You are on page 1of 1328

SCHRIER’SDISEASES

OF THE KIDNEY
N IN TH ED I TIO N
SCHRIER’S DISEASES
OF THE KIDNEY
N I N TH ED I TI O N

VO LU M E I

ED I TED BY

THOMAS M. COFFMAN, MD BRUCE A. MOLITORIS, MD


James R. Clapp Professor of Medicine Professor of Medicine
Chief, Division of Nephrology Indiana University
Duke University and Durham VA Medical Centers Roudebush VA Medical Center
Durham, North Carolina Indianapolis, Indiana
Program Director in Cardiovascular and
Metabolic Disorders
Duke-NUS Graduate Medical School
ERIC G. NEILSON, MD
Singapore Vice President for Medical Affairs
Lewis Landsberg Dean
Professor of Medicine and Cell and
RONALD J. FALK, MD Molecular Biology
Allen Brewster Distinguished Feinberg School of Medicine
Professor of Medicine Northwestern University
Director, UNC Kidney Center Chicago, Illinois
Director, UNC Center for Transplant Care
Chief, Division of Nephrology and Hypertension
University of North Carolina
ROBERT W. SCHRIER, MD,
Chapel Hill, North Carolina
MACP, Ho n o ra r y MRCP (UK)
Professor Emeritus
University of Colorado School of Medicine
Aurora, Colorado
Acquisitions Editor: Julie Goolsby
Product Manager: Tom Gibbons
Vendor Manager: Alicia Jackson
Senior Manufacturing Manager: Benjamin Rivera
Marketing Manager: Kimberly Schonberger
Design Coordinator: Steve Druding
Production Service: Absolute Service, Inc.

© 2013 by LIPPINCOTT WILLIAMS & WILKINS, a WOLTERS KLUWER business


Two Commerce Square
2001 Market Street
Philadelphia, PA 19103 USA
LWW.com

© 2007 by Lippincott Williams & Wilkins. All rights reserved. This book is protected by copyright. No
part of this book may be reproduced in any form by any means, including photocopying, or utilized by
any information storage and retrieval system without written permission from the copyright owner, except
for brief quotations embodied in critical articles and reviews. Materials appearing in this book prepared
by individuals as part of their of cial duties as U.S. government employees are not covered by the above-
mentioned copyright.

Printed in China

Library of Congress Cataloging-in-Publication Data


[978-1-4511-1075-3]
[1-4511-1075-8]
Schrier’s diseases of the kidney. – