readily recycle back to DVR in vascular bundles using UTB in the RBC membrane and DVR endothelium. The UTA1, A3 and A4 collecting duct urea transporters conduct urea from the lumen to the inner medullary interstitium. C, cortex; OS, outer stripe of outer medulla; IS, inner stripe of outer medulla; IM, inner medulla.

Reproduced, with permission, from Yang and Bankir 706.


Figure 15.

Effect of AQP1 deletion on predictions of renal medullary interstitial osmolality. A mathematical simulation of the renal medulla was solved to predict interstitial osmolality. Interstitial osmolality is shown as a function of corticomedullary axis (x/liter = 0 is the corticomedullary junction, x/liter = 1 is the papillary tip). Various curves denote predictions for different values of Pf [descending vasa recta (DVR) osmotic water permeability]. Pf was varied between 0 (equivalent to AQP1 deletion) and 2000 μm/s. AQP1 expression in DVR is predicted to enhance concentrating ability by conducting water flux from DVR to interstitium where it is then taken up by ascending vasa recta. The net result is a secondary reduction of blood flow in the deepest regions of the inner medulla (papillary tip).

Reproduced, with permission, from Pallone et al. 501.


Figure 16.

Predicted oxygen tension (PO2) profiles in the rat outer medullary interstitium. The structural organization of the OM is represented by means of four concentric regions centered on a vascular bundle: an innermost region containing the central vascular bundle (R1), where long descending vasa recta are sequestered; a peripheral region of the vascular bundle (R2); a region neighboring the vascular bundle (R3), where most thick ascending limbs reside; and the region most distant from the vascular bundle (R4), where collecting ducts are located. Vertical dotted lines mark the boundary between the outer stripe (OS) and the inner stripe (IS), and x/liter denotes the ratio of the axial coordinate to total length of outer medulla. Model suggests that the OM anatomy has a significant impact on the radial distribution of oxygen. It preserves O2 delivery to the inner medulla, but limits active Na+ reabsorption across medullary thick ascending limbs and thereby decreases the OM urinary concentrating capacity.

Reproduced, with permission, from Chen et al. 89.


Figure 17.

Distribution of hematocrit in the kidney. 51Cr‐RBCs and 131I‐IgM (plasma volume marker) were simultaneously infused into the kidney. An equilibration period of either 1 or 10 min followed before ligation of the renal artery and vein. The distribution of RBCs and plasma was inferred by measuring activity of the isotopes in tissue and dividing their ratio by the systemic ratio. Results show that the hematocrit of inner medullary blood is lower than that of either whole kidney, cortex, or outer medulla.

Data redrawn, with permission, from Rasmussen 561.


Figure 18.

Connexin staining in descending vasa recta. Immunostaining of Cx40. Immunostaining is shown with antibody directed against α‐smooth muscle actin (SMA red) or Cx40 (green). A merged image is shown along with a corresponding white light micrograph. Cx40 showed linear staining confined to the endothelium with very little SMA colocalization. Bar = 10 μm.

Reproduced, with permission, from Zhang et al. 725.


Figure 19.

Effect of arginine vasopressin (AVP) on inner medullary blood flow and urine osmolality. To control plasma vasopressin concentrations, decerebrate rats were infused with AVP. Increasing AVP concentration within the physiological range caused a reduction of inner medullary blood flow and an improvement in urinary concentration. * indicates significance from control period (P < .05).

Reproduced, with permission, from Franchini and Cowley 189.


Figure 20.

In vitro microperfusion. (A) An afferent arteriole is cannulated with concentric pipettes and perfused toward the glomerulus.

Reproduced, with permission, from Hansen et al. (B and C) Perfusion of an isolated descending vasa recta (DVR). Images obtained before (B) and after (C) exposure to angiotensin II (AngII). Compare the thick smooth muscle layer of the afferent arteriole to the sporadically distributed pericyte cell bodies of DVR. Reproduced, with permission, from Hansen et al. and Pallone et al. 233,511.


Figure 21.

Arachidonic acid (AA) metabolites. (A) Abbreviated schematic showing signaling molecules generated from arachadonic acid by cyclo‐oxygenase (COX). Prostaglandins (PG) and thromboxanes (Tx) are vasoactive end products. B. Abbreviated schematic showing signaling molecules generated from the cytochrome P450 metabolism of AA. Hydrolases generate the potent constrictor 20‐hydroxyeicosatetraenoic acid (20‐HETE) and epoxygenases generate the epoxyeicosatrienoic acids that most often function as vasodilators. The lipoxygenase pathway of arachidonic acid metabolism is not shown.

Reproduced, with permission, from Pallone and Cao 499.


Figure 22.

Effect of intrarenal infusion of adenosine receptor A1 or A2 subtype agonists. Top and bottom panels show the effect of adenosine agonist infusions on intrarenal blood flow. Cortical and medullary measurements were obtained using laser‐Doppler flowmetry with optical fibers placed on the kidney surface or inserted into the renal parenchyma, respectively. Left and right panels show the respective effects of either A1 or A2 receptor stimulation with subtype‐specific agonists. At time = 0, the A1 agonist N6‐cyclopentyladenosine (left panels) or the A2 agonist CGS‐21680C (right panels) was transiently infused (1 min) into the renal parenchyma. The A1 agonist transiently reduced both cortical and medullary blood flow, while the A2 agonist caused a preferential increase in blood flow to the medulla.

Reproduced, with permission, from Agmon et al. 5.


Figure 23.

Autoregulation in different regions of the kidney. An electromagnetic flow device on the renal artery was used to measure total renal blood flow (RBF). Laser‐Doppler flow probes were inserted into the renal parenchyma at various depths to measure regional blood flow in the outer and inner medulla (IM). Total RBF and cortical tissue blood flow shows intact autoregulation (stability of blood flow over a range of perfusion pressure). In contrast, in these volume‐expanded rats (see text), the small fraction of blood flow that reaches the outer or IM is not autoregulated.

Reproduced, with permission, from Mattson et al. 410.


Figure 24.

Pressure natriuresis. (A) Medullary blood flow is autoregulated in hydropenic but not volume expanded rats. (B) Renal interstitial hydrostatic pressure (RIHP) is higher and increases to a greater degree with renal perfusion pressure in volume expanded animals. (C) When renal perfusion pressure is increased, urinary sodium excretion (UNaV) increases much more markedly in volume expanded than in hydropenic animals.

Reproduced, with permission, from Cowley 108.


Figure 25.

Acute NOS inhibition in the renal medulla. Line graph (left) showing changes in renal cortical and medullary blood flow (measured by laser‐Doppler flowmetry) in response to renal medullary interstitial infusion of the nitric oxide synthase inhibitor NG‐nitro‐l‐arginine methyl ester (L‐NAME, 120 μg/h). Bar graph (right) shows changes in urine volume (UVol) and sodium excretion (UNaV) accompanying medullary interstitial infusion of L‐NAME in the infused kidney and the noninfused contralateral kidney. * indicates significance from control (P < 0.05).

Reproduced, with permission, from Mattson et al. 415.


Figure 26.

Chronic influence of renal medullary interstitial infusion of the nitric oxide synthase inhibitor NG‐nitro‐l‐arginine methyl ester (L‐NAME, 8.6 mg/kg/day) on renal medullary blood flow (top), daily sodium balance (middle), and mean arterial blood pressure (bottom) in conscious Sprague Dawley rats. Vertical‐hashed marks indicate the L‐NAME infusion period. * indicates significance from control (P < 0.05).

Reproduced, with permission, from Mattson et al. 409.


Figure 27.

Hypothetical mechanisms whereby changes in renal medullary perfusion may lead to an alteration in arterial blood pressure.

Reproduced, with permission, from Mattson 404.
References
 1. Abe M, O'Connor P, Kaldunski M, Liang M, Roman RJ, Cowley AW Jr. Effect of sodium delivery on superoxide and nitric oxide in the medullary thick ascending limb. Am J Physiol Renal Physiol 291: F350‐F357, 2006.
 2. Abraham NG, Botros FT, Rezzani R, Rodella L, Bianchi R, Goodman AI. Differential effect of cobalt protoporphyrin on distributions of heme oxygenase in renal structure and on blood pressure in SHR. Cell Mol Biol (Noisy ‐le‐grand) 48: 895‐902, 2002.
 3. Abraham NG, Cao J, Sacerdoti D, Li X, Drummond GS. Heme oxygenase: The key to renal function regulation. Am J Physiol Renal Physiol 297: F1137‐F1152, 2009.
 4. Abraham NG, Kappas A. Heme oxygenase and the cardiovascular‐renal system. Free Radic Biol Med 39: 1‐25, 2005.
 5. Agmon Y, Dinour D, Brezis M. Disparate effects of adenosine A1‐ and A2‐receptor agonists on intrarenal blood flow. Am J Physiol 265: F802‐F806, 1993.
 6. Agmon Y, Peleg H, Greenfeld Z, Rosen S, Brezis M. Nitric oxide and prostanoids protect the renal outer medulla from radiocontrast toxicity in the rat. J Clin Invest 94: 1069‐1075, 1994.
 7. Akagi R, Takahashi T, Sassa S. Cytoprotective effects of heme oxygenase in acute renal failure. Contrib Nephrol 148: 70‐85, 2005.
 8. Aki Y, Tamaki T, Kiyomoto H, He H, Yoshida H, Iwao H, Abe Y. Nitric oxide may participate in V2 vasopressin‐receptor‐mediated renal vasodilation. J Cardiovascr Pharmacol 23: 331‐336, 1994.
 9. Albert AP, Large WA. Store‐operated Ca2+‐permeable non‐selective cation channels in smooth muscle cells. Cell Calcium 33: 345‐356, 2003.
 10. Alonso‐Galicia M, Sun CW, Falck JR, Harder DR, Roman RJ. Contribution of 20‐HETE to the vasodilator actions of nitric oxide in renal arteries. Am J Physiol 275: F370‐F378, 1998.
 11. Aperia AC, Broberger CGO, Söderlund S. Relationship between renal artery perfusion pressure and tubular reabsorption. Am J Physiol 220: 1205‐1212, 1971.
 12. Araujo M, Welch WJ. Oxidative stress and nitric oxide in kidney function. Curr Opin Nephrol Hypertens 15: 72‐77, 2006.
 13. Arendshorst WJ, Brannstrom K, Ruan X. Actions of angiotensin II on the renal microvasculature. J Am Soc Nephrol 10(Suppl 11): S149‐S161, 1999.
 14. Arendshorst WJ, Navar LG. Renal circulation and glomerular hemodynamics. In: Schrier RW, editor. Diseases of the Kidney and Urinary Tract (7th ed). Philadelphia: Lippincott Williams & Wilkins, 2001, p. 59‐107
 15. Arendshorst WJ, Navar LG. Renal circulation and glomerular hemodynamics. In: Schrier RW, editor. Diseases of the Kidney and Urinary Tract (8th ed). Philadelphia: Lippincott Williams & Wilkins, 2007, p. 54‐95.
 16. Arima S, Endo Y, Yaoita H, Omata K, Ogawa S, Tsunoda K, Abe M, Takeuchi K, Abe K, Ito S. Possible role of P‐450 metabolite of arachidonic acid in vasodilator mechanism of angiotensin II type 2 receptor in the isolated microperfused rabbit afferent arteriole. J Clin Invest 100: 2816‐2823, 1997.
 17. Aukland, K. Methods for measuring renal blood flow: Total flow and regional distribution. Ann Rev Physiol 42, 543‐555, 1980a.
 18. Aukland K. “Redistribution” of intrarenal blood flow: Facts or methodological artifacts? In: Leaf A, et al. editors. Renal Pathophysiology. New York: Raven Press, , 1980b; p. 145‐154.
 19. Aukland K, Bower BF, Berliner RW. Measurement of local blood flow with hydrogen gas. Circ Res 14: 164‐187, 1964.
 20. Badzynska B, Grzelec‐Mojzesowicz M, Dobrowolski L, Sadowski J. Differential effect of angiotensin II on blood circulation in the renal medulla and cortex of anaesthetised rats. J Physiol 538: 159‐166, 2002.
 21. Badzynska B, Grzelec‐Mojzesowicz M, Sadowski J. Prostaglandins but not nitric oxide protect renal medullary perfusion in anaesthetised rats receiving angiotensin II. J Physiol 548: 875‐880, 2003.
 22. Badzynska B, Sadowski J. Opposed effects of prostaglandin E2 on perfusion of rat renal cortex and medulla: Interactions with the renin‐angiotensin system. Exp Physiol 93: 1292‐1302, 2008.
 23. Bagnasco SM. Role and regulation of urea transporters. Pflugers Arch 450: 217‐226, 2005.
 24. Bagnasco SM. The erythrocyte urea transporter UT‐B. J Membr Biol 212: 133‐138, 2006.
 25. Balint K, Bartha F, Fekete A. Intrarenal distribution of blood flow in the dog. Acta Physiol Acad Sci Hung 36: 1‐11, 1969.
 26. Bankir L. Antidiuretic action of vasopressin: Quantitative aspects and interaction between V1a and V2 receptor‐mediated effects. Cardiovasc Res 51: 372‐390, 2001.
 27. Bankir L, Chen K, Yang B. Lack of UT‐B in vasa recta and red blood cells prevents urea‐induced improvement of urinary concentrating ability. Am J Physiol Renal Physiol 286: F144‐F151, 2004.
 28. Bankir L, de Rouffignac C. Urinary concentrating ability: Insights from comparative anatomy. Am J Physiol 249: R643‐R666, 1985.
 29. Bankir L, Kaissling B, de Rouffignac C, Kriz W. The vascular organization of the kidney of Psammomys obesus. Anat Embryol (Berl) 155: 149‐160, 1979.
 30. Barajas L, Liu L, Tucker M. Localization of connexin43 in rat kidney. Kidney Int 46: 621‐626, 1994.
 31. Baranowski RL, Westernfelder C. Estimation of renal interstitial adenosine and purine metabolites by microdialysis. Am J Physiol 267: F174‐F182, 1994.
 32. Barber RD, Woolf AS, Henderson RM. Potassium conductances and proliferation in conditionally immortalized renal glomerular mesangial cells from the H‐2Kb‐tsA58 transgenic mouse. Biochim Biophys Acta 1355: 191‐203, 1997.
 33. Bascands J‐L, Pecher C, Bompart G, Rakotoarivony J, Tack JL, Girolami J‐P. Bradykinin‐induced in vitro contraction of rat mesangial cells via a B2 receptor type. Am J Physiol 267: F871‐F878, 1994.
 34. Bascands J‐L, Pecher C, Rouaud S, Emond C, Tack JL, Bastie MJ, Burch R, Regoli D, Girolami J‐P. Evidence for existence of two distinct bradykinin receptors on rat mesangial cells. Am J Physiol 264: F548‐F556, 1993.
 35. Bayle F, Eloy L, Trinh‐Trang‐Tan MM, Grunfeld JP, Bankir L. Papillary plasma flow in rats. I. Relation to urine osmolality in normal and Brattleboro rats with hereditary diabetes insipidus. Pflugers Arch 394: 211‐216, 1982.
 36. Beach RE, Good DW. Effects of adenosine on ion transport in rat medullary thick ascending limb. Am J Physiol 263: F482‐F487, 1992.
 37. Beach RE, Watts IBA, Good DW, Benedict CR, Dubose TD Jr.. Effects of graded oxygen tension on adenosine release by renal medullary and thick ascending limb suspensions. Kidney Int 39: 836‐842, 1991.
 38. Beech DJ, Muraki K, Flemming R. Non‐selective cationic channels of smooth muscle and the mammalian homologues of Drosophila TRP. J Physiol 559: 685‐706, 2004.
 39. Beeuwkes R, III. The vascular organization of the kidney. Ann Rev Physiol 42: 531‐542, 1980.
 40. Beeuwkes R, III, Bonventre JV. Tubular organization and vascular‐tubular relations in the dog kidney. Am J Physiol 229: 695‐713, 1975.
 41. Bell RD, Keyl MJ, Shrader FR, Jones EW, Henry LP. Renal lymphatics: The internal distribution. Nephron 5: 454‐463, 1968.
 42. Bellien J, Thuillez C, Joannides R. Contribution of endothelium‐derived hyperpolarizing factors to the regulation of vascular tone in humans. Fundam Clin Pharmacol 22: 363‐377, 2008.
 43. Bergström G, Gothberg G, Karlström G, Rudenstam J. Renal medullary blood flow and renal medullary antihypertensive mechanisms. Clin Exp Hypertension 20: 1‐26, 1998.
 44. Betts LC, Kozlowski RZ. Electrophysiological effects of endothelin‐1 and their relationship to contraction in rat renal arterial smooth muscle. Br J Pharmacol 130: 787‐796, 2000.
 45. Biondi ML, Dousa T, Vanhoutte P, Romero JC. Evidences for the existence of endothelium‐derived relaxing factor in the renal medulla. Am J Hypertens 3: 876‐878, 1990.
 46. Birck R, Krzossok S, Knoll T, Braun C, Der Woude FJ, Rohmeiss P. Preferential COX‐2 inhibitor, meloxicam, compromises renal perfusion in euvolemic and hypovolemic rats. Exp Nephrol 8: 173‐180, 2000.
 47. Bloom IT, Bentley FR, Wilson MA, Garrison RN. In vivo effects of endothelin on the renal microcirculation. J Surg Res 54: 274‐280, 1993.
 48. Boesen EI, Pollock DM. Acute increases of renal medullary osmolality stimulate endothelin release from the kidney. Am J Physiol Renal Physiol 292: F185‐F191, 2006.
 49. Bohm F, Pernow J. The importance of endothelin‐1 for vascular dysfunction in cardiovascular disease. Cardiovasc Res 76: 8‐18, 2007.
 50. Bonner RF, Nossal R. Principles of laser‐Doppler flowmetry. In: Shepard AP, Oberg PA, editors. Laser Doppler Flowmetry. Boston: Kluwer Acad. Pub., 1990, p. 17‐45
 51. Braunstein TH, Sorensen CM, Holstein‐Rathlou NH. Connexin abundance in resistance vessels from the renal microcirculation in normo‐ and hypertensive rats. APMIS 117: 268‐276, 2009.
 52. Brezis M, Agmon Y, Epstein FH. Determinants of intrarenal oxygenation. I. Effects of diuretics. Am J Physiol 267: F1059‐F1062, 1994.
 53. Brezis M, Heyman SN, Dinour D, Epstein FH, Rosen S. Role of nitric oxide in renal medullary oxygenation: Studies in isolated and intact rat kidneys. J Clin Invest 88: 390‐395, 1991.
 54. Brezis M, Heyman SN, Epstein FH. Determinants of intrarenal oxygenation II. Hemodynamic effects. Am J Physiol 267: F1063‐F1068, 1994.
 55. Brezis M, Rosen S. Hypoxia of the renal medulla—its implications for disease. N Engl J Med 332: 647‐655, 1995.
 56. Brezis M, Rosen S, Silva P, Epstein FH. Renal ischemia: A new perspective. Kidney Int 26: 375‐383, 1984.
 57. Brisset AC, Isakson BE, Kwak BR. Connexins in vascular physiology and pathology. Antioxid Redox Signal 11: 267‐282, 2009.
 58. Bugaj V, Pochynyuk O, Mironova E, Vandewalle A, Medina JL, Stockand JD. Regulation of the epithelial Na+ channel by endothelin‐1 in rat collecting duct. Am J Physiol Renal Physiol 295: F1063‐F1070, 2008.
 59. Buhrle CP, Nobiling R, Mannek E, Schneider D, Hackenthal E, Taugner R. The afferent glomerular arteriole: Immunocytochemical and electrophysiological investigations. J Cardiovasc Pharmacol 6(Suppl 2): S383‐S393, 1984.
 60. Busse R, Edwards G, Feletou M, Fleming I, Vanhoutte PM, Weston AH. EDHF: Bringing the concepts together. Trends Pharmacol Sci 23: 374‐380, 2002.
 61. Bussemaker E, Popp R, Binder J, Busse R, Fleming I. Characterization of the endothelium‐derived hyperpolarizing factor (EDHF) response in the human interlobar artery. Kidney Int 63: 1749‐1755, 2003.
 62. Campean V, Theilig F, Paliege A, Breyer M, Bachmann S. Key enzymes for renal prostaglandin synthesis: Site‐specific expression in rodent kidney (rat, mouse). Am J Physiol Renal Physiol 285: F19‐F32, 2003.
 63. Cao C, Goo JH, Lee‐Kwon W, Pallone TL. Vasa recta pericytes express a strong inward rectifier K +conductance. Am J Physiol Regul Integr Comp Physiol 290: R1601‐R1607, 2006.
 64. Cao C, Lee‐Kwon W, Payne K, Edwards A, Pallone TL. Descending vasa recta endothelia express inward rectifier potassium channels. Am J Physiol Renal Physiol 293: F1248‐F1255, 2007.
 65. Cao C, Lee‐Kwon W, Silldorff EP, Pallone TL. KATP channel conductance of descending vasa recta pericytes. Am J Physiol Renal Physiol 289: F1235‐F1245, 2005.
 66. Cao C, Payne K, Lee‐Kwon W, Zhang Z, Lim SW, Hamlyn J, Blaustein MP, Kwon HM, Pallone TL. Chronic ouabain treatment induces vasa recta endothelial dysfunction in the rat. Am J Physiol Renal Physiol 296: F98‐F106, 2009.
 67. Capdevila JH. Regulation of ion transport and blood pressure by cytochrome p450 monooxygenases. Curr Opin Nephrol Hypertens 16: 465‐470, 2007.
 68. Capdevila JH, Falck JR, Imig JD. Roles of the cytochrome P450 arachidonic acid monooxygenases in the control of systemic blood pressure and experimental hypertension. Kidney Int 72: 683‐689, 2007.
 69. Carey RM. Update on the role of the AT2 receptor. Curr Opin Nephrol Hypertens 14: 67‐71, 2005.
 70. Carey RM, Jin X, Wang Z, Siragy HM. Nitric oxide: A physiological mediator of the type 2 (AT2) angiotensin receptor. Acta Physiol Scand 168: 65‐71, 2000.
 71. Carmines PK. Segment‐specific effect of chloride channel blockade on rat renal arteriolar contractile responses to angiotensin II. Am J Hypertens 8: 90‐94, 1995.
 72. Carmines PK, Inscho EW. Renal arteriolar angiotensin responses during varied adenosine receptor activation. Hypertension 23(suppl I): I‐114‐I‐119, 1994.
 73. Carmines PK, Navar LG. Disparate effects of Ca channel blockade on afferent and efferent arteriolar responses to ANG II. Am J Physiol 256: F1015‐F1020, 1989.
 74. Carmines PK, Fowler BC, Bell PD. Segmentally distinct effects of depolarization on intracellular [Ca2+] in renal arterioles. Am J Physiol 265: F677‐F685, 1993.
 75. Carmines PK, Morrison TK, Navar LG. Angiotensin II effects on microvascular diameters of in vitro blood‐perfused juxtamedullary nephrons. Am J Physiol 251: F610‐F618, 1986.
 76. Carone FA, Everett BA, Blondeel NJ, Stolarczyk J. Renal localization of albumin and its function in the concentrating mechanism. Am J Physiol 212: 387‐393, 1967.
 77. Casellas D, Mimran A. Shunting in renal microvasculature of the rat: A scanning electron microscopic study of corrosion casts. Anat Rec 201: 237‐248, 1981.
 78. Catterall WA, Cestele S, Yarov‐Yarovoy V, Yu FH, Konoki K, Scheuer T. Voltage‐gated ion channels and gating modifier toxins. Toxicon 49: 124‐141, 2007.
 79. Catterall WA, Striessnig J, Snutch TP, Perez‐Reyes E. International Union of Pharmacology. XL. Compendium of voltage‐gated ion channels: Calcium channels. Pharmacol Rev 55: 579‐581, 2003.
 80. Cavarape A, Bartoli E. Effects of BQ‐123 on systemic and renal hemodynamic responses to endothelin‐1 in the rat split hydronephrotic kidney. J Hypertens 16: 1449‐1458, 1998.
 81. Cermak R, Kleta R, Forssmann WG, Schlatter E. Natriuretic peptides increase a K+ conductance in rat mesangial cells. Pflugers Arch 431: 571‐577, 1996.
 82. Chabrashvili T, Kitiyakara C, Blau J, Karber A, Aslam S, Welch WJ, Wilcox CS. Effects of ANG II type 1 and 2 receptors on oxidative stress, renal NADPH oxidase, and SOD expression. Am J Physiol Regul Integr Comp Physiol 285: R117‐R124, 2003.
 83. Chatziantoniou C, Arendshorst WJ. Prostaglandin interactions with angiotensin, norepinephrine, and thromboxane in rat renal vasculature. Am J Physiol 262: F68‐F76, 1992.
 84. Chen J, Edwards A, Layton AT. A mathematical model of O2 transport in the rat outer medulla. II. Impact of outer medullary architecture. Am J Physiol Renal Physiol 297: F537‐F548, 2009.
 85. Chen J, Layton AT, Edwards A. A mathematical model of O2 transport in the rat outer medulla. I. Model formulation and baseline results. Am J Physiol Renal Physiol 297: F517‐F536, 2009.
 86. Chen PY, Sander PW. L‐Arginine abrogates salt‐senitive hypertension in Dahl/Rapp rats . J Clin Invest 88: 1559‐1567, 1991.
 87. Chen YF, Cowley AW Jr., Zou AP. Increased H(2)O(2) counteracts the vasodilator and natriuretic effects of superoxide dismutation by tempol in renal medulla. Am J Physiol Regul Integr Comp Physiol 285: R827‐R833, 2003.
 88. Cheng HF, Harris RC. Cyclooxygenases, the kidney, and hypertension. Hypertension 43: 525‐530, 2004.
 89. Cheng MK, Doumad AB, Jiang H, Falck JR, McGiff JC, Carroll MA. Epoxyeicosatrienoic acids mediate adenosine‐induced vasodilation in rat preglomerular microvessels (PGMV) via A2A receptors. Br J Pharmacol 141: 441‐448, 2004.
 90. Chilton L, Loutzenhiser R. Functional evidence for an inward rectifier potassium current in rat renal afferent arterioles. Circ Res 88: 152‐158, 2001.
 91. Chin SY, Wang C‐T, Majid DSA, Navar LG. Renoprotective effects of nitric oxide in angiotensin II‐induced hypertension in the rat. Am J Physiol 274: F876‐F882, 1998.
 92. Chipperfield AR, Harper AA. Chloride in smooth muscle. Prog Biophys Mol Biol 74: 175‐221, 2000.
 93. Chou CL, Knepper MA. In vitro perfusion of chinchilla thin limb segments: Segmentation and osmotic water permeability. Am J Physiol 263: F417‐F426, 1992.
 94. Chou CL, Nielsen S, Knepper MA. Structural‐functional correlation in chinchilla long loop of Henle thin limbs: A novel papillary subsegment. Am J Physiol 265: F863‐F874, 1993.
 95. Chou S‐Y, Spitalewitz S, Faubert PF, Park IY, Porush JG. Inner medullary hemodynamics in chronic salt‐depleted dogs. Am J Physiol 246: F146‐F154, 1984.
 96. Chuang EL, Reineck HJ, Osgood RW, Kunau RT, Stein JH. Studies on the mechanism of reduced urinary osmolality after exposure of the renal papilla. J Clin Invest 61: 633‐639, 1978.
 97. Clapham DE. TRP channels as cellular sensors. Nature 426: 517‐524, 2003.
 98. Coelho JB. Heterogeneity of intracortical peritubular plasma flow in the rat kidney. Am J Physiol 245: F333‐F341, 1977.
 99. Coffman TM, Crowley SD. Kidney in hypertension: Guyton redux. Hypertension 51: 811‐816, 2008.
 100. Correia AG, Bergstrom G, Jia J, Anderson WP, Evans RG. Dominance of pressure natriuresis in acute depressor responses to increased renal artery pressure in rabbits and rats. J Physiol 538: 901‐910, 2002.
 101. Correia AG, Denton KM, Evans RG. Effects of activation of vasopressin‐V1‐receptors on regional kidney blood flow and glomerular arteriole diameters. J Hypertens 19: 649‐657, 2001.
 102. Cowley AW Jr. Long‐term control of arterial pressure. Physiol Rev 72: 231‐300, 1992.
 103. Cowley AW Jr. Role of the renal medulla in volume and arterial pressure regulation. Am J Physiol 273: R1‐R15, 1997.
 104. Cowley AW Jr. Control of the renal medullary circulation by vasopressin V1 and V2 receptors in the rat. Exp Physiol 85 Spec No: 223S‐231S, 2000.
 105. Cowley AW Jr., Mori T, Mattson D, Zou AP. Role of renal NO production in the regulation of medullary blood flow. Am J Physiol Regul Integr Comp Physiol 284: R1355‐R1369, 2003.
 106. Cowley AW Jr., Roman RJ. The role of the kidney in hypertension. JAMA 275: 1581‐1589, 1996.
 107. Cowley AW Jr., Roman RJ. Renal mechanisms in hypertension. In: Zanchetti A, Mancia G, editors. Handbook of Hypertension, Vol. 17: Pathophysiology of Hypertension. Amsterdam, The Netherlands: Elsevier, 1997.
 108. Cribbs LL. T‐type Ca2+ channels in vascular smooth muscle: Multiple functions. Cell Calcium 40: 221‐230, 2006.
 109. Crowley SD, Gurley SB, Herrera MJ, Ruiz P, Griffiths R, Kumar AP, Kim H‐S, Smithies O, Le TH, Coffman TM. Angiotensin II causes hypertension and cardiac hypertrophy through its receptors in the kidney. Proc Natl Acad Sci U S A 103: 17985‐17990, 2006.
 110. Cupples WA. Renal medullary blood flow: Its measurement and physiology. Can J Physiol Pharmacol 64, 873‐880, 1986.
 111. Cupples WA, Marsh DJ. Autoregulation of blood flow in renal medulla of the rat: No role for angiotensin II. Can J Physiol Pharmacol 66: 833‐836, 1988.
 112. Cupples WA, Sakai T, Marsh DJ. Angiotensin II and prostaglandins in control of vasa recta blood flow. Am J Physiol 254: F417‐F424, 1988.
 113. Curtis JJ, Luke RG, Dustan HP, Kashgarian M, Whelchel JD, Jones P, Dieltham AG. Remission of essential hypertension after renal tranplantation. N Engl J Med 309: 1009‐1015, 1983.
 114. Curtis LM, Agarwal A. HOpe for contrast‐induced acute kidney injury. Kidney Int 72: 907‐909, 2007.
 115. de NG, Thomas R, Orleans‐Juste P, Antunes E, Walder C, Warner TD, Vane JR. Pressor effects of circulating endothelin are limited by its removal in the pulmonary circulation and by the release of prostacyclin and endothelium‐derived relaxing factor. Proc Natl Acad Sci U S A 85: 9797‐9800, 1988.
 116. De Wit C. Connexins pave the way for vascular communication. News Physiol Sci 19: 148‐153, 2004.
 117. de Wit C, Boettcher M, Schmidt VJ. Signaling across myoendothelial gap junctions–fact or fiction? Cell Commun Adhes 15: 231‐245, 2008.
 118. de Wit C, Roos F, Bolz SS, Kirchhoff S, Kruger O, Willecke K, Pohl U. Impaired conduction of vasodilation along arterioles in connexin40‐deficient mice. Circ Res 86: 649‐655, 2000.
 119. de Wit C, Roos F, Bolz SS, Pohl U. Lack of vascular connexin 40 is associated with hypertension and irregular arteriolar vasomotion. Physiol Genomics 13: 169‐177, 2003.
 120. de Wit C, Wolfle SE. EDHF and gap junctions: Important regulators of vascular tone within the microcirculation. Curr Pharm Biotechnol 8: 11‐25, 2007.
 121. Deng A, Wead LM, Blantz RC. Temporal adaptation of tubuloglomerular feedback: Effects of COX‐2. Kidney Int 66: 2348‐2353, 2004.
 122. Denton KM, Shweta A, Finkelstein L, Flower RL, Evans RG. Effect of endothelin‐1 on regional kidney blood flow and renal arteriole calibre in rabbits. Clin Exp Pharmacol Physiol 31: 494‐501, 2004.
 123. Dhein S. Peptides acting at gap junctions. Peptides 23: 1701‐1709, 2002.
 124. DiBona GF, Kopp UC. Neural control of renal function. Physiol Rev 77: 75‐97, 1997.
 125. Dickhout JG, Mori T, Cowley AW Jr. Tubulovascular nitric oxide crosstalk: Buffering of angiotensin II‐induced medullary vasoconstriction. Circ Res 91: 487‐493, 2002.
 126. Dinour D, Brezis M. Effects of adenosine on intrarenal oxygenation. Am J Physiol 261: F787‐F791, 1991.
 127. Dobrowolski L, Kompanowska‐Jezierska E, Walkowska A, Sadowski J. Sodium intake determines the role of adenosine A2 receptors in control of renal medullary perfusion in the rat. Nephrol Dial Transplant 22: 2805‐2809, 2007.
 128. Dobrucki LW, Sinusas AJ. Cardiovascular molecular imaging. Semin Nuclear Med 35: 73‐81, 2005.
 129. Duke LM, Eppel GA, Widdop RE, Evans RG. Disparate roles of AT2 receptors in the renal cortical and medullary circulations of anesthetized rabbits. Hypertension 42: 200‐205, 2003.
 130. Earley S, Resta TC, Walker BR. Disruption of smooth muscle gap junctions attenuates myogenic vasoconstriction of mesenteric resistance arteries. Am J Physiol Heart Circ Physiol 287: H2677‐H2686, 2004.
 131. Eckman DM, Nelson MT. Potassium ions as vasodilators: Role of inward rectifier potassium channels. Circ Res 88: 132‐133, 2001.
 132. Edwards A, Delong MJ, Pallone TL. Interstitial water and solute recovery by inner medullary vasa recta. Am J Physiol Renal Physiol 278: F257‐F269, 2000.
 133. Edwards A, Pallone TL. Facilitated transport in vasa recta: Theoretical effects on solute exchange in the medullary microcircualtion. Am J Physiol 272: F505‐F514, 1997.
 134. Edwards A, Pallone TL. A multiunit model of solute and water removal by inner medullary vasa recta. Am J Physiol 274: H1202‐H1210, 1998.
 135. Edwards A, Pallone TL. Mechanisms underlying angiotensin II‐induced calcium oscillations. Am J Physiol Renal Physiol 295: F568‐F584, 2008.
 136. Edwards G, Weston AH. Potassium and potassium clouds in endothelium‐dependent hyperpolarizations. Pharmacol Res 49: 535‐541, 2004.
 137. Edwards RM, Trizna W, Kinter LB. Renal microvascular effects of vasopressin and vasopressin antagonists. Am J Physiol 256: F274‐F278, 1989.
 138. Edwards RM, Trizna W, Ohlstein EH. Renal microvascular effects of endothelin. Am J Physiol 259: F217‐F221, 1990.
 139. Eggermont J. Calcium‐activated chloride channels: (un)known, (un)loved? Proc Am Thorac Soc 1: 22‐27, 2004.
 140. Endlich K, Hoffend J, Steinhausen M. Localization of endothelin ETA and ETB receptor‐mediated constriction in the renal microcirculation of rats. J Physiol (Lond) 497: 211‐218, 1996.
 141. Endo Y, Arima S, Yaoita H, Omata K, Tsunoda K, Takeuchi K, Abe K, Ito S. Function of angiotensin II type 2 receptor in the postglomerular efferent arteriole. Kidney Int Suppl 63: S205‐S207, 1997.
 142. Endo Y, Arima S, Yaoita H, Tsunoda K, Omata K, Ito S. Vasodilation mediated by angiotensin II type 2 receptor is impaired in afferent arterioles of young spontaneously hypertensive rats. J Vasc Res 35: 421‐427, 1998.
 143. Eppel GA, Bergstrom G, Anderson WP, Evans RG. Autoregulation of renal medullary blood flow in rabbits. Am J Physiol Regul Integr Comp Physiol 284: R233‐R244, 2003.
 144. Eppel GA, Ventura S, Evans RG. Regional vascular responses to ATP and ATP analogues in the rabbit kidney in vivo: Roles for adenosine receptors and prostanoids. Br J Pharmacol 149: 523‐531, 2006.
 145. Epstein FH, Balaban RS, Ross BD. Redox state of cytochrome aa3 in isolated perfused rat kidney. Am J Physiol 243: F356‐F363, 1982.
 146. Evan AP, Dail WG Jr. Efferent arterioles in the cortex of the rat kidney. Anat Rec 187: 135‐145, 1977.
 147. Evans RG, Gardiner BS, Smith DW, O’Connor PM. Intrarenal oxygenation: Unique challenges and the biophysical basis of homeostasis. Am J Physiol Renal Physiol 295: F1259‐F1270, 2008a.
 148. Evans RG, Gardiner BS, Smith DW, O’Connor PM. Methods for studying the physiology of kidney oxygenation. Clin Exp Pharmacol Physiol 35: 1405‐1412, 2008b.
 149. Evans RG, Head GA, Eppel GA, Burke SL, Rajapakse NW. Angiotensin II and neurohumoral control of the renal medullary circulation. Clin Exp Pharmacol Physiol 37: e58‐e69, 2010.
 150. Evans RG, Madden AC, Denton KM. Diversity of responses of renal cortical and medullary blood flow to vasoconstrictors in conscious rabbits. Acta Physiol Scand 169: 297‐308, 2000.
 151. Facemire CS, Mohler PJ, Arendshorst WJ. Expression and relative abundance of short transient receptor potential channels in the rat renal microcirculation. Am J Physiol Renal Physiol 286: F546‐F551, 2004.
 152. Fadem SZ, Hernandez‐Llamas G, Patak RV, Rosenblatt SG, Lifschitz MD, Stein JH. Studies on the mechanism of sodium excretion during drug‐induced vasodilation in the dog. J Clin Invest 69: 604‐610, 1982.
 153. Fahraeus R. The suspension stability of blood. Physiol Rev 9: 241‐274, 1929.
 154. Fallet RW, Bast JP, Fujiwara K, Ishii N, Sansom SC, Carmines PK. Influence of Ca2+‐activated K+ channels on rat renal arteriolar responses to deolarizing agonists. Am J Physiol Renal Physiol 280: F583‐F591, 2001.
 155. Farrugia E, Lockart LC, Larson TS. Relationship between vasa recta blood flow and renal interstitial hydrostatic pressure during pressure natriuresis. Circ Res 71: 1153‐1158, 1993.
 156. Faubert PF, Chou S‐Y, Porush JG. Regulation of papillary plasma flow by angiotensin II. Kidney Int 32: 472‐478, 1987.
 157. Fellner SK, Arendshorst WJ. Capacitative calcium entry in smooth muscle cells from preglomerular vessels. Am J Physiol 277: F533‐F542, 1999.
 158. Fellner SK, Arendshorst WJ. Store‐operated Ca2+ entry is exaggerated in fresh preglomerular vascular smooth muscle cells of SHR. Kidney Int 61: 2132‐2141, 2002.
 159. Fellner SK, Arendshorst WJ. Endothelin A and B receptors of preglomerular vascular smooth muscle cells. Kidney Int 65: 1810‐1817, 2004.
 160. Feng JJ, Arendshorst WJ. Calcium signaling mechanisms in renal vascular responses to vasopressin in genetic hypertension. Hypertension 30: 1223‐1231, 1997.
 161. Feng MG, Li M, Navar LG. T‐type calcium channels in the regulation of afferent and efferent arterioles in rats. Am J Physiol Renal Physiol 286: F331‐F337, 2004.
 162. Feng MG, Navar LG. Angiotensin II‐mediated constriction of afferent and efferent arterioles involves T‐type Ca2+ channel activation. Am J Nephrol 24: 641‐648, 2004.
 163. Feng MG, Navar LG. Nitric oxide synthase inhibition activates L‐ and T‐type Ca2+ channels in afferent and efferent arterioles. Am J Physiol Renal Physiol 290: F873‐F879, 2006.
 164. Fenton RA. Urea transporters and renal function: Lessons from knockout mice. Curr Opin Nephrol Hypertens 17: 513‐518, 2008.
 165. Fenton RA, Knepper MA. Urea and renal function in the 21st century: Insights from knockout mice. J Am Soc Nephrol 18: 679‐688, 2007.
 166. Fenton RA, Smith CP, Knepper MA. Role of collecting duct urea transporters in the kidney—insights from mouse models. J Membr Biol 212: 119‐131, 2006.
 167. Fergus DJ, Martens JR, England SK. Kv channel subunits that contribute to voltage‐gated K +current in renal vascular smooth muscle. Pflugers Arch 445: 697‐704, 2003.
 168. Figueroa CD, Gonzalez CB, Grigoriev S, Alla SA, Haasemann M, Jarnagin K, Muller‐Esterl W. Probing for the bradykinin B2 receptor in rat kidney by anti‐peptide and anti‐ligand antibodies. J Histochem Cytochem 43: 137‐148, 1995.
 169. Figueroa CD, Maclver AG, Mackenzie JC, Bhoola KD. Localisation of immunoreactive kininogen and tissue kallikrein in the human nephron. Histochemistry 89: 437‐442, 1988.
 170. Figueroa XF, Duling BR. Gap junctions in the control of vascular function. Antioxid Redox Signal 11: 251‐266, 2009.
 171. Figueroa XF, Isakson BE, Duling BR. Connexins: Gaps in our knowledge of vascular function. Physiology (Bethesda) 19: 277‐284, 2004.
 172. Figueroa XF, Paul DL, Simon AM, Goodenough DA, Day KH, Damon DN, Duling BR. Central role of connexin40 in the propagation of electrically activated vasodilation in mouse cremasteric arterioles in vivo. Circ Res 92: 793‐800, 2003.
 173. Fishman MC. Membrane potential of juxtaglomerular cells. Nature 260: 542‐544, 1976.
 174. Fleming I. Cytochrome p450 and vascular homeostasis. Circ Res 89: 753‐762, 2001.
 175. Fleming I. Cytochrome P450 epoxygenases as EDHF synthase(s). Pharmacol Res 49: 525‐533, 2004.
 176. Fleming JT, Parekh N, Steinhausen M. Calcium antagonists preferentially dilate preglomerular vessels of hydronephrotic kidney. Am J Physiol 253: F1157‐F1163, 1987.
 177. Fourman J, Moffat DB. The effect of intra‐arterial cushions on plasma skimming in small arteries. J Physiol 158: 374‐380, 1961.
 178. Fourman J, Moffat DB. The Blood Vessels of the Kidney. Oxford, UK: Blackwell Scientific, 1971.
 179. Franchini KG, Cowley Jr. AW. Renal cortical and medullary blood flow responses during water restriction: Role of vasopressin. Am J Physiol 270: R1257‐R1264, 1996a.
 180. Franchini KG, Cowley Jr. AW. Sensitivity of the renal medullary circulation to plasma vasopressin. Am J Physiol Regul Integr Comp Physiol 271: R647‐R653, 1996b.
 181. Friis UG, Jorgensen F, Andreasen D, Jensen BL, Skott O. Molecular and functional identification of cyclic AMP‐sensitive BKCa potassium channels (ZERO variant) and L‐type voltage‐dependent calcium channels in single rat juxtaglomerular cells. Circ Res 93: 213‐220, 2003.
 182. Fujino T, Nakagawa N, Yuhki K, Hara A, Yamada T, Takayama K, Kuriyama S, Hosoki Y, Takahata O, Taniguchi T, Fukuzawa J, Hasebe N, Kikuchi K, Narumiya S, Ushikubi F. Decreased susceptibility to renovascular hypertension in mice lacking the prostaglandin I2 receptor IP. J Clin Invest 114: 805‐812, 2004.
 183. Fukao M, Mason HS, Kenyon JL, Horowitz B, Keef KD. Regulation of BK(Ca) channels expressed in human embryonic kidney 293 cells by epoxyeicosatrienoic acid. Mol Pharmacol 59: 16‐23, 2001.
 184. Fuller AJ, Hauschild BC, Gonzalez‐Villalobos R, Awayda MS, Imig JD, Inscho EW, Navar LG. Calcium and chloride channel activation by angiotensin II‐AT1 receptors in preglomerular vascular smooth muscle cells. Am J Physiol Renal Physiol 289: F760‐F767, 2005.
 185. Gaehtgens P. Flow of blood through narrow capillaries: Rheological mechanisms determining capillary hematocrit and apparent viscosity. Biorheology 17: 183‐189, 1980.
 186. Ganguli M, Tobian L. Renal medullary plasma flow in hypertension (abstract) Federation Proc 31: 394, 1972.
 187. Ganguli M, Tobian L. Does the kidney autoregulate papillary plasma flow in chronic “post‐salt” hypertension? Am J Physiol 226: 330‐333, 1977.
 188. Garcia‐Estan J, Roman RJ. Role of renal interstitial hydrostatic pressure in the pressure diuresis response. Am J Physiol 256: F63‐F70, 1989.
 189. Gariepy CE, Ohuchi T, Williams SC, Richardson JA, Yanagisawa M. Salt‐sensitive hypertension in endothelin‐B receptor‐deficient rats. J Clin Invest 105: 925‐933, 2000.
 190. Gariepy CE, Williams SC, Richardson JA, Hammer RE, Yanagisawa M. Transgenic expression of the endothelin‐B receptor prevents congenital intestinal aganglionosis in a rat model of Hirschsprung disease. J Clin Invest 102: 1092‐1101, 1998.
 191. Garvin JL, Ortiz PA. The role of reactive oxygen species in the regulation of tubular function. Acta Physiol Scand 179: 225‐232, 2003.
 192. Ge Y, Bagnall A, Stricklett PK, Webb D, Kotelevtsev Y, Kohan DE. Combined knockout of collecting duct endothelin A and B receptors causes hypertension and sodium retention. Am J Physiol Renal Physiol 295: F1635‐F1640, 2008.
 193. Gebremedhin D, Kaldunski M, Jacobs ER, Harder DR, Roman RJ. Coexistence of two types of Ca2+‐activated K+ channels in rat renal arterioles. Am J Physiol 270: F69‐F81, 1996.
 194. Gelband CH, Hume JR. Ionic currents in single smooth muscle cells of the canine renal artery. Circ Res 71: 745‐758, 1992.
 195. Gelband CH, Hume JR. [Ca2+]i inhibition of K+ channels in canine renal artery. Novel mechanism for agonist‐induced membrane depolarization. Circ Res 77: 121‐130, 1995.
 196. Gelband CH, Ishikawa T, Post JM, Keef KD, Hume JR. Intracellular divalent cations block smooth muscle K+ channels. Circ Res 73: 24‐34, 1993.
 197. Gomez SI, Strick DM, Romero JC. Role of nitric oxide and prostaglandin in the maintenance of cortical and renal medullary blood flow. Braz J Med Biol Res 41: 170‐175, 2008.
 198. Goodman AI, Olszanecki R, Yang LM, Quan S, Li M, Omura S, Stec DE, Abraham NG. Heme oxygenase‐1 protects against radiocontrast‐induced acute kidney injury by regulating anti‐apoptotic proteins. Kidney Int 72: 945‐953, 2007.
 199. Goransson A, Sjoquist M, Ulfendahl HR. Superficial and juxtamedullary nephron function during converting enzyme inhibition. Am J Physiol 251: F25‐F33, 1986.
 200. Gordienko DV, Clausen C, Goligorsky MS. Ionic currents and endothelin signaling in smooth muscle cells from rat renal resistance arteries. Am J Physiol 266: F325‐F341, 1994.
 201. Goto M, Mukoyama M, Sugawara A, Suganami T, Kasahara M, Yahata K, Makino H, Suga S, Tanaka I, Nakao K. Expression and role of angiotensin II type 2 receptor in the kidney and mesangial cells of spontaneously hypertensive rats. Hypertens Res 25: 125‐133, 2002.
 202. Gottschalk CW, Mylle M. Micropuncture study of the mammalian urinary concentrating mechansm: Evidence for the countercurrent hypothesis. Am J Physiol 196: 927‐936, 1959.
 203. Grollman A, Muirhead EE, Vanatta J. Role of the kidney in pathogenesis of hypertension as determined by a study of the effects of bilateral nephrectomy and other experimental procedures on the blood pressure of the dog. Am J Physiol 157: 21‐30, 1949.
 204. Gross V, Kurth TM, Skelton MM, Mattson DL, Cowley AW Jr. Effects of daily sodium intake and angiotensin II upon cortical and medullary renal blood flow in conscious rats. Am J Physiol 274: R1317‐R1323, 1998.
 205. Guidi E, Menghetti D, Milani S, Montagnino G, Palazzi P, Bianchi G. Hypertension may be transplanted with the kidney in humans: A long‐term historical prospective follow‐up of recipients grafted with kidneys coming from donors with or without hypertension in their families. J Am Soc Nephrol 7: 1131‐1138, 1996.
 206. 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.
 207. Guo X, Yang T. Endothelin B receptor antagonism in the rat renal medulla reduces urine flow rate and sodium excretion. Exp Biol Med (Maywood) 231: 1001‐1005, 2006.
 208. Gurbanov K, Rubinstein I, Hoffman A, Abassi Z, Better OS, Winaver J. Differential regulation of renal regional blood flow by endothelin‐1. Am J Physiol 271: F1166‐F1172, 1996.
 209. Gussis GL, Robertson CR, Jamison RL. Erythrocyte velocity in vasa recta: Effect of antidiuretic hormone and saline loading. Am J Physiol 237: F326‐F332, 1979.
 210. Guyton AC, Coleman TG, Cowley AW Jr., Manning RD Jr., Norman RA Jr., Ferguson JD. A systems analysis approach to understanding long‐range arterial blood pressure control and hypertension. Circ Res 35: 159‐176, 1974.
 211. Ha H, Hwang IA, Park JH, Lee HB. Role of reactive oxygen species in the pathogenesis of diabetic nephropathy. Diabetes Res Clin Pract 82(Suppl 1): S42‐S45, 2008.
 212. Haas JA, Granger JP, Knox FG. Effect of renal perfusion pressure on sodium reabsorption from proximal tubules of superficial and deep nephrons. Am J Physiol 250: F425‐F429, 1986.
 213. Haas JA, Khraibi AA, Perrella MA, Knox FG. Role of renal interstitial hydrostatic pressure in natriuresis of systemic nitric oxide inhibition. Am J Physiol 164: F411‐F414, 1993.
 214. Haefliger J‐A, Demotz S, Braissant O, Suter E, Waeber B, Nicod P, Meda P. Connexins 40 and 43 are differentially regulated within the kdineys of rats with renovascular hypertension. Kidney Int 60: 190‐201, 2001.
 215. Haefliger JA, Krattinger N, Martin D, Pedrazzini T, Capponi A, Doring B, Plum A, Charollais A, Willecke K, Meda P. Connexin43‐dependent mechanism modulates renin secretion and hypertension. J Clin Invest 116: 405‐413, 2006.
 216. Haefliger JA, Nicod P, Meda P. Contribution of connexins to the function of the vascular wall. Cardiovasc Res 62: 345‐356, 2004.
 217. Hall DA, Carmines PK, Sansom SC. Dihydropyridine‐sensitive Ca(2+) channels in human glomerular mesangial cells. Am J Physiol 278: F97‐F103, 2000.
 218. Hall JM. Bradykinin receptors: Pharmacological properties and biological roles. Pharmacol Ther 56: 131‐190, 1992.
 219. Hansen PB, Castrop H, Briggs J, Schnermann J. Adenosine induces vasoconstriction through Gi‐dependent activation of phospholipase C in isolated perfused afferent arterioles of mice. J Am Soc Nephrol 14: 2457‐2465, 2003.
 220. Hansen PB, Hashimoto S, Briggs J, Schnermann J. Attenuated renovascular constrictor responses to angiotensin II in adenosine 1 receptor knockout mice. Am J Physiol 285: R44‐R49, 2003.
 221. Hansen PB, Jensen BL, Andreasen D, Friis UG, Skott O. Vascular smooth muscle cells express the alpha(1A) subunit of a P‐/Q‐type voltage‐dependent Ca(2+)Channel, and It is functionally important in renal afferent arterioles. Circ Res 87: 896‐902, 2000.
 222. Hansen PB, Jensen BL, Andreasen D, Skott O. Differential expression of T‐ and L‐type voltage‐dependent calcium channels in renal resistance vessels. Circ Res 89: 630‐638, 2001.
 223. Hansen PB, Jensen BL, Skott O. Chloride regulates afferent arteriolar contraction in response to depolarization. Hypertension 32: 1066‐1070, 1998.
 224. Hansen PB, Schnermann J. Vasoconstrictor and vasodilator effects of adenosine in the kidney. Am J Physiol 285: F590‐F599, 2003.
 225. Hao CM, Breyer MD. Physiologic and pathophysiologic roles of lipid mediators in the kidney. Kidney Int 71: 1105‐1115, 2007.
 226. Harder DR, Campbell WB, Roman RJ. Role of cytochrome P‐450 enzymes and metabolites of arachidonic acid in the control of vascular tone. J Vasc Res 32: 79‐92, 1995.
 227. Harder DR, Gilbert R, Lombard JH. Vascular muscle cell deoplarization and activation in renal arteries on elevation of transmural pressure. Am J Physiol 253: F778‐F781, 1987.
 228. Harris RC, Breyer MD. Physiological regulation of cyclooxygenase‐2 in the kidney. Am J Physiol 281: F1‐F11, 2001.
 229. Harris RC, Zhang MZ, Cheng HF. Cyclooxygenase‐2 and the renal renin‐angiotensin system. Acta Physiol Scand 181: 543‐547, 2004.
 230. Harrison‐Bernard LM, Carmines PK. Juxtamedullary microvascular responses to arginine vasopressin in rat kidney. Am J Physiol 267: F249‐F256, 1994.
 231. Harrison‐Bernard LM, Carmines PK. Impact of cyclooxygenase blockade on juxtamedullary microvascular responses to angiotensin II in rat kidney. Clin Exp Pharmacol Physiol 22: 732‐738, 1995.
 232. Harrison‐Bernard LM, Cook AK, Oliverio MI, Coffman TM. Renal segmental microvascular responses to ANG II in AT1A receptor null mice. Am J Physiol Renal Physiol 284: F538‐F545, 2003.
 233. Harsing L, Pelly K. die Bestimmung der Nierenmarkdurch‐blutung auf Grund der Ablagerungen und Verteilung von 86Rb. Pfluegers Arch 285: 302‐312, 1965.
 234. Hartzell C, Putzier I, Arreola J. Calcium‐activated chloride channels. Annu Rev Physiol 67: 719‐758, 2005.
 235. Hayashi K, Ozawa Y, Fujiwara K, Wakino S, Kumagai H, Saruta T. Role of actions of calcium antagonists on efferent arterioles–with special references to glomerular hypertension. Am J Nephrol 23: 229‐244, 2003.
 236. Hayashi K, Ozawa Y, Wakino S, Kanda T, Homma K, Takamatsu I, Tatematsu S, Saruta T. Cellular mechanism for mibefradil‐induced vasodilation of renal microcirculation: Studies in the isolated perfused hydronephrotic kidney. J Cardiovasc Pharmacol 42: 697‐702, 2003.
 237. Hayashi K, Wakino S, Sugano N, Ozawa Y, Homma K, Saruta T. Ca2+ channel subtypes and pharmacology in the kidney. Circ Res 100: 342‐353, 2007.
 238. Hebert SC, Andreoli TE. Control of NaCl transport in the thick ascending limb. Am J Physiol 246: F745‐F756, 1984.
 239. Helou CM, Imbert‐Teboul M, Doucet A, Rajerison R, Chollet C, Alhenc‐Gelas F, Marchetti J. Angiotensin receptor subtypes in thin and muscular juxtamedullary efferent arterioles of rat kidney. Am J Physiol Renal Physiol 285: F507‐F514, 2003.
 240. Helou CM, Marchetti J. Morphological heterogeneity of renal glomerular arterioles and distinct [Ca2+]i responses to ANG II. Am J Physiol 273: F84‐F96, 1997.
 241. Hercule HC, Oyekan AO. Cytochrome P450 omega/omega‐1 hydroxylase‐derived eicosanoids contribute to endothelin(A) and endothelin(B) receptor‐mediated vasoconstriction to endothelin‐1 in the rat preglomerular arteriole. J Pharmacol Exp Ther 292: 1153‐1160, 2000.
 242. Hermann A, Braun A, Figueroa CD, Müller‐Esterl W, Fritz H, Rehbock J. Expression and cellular localization of kininogens in the human kidney. Kidney Int 50: 79‐84, 1996.
 243. Herrera M, Ortiz PA, Garvin JL. Regulation of thick ascending limb transport: Role of nitric oxide. Am J Physiol Renal Physiol 290: F1279‐F1284, 2006.
 244. Hervy S, Thomas SR. Inner medullary lactate production and urine‐concentrating mechanism: A flat medullary model. Am J Physiol Renal Physiol 284: F65‐F81, 2003.
 245. Heyman SN, Brezis M, Epstein FH, Spokes K, Silva P, Rosen S. Early renal medullary hypoxic injury from radiocontrast and indomethacin. Kidney Int 40: 632‐642, 1991.
 246. Heyman SN, Fuchs S, Jaffe R, Shina A, Ellezian L, Brezis M, Rosen S. Renal microcirculation and tissue damage during acute ureteral obstruction in the rat: Effect of saline infusion, indomethacin and radiocontrast. Kidney Int 51: 653‐663, 1997.
 247. Heyman SN, Rosen S, Darmon D, Goldfarb M, Bitz H, Shina A, Brezis M. Endotoxin‐induced renal failure. II. A role for tubular hypoxic damage. Exp Nephrol 8: 275‐282, 2000.
 248. Hillis GS, Duthie LA, Mlynski R, McKay NG, Mistry S, MacLeod AM, Simpson JG, Haites NE. The expression of connexin 43 in human kidney and cultured renal cells. Nephron 75: 458‐463, 1997.
 249. Hoffman A, Abassi ZA, Brodsky S, Ramadan R, Winaver J. Mechanisms of big endothelin‐1‐induced diuresis and natriuresis : Role of ET(B) receptors. Hypertension 35: 732‐739, 2000.
 250. Holliger C, Lemley KV, Schmitt SL, Thomas FC, Robertson CR, Jamison RL. Direct determination of vasa recta blood flow in the rat renal papilla. Circ Res 53: 401‐413, 1983.
 251. Hsu YJ, Hoenderop JG, Bindels RJ. TRP channels in kidney disease. Biochim Biophys Acta 1772: 928‐936, 2007.
 252. Huang C, Davis G, Johns EJ. Effect of nitrendipine on autoregulation of perfusion in the renal cortex and papilla of the kidneys from Wistar and stroke‐prone spontaneously hypertensive rats. Br J Pharmacol 111: 111‐116, 1994.
 253. Hughes AK, Barry WH, Kohan DE. Identification of a contractile function for renal medullary interstitial cells. J Clin Invest 96: 411‐416, 1995.
 254. Hwan SK, Beyer EC. Heterogeneous localization of connexin40 in the renal vasculature. Microvasc Res 59: 140‐148, 2000.
 255. Ichihara A, Imig JD, Inscho EW, Navar L. Cyclooxygenase‐2 participates in tubular flow‐dependent afferent arteriolar tone: Interaction with neuronal NOS. Am J Physiol 275: F605‐F612, 1998.
 256. Imig JD. Epoxyeicosatrienoic acids. Biosynthesis, regulation, and actions. Methods Mol Biol 120: 173‐192, 1999.
 257. Imig JD. Eicosanoid regulation of the renal vasculature. Am J Physiol Renal Physiol 279: F965‐F981, 2000.
 258. Imig JD. Eicosanoids and renal vascular function in diseases. Clin Sci (Lond) 111: 21‐34, 2006.
 259. Imig JD, Dimitropoulou C, Reddy DS, White RE, Falck JR. Afferent arteriolar dilation to 11, 12‐EET analogs involves PP2A activity and Ca2+‐activated K +Channels. Microcirculation 15: 137‐150, 2008.
 260. Imig JD, Falck JR, Wei S, Capdevila JH. Epoxygenase metabolites contribute to nitric oxide‐independent afferent arteriolar vasodilation in response to bradykinin. J Vasc Res 38: 247‐255, 2001.
 261. Imig JD, Inscho EW, Deichmann PC, Reddy KM, Falck JR. Afferent arteriolar vasodilation to the sulfonimide analog of 11,12‐epoxygeicosatrienoic acid involves protein kinase A. Hypertension 33 [ part II]: 408‐413, 1999.
 262. Imig JD, Navar LG, Roman RJ, Reddy KK, Falck JR. Actions of epoxygenase metabolites on the preglomerular vasculature. J Am Soc Nephrol 7: 2364‐2370, 1996.
 263. Imig JD, Pham BT, LeBlanc EA, Reddy KM, Falck JR, Inscho EW. Cytochrome P450 and cyclooxygenase metabolites contribute to the endothelin‐1 afferent arteriolar vasoconstrictor and calcium responses. Hypertension 35: 307‐312, 2000.
 264. Imig JD, Zhao X, Capdevila JH, Morisseau C, Hammock BD. Soluble epoxide hydrolase inhibition lowers arterial blood pressure in angiotensin II hypertension. Hypertension 39: 690‐694, 2002.
 265. Imig JD, Zhao X, Falck JR, Wei S, Capdevila JH. Enhanced renal microvascular reactivity to angiotensin II in hypertension is ameliorated by the sulfonimide analog of 11,12‐epoxyeicosatrienoic acid. J Hypertens 19: 935‐992, 2001.
 266. Inscho EW, Mason MJ, Schroeder AC, Deichmann PC, Stiegler ID, Imig JD. Agonist‐induced calcium regulation in freshly isolated renal microvascular smooth muscle cells. J Am Soc Nephrol 8: 569‐579, 1997.
 267. Inscho EW, Schroeder AC, Deichmann PC, Imig JD. ATP‐mediated Ca2 +signaling in preglomerular smooth muscle cells. Am J Physiol 276: F450‐F456, 1999.
 268. Ito S, Arima S, Ren YL, Juncos LA, Carretero OA. Endothelium‐derived relaxing factor/nitric oxide modulates angiotensin II action in the isolated microperfused rabbit afferent but not efferent arteriole. J Clin Invest 91: 2012‐2019, 1993.
 269. Ito S, Johnson CS, Carretero OA. Modulation of angiotensin II‐induced vasoconstriction by endothelium‐derived relaxing factor in the isolated microperfused rabbit afferent arteriole. J Clin Invest 87: 1656‐1663, 1991.
 270. Ito S, Juncos LA, Nushiro N, Johnson CS, Carretero OA. Endothelium‐derived relaxing factor modulates endothelin action in afferent arterioles. Hypertension 17: 1052‐1056, 1991.
 271. Itskovitz HD, Stemper J, Pacholczyk D, McGiff JC. Renal prostaglandins: Determinants of intrarenal distribution of blood flow in the dog. Clin Sci Mol Med Suppl 45(Suppl 1): 321s‐4, 1973.
 272. Iversen BM, Arendshorst WJ. Exaggerated Ca2+ signaling in preglomerular arteriolar smooth muscle cells of genetically hypertensive rats. Am J Physiol 276: F260‐F270, 1999.
 273. Jackson EK, Dubey RK. Role of the extracellular cAMP‐adenosine pathway in renal physiology. Am J Physiol Renal Physiol 281: F597‐F612, 2001.
 274. Jackson EK, Zhu C, Tofovic SP. Expression of adenosine receptors in the preglomerular microcirculation. Am J Physiol Renal Physiol 283: F41‐F51, 2002.
 275. Jackson WF. Potassium channels in the peripheral microcirculation. Microcirculation 12: 113‐127, 2005.
 276. James PE, Bacic G, Grinberg OY, Goda F, Dunn JF, Jackson SK, Swartz HM. Endotoxin‐induced changes in intrarenal pO2, measured by in vivo electron paramagnetic resonance oximetry and magnetic resonance imaging. Free Radic Biol Med 21: 25‐34, 1996.
 277. Jamison RL, Kriz W. Urinary Concentrating Mechanism: Structure and Function. New York, Oxford: Oxford University Press, 1982.
 278. Jamison RL, Work J, Schafer JA. New pathways for potassium transport in the kidney. Am J Physiol 242: F297‐F312, 1982.
 279. Jensen BL, Ellekvist P, Skott O. Chloride is essential for contraction of afferent arterioles after agonists and potassium. Am J Physiol 272: F389‐F396, 1997.
 280. Jensen BL, Friis UG, Hansen PB, Andreasen D, Uhrenholt T, Schjerning J, Skott O. Voltage‐dependent calcium channels in the renal microcirculation. Nephrol Dial Transplant 19: 1368‐1373, 2004.
 281. Jensen BL, Skott O. Blockade of chloride channels by DIDS stimulates renin release and inhibits contraction of afferent arterioles. Am J Physiol 270: F718‐F727, 1996.
 282. Jin C, Hu C, Polichnowski A, Mori T, Skelton M, Ito S, Cowley AW Jr. Effects of renal perfusion pressure on renal medullary hydrogen peroxide and nitric oxide production. Hypertension 53: 1048‐1053, 2009.
 283. Johnston PA, Battilana CA, Lacy FB, Jamison RL. Evidence for a concentration gradient favoring outward movement of sodium from the thin loop of Henle. J Clin Invest 59: 234‐240, 1977.
 284. Jung JY, Madsen KM, Han KH, Yang CW, Knepper MA, Sands JM, Kim J. Expression of urea transporters in potassium‐depleted mouse kidney. Am J Physiol Renal Physiol 285: F1210‐F1224, 2003.
 285. Just A, Kurtz L, de WC, Wagner C, Kurtz A, Arendshorst WJ. Connexin 40 mediates the tubuloglomerular feedback contribution to renal blood flow autoregulation. J Am Soc Nephrol 20: 1577‐1585, 2009.
 286. Just A, Olson AJ, Arendshorst WJ. Dual constrictor and dilator actions of ET(B) receptors in the rat renal microcirculation: Interactions with ET(A) receptors. Am J Physiol Renal Physiol 286: F660‐F668, 2004.
 287. Kaide JI, Zhang F, Wei Y, Jiang H, Yu C, Wang WH, Balazy M, Abraham NG, Nasjletti A. Carbon monoxide of vascular origin attenuates the sensitivity of renal arterial vessels to vasoconstrictors. J Clin Invest 107: 1163‐1171, 2001.
 288. Kakoki M, Kim HS, Arendshorst WJ, Mattson DL. L‐Arginine uptake affects nitric oxide production and blood flow in the renal medulla. Am J Physiol Regul Integr Comp Physiol 287: R1478‐R1485, 2004.
 289. Kakoki M, Kim HS, Edgell CJ, Maeda N, Smithies O, Mattson DL. Amino acids as modulators of endothelium‐derived nitric oxide. Am J Physiol Renal Physiol 291: F297‐F304, 2006.
 290. Kakoki M, Wang W, Mattson DL. Cationic amino acid transport in the renal medulla and blood pressure regulation. Hypertension 39: 287‐292, 2002.
 291. Kakoki M, Zou AP, Mattson DL. The influence of nitric oxide synthase 1 on blood flow and interstitial nitric oxide in the kidney. Am J Physiol Regul Integr Comp Physiol 281: R91‐R97, 2001.
 292. Karlberg L, Kallskog O, Ojteg G, Wolgast M. Renal medullary blood flow studied with the 86‐Rb extraction method. Methodological consideration. Acta Physiol Scand 115: 11‐18, 1982.
 293. Karlström G, Arnman V, Folkow B, Göthberg G. Activation of the humoral antihypertensive system of the kidney increases diuresis. Hypertension 11: 597‐601, 1988.
 294. Kedem O, Katchalsky A. Thermodynamic analysis of the permeability of biological membranes to non‐electrolytes. Biochim Biophys Acta 27: 229‐246, 1958.
 295. Kiberd B, Robertson CR, Larson T, Jamison RL. Effect of V2‐receptor‐mediated changes on inner medullary blood flow induced by AVP. Am J Physiol‐Renal Physiol 253: F576‐F581, 1987.
 296. Kim D, Klein JD, Racine S, Murrell BP, Sands JM. Urea may regulate urea transporter protein abundance during osmotic diuresis. Am J Physiol Renal Physiol 288: F188‐F197, 2005.
 297. Kirton CA, Loutzenhiser R. Alterations in basal protein kinase C activity modulate renal afferent arteriolar myogenic reactivity. Am J Physiol 275: H467‐H475, 1998.
 298. Kitamura K, Tanaka T, Kato J, Eto T, Tanaka K. Regional distribution of immunoreactive endothelin in porcine tissue: Abundance in inner medulla of kidney. Biochem Biophys Res Commun 161: 348‐352, 1989.
 299. Kitamura K, Yamazaki J. Chloride channels and their functional roles in smooth muscle tone in the vasculature. Jpn J Pharmacol 85: 351‐357, 2001.
 300. Klein JD, Gunn RB, Roberts BR, Sands JM. Down‐regulation of urea transporters in the renal inner medulla of lithium‐fed rats. Kidney Int 61: 995‐1002, 2002.
 301. Knepper MA, Danielson RA, Saidel GM, Post RS. Quantitative analysis of renal medullary anatomy in rats and rabbits. Kidney Int 12: 313‐323, 1977.
 302. Knox FG, Ritman EL, Romero JC. Intrarenal distribution of blood flow: Evolution of a new approach to measurement. Kidney Int 25: 473‐479, 1984.
 303. Koepsell H, Nicholson WA, Kriz W, Hohling HJ. Measurements of exponential gradients of sodium and chlorine in the rat kidney medulla using the electron microprobe. Pflugers Arch 350: 167‐184, 1974.
 304. Kohagura K, Arima S, Endo Y, Chiba Y, Ito O, Abe M, Omata K, Ito S. Involvement of cytochrome P450 metabolites in the vascular action of angiotensin II on the afferent arterioles. Hypertens Res 24: 551‐557, 2001.
 305. Kohagura K, Endo Y, Ito O, Arima S, Omata K, Ito S. Endogenous nitric oxide and epoxyeicosatrienoic acids modulate angiotensin II‐induced constriction in the rabbit afferent arteriole. Acta Physiol Scand 168: 107‐112, 2000.
 306. Kohan DE. The renal medullary endothelin system in control of sodium and water excretion and systemic blood pressure. Curr Opin Nephrol Hypertens 15: 34‐40, 2006.
 307. Kohan DE. Biology of endothelin receptors in the collecting duct. Kidney Int 76: 481‐486, 2009.
 308. Kone BC, Baylis C. Biosynthesis and homeostatic roles of nitric oxide in the normal kidney. Am J Physiol 272: F561‐F578, 1997.
 309. Konishi F, Okada Y, Takaoka M, Gariepy CE, Yanagisawa M, Matsumura Y. Role of endothelin ET(B) receptors in the renal hemodynamic and excretory responses to big endothelin‐1. Eur J Pharmacol 451: 177‐184, 2002.
 310. Kotelevtsev Y, Webb DJ. Endothelin as a natriuretic hormone: The case for a paracrine action mediated by nitric oxide. Cardiovasc Res 51: 481‐488, 2001.
 311. Kramer K, Thurau K, Deetjen P. Hemodynamics of kidney medullary substance. Part I. Capillary passage time, blood volume, circulation, tissue hematocrit and oxygen consumption of kidney medullary substance in situ. Pflugers Arch Gesamte Physiol Menschen Tiere 270: 251‐269, 1960.
 312. Kreisberg MS, Silldorff EP, Pallone TL. Localization of adenosine‐receptor subtype mRNA in rat outer medullary descending vasa recta by RT‐PCR. Am J Physiol 272: H1231‐H1238, 1997.
 313. Kriz W. Structural organization of the renal medulla: Comparative and functional aspects. Am J Physiol Regul Integr Comp Physiol 241: R3‐R16, 1981.
 314. Kriz W, Dieterich HJ. The lymphatic system of the kidney in some mammals. Light and electron microscopic investigations. Z Anat Entwicklungsgesch 131: 111‐147, 1970.
 315. Kriz W, Napiwotzky P. Structural and functional aspects of the renal interstitium. Contrib Nephrol 16: 104‐108, 1979.
 316. Krum H, Viskoper RJ, Lacourciere Y, Budde M, Charlon V. The effect of an endothelin‐receptor antagonist, bosentan, on blood pressure in patients with essential hypertension. Bosentan Hypertension Investigators. N Engl J Med 338: 784‐790, 1998.
 317. Kuczeriszka M, Badzynska B, Kompanowska‐Jezierska E. Cytochrome P‐450 monooxygenases in control of renal haemodynamics and arterial pressure in anaesthetized rats. J Physiol Pharmacol 57(Suppl 11): 179‐185, 2006.
 318. Kudlacek PE, Pluznick JL, Ma R, Padanilam B, Sansom SC. Role of hbeta1 in activation of human mesangial BK channels by cGMP kinase. Am J Physiol Renal Physiol 285: F289‐F294, 2003.
 319. Kuhn W, Ryffel R. Herstellung konzentrierter Losungen aus verdunnten durch blosse Membranwirkung. (Ein Modellversuch zur Function der Niere). Hoppe‐Seylers Z Physiol Chem 276: 145‐178, 1942.
 320. Kurtz L, Janssen‐Bienhold U, Kurtz A, Wagner C. Connexin expression in renin‐producing cells. J Am Soc Nephrol 20: 506‐512, 2009.
 321. Kurtz A, Penner R. Angiotensin II induces oscillations of intracellular calcium and blocks anomalous inward rectifying potassium current in mouse renal juxtaglomerular cells. Proc Natl Acad Sci U S A 86: 3423‐3427, 1989.
 322. Kurtz L, Schweda F, De Wit C, Kriz W, Witzgall R, Warth R, Sauter A, Kurtz A, Wagner C. Lack of connexin 40 causes displacement of renin‐producing cells from afferent arterioles to the extraglomerular mesangium. J Am Soc Nephrol 18: 1103‐1111, 2007.
 323. Lameire N, Vanholder R, Ringoir S, Leusen I. Role of medullary hemodynamics in the natriuresis of drug‐induced renal vasodilation in the rat. Circ Res 47: 839‐844, 1980.
 324. Landis EM, Pappenheimer JR. Exchange of substances through the capillary wall. In: Handbook of Physiology. Circulation, Washington, DC: American Physiological Society, 1963, p. 962‐1034.
 325. Large WA, Wang Q. Characteristics and physiological role of the Ca(2+)‐activated Cl‐ conductance in smooth muscle. Am J Physiol 271: C435‐C454, 1996.
 326. Larson TS, Lockhart JC. Restoration of vasa recta hemodynamics and pressure natriuresis in SHR by L‐arginine. Am J Physiol‐Renal Physiol 268: F907‐F912, 1995.
 327. Larsson C, Anggard E. Increased juxtamedullary blood flow on stimulation of intrarenal prostaglandin biosynthesis. Eur J Pharmacol 25: 326‐334, 1974.
 328. Lassen NA, Longley JB, Lilienfield LS. Concentration of albumin in renal papilla. Science 128: 720‐721, 1958.
 329. Layton AT. Role of UTB urea transporters in the urine concentrating mechanism of the rat kidney. Bull Math Biol 69: 887‐929, 2007.
 330. Layton HE. Mathematical models of the mammalian urine concentrating mechanism. In: Layton HE, Weinstein AM, editors. Membrane Transport and Renal Physiology, The IMA Volumes in Mathematics and Its Applications New York: Springer‐Verlag, 2009, p. 233‐272.
 331. Layton AT, Layton HE. A region‐based mathematical model of the urine concentrating mechanism in the rat outer medulla. I. Formulation and base‐case results. Am J Physiol Renal Physiol 289: F1346‐F1366, 2005a.
 332. Layton AT, Layton HE. A region‐based mathematical model of the urine concentrating mechanism in the rat outer medulla. II. Parameter sensitivity and tubular inhomogeneity. Am J Physiol Renal Physiol 289: F1367‐F1381, 2005b.
 333. Layton AT, Pannabecker TL, Dantzler WH, Layton HE. Two modes for concentrating urine in rat inner medulla. Am J Physiol Renal Physiol 287: F816‐F839, 2004.
 334. LeBrie SJ. Renal peritubular capillary permeability to macromolecules. Am J Physiol 213: 1225‐1232, 1967.
 335. Lee‐Kwon W, Goo JH, Zhang Z, Silldorff EP, Pallone TL. Vasa recta voltage‐gated Na+ channel Nav1.3 is regulated by calmodulin. Am J Physiol Renal Physiol 292: F404‐F414, 2007.
 336. Lee‐Kwon W, Wade JB, Zhang Z, Pallone TL, Weinman E. Expression of TRPC 4 channel protein that interacts with NHERF‐2 in rat descending vasa recta. Am J Physiol Cell Physiol 288: C942‐C949, 2005.
 337. Leichtle A, Rauch U, Albinus M, Benohr P, Kalbacher H, Mack AF, Veh RW, Quast U, Russ U. Electrophysiological and molecular characterization of the inward rectifier in juxtaglomerular cells from rat kidney. J Physiol 560: 365‐376, 2004.
 338. Lemley KV, Kriz W. Cycles and separations: The histotopography of the urinary concentrating process. Kidney Int 31: 538‐548, 1987.
 339. Lemley KV, Kriz W. Anatomy of the renal interstitium. Kidney Int 39: 370‐381, 1991.
 340. Lemley KV, Schmitt SL, Holliger C, Dunn MJ, Robertson CR, Jamison RL. Prostaglandin synthesis inhibitors and vasa recta erythrocyte velocities in the rat. Am J Physiol 247: F562‐F567, 1984.
 341. Leonard BL, Evans RG, Navakatikyan MA, Malpas SC. Differential neural control of intrarenal blood flow. Am J Physiol Reg Int Comp Physiol 279: R907‐R916, 2000.
 342. Leong CL, Anderson WP, O’Connor PM, Evans RG. Evidence that renal arterial‐venous oxygen shunting contributes to dynamic regulation of renal oxygenation. Am J Physiol Renal Physiol 292: F1726‐F1733, 2007.
 343. Lerman LO, Rodriguez‐Porcal M, Romero JC. The development of x‐ray imaging to study renal function. Kidney Int 55: 400‐416, 1999.
 344. Li C, Klein JD, Wang W, Knepper MA, Nielsen S, Sands JM, Frokiaer J. Altered expression of urea transporters in response to ureteral obstruction. Am J Physiol Renal Physiol 286: F1154‐F1162, 2004.
 345. Li L, Wu J, Jiang C. Differential expression of Kir6.1 and SUR2B mRNAs in the vasculature of various tissues in rats. J Membr Biol 196: 61‐69, 2003.
 346. Li N, Yi F, dos Santos EA, Donley DK, Li PL. Role of renal medullary heme oxygenase in the regulation of pressure natriuresis and arterial blood pressure. Hypertension 49: 148‐154, 2007.
 347. Liao Y, Day KH, Damon DN, Duling BR. Endothelial cell‐specific knockout of connexin 43 causes hypotension and bradycardia in mice. Proc Natl Acad Sci U S A 98: 9989‐9994, 2001.
 348. Liard JF. L‐NAME antagonizes vasopressin V2‐induced vasodilatation in dogs. Am J Physiol 266: H99‐106, 1994.
 349. Lilienfield LS, Maganzini HC, Bauer MH. Blood flow in the renal medulla. Circ Res 9: 614‐617, 1961.
 350. Lilienfield LS, Rose JC, Lassen NA. Diverse distribution of red cells and albumin in the dog kidney. Circ Res 6: 810‐815, 1958.
 351. Lim SW, Li C, Sun BK, Han KH, Kim WY, Oh YW, Lee JU, Kador PF, Knepper MA, Sands JM, Kim J, Yang CW. Long‐term treatment with cyclosporine decreases aquaporins and urea transporters in the rat kidney. Am J Physiol Renal Physiol 287: F139‐F151, 2004.
 352. Little TL, Beyer EC, Duling BR. Connexin 43 and connexin 40 gap junctional proteins are present in arteriolar smooth muscle and endothelium in vivo. Am J Physiol 268: H729‐H739, 1995.
 353. Liu KL. Regulation of renal medullary circulation by the Renin‐Angiotensin system in genetically hypertensive rats. Clin Exp Pharmacol Physiol 36: 455‐461, 2009.
 354. Lopez R, Llinas MT, Roig F, Salazar FJ. Role of nitric oxide and cyclooxygenase‐2 in regulating the renal hemodynamic response to norepinephrine. Am J Physiol Regul Integr Comp Physiol 284: R488‐R493, 2003.
 355. Lorenz JN, Schnermann J, Brosius FC, Briggs JP, Furspan PB. Intracellular ATP can regulate afferent arteriolar tone via ATP‐sensitive K+ channels in the rabbit. J Clin Invest 90: 733‐740, 1992.
 356. Loutzenhiser K, Loutzenhiser R. Angiotensin II‐induced Ca(2+) influx in renal afferent and efferent arterioles: Differing roles of voltage‐gated and store‐operated Ca(2+) entry. Circ Res 87: 551‐557, 2000.
 357. Loutzenhiser R. Inward rectifier currents in pericytes. Am J Physiol Regul Integr Comp Physiol 290: R1598‐R1600, 2006.
 358. Loutzenhiser R, Chilton L, Trottier G. Membrane potential measurements in renal afferent and efferent arterioles: Actions of angiotensin II. Am J Physiol‐Renal Physiol 273: F307‐F314, 1997.
 359. Loutzenhiser R, Epstein M, Hayashi K, Horton C. Direct visualization of effects of endothelin on the renal microvasculature. Am J Physiol 258: F61‐F68, 1990.
 360. Loutzenhiser R, Epstein M, Horton C. Inhibition by diltiazem of pressure‐induced afferent vasoconstriction in the isolated perfused rat kidney. Am J Cardiol 59: 72A‐75A, 1987.
 361. Loutzenhiser R, Hayashi K, Epstein M. Divergent effects of KCl‐induced depolarization on afferent and efferent arterioles. Am J Physiol 257: F561‐F564, 1989.
 362. Loutzenhiser RD, Parker MJ. Hypoxia inhibits myogenic reactivity of renal afferent arterioles by activating ATP‐sensitive K+ channels. Circ Res 74: 861‐869, 1994.
 363. Lu SH, Mattson DL, Cowley AW Jr. Renal medullary captopril delivery lowers blood pressure in spontaneously hypertensive rats. Hypertension 23: 337‐345, 1994.
 364. Lu SH, Roman RJ, Mattson DL, Cowley AW Jr. Renal medullary interstitial infusion of diltiazem alters sodium and water excretion in rats. Am J Physiol Reg Int Comp Physiol 263: R1064‐R1070, 1992.
 365. Ma R, Du J, Sours S, Ding M. Store‐operated Ca2+ channel in renal microcirculation and glomeruli. Exp Biol Med (Maywood) 231: 145‐153, 2006.
 366. Ma YH, Gebremedhin D, Schwartzman ML, Falck JR, Clark JE, Masters BS, Harder DR, Roman RJ. 20‐Hydroxyeicosatetraenoic acid is an endogenous vasoconstrictor of canine renal arcuate arteries. Circ Res 72: 126‐136, 1993.
 367. Macey RI, Yousef LW. Osmotic stability of red cells in renal circulation requires rapid urea transport. Am J Physiol 254: C669‐C674, 1988.
 368. MacPhee PJ, Michel CC. Fluid uptake from the renal medulla into the ascending vasa recta in anaesthetized rats. J Physiol 487: 169‐183, 1995a.
 369. MacPhee PJ, Michel CC. Subatmospheric closing pressures in individual microvessels of rats and frogs. J Physiol 484(Pt 1): 183‐187, 1995b.
 370. Maier KG, Roman RJ. Cytochrome P450 metabolites of arachidonic acid in the control of renal function. Curr Opin Nephrol Hypertens 10: 81‐87, 2001.
 371. Majid DS, Williams A, Navar LG. Inhibition of nitric oxide synthesis attenuates pressure‐induced natriuretic responses in anesthetized dogs. Am J Physiol Renal Physiol 264: F79‐F87, 1993.
 372. Majid DSA, Godfrey M, Navar LG. Pressure natriuresis and renal medullary blood flow in dogs. Hypertension 29: 1051‐1057, 1997.
 373. Majid DSA, Godfrey M, Omoro S. Pressure natriuresis and autoregulation of inner medullary blood flow in canine kidney. Hypertension 29 [part 2]: 210‐215, 1997.
 374. Majid DSA, Navar LG. Medullary blood flow responses to changes in arterial pressure in canine kidney. Am J Physiol 270: F833‐F838, 1996.
 375. Majid DSA, Omoro SA, Chin SY, Navar LG. Intrarenal nitric oxide activity and pressure natriuresis in anesthetized dogs. Hypertension 32: 266‐272, 1998.
 376. Majid DSA, Said KE, Omoro SA, Navar LG. Nitric oxide dependency of arterial pressure‐induced changes in renal interstitial hydrostatic pressure in dogs. Circ Res 88: 347‐351, 2001.
 377. Makino A, Skelton MM, Zou AP, Cowley AW Jr. Increased renal medullary H2O2 leads to hypertension. Hypertension 42: 25‐30, 2003.
 378. Makino A, Skelton MM, Zou AP, Roman RJ, Cowley AW Jr. Increased renal medullary oxidative stress produces hypertension. Hypertension 39: 667‐672, 2002.
 379. Marchand GR, Ott CE, Lang FC, Greger RF, Knox FG. Effect of secretin on renal blood flow, interstitial pressure, and sodium excretion. Am J Physiol Renal Physiol 232: F147‐F151, 1977.
 380. Marchetti J, Praddaude F, Rajerison R, Ader JL, Alhenc‐Gelas F. Bradykinin attenuates the [Ca(2+)](i) response to angiotensin II of renal juxtamedullary efferent arterioles via an EDHF. Br J Pharmacol 132: 749‐759, 2001.
 381. Marsh DJ, Azen SP. Mechanism of NaCl reabsorption by hamster thin ascending limbs of Henle's loop. Am J Physiol 228: 71‐79, 1975.
 382. Marsh DJ, Segel LA. Analysis of countercurrent diffusion exchange in blood vessels of the renal medulla. Am J Physiol 221: 817‐828, 1971.
 383. Martens JR, Gelband CH. Alterations in rat interlobar artery membrane potential and K +channels in genetic and nongenetic hypertension. Circ Res 79: 295‐301, 1996.
 384. Martens JR, Gelband CH. Ion channels in vascular smooth muscle: Alterations in essential hypertension. Proc Soc Exp Biol Med 218: 192‐203, 1998.
 385. Matsuda H, Hayashi K, Arakawa K, Kubota E, Honda M, Tokuyama H, Suzuki H, Yamamoto T, Kajiya F, Saruta T. Distinct modulation of superficial and juxtamedullary arterioles by prostaglandin in vivo. Hypertens Res 25: 901‐910, 2002.
 386. Matsunaga H, Yamashita N, Miyajima Y, Okuda T, Chang H, Ogata E, Kurokawa K. Ion channel activities of cultured rat mesangial cells. Am J Physiol 251: F808‐F814, 1991.
 387. Mattson DL. Importance of the renal medullary circulation in the control of sodium excretion and blood pressure. Am J Physiol Regul Integr Comp Physiol 284: R13‐R27, 2003.
 388. Mattson DL, Bellehumeur TG. Neural nitric oxide synthase in the renal medulla and blood pressure regulation. Hypertension 28: 297‐303, 1996.
 389. Mattson DL, Cowley AW Jr. Kinin actions on renal papillary blood flow and sodium excretion. Hypertension 21: 961‐965, 1993.
 390. Mattson DL, Cowley AW Jr. Renal mechanisms of hypertension. Current Opin Nephrol Hypertens 8: 217‐224, 1999.
 391. Mattson DL, Lu S, Cowley AW Jr. Role of nitric oxide in the control of the renal medullary circulation. Clin Exp Pharmacol Physiol 24: 587‐590, 1997.
 392. Mattson DL, Lu S, Nakanishi K, Papanek PE, Cowley AW Jr. Effect of chronic renal medullary nitric oxide inhibition on blood pressure. Am J Physiol 266: H1918‐H1926, 1994.
 393. Mattson DL, Lu S, Roman RJ, Cowley AW Jr. Relationship between renal perfusion pressure and blood flow in different regions of the kidney. Am J Physiol Regul Integr Comp Physiol 264: R578‐R583, 1993.
 394. Mattson DL, Maeda CY, Bachman TD, Cowley AW Jr. Inducible nitric oxide synthase and blood pressure. Hypertension 31(part 1): 15‐20, 1998.
 395. Mattson DL, Meister CJ. Renal cortical and medullary blood flow responses to L‐NAME and ANG II in wild‐type, nNOS null mutant, and eNOS null mutant mice. Am J Physiol Regul Integr Comp Physiol 289: R991‐R997, 2005.
 396. Mattson DL, Meister CJ. Sodium sensitivity of arterial blood pressure in L‐NAME hypertensive but not eNOS knockout mice. Am J Hypertens 19: 327‐329, 2006.
 397. Mattson DL, Raff H, Roman RJ. Influence of angiotensin II on pressure natriuresis and renal hemodynamics in volume‐expanded rats. Am J Physiol Reg Int Comp Physiol 260: R1200‐R1209, 1991.
 398. Mattson DL, Roman RJ, Cowley AW Jr. Role of nitric oxide in renal papillary blood flow and sodium excretion in the rat. Hypertension 19: 766‐769, 1992.
 399. Mattson DL, Wu F. Control of arterial blood pressure and renal sodium excretion by nitric oxide synthase in the renal medulla. Acta Physiol Scand 168: 149‐154, 2000a.
 400. Mattson DL, Wu F. Nitric oxide synthase activity and isoforms in rat renal vasculature. Hypertension 35: 337‐341, 2000b.
 401. McDonough AA. Mechanisms of proximal tubule sodium transport regulation that link extracellular fluid volume and blood pressure. Am J Physiol Reg Int Comp Physiol 298: R851‐R861, 2010.
 402. McCoy DE, Bhattacharya S, Olson BA, Levier DG, Arend LJ, Speilman WS. The renal adenosine system: Structure, function and regulation. Sem Nephrol 13: 31‐40, 1993.
 403. McKee M, Scavone C, Nathanson JA. Nitric oxide, cGMP, and hormone regulation of active sodium transport. Proc Natl Acad Sci U S A 91: 12056‐12060, 1994.
 404. McNeely EA, Pallone TL, Deen WM, Robertson CR. Models of the medullary microcirculation. Kidney Int 31: 662‐667, 1987.
 405. Mendez RE, Dunn BR, Troy JL, Brenner BM. Atrial natriuretic peptide and furosemide effects on hydraulic pressure in the renal papilla. Kidney Int 34: 36‐42, 1988.
 406. Meng S, Roberts LJ, Cason GW, Curry TS, Manning RD Jr. Superoxide dismutase and oxidative stress in Dahl salt‐sensitive and ‐resistant rats. Am J Physiol Regul Integr Comp Physiol 283: R732‐R738, 2002.
 407. Metzger F, Quast U. Binding of [3H]‐P1075, an opener of ATP‐sensitive K+ channels, to rat glomerular preparations. Naunyn Schmiedebergs Arch Pharmacol 354: 452‐459, 1996.
 408. Michel CC. Renal medullary microcirculation: Architecture and exchange. Microcirculation 2: 125‐139, 1995.
 409. Mink D, Schiller A, Kriz W, Taugner R. Interendothelial junctions in kidney vessels. Cell Tissue Res 236: 567‐576, 1984.
 410. Mitchell KD, Navar LG. Tubuloglomerular feedback responses during peritubular infusions of calcium channel blockers. Am J Physiol 258: F537‐F544, 1990.
 411. Miyamoto M, Yagil Y, Larson T, Robertson C, Jamison RL. Effects of intrarenal adenosine on renal function and medullary blood flow in the rat. Am J Physiol 255: F1230‐F1234, 1988.
 412. Miyata A, Park F, Li XF, Cowley AW Jr. Distribution of angiotensin AT1 and AT2 receptor subtypes in the rat kidney. Am J Physiol 277: F437‐F446, 1999.
 413. Miyata N, Cowley AW Jr. Renal intramedullary infusion of L‐arginine prevents reduction of medullary blood flow and hypertension in Dahl salt‐sensitive rats. Hypertension 33 [part II]: 446‐450, 1999.
 414. Miyata N, Zou AP, Mattson DL, Cowley AW Jr. Renal medullary interstitial infusion of L‐arginine prevents hypertension in Dahl salt‐sensitive rats. Am J Physiol 275: R1667‐R1673, 1998.
 415. Moffat DB. Extravascular protein in the renal medulla. Q J Exp Physiol Cogn Med Sci 54: 60‐67, 1969.
 416. Moffat DB, Creasey M. The fine structure of the intra‐arterial cushions at the origins of the juxtamedullary afferent arterioles in the rat kidney. J Anat 110: 409‐419, 1971.
 417. Moffat DB, Fourman J. The vascular pattern of the rat kidney. J Anatomy 97: 543‐553, 1963.
 418. Molero MM, Giulumian AD, Reddy VB, Ludwig LM, Pollock JS, Pollock DM, Rusch NJ, Fuchs LC. Decreased endothelin binding and [Ca2+]i signaling in microvessels of DOCA‐salt hypertensive rats. J Hypertens 20: 1799‐1805, 2002.
 419. Morgan T, Berliner RW. Permeability of the loop of Henle, vasa recta, and collecting duct to water, urea, and sodium. Am J Physiol 215: 108‐115, 1968.
 420. Mori T, Cowley AW Jr. Angiotensin II‐NAD(P)H oxidase‐stimulated superoxide modifies tubulovascular nitric oxide cross‐talk in renal outer medulla. Hypertension 42: 588‐593, 2003.
 421. Mori T, Cowley AW Jr., Ito S. Molecular mechanisms and therapeutic strategies of chronic renal injury: Physiological role of angiotensin II‐induced oxidative stress in renal medulla. J Pharmacol Sci 100: 2‐8, 2006.
 422. Mori T, Dickhout JG, Cowley AW Jr. Vasopressin increases intracellular NO concentration via Ca(2+) signaling in inner medullary collecting duct. Hypertension 39: 465‐469, 2002.
 423. Mori T, O'Connor PM, Abe M, Cowley AW Jr. Enhanced superoxide production in renal outer medulla of Dahl salt‐sensitive rats reduces nitric oxide tubular‐vascular cross‐talk. Hypertension 49: 1336‐1341, 2007.
 424. Moridani BA, Kline RL. Effect of endogenous L‐arginine on the measurement of nitric oxide synthase activity in the rat kidney. Canadian Jf Physiol Pharmacol 74: 1210‐1214, 1996.
 425. Mule S, De Cesare A, Lucidarme O, Frouin F, Herment A. Regularized estimation of contrast agent attenuation to improve the imaging of microbubbles in small animal studies. Ultrasound Med Biol 34: 938‐948, 2008.
 426. Muirhead EE. The medullipin system of blood pressure control. Am J Hypertension 4: 556S‐568S, 1991.
 427. Muirhead EE, Germain GS, Armstrong FB, Brooks B, Leach BE, Byers LW, Pitcock JA, Brown P. Endocrine‐type antihypertensive function of renomedullary interstitial cells. Kidney Int 8: S271‐S282, 1975.
 428. Muirhead EE, Stirman JA, Jones F. Renal autoexplantation and protection against renoprival hypertensive cardiovascular disease and hemolysis. J Clin Invest 39: 266‐281, 1960.
 429. Naicker S, Bhoola KD. Endothelins: Vasoactive modulators of renal function in health and disease. Pharmacol Ther 90: 61‐88, 2001.
 430. Naitoh M, Suzuki H, Murakami M, Matsumoto A, Ichihara A, Nakamoto H, Yamamura Y, Saruta T. Arginine vasopressin produces renal vasodilation via V2 receptors in conscious dogs. Am J Physiol Regul Integr Comp Physiol 265: R934‐R942, 1993.
 431. Nakanishi K, Mattson DL, Cowley AW Jr. Role of renal medullary blood flow in the development of L‐NAME hypertension in rats. Am J Physiol Regul Integr Comp Physiol 268: R317‐R323, 1995.
 432. Nakanishi K, Mattson DL, Gross V, Roman RJ, Cowley AW Jr. Control of renal medullary blood flow by vasopressin V1 and V2 receptors. Am J Physiol Regul Integr Comp Physiol 269: R193‐R200, 1995.
 433. Nakano D, Pollock JS, Pollock DM. Renal medullary ETB receptors produce diuresis and natriuresis via NOS1. Am J Physiol Renal Physiol 294: F1205‐F1211, 2008.
 434. Nasrallah R, Clark J, Hebert RL. Prostaglandins in the kidney: Developments since Y2K. Clin Sci (Lond) 113: 297‐311, 2007.
 435. Navar LG. The intrarenal renin‐angiotensin system in hypertension. Kidney Int 65: 1522‐1532, 2004.
 436. Navar LG, Arendshorst WJ, Pallone TL, Inscho EW, Imig JD, Bell PD. The renal microcirculation, Part V. In: Tuma RF, Duran WN, Ley K, editors. Handbook of Physiology. San Diego, CA: American Physiological Society Press, 2008, Chpt 13, p. 550‐683.
 437. Navar LG, Bell PD, Evan AP. The regulation of glomerular filtration rate in mammalian kidneys. In: Andreoli TE, Hoffman J, Fanestil D, Shultz SG, editors. Physiology of Membrane Disorders. New York: Plenum Medical Book Co., 1986, p. 637‐667.
 438. Navar LG, Champion WJ, Thomas CE. Effects of calcium channel blockade on renal vascular resistance responses to changes in perfusion pressure and angiotensin‐converting enzyme inhibition in dogs. Circ Res 58: 874‐881, 1986.
 439. Navar LG, Evan AP, Rosivall L. Microcirculation of the kidneys. In: Mortillaro N, editor. The Physiology and Pharmacology of the Microcirculation. New York: Academic Press, 1983, p. 397‐488.
 440. Navar LG, Ichihara A, Chin SY, Imig JD. Nitric oxide‐angiotensin II interactions in angiotensin II‐dependent hypertension. Acta Physiol Scand 168: 139‐147, 2000.
 441. Navar LG, Inscho EW, Imig JD, Mitchell KD. Heterogeneous activation mechanisms in the renal microvasculature. Kidney Int Suppl 67: S17‐S21, 1998.
 442. Navar LG, Inscho EW, Majid SA, Imig JD, Harrison‐Bernard LM, Mitchell KD. Paracrine regulation of the renal microcirculation. Physiol Rev 76: 425‐536, 1996.
 443. Navar LG, Nishiyama A. Why are angiotensin concentrations so high in the kidney? Curr Opin Nephrol Hypertens 13: 107‐115, 2004.
 444. Navar LG, Prieto‐Carrasquero MC, Kobori H. Molecular aspects of the renal renin‐angiotensin system. In: Re R, DiPette DJ, Schiffrin EL, Sowers JR, editors. Molecular Mechanisms in Hypertension. Taylor & Francis Group, 2006, p. 3‐14.
 445. Nelson MT, Quayle JM. Physiological roles and properties of potassium channels in arterial smooth muscle. Am J Physiol 268: C799‐C822, 1995.
 446. 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.
 447. 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.
 448. Nilius B, Droogmans G, Wondergem R. Transient receptor potential channels in endothelium: Solving the calcium entry puzzle? Endothelium 10: 5‐15, 2003.
 449. Nishiyama A, Inscho EW, Navar LG. Interactions of adenosine A1 and A2a receptors on renal microvascular reactivity. Am J Physiol Renal Physiol 280: F406‐F414, 2001.
 450. Nishiyama A, Majid DSA, Walker III, M, Miyatake A, Navar LG. Renal interstitial ATP responses to changes in arterial pressure during alterations in tubuloglomerular feedback activity. Hypertension 37: 753‐759, 2001.
 451. Nishiyama A, Seth DM, Navar LG. Renal interstitial fluid angiotensin I and angiotensin II concentrations during local angiotensin‐converting enzyme inhibition. J Am Soc Nephrol 13: 2207‐2212, 2002.
 452. Nishiyama A, Seth DM, Navar LG. Angiotensin II type 1 receptor‐mediated augmentation of renal interstitial fluid angiotensin II in angiotensin II‐induced hypertension. J Hypertens 21: 1897‐1903, 2003.
 453. Nitescu N, Grimberg E, Herlitz H, Guron G. Role of endothelin ET(A) and ET(B) receptor subtypes in the regulation of intrarenal blood flow and oxygen tension in rats. Clin Exp Pharmacol Physiol 35: 1227‐1232, 2008.
 454. Nobiling R, Buhrle CP, Hackenthal E, Helmchen U, Steinhausen M, Whalley A, Taugner R. Ultrastructure, renin status, contractile and electrophysiological properties of the afferent glomerular arteriole in the rat hydronephrotic kidney. Virchows Archiv A Pathol Anat 410: 31‐42, 1986.
 455. O'Connor PM, Cowley AW Jr. Vasopressin‐induced nitric oxide production in rat inner medullary collecting duct is dependent on V2 receptor activation of the phosphoinositide pathway. Am J Physiol Renal Physiol 293: F526‐F532, 2007.
 456. O'Connor PM, Kett MM, Anderson WP, Evans RG. Renal medullary tissue oxygenation is dependent on both cortical and medullary blood flow. Am J Physiol Renal Physiol 290: F688‐F694, 2006.
 457. Ogungbade GO, Akinsanmi LA, Jiang H, Oyekan AO. Role of epoxyeicosatrienoic acids in renal functional response to inhibition of NO production in the rat. Am J Physiol Renal Physiol 285: F955‐F964, 2003.
 458. Oliver JJ, Eppel GA, Rajapakse NW, Evans RG. Lipoxygenase and cyclo‐oxygenase products in the control of regional kidney blood flow in rabbits. Clin Exp Pharmacol Physiol 30: 812‐819, 2003.
 459. Omoro SA, Majid DSA, El‐Dahr S, Navar LG. Roles of ANG II bradykinin in the renal regional blood flow responses to ACE inhibition in sodium‐depleted dogs. Am J Physiol Renal Physiol 279: F289‐F293, 2000.
 460. Omoro SA, Majid DSA, El‐Dahr SS, Navar LG. Kinin influences on renal regional blood flow responses to angiotensin‐converting enzyme inhibition in dogs. Am J Physiol‐Renal Physiol 276: F271‐F277, 1999.
 461. Ortiz PA, Garvin JL. Role of nitric oxide in the regulation of nephron transport. Am J Physiol Renal Physiol 282: F777‐F784, 2002a.
 462. Ortiz PA, Garvin JL. Superoxide stimulates NaCl absorption by the thick ascending limb. Am J Physiol Renal Physiol 283: F957‐F962, 2002b.
 463. Ott CE, Knox FG. Tissue pressures and fluid dynamics in the kidney. Federation Proceedings 35: 1872‐1875, 1976.
 464. Ott CE, Navar LG, Guyton AC. Pressures in static and dynamic states from capsules implanted in the kidney. Am J Physiol 221: 394‐400, 1971.
 465. Oyekan AO. Contributions of nitric oxide and prostanoids and their signaling pathways to the renal medullary vasodilator effect of U46619 (9‐11‐dideoxy‐11 alpha,9a‐epoxymethano‐prostaglandin F(2a)) in the rat. J Pharmacol Exp Ther 304: 507‐512, 2003.
 466. Oyekan AO. Differential effects of 20‐hydroxyeicosatetraenoic acid on intrarenal blood flow in the rat. J Pharmacol Exp Ther 313: 1289‐1295, 2005.
 467. Ozawa Y, Hayashi K, Nagahama T, Fujiwara K, Saruta T. Effect of T‐type selective calcium antagonist on renal microcirculation: Studies in the isolated perfused hydronephrotic kidney. Hypertension 38: 343‐347, 2001.
 468. Ozawa Y, Hayashi K, Wakino S, Kanda T, Homma K, Takamatsu I, Tatematsu S, Yoshioka K, Saruta T. Free radical activity depends on underlying vasoconstrictors in renal microcirculation. Clin Exp Hypertens 26: 219‐229, 2004.
 469. Paliege A, Mizel D, Medina C, Pasumarthy A, Huang YG, Bachmann S, Briggs JP, Schnermann JB, Yang T. Inhibition of nNOS expression in the macula densa by COX‐2‐derived prostaglandin E(2). Am J Physiol Renal Physiol 287: F152‐F159, 2004.
 470. Pallone TL. Resistance of ascending vasa recta to transport of water. Am J Physiol 260: F303‐F310, 1991a.
 471. Pallone TL. Transport of sodium chloride and water in rat ascending vasa recta. Am J Physiol 261: F519‐F525, 1991b.
 472. Pallone TL. Effect of sodium chloride gradients on water flux in rat descending vasa recta. J Clin Invest 87: 12‐19, 1991c.
 473. Pallone TL. Molecular sieving of albumin by the ascending vasa recta wall. J Clin Invest 90: 30‐34, 1992.
 474. Pallone TL. Characterization of the urea transporter in outer medullary descending vasa recta. Am J Physiol 267: R260‐R267, 1994a.
 475. Pallone TL. Extravascular protein in the renal medulla: Analysis by two methods. Am J Physiol Regul Integr Comp Physiol 266: R1429‐R1436, 1994b.
 476. Pallone TL. Vasoconstriction of outer medullary vasa recta by angiotensin II is modulated by prostaglandin E2. Am J Physiol 266: F850‐F857, 1994c.
 477. Pallone TL. Control of renal Na+ excretion by heme oxygenase. Hypertension 49: 23‐24, 2007a.
 478. Pallone TL. Aquaporin 1, urea transporters, and renal vascular bundles. J Am Soc Nephrol 18: 2798‐2800, 2007b.
 479. Pallone TL, Cao C. Renal cortical and medullary microcirculations: Structure and function. In: Alpern RJ, Hebert SC, editors. Seldin and Giebisch's The Kidney: Physiology and Pathophysiology, (4th ed). Elsevier, 2008, chapt. 23, p. 627‐670
 480. Pallone TL, Cao C, Zhang Z. Inhibition of K+ conductance in descending vasa recta pericytes by ANG II. Am J Physiol Renal Physiol 287: F1213‐F1222, 2004.
 481. Pallone TL, Edwards A, Ma T, Silldorff EP, Verkman AS. Requirement of aquaporin‐1 for NaCl‐driven water transport across descending vasa recta. J Clin Invest 105: 215‐222, 2000.
 482. Pallone TL, Huang JM. Control of descending vasa recta pericyte membrane potential by angiotensin II. Am J Physiol Renal Physiol 282: F1064‐F1074, 2002.
 483. Pallone TL, Jamison RL. Effect of ureteral excision on inner medullary solute concentration in rats. Am J Physiol 255: F1225‐F1229, 1988.
 484. Pallone TL, Kishore BK, Nielsen S, Agre P, Knepper MA. Evidence that aquaporin‐1 mediates NaCl‐induced water flux across descending vasa recta. Am J Physiol 272: F587‐F596, 1997.
 485. Pallone TL, Mattson DL. Role of nitric oxide in regulation of the renal medulla in normal and hypertensive kidneys. Curr Opin Nephrol Hypertens 11: 93‐98, 2002.
 486. Pallone TL, Morgenthaler TI, Deen WM. Analysis of microvascular water and solute exchanges in the renal medulla. Am J Physiol 247: F303‐F315, 1984.
 487. Pallone TL, Nielsen S, Silldorff EP, Yang S. Diffusive transport of solute in the rat medullary microcirculation. Am J Physiol 269: F55‐F63, 1995.
 488. Pallone TL, Robertson CR, Jamison RL. Renal medullary microcirculation. Physiol Rev 70: 885‐920, 1990.
 489. Pallone TL, Silldorff EP. Pericyte regulation of renal medullary blood flow. Exp Nephrol 9: 165‐170, 2001.
 490. Pallone TL, Silldorff EP, Cheung JY. Response of isolated rat descending vasa recta to bradykinin. Am J Physiol 274: H752‐H759, 1998.
 491. Pallone TL, Silldorff EP, Zhang Z. Inhibition of calcium signaling in descending vasa recta endothelia by ANG II. Am J Physiol Heart Circ Physiol 278: H1248‐H1255, 2000.
 492. Pallone TL, Turner MR. Molecular sieving of small solutes by outer medullary descending vasa recta. Am J Physiol 272: F579‐F586, 1997.
 493. Pallone TL, Turner MR. Ion channel architecture of the renal microcirculation. Current Hypertension Reviews 2: 69‐81, 2006.
 494. Pallone TL, Turner MR, Edwards A, Jamison RL. Countercurrent exchange in the renal medulla. Am J Physiol Regul Integr Comp Physiol 284: R1153‐R1175, 2003.
 495. Pallone TL, Work J, Jamison RL. Resistance of descending vasa recta to the transport of water. Am J Physiol 259: F688‐F697, 1990.
 496. Pallone TL, Work J, Myers RL, Jamison RL. Transport of sodium and urea in outer medullary descending vasa recta. J Clin Invest 93: 212‐222, 1994.
 497. Pallone TL, Yagil Y, Jamison RL. Effect of small‐solute gradients on transcapillary fluid movement in renal inner medulla. Am J Physiol 257: F547‐F553, 1989.
 498. Pallone TL, Zhang Z, Rhinehart K. Physiology of the renal medullary microcirculation. Am J Physiol Renal Physiol 284: F253‐F266, 2003.
 499. 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.
 500. Pannabecker TL, Dahlmann A, Brokl OH, Dantzler WH. Mixed descending‐ and ascending‐type thin limbs of Henle's loop in mammalian renal inner medulla. Am J Physiol Renal Physiol 278: F202‐F208, 2000.
 501. Pannabecker TL, Dantzler WH. Three‐dimensional lateral and vertical relationships of inner medullary loops of Henle and collecting ducts. Am J Physiol Renal Physiol 287: F767‐F774, 2004.
 502. Pannabecker TL, Dantzler WH. Three‐dimensional architecture of inner medullary vasa recta. Am J Physiol Renal Physiol 290: F1355‐F1366, 2006.
 503. Pannabecker TL, Dantzler WH, Layton HE, Layton AT. Role of three‐dimensional architecture in the urine concentrating mechanism of the rat renal inner medulla. Am J Physiol Renal Physiol 295: F1271‐F1285, 2008.
 504. Pappenheimer JR, Kinter WB. Hematocrit ratio of blood within mammalian kidney and its significance for renal hemodynamics. Am J Physiol 185: 377‐390, 1956.
 505. Parekh AB. Store‐operated Ca2+ entry: Dynamic interplay between endoplasmic reticulum, mitochondria and plasma membrane. J Physiol 547: 333‐348, 2003.
 506. Parekh AB, Putney JW Jr. Store‐operated calcium channels. Physiol Rev 85: 757‐810, 2005.
 507. Parekh N, Zou AP. Role of prostaglandins in renal medullary circulation: Response to different vasoconstrictors. Am J Physiol 271: F653‐F658, 1996.
 508. Park F, Mattson DL, Roberts LA, Cowley AW Jr. Evidence for the presence of smooth muscle alpha‐actin within pericytes of the renal medulla. Am J Physiol 273: R1742‐R1748, 1997.
 509. Park F, Mattson DL, Skelton MM, Cowley AW Jr. Localization of the vasopressin V1a and V2 receptors within the renal cortical and medullary circulation. Am J Phyisol 273: R243‐R251, 1997.
 510. Park F, Zou A‐P, Cowley AW Jr. Arginine vasopressin‐mediated stimulation of nitric oxide within the rat renal medulla. Hypertension 32: 896‐901, 1998.
 511. Park WS, Han J, Earm YE. Physiological role of inward rectifier K(+) channels in vascular smooth muscle cells. Pflugers Arch 457: 137‐147, 2008.
 512. Patel AS, Layne S, Watts D, Kirchner KA. L‐arginine improves transmission of perfusion pressure to the renal interstitium of Dahl salt‐sensitive rats. Am J Physiol Reg Int Comp 266: R1730‐R1735, 1994.
 513. Patzak A, Kleinmann F, Lai EY, Kupsch E, Skelweit A, Mrowka R. Nitric oxide counteracts angiotensin II induced contraction in efferent arterioles in mice. Acta Physiol Scand 181: 439‐444, 2004.
 514. Patzak A, Lai EY, Mrowka R, Steege A, Persson PB, Persson AE. AT1 receptors mediate angiotensin II‐induced release of nitric oxide in afferent arterioles. Kidney Int 66: 1949‐1958, 2004.
 515. Patzak A, Persson AE. Angiotensin II‐nitric oxide interaction in the kidney. Curr Opin Nephrol Hypertens 16: 46‐51, 2007.
 516. Perez‐Reyes E. Molecular physiology of low‐voltage‐activated T‐type calcium channels. Physiol Rev 83: 117‐161, 2003.
 517. Perez‐Reyes E. Molecular characterization of T‐type calcium channels. Cell Calcium 40: 89‐96, 2006.
 518. Peti‐Peterdi J, Komlosi P, Fuson AL, Guan Y, Schneider A, Qi Z, Redha R, Rosivall L, Breyer MD, Bell PD. Luminal NaCl delivery regulates basolateral PGE2 release from macula densa cells. J Clin Invest 112: 76‐82, 2003.
 519. Pittner J, Rhinehart K, Pallone TL. Ouabain modulation of endothelial calcium signaling in descending vasa recta. Am J Physiol Renal Physiol 291: F761‐F769, 2006.
 520. Plato CF, Garvin JL. Nitric oxide, endothelin and nephron transport: Potential interactions. Clin Exp Pharmacol Physiol 26: 262‐268, 1999.
 521. Pollock DM. Renal endothelin in hypertension. Curr Opin Nephrol Hypertens 9: 157‐164, 2000.
 522.Pollock DM. Contrasting pharmacological ETB receptor blockade with genetic ETB deficiency in renal responses to big ET‐1. Physiol Genomics 6: 39‐43, 2001.
 523. Pollock DM, Jenkins JM, Cook AK, Imig JD, Inscho EW. L‐type calcium channels in the renal microcirculatory response to endothelin. Am J Physiol Renal Physiol 288: F771‐F777, 2005.
 524. Pomerantz RM, Slotkoff LM, Lilienfield LS. Histochemical and microanatomical differences between renal cortical and medullary interstitium. In: Kass EH, editor. Progress in Pyelonephritis. Philadelphia: F.A. Davis Co., 1965, p. 434.
 525. Popp R, Brandes RP, Ott G, Busse R, Fleming I. Dynamic modulation of interendothelial gap junctional communication by 11,12‐epoxyeicosatrienoic acid. Circ Res 90: 800‐806, 2002.
 526. Pries AR, Gaehtgens P. Generalization of the Fahraeus principle for microvessel networks. Am J Physiol Heart Circ Physiol 251: H1324‐H1332, 1986.
 527. Prior HM, Yates MS, Beech DJ. Functions of large conductance Ca2+‐activated (BKCa), delayed rectifier (KV) and background K+ channels in the control of membrane potential in rabbit renal arcuate artery. J Physiol 511(Pt 1): 159‐169, 1998.
 528. Prior HM, Yates MS, Beech DJ. Role of K+ channels in A2A adenosine receptor‐mediated dilation of the pressurized renal arcuate artery. British J Pharmacol 126: 494‐500, 1999.
 529. Promeneur D, Bankir L, Hu MC, Trinh‐Trang‐Tan MM. Renal tubular and vascular urea transporters: Influence of antidiuretic hormone on messenger RNA expression in Brattleboro rats. J Am Soc Nephrol 9: 1359‐1366, 1998.
 530. Promeneur D, Rousselet G, Bankir L, Bailly P, Cartron JP, Ripoche P, Trinh‐Trang‐Tan MM. Evidence for distinct vascular and tubular urea transporters in the rat kidney. J Am Soc Nephrol 7: 852‐860, 1996.
 531. Proud D, Knepper MA, Pisano JJ. Distribution of immunoreactive kallikrein along the rat nephron. Am J Physiol 244: F510‐F515, 1983.
 532. Qi Z, Hao CM, Langenbach RI, Breyer RM, Redha R, Morrow JD, Breyer MD. Opposite effects of cyclooxygenase‐1 and ‐2 activity on the pressor response to angiotensin II. J Clin Invest 110: 61‐69, 2002.
 533. Quayle JM, Nelson MT, Standen NB. ATP‐sensitive and inwardly rectifying potassium channels in smooth muscle. Physiol Rev 77: 1165‐1232, 1997.
 534. Quilley J, Fulton D, McGiff JC. Hyperpolarizing factors. Biochem Pharmacol 54: 1059‐1070, 1997.
 535. Rajapakse NW, Mattson DL. Role of L‐arginine in nitric oxide production in health and hypertension. Clin Exp Pharmacol Physiol 36: 249‐255, 2009.
 536. Rajapakse NW, Roman RJ, Falck JR, Oliver JJ, Evans RG. Modulation of V1‐receptor‐mediated renal vasoconstriction by epoxyeicosatrienoic acids. Am J Physiol Regul Integr Comp Physiol 287: R181‐R187, 2004.
 537. Rasmussen SN. Step function input to the rat kidney by shifting between auto‐ and alloperfusion. Am J Physiol‐Heart Circul Physiol 233: H488‐H492, 1977.
 538. Rasmussen SN. Red cell and plasma volume flows to the inner medulla of the rat kidney: Determinations by means of a step function input indicator technique. Pflugers Arch 373: 153‐159, 1978.
 539. Renkin EM. Capillary transport of macromolecules: Pores and other endothelial pathways. J Appl Physiol 58: 315‐325, 1985.
 540. Reslerova M, Loutzenhiser R. Divergent mechanisms of ATP‐sensitive K+ channel‐induced vasodilation in renal afferent and efferent arterioles. Evidence of L‐type Ca2+ channel‐dependent and ‐independent actions of pinacidil. Circ Res 77: 1114‐1120, 1995.
 541. Reslerova M, Loutzenhiser R. Renal microvascular actions of calcitonin gene‐related peptide. Am J Physiol 274: F1078‐F1085, 1998.
 542. Rettig R, Bandelow N, Patschan O, Kuttler B, Frey B, Uber A. The importance of the kidney in primary hypertension: Insights from cross‐transplantation. J Human Hypertension 10: 641‐644, 1996.
 543. Rhinehart K, Handelsman CA, Silldorff EP, Pallone TL. ANG II AT2 receptor modulates AT1 receptor‐mediated descending vasa recta endothelial Ca2+ signaling. Am J Physiol Heart Circ Physiol 284: H779‐H789, 2003.
 544. Rhinehart KL, Pallone TL. Nitric oxide generation by isolated descending vasa recta. Am J Physiol Heart Circ Physiol 281: H316‐H324, 2001.
 545. Rhinehart K, Zhang Z, Pallone TL. Ca(2+) signaling and membrane potential in descending vasa recta pericytes and endothelia. Am J Physiol Renal Physiol 283: F852‐F860, 2002.
 546. Roman RJ. Altered pressure‐natriuresis relationship in young spontaneously hypertensive rats. Hypertension 9: III‐130‐III‐136, 1987.
 547. Roman RJ. Pressure‐diuresis in volume‐expanded rats: Tubular reabsorption in superficial and deep nephrons. Hypertension 12: 177‐183, 1988.
 548. Roman RJ. Blood flow measurements in the renal circulation. In: Shepherd AP, Oberg PA, editors. Laser Doppler Flowmetry. Norwell, MA: Kluwer Academic, 1990, p. 289‐304.
 549. Roman RJ, Carmines PK, Loutzenhiser R, Conger JD. Direct studies on the control of the renal microcirculation. J Am Soc Nephrol 2: 136‐149, 1991.
 550. Roman RJ, Cowley AW Jr. Characterization of a new model for the study of pressure natriuresis in the rat. Am J Physiol Renal Physiol 248: F190‐F198, 1985.
 551. Roman RJ, Cowley AW Jr., Garcia‐Estan J, Lombard JH. Pressure‐diuresis in volume‐expanded rats: Cortical and medullary hemodynamics. Hypertension 12: 168‐176, 1988.
 552. Roman RJ, Kaldunski ML. Renal cortical and papillary blood flow in spontaneously hypertensive rats. Hypertension 11: 657‐663, 1988.
 553. Roman RJ, Kaldunski ML, Scicli AG, Carretero OA. Influence of kinins and angiotensin II on the regulation of papillary blood flow. Am J Physiol 255: F690‐F698, 1988.
 554. Roman RJ, Lianos E. Influence of prostaglandins on papillary blood flow and pressure‐natriuretic response. Hypertension 15: 29‐35, 1990.
 555. Roman RJ, Maier KG, Sun CW, Harder DR, Alonso‐Galicia M. Renal and cardiovascular actions of 20‐hydroxyeicosatetraenoic acid and epoxyeicosatrienoic acids. Clin Exp Pharmacol Physiol 27: 855‐865, 2000.
 556. Roman RJ, Mattson DL, Cowley AW Jr. Measurement of regional blood flow in the kidney using laser‐Doppler flowmetry. In: Wang DH, editor. Methods in Molecular Medicine, vol 51: Angiotensin Protocols. Totowa, NJ: Humana Press Inc., 2000.
 557. Roman RJ, Smits C. Laser‐Doppler determination of papillary blood flow in young and adult rats. Am J Physiol 25, F115‐F124, 1985.
 558. Roman RJ, Zou AP. Influence of the renal medullary circulation on the control of sodium excretion. Am J Physiol Reg Int Comp Physiol 265: R963‐R973, 1993.
 559. Romero JC, Knox FG. Mechanisms underlying pressure‐related natriuresis: The role of the renin‐angiotensin and prostaglandin systems: State of the art lecture. Hypertension 11: 724‐738, 1988.
 560. Rudenstam J, Bergström G, Taghipour K, Göthberg G, Karlström G. Efferent renal sympathetic nerve stimulation in vivo. Effects on regional renal hemodynamics in the Wistar rat studied by laser‐Doppler technique. Acta Physiol Scand 154: 387‐394, 1995.
 561. Rudichenko VM, Beierwaltes WH. Arginine vasopressin‐induced renal vasodilation mediated by nitric oxide. J Vasc Res 32: 100‐105, 1995.
 562. Sadowski J, Badzynska B. Specific features and roles of renal circulation: Angiotensin II revisited. J Physiol Pharmacol 57(Suppl 11): 169‐178, 2006.
 563. Sadowski J, Badzynska B. Intrarenal vasodilator systems: NO, prostaglandins and bradykinin. An integrative approach. J Physiol Pharmacol 59(Suppl 9): 105‐119, 2008.
 564. Sadowski J, Kompanowska‐Jezierska E, Dobrowolski L, Walkowska A, Badzynska B. Simultaneous recording of tissue ion content and blood flow in rat renal medulla: Evidence on interdependence. Am J Physiol 273: F658‐F662, 1997.
 565. Salomonsson M, Arendshorst WJ. Calcium recruitment in renal vasculature: NE effects on blood flow and cytosolic calcium concentration. Am J Physiol 276: F700‐F710, 1999.
 566. Salomonsson M, Arendshorst WJ. Norepinephrine‐induced calcium signaling pathways in afferent arterioles of genetically hypertensive rats. Am J Physiol Renal Physiol 281: F264‐F272, 2001.
 567. Salomonsson M, Gustafsson F, Andreasen D, Jensen BL, Holstein‐Rathlou NH. Local electric stimulation causes conducted calcium response in rat interlobular arteries. Am J Physiol Renal Physiol 283: F473‐F480, 2002.
 568. Salomonsson M, Sorensen CM, Arendshorst WJ, Steendahl J, Holstein‐Rathlou NH. Calcium handling in afferent arterioles. Acta Physiol Scand 181: 421‐429, 2004.
 569. Sander S, Ehrig B, Rettig R. Role of the native kidney in experimental post‐transplant hypertension. Pflugers Arch 431: 971‐976, 1996.
 570. Sanders KM. Invited review: Mechanisms of calcium handling in smooth muscles. J Appl Physiol 91: 1438‐1449, 2001.
 571. Sands JM. Renal urea transporters. Curr Opin Nephrol Hypertens 13: 525‐532, 2004.
 572. Sands JM, Layton HE. The urine concentrating mechanism and urea transporters. In: Seldin DW, Giebisch G, editors. The Kidney: Physiology and Pathophysiogy. Philadelphia: Lippincott, Williams & Wilkins, 2008, p. 1143‐1178.
 573. Sanjana VM, Johnston PA, Deen WM, Robertson CR, Brenner BM, Jamison RL. Hydraulic and oncotic pressure measurements in inner medulla of mammalian kidney. Am J Physiol 228: 1921‐1926, 1975.
 574. Sanjana VM, Johnston PA, Robertson CR, Jamison RL. An examination of transcapillary water flux in renal inner medulla. Am J Physiol 231: 313‐318, 1976.
 575. Sansom SC, Ma R, Carmines PK, Hall DA. Regulation of Ca(2+)‐activated K(+) channels by multifunctional Ca(2+)/calmodulin‐dependent protein kinase. Am J Physiol Renal Physiol 279: F283‐F288, 2000.
 576. Sansom SC, Stockand JD. Physiological role of large, Ca2+‐activated K +channels in human glomerular mesangial cells. Clin Exp Pharmacol Physiol 23: 76‐82, 1996.
 577. Sansom SC, Stockand JD, Hall D, Williams B. Regulation of large calcium‐activated potassium channels by protein phosphatase 2A. J Biol Chem 272: 9902‐9906, 1997.
 578. Sasser JM, Pollock JS, Pollock DM. Renal endothelin in chronic angiotensin II hypertension. Am J Physiol Regul Integr Comp Physiol 283: R243‐R248, 2002.
 579. Schmidt VJ, Wolfle SE, Boettcher M, de WC. Gap junctions synchronize vascular tone within the microcirculation. Pharmacol Rep 60: 68‐74, 2008.
 580. Schnackenberg CG. Physiological and pathophysiological roles of oxygen radicals in the renal microvasculature. Am J Physiol Regul Integr Comp Physiol 282: R335‐R342, 2002.
 581. Schnackenberg CG, Welch WJ, Wilcox CS. Normalization of blood pressure and renal vascular resistance in SHR with a membrane‐permeable superoxide dismutase mimetic. Role of nitric oxide. Hypertension 32: 59‐64, 1998.
 582. Schnackenberg CG, Welch WJ, Wilcox CS. TP receptor‐mediated vasoconstriction in microperfused afferent arterioles: Roles of O2− and NO. Am J Physiol Renal Physiol 279: F302‐F308, 2000.
 583. Schnackenberg CG, Wilcox CS. Two‐week administration of tempol attenuates both hypertension and renal excretion of I‐iso prostaglandin F2a. Hypertension 33 [ part II]: 424‐428, 1999.
 584. Scholander PF. The wonderful net. Scient Am 196: 96‐107, 1957.
 585. Schoonmaker GC, Fallet RW, Carmines PK. Superoxide anion curbs nitric oxide modulation of afferent arteriolar ANG II responsiveness in diabetes mellitus. Am J Physiol Renal Physiol 278: F302‐F309, 2000.
 586. Schroeder AC, Imig JD, LeBlanc EA, Pham BT, Pollock DM, Inscho EW. Endothelin‐mediated calcium signaling in preglomerular smooth muscle cells. Hypertension 35: 280‐286, 2000.
 587. Schwartz MM, Karnovsky MJ, Vehkatachalam MA. Ultrastructural differences between rat inner medullary descending and ascending vasa recta. Lab Invest 35: 161‐170, 1976.
 588. Seikaly MG, Arant BS Jr., Seney FD Jr. Endogenous angiotensin concentrations in specific intrarenal fluid compartments of the rat. J Clin Invest 86: 1352‐1357, 1990.
 589. Seino M, Abe K, Nushiro N, Omata K, Kasai Y, Yoshinaga K. Contribution of bradykinin to maintenance of blood pressure and renal blood flow in anaesthetized spontaneously hypertensive rats. J Hypertens Suppl 6: S401‐S403, 1988.
 590. Shayakul C, Knepper MA, Smith CP, DiGiovanni SR, Hediger MA. Segmental localization of urea transporter mRNAs in rat kidney. Am J Physiol 272: F654‐F660, 1997.
 591. Shayakul C, Steel A, Hediger MA. Molecular cloning and characterization of the vasopressin‐regulated urea transporter of rat kidney collecting ducts. J Clin Invest 98: 2580‐2587, 1996.
 592. Shimamura T, Morrison AB. Vascular permeability of the renal medullary vessels in the mouse and rat. Am J Pathol 71: 155‐163, 1973.
 593. Silldorff EP, Kreisberg MS, Pallone TL. Adenosine modulates vasomotor tone in outer medullary descending vasa recta of the rat. J Clin Invest 98: 18‐23, 1996.
 594. Silldorff EP, Pallone TL. Adenosine signaling in outer medullary descending vasa recta. Am J Physiol Regul Integr Comp Physiol 280: R854‐R861, 2001.
 595. Silldorff EP, Yang S, Pallone TL. Prostaglandin E2 abrogates endothelin induced vasoconstriction in renal outer medullary descending vasa recta of the rat. J Clin Invest 95: 2734‐2740, 1995.
 596. Silverstein DM, Thornhill BA, Leung JC, Vehaskari VM, Craver RD, Trachtman HA, Chevalier RL. Expression of connexins in the normal and obstructed developing kidney. Pediatr Nephrol 18: 216‐224, 2003.
 597. Ska BE, Sadowski J. Differential action of bradykinin on intrarenal regional perfusion in the rat: Waning effect in the cortex and major impact in the medulla. J Physiol 587: 3943‐3953, 2009.
 598. Slotkoff LM, Lilienfield LS. Extravascular renal albumin. Am J Physiol 212: 400‐406, 1967.
 599. Solez K, Kramer EC, Fox JA, Heptinstall RH. Medullary plasma flow and intravascular leukocyte accumulation in acute renal failure. Kidney Int 6: 24‐37, 1974.
 600. Spector AA. Arachidonic acid cytochrome P450 epoxygenase pathway. J Lipid Res 50 Suppl: S52‐S56, 2009.
 601. Spiecker M, Liao JK. Vascular protective effects of cytochrome p450 epoxygenase‐derived eicosanoids. Arch Biochem Biophys 433: 413‐420, 2005.
 602. Steendahl J, Holstein‐Rathlou NH, Sorensen CM, Salomonsson M. Effects of chloride channel blockers on rat renal vascular responses to angiotensin II and norepinephrine. Am J Physiol Renal Physiol 286: F323‐F330, 2004.
 603. Steiner SH, King RD. Nutrient renal blood flow and its ditribution in the unanesthetized dog. J Surg Res 10: 133‐146, 1970.
 604. Steinhausen M, Kucherer H, Parekh N, Weis S, Wiegman DL, Wilhelm KR. Angiotensin II control of the renal microcirculation: Effect of blockade by saralasin. Kidney Int 30: 56‐61, 1986.
 605. Stephenson JL. Concentration of urine in a central core model of the renal counterflow system. Kidney Int 2: 85‐94, 1972.
 606. Stern MD, Bowen PD, Parma R, Osgood RW, Bowman RL, Stein JH. Measurement of renal cortical and medullary blood flow by laser‐Doppler spectroscopy in the rat. Am J Physiol Renal Physiol 236: F80‐F87, 1979.
 607. Stockand JD, Sansom SC. Large Ca(2+)‐activated K+ channels responsive to angiotensin II in cultured human mesangial cells. Am J Physiol 267: C1080‐C1086, 1994.
 608. Stockand JD, Sansom SC. Role of large Ca(2+)‐activated K+ channels in regulation of mesangial contraction by nitroprusside and ANP. Am J Physiol 270: C1773‐C1779, 1996a.
 609. Stockand JD, Sansom SC. Activation by methylene blue of large Ca(2+)‐activated K +channels. Biochim Biophys Acta 1285: 123‐126, 1996b.
 610. Stockand JD, Sansom SC. Glomerular mesangial cells: Electrophysiology and regulation of contraction. Physiol Rev 78: 723‐744, 1998.
 611. Strandgaard S, Hansen U. Hypertension in renal allograft recipients may be conveyed by cadaveric kidneys from donors with subarachnoid haemorrhage. BMJ 292: 1041‐1044, 1986.
 612. Strick DM, Ficksen‐Olsen MJ, Lockart JC, Roman RJ, Romero JC. Direct measurement of renal medullary blood flow in the dog. Am J Physiol Reg Int Comp Physiol 257: R253‐R259, 1994.
 613. Stricklett PK, Hughes AK, Kohan DE. Endothelin‐1 stimulates NO production and inhibits cAMP accumulation in rat inner medullary collecting duct through independent pathways. Am J Physiol Renal Physiol 290: F1315‐F1319, 2006.
 614. Striessnig J, Grabner M, Mitterdorfer J, Hering S, Sinnegger MJ, Glossmann H. Structural basis of drug binding to L Ca2+ channels. Trends Pharmacol Sci 19: 108‐115, 1998.
 615. Sun CW, Alonso‐Galicia M, Taheri MR, Falck JR, Harder DR, Roman RJ. Nitric oxide‐20‐hydroxygeicosatetraenoic acid interaction in the regulation of K+ channel activity and vascular tone in renal arterioles. Circ Res 83: 1069‐1079, 1998.
 616. Sun CW, Falck JR, Harder DR, Roman RJ. Role of tyrosine kinase and PKC in the vasoconstrictor response to 20‐HETE in renal arterioles. Hypertension 33 [ part II]: 414‐418, 1999.
 617. Sun X, Cao K, Yang G, Huang Y, Hanna ST, Wang R. Selective expression of Kir6.1 protein in different vascular and non‐vascular tissues. Biochem Pharmacol 67: 147‐156, 2004.
 618. Szamosfalvi B, Cortes P, Alviani R, Asano K, Riser BL, Zasuwa G, Yee J. Putative subunits of the rat mesangial KATP: A type 2B sulfonylurea receptor and an inwardly rectifying K+ channel. Kidney Int 61: 1739‐1749, 2002.
 619. Szentivanyi M Jr., Park F, Maeda CY, Cowley AW Jr. Nitric oxide in the renal medulla protects from vasopressin‐induced hypertension. Hypertension 35: 740‐745, 2000.
 620. Szentivanyi M Jr., Zou AP, Maeda CY, Mattson DL, Cowley AW Jr. Increase in renal medullary nitric oxide synthase activity protects from norepinephrine‐induced hypertension. Hypertension 35: 418‐423, 2000.
 621. Takahashi‐Iwanaga H. The three‐dimensional cytoarchitecture of the interstitial tissue in the rat kidney. Cell Tissue Res 264: 269‐281, 1991.
 622. Takenaka T, Epstein M, Forster H, Landry DW, Iijima K, Goligorsky MS. Attenuation of endothelin effects by a chloride channel inhibitor, indanyloxyacetic acid. Am J Physiol 262: F799‐F806, 1992.
 623. Takenaka T, Harrison‐Bernard LM, Inscho EW, Carmines PK, Navar LG. Autoregulation of afferent arteriolar blood flow in juxtamedullary nephrons. Am J Physiol 267: F879‐F887, 1994.
 624. Takenaka T, Inoue T, Kanno Y, Okada H, Hill CE, Suzuki H. Connexins 37 and 40 transduce purinergic signals mediating renal autoregulation. Am J Physiol Regul Integr Comp Physiol 294: R1‐R11, 2008.
 625. Takenaka T, Inoue T, Kanno Y, Okada H, Meaney KR, Hill CE, Suzuki H. Expression and role of connexins in the rat renal vasculature. Kidney Int 73: 415‐422, 2008.
 626. Takenaka T, Kanno Y, Kitamura Y, Hayashi K, Suzuki H, Saruta T. Role of chloride channels in afferent arteriolar constriction. Kidney Int 50: 864‐872, 1996.
 627. Talavera K, Nilius B. Biophysics and structure‐function relationship of T‐type Ca2 +channels. Cell Calcium 40: 97‐114, 2006.
 628. Tamaki T, Kiyomoto K, He H, Tomohiro A, Nishiyama A, Aki Y, Kimura S, Abe Y. Vasodilation induced by vasopressin V2 receptor stimulation in afferent arterioles. Kidney Int 49: 722‐729, 1996.
 629. Tang L, Loutzenhiser K, Loutzenhiser R. Biphasic actions of prostaglandin E(2) on the renal afferent arteriole: Role of EP(3) and EP(4) receptors. Circ Res 86: 663‐670, 2000.
 630. Tang L, Parker M, Fei Q, Loutzenhiser R. Afferent arteriolar adenosine A2a receptors are coupled to KATP in in vitro perfused hydronephrotic rat kidney. Am J Physiol 277: F926‐F933, 1999.
 631. Taylor NE, Cowley AW Jr. Effect of renal medullary H2O2 on salt‐induced hypertension and renal injury. Am J Physiol Regul Integr Comp Physiol 289: R1573‐R1579, 2005.
 632. Taylor NE, Glocka P, Liang M, Cowley AW Jr. NADPH oxidase in the renal medulla causes oxidative stress and contributes to salt‐sensitive hypertension in Dahl S rats. Hypertension 47: 692‐698, 2006.
 633. Terada Y, Tomita K, Nonoguchi H, Marumo F. Different localization of two types of endothelin receptor mRNA in microdissected rat nephron segments using reverse transcription and polymerase chain reaction assay. J Clin Invest 90: 107‐112, 1992.
 634. Thomas CJ, Woods RL, Evans RG, Alcorn D, Christy IJ, Anderson WP. Evidence for a renomedullary vasodepressor hormone. Clin Exp Pharmacol Physiol 23: 777‐785, 1996.
 635. Thomas SR. Cycles and separations in a model of the renal medulla. Am J Physiol 275: F671‐F690, 1998.
 636. Thomas SR. Inner medullary lactate production and accumulation: A vasa recta model. Am J Physiol Renal Physiol 279: F468‐F481, 2000.
 637. Thomas SR, Layton AT, Layton HE, Moore LC. Kidney modelling: Status and perspectives. Proceedings of IEEE 94: 740‐752, 2006.
 638. Thornburn GD, Kopald HH, Herd JA, Hollenberg M, O’Morchoe CCC, Barger AC. Intrarenal distribution of nutrient blood flow determined with krypton85 in the unanesthetized dog. Circ Res 13: 290‐307, 1963.
 639. Thurau K. Renal hemodynamics. Am J Med 36: 698‐719, 1964.
 640. Thurau K, Sugiura T, Lilienfeld LS. Micropuncture of renal vasa recta in hydropenic hamsters. Circ Res 8: 383, 1960.
 641. Tian W, Bonkovsky HL, Shibahara S, Cohen DM. Urea and hypertonicity increase expression of heme oxygenase‐1 in murine renal medullary cells. Am J Physiol Renal Physiol 281: F983‐F991, 2001.
 642. Tobian L, Lange J, Azar S, Iwai J, Koop D, Coffee K, Johnson MA. Reduction of natriuretic capacity and renin release in isolated blood‐perfused kidneys of Dahl hypertension‐prone rats. Circ Res 43: I‐92‐I‐98, 1978.
 643. Trinh‐Trang‐Tan MM, Lasbennes F, Gane P, Roudier N, Ripoche P, Cartron JP, Bailly P. UT‐B1 proteins in rat: Tissue distribution and regulation by antidiuretic hormone in kidney. Am J Physiol Renal Physiol 283: F912‐F922, 2002.
 644. Turner MR, Pallone TL. Hydraulic and diffusional permeabilities of isolated outer medullary descending vasa recta from the rat. Am J Physiol 272: H392‐H400, 1997a.
 645. Turner MR, Pallone TL. Vasopressin constricts outer medullary descending vasa recta isolated from rat kidneys. Am J Physiol 272: F147‐F151, 1997b.
 646. Vassileva I, Mountain C, Pollock DM. Functional role of ETB receptors in the renal medulla. Hypertension 41: 1359‐1363, 2003.
 647. 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.
 648. Vera T, Kelsen S, Stec DE. Kidney‐specific induction of heme oxygenase‐1 prevents angiotensin II hypertension. Hypertension 52: 660‐665, 2008.
 649. Vera T, Kelsen S, Yanes LL, Reckelhoff JF, Stec DE. HO‐1 induction lowers blood pressure and superoxide production in the renal medulla of angiotensin II hypertensive mice. Am J Physiol Regul Integr Comp Physiol 292: R1472‐R1478, 2007.
 650. Vio CP, Figueroa CD. Subcellular localization of renal kallikrein by ultrastructural immunocytochemistry. Kidney Int 28: 36‐42, 1985.
 651. Vio CP, Figueroa CD. Evidence for a stimulatory effect of high potassium diet on renal kallikrein. Kidney Int 31: 1327‐1334, 1987.
 652. Vitzthum H, Weiss B, Bachleitner W, Kramer BK, Kurtz A. Gene expression of adenosine receptors along the nephron. Kidney Int 65: 1180‐1190, 2004.
 653. Wagner C. Function of connexins in the renal circulation. Kidney Int 73: 547‐555, 2008.
 654. Wagner C, de WC, Kurtz L, Grunberger C, Kurtz A, Schweda F. Connexin40 is essential for the pressure control of renin synthesis and secretion. Circ Res 100: 556‐563, 2007.
 655. Wang D, Borrego‐Conde LJ, Falck JR, Sharma KK, Wilcox CS, Umans JG. Contributions of nitric oxide, EDHF, and EETs to endothelium‐dependent relaxation in renal afferent arterioles. Kidney Int 63: 2187‐2193, 2003.
 656. Wang MC, Dolphin A, Kitmitto A. L‐type voltage‐gated calcium channels: Understanding function through structure. FEBS Lett 564: 245‐250, 2004.
 657. Wang W, Michel CC. Effects of anastomoses on solute transcapillary exchange in countercurrent systems. Microcirculation 4: 381‐390, 1997.
 658. Wang W, Michel CC. Modeling exchange of plasma proteins between microcirculation and interstitium of the renal medulla. Am J Physiol Renal Physiol 279: F334‐F344, 2000.
 659. Wang W, Parker KH, Michel CC. Theoretical studies of steady‐state transcapillary exchange in countercurrent systems. Microcirculation 3: 301‐311, 1996.
 660. Wang X, Trottier G, Loutzenhiser R. Determinants of renal afferent arteriolar actions of bradykinin: Evidence that multiple pathways mediate responses attributed to EDHF. Am J Physiol Renal Physiol 285: F540‐F549, 2003.
 661. Wang YX, Betton G, Floettmann E, Fantham E, Ridgwell G. Imaging kidney in conscious rats with high‐frequency ultrasound and detection of two cases of unilateral congenital hydronephrosis. Ultasound Med Biol 33: 483‐486, 2007.
 662. Warden DH, Stokes JB. EGF and PGE2 inhibit rabbit CCD Na+ transport by different mechanisms: PGE2 inhibits Na(+)‐K+ pump. Am J Physiol 264: F670‐F677, 1993.
 663. Webb RC. Smooth muscle contraction and relaxation. Adv Physiol Educ 27: 201‐206, 2003.
 664. Weihprecht H, Lorenz JN, Briggs JP, Schnermann J. Vasoconstrictor effect of angiotensin and vasopressin in isolated rabbit afferent arterioles. Am J Physiol 261: F273‐F282, 1991.
 665. Weihprecht H, Lorenz JN, Briggs JP, Schnermann J. Vasomotor effects of purinergic agonists in isolated rabbit afferent arterioles. Am J Physiol 263: F1026‐F1033, 1992.
 666. Weinstein AM. Mathematical models of renal fluid and electrolyte transport: Acknowledging our uncertainty. Am J Physiol Renal Physiol 284: F871‐F884, 2003.
 667. Welch WJ. Angiotensin II‐dependent superoxide: Effects on hypertension and vascular dysfunction. Hypertension 52: 51‐56, 2008.
 668. Welch WJ, Baumgartl H, Lubbers D, Wilcox CS. Renal oxygenation defects in the spontaneously hypertensive rat: Role of AT1 receptors. Kidney Int 63: 202‐208, 2003.
 669. Welch WJ, Blau J, Xie H, Chabrashvili T, Wilcox CS. Angiotensin‐induced defects in renal oxygenation: Role of oxidative stress. Am J Physiol Heart Circ Physiol 288: H22‐H28, 2005.
 670. Wilcox CS, Pearlman A. Chemistry and antihypertensive effects of tempol and other nitroxides. Pharmacol Rev 60: 418‐469, 2008.
 671. Wilde WS, Vorburger C. Albumin multiplier in kidney vasa recta analyzed by microspectrophotometry of T‐1824. Am J Physiol 213: 1233‐1243, 1967.
 672. Wolgast M. Studies on the regional blood flow with P32‐labelled red cells an small beta‐sensitive semiconductor detectors. Acta Physiol Scand Suppl 313: 1‐109, 1968.
 673. Wray S, Burdyga T, Noble K. Calcium signalling in smooth muscle. Cell Calcium 38: 397‐407, 2005.
 674. Wu F, Cholewa B, Mattson DL. Characterization of L‐arginine transporters in rat renal inner medullary collecting duct. Am J Physiol Regul Integr Comp Physiol 278: R1506‐R1512, 2000.
 675. Xu Y, Olives B, Bailly P, Fischer E, Ripoche P, Ronco P, Cartron JP, Rondeau E. Endothelial cells of the kidney vasa recta express the urea transporter HUT11. Kidney Int 51: 138‐146, 1997.
 676. Yang B, Bankir L. Urea and urine concentrating ability: New insights from studies in mice. Am J Physiol Renal Physiol 288: F881‐F896, 2005.
 677. Yang B, Bankir L, Gillespie A, Epstein CJ, Verkman AS. Urea‐selective concentrating defect in transgenic mice lacking urea transporter UT‐B. J Biol Chem 277: 10633‐10637, 2002.
 678. Yang B, Verkman AS. Urea transporter UT3 functions as an efficient water channel. Direct evidence for a common water/urea pathway. J Biol Chem 273: 9369‐9372, 1998.
 679. Yang B, Verkman AS. Analysis of double knockout mice lacking aquaporin‐1 and urea transporter UT‐B. Evidence for UT‐B‐facilitated water transport in erythrocytes. J Biol Chem 277: 36782‐36786, 2002.
 680. Yang S, Silldorff EP, Pallone TL. Effect of norepinephrine and acetylcholine on outer medullary descending vasa recta. Am J Physiol 269: H710‐H716, 1995.
 681. Yang T. Regulation of cyclooxygenase‐2 in renal medulla. Acta Physiol Scand 177: 417‐421, 2003.
 682. Yang ZZ, Zhang AY, Yi FX, Li PL, Zou AP. Redox regulation of HIF‐1alpha levels and HO‐1 expression in renal medullary interstitial cells. Am J Physiol Renal Physiol 284: F1207‐F1215, 2003.
 683. Yang ZZ, Zou AP. Transcriptional regulation of heme oxygenases by HIF‐1alpha in renal medullary interstitial cells. Am J Physiol Renal Physiol 281: F900‐F908, 2001.
 684. Yarger WE, Boyd MA, Schrader NW. Evaluation of methods of measuring glomerular and nutrient blood flow in rat kidneys. Am J Physiol‐Heart Circ Physiol 235: H592‐H600, 1978.
 685. Yu Z, Huse LM, Adler P, Graham L, Ma J, Zeldin DC, Kroetz DL. Increased CYP2J expression and epoxyeicosatrienoic acid formation in spontaneously hypertensive rat kidney. Mol Pharmacol 57: 1011‐1020, 2000.
 686. Yu Z, Xu F, Huse LM, Morisseau C, Draper AJ, Newman JW, Parker C, Graham L, Engler MM, Hammock BD, Zeldin DC, Kroetz DL. Soluble epoxide hydrolase regulates hydrolysis of vasoactive epoxyeicosatrienoic acids. Circ Res 87: 992‐998, 2000.
 687. Yuan B, Cowley AW Jr. Evidence that reduced renal medullary nitric oxide synthase activity of dahl s rats enables small elevations of arginine vasopressin to produce sustained hypertension. Hypertension 37: 524‐528, 2001.
 688. Yuan BH, Robinette JB, Conger JD. Effect of angiotensin II and norepinephrine on isolated rat afferent and efferent arterioles. Am J Physiol 258: F741‐F750, 1990.
 689. Zewde T, Mattson DL. Inhibition of cyclooxygenase‐2 in the rat renal medulla leads to sodium‐sensitive hypertension. Hypertension 44: 424‐428, 2004.
 690. Zewde T, Wu F, Mattson DL. Influence of dietary NaCl on L‐arginine transport in the renal medulla. Am J Physiol Regul Integr Comp Physiol 286: R89‐R93, 2004.
 691. 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.
 692. 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.
 693. Zhang J, Hill CE. Differential connexin expression in preglomerular and postglomerular vasculature: Accentuation during diabetes. Kidney Int 68: 1171‐1185, 2005.
 694. Zhang MZ, Sanchez LP, McKanna JA, Harris RC. Regulation of cyclooxygenase expression by vasopressin in rat renal medulla. Endocrinology 145: 1402‐1409, 2004.
 695. Zhang Q, Cao C, Mangano M, Zhang Z, Silldorff EP, Lee‐Kwon W, Payne K, Pallone TL. Descending vasa recta endothelium is an electrical syncytium. Am J Physiol Regul Integr Comp Physiol 291: R1688‐R1699, 2006.
 696. Zhang Q, Cao C, Zhang Z, Wier WG, Edwards A, Pallone TL. Membrane current oscillations in descending vasa recta pericytes. Am J Physiol Renal Physiol 294: F656‐F666, 2008.
 697. Zhang W, Edwards A. Transport of plasma proteins across vasa recta in the renal medulla. Am J Physiol Renal Physiol 281: F478‐F492, 2001.
 698. Zhang W, Edwards A. Theoretical effects of UTB urea transporters in the renal medullary microcirculation. Am J Physiol Renal Physiol 285: F731‐F747, 2003.
 699. Zhang W, Edwards A. A model of glucose transport and conversion to lactate in the renal medullary microcirculation. Am J Physiol Renal Physiol 290: F87‐F102, 2006a.
 700. Zhang W, Edwards A. Mathematical model of nitric oxide convection and diffusion in a renal medullary vas rectum. J Math Biol 53: 385‐420, 2006b.
 701. Zhang W, Edwards A. A model of nitric oxide tubulovascular cross talk in a renal outer medullary cross section. Am J Physiol Renal Physiol 292: F711‐F722, 2007.
 702. Zhang W, Pibulsonggram T, Edwards A. Determinants of basal nitric oxide concentration in the renal medullary microcirculation. Am J Physiol Renal Physiol 287: F1189‐F1203, 2004.
 703. Zhang Z, Cao C, Lee‐Kwon W, Pallone TL. Descending vasa recta pericytes express voltage operated Na+ conductance in the rat. J Physiol 567: 445‐457, 2005.
 704. Zhang Z, Huang JM, Turner MR, Rhinehart KL, Pallone TL. Role of chloride in constriction of descending vasa recta by angiotensin II. Am J Physiol Regul Integr Comp Physiol 280: R1878‐R1886, 2001.
 705. Zhang Z, Pallone TL. Response of descending vasa recta to luminal pressure. Am J Physiol Renal Physiol 287: F535‐F542, 2004.
 706. Zhang Z, Rhinehart K, Kwon W, Weinman E, Pallone TL. ANG II signaling in vasa recta pericytes by PKC and reactive oxygen species. Am J Physiol Heart Circ Physiol 287: H773‐H781, 2004.
 707. Zhang Z, Rhinehart K, Pallone TL. Membrane potential controls calcium entry into descending vasa recta pericytes. Am J Physiol Regul Integr Comp Physiol 283: R949‐R957, 2002.
 708. Zhang Z, Rhinehart K, Solis G, Pittner J, Lee‐Kwon W, Welch WJ, Wilcox CS, Pallone TL. Chronic ANG II infusion increases NO generation by rat descending vasa recta. Am J Physiol Heart Circ Physiol 288: H29‐H36, 2005.
 709. Zhao X, Pollock DM, Inscho EW, Zeldin DC, Imig JD. Decreased renal cytochrome P450 2C enzymes and impaired vasodilation are associated with angiotensin salt‐sensitive hypertension. Hypertension 41: 709‐714, 2003.
 710. Zhao X, Yamamoto T, Newman JW, Kim IH, Watanabe T, Hammock BD, Stewart J, Pollock JS, Pollock DM, Imig JD. Soluble epoxide hydrolase inhibition protects the kidney from hypertension‐induced damage. J Am Soc Nephrol 15: 1244‐1253, 2004.
 711. Zhuo J, Dean R, Maric C, Aldred PG, Harris P, Alcorn D, Mendelsohn FA. Localization and interactions of vasoactive peptide receptors in renomedullary interstitial cells of the kidney. Kidney Int Suppl 67: S22‐S28, 1998.
 712. Zhuo JL. Renomedullary interstitial cells: A target for endocrine and paracrine actions of vasoactive peptides in the renal medulla. Clin Exp Pharmacol Physiol 27: 465‐473, 2000.
 713. Zimmerhackl B, Dussel R, Steinhausen M. Erythrocyte flow and dynamic hematocrit in the renal papilla of the rat. Am J Physiol 249: F898‐F902, 1985.
 714. Zimmerhackl B, Robertson CR, Jamison RL. Fluid uptake in the renal papilla by vasa recta estimated by two methods simultaneously. Am J Physiol 248: F347‐F353, 1985a.
 715. Zimmerhackl B, Robertson CR, Jamison RL. Effect of arginine vasopressin on renal medullary blood flow. A videomicroscopic study in the rat. J Clin Invest 76: 770‐778, 1985b.
 716. Zou AP, Billington H, Su N, Cowley AW Jr. Expression and actions of heme oxygenase in the renal medulla of rats. Hypertension 35: 342‐347, 2000.
 717. Zou AP, Cowley AW Jr. Nitric oxide in renal cortex and medulla. An in vivo microdialysis study. Hypertension 29(part 2): 194‐198, 1997.
 718. Zou AP, Cowley AW Jr. Alpha(2)‐adrenergic receptor‐mediated increase in NO production buffers renal medullary vasoconstriction. Am J Physiol Regul Integr Comp Physiol 279: R769‐R777, 2000.
 719. Zou AP, Cowley AW Jr. Reactive oxygen species and molecular regulation of renal oxygenation. Acta Physiol Scand 179: 233‐241, 2003.
 720. Zou AP, Fleming JT, Falck JR, Jacobs ER, Gebremedhin D, Harder DR, Roman RJ. Stereospecific effects of epoxyeicosatrienoic acid on renal vascular tone and K+channel activity. Am J Physiol‐Renal Physiol 270: F822‐F832, 1996a.
 721. Zou AP, Fleming JT, Falck JR, Jacobs ER, Gebremedhin D, Harder DR, Roman RJ. 20‐HETE is an endogenous inhibitor of the large‐conductance Ca2+‐activated K+ channel in renal arterioles. Am J Physiol Regul Integr Comp Physiol 270: R228‐R237, 1996b.
 722. Zou AP, Li N, Cowley AW Jr. Production and actions of superoxide in the renal medulla. Hypertension 37: 547‐553, 2001.
 723. Zou AP, Wu F, Cowley AW Jr. Protective effect of angiotensin II‐induced increase in nitric oxide in the renal medullary circulation. Hypertension 31(part 2): 271‐276, 1998.
 724. Zou AP, Wu F, Li PL, Cowley AW Jr. Effect of chronic salt loading on adenosine metabolism and receptor expression in renal cortex and medulla in rats. Hypertension 33: 511‐516, 1999.
 725. Zusman RM, Keiser HR. Prostaglandin biosynthesis by rabbit renomedullary interstitial cells in tissue culture. Stimulation by angiotensin II, bradykinin and arginine vasopressin. J Clin Invest 60: 215‐222, 1977.

Contact Editor

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

* Required Field

How to Cite

Thomas L. Pallone, Aurélie Edwards, David L. Mattson. Renal Medullary Circulation. Compr Physiol 2012, 2: 97-140. doi: 10.1002/cphy.c100036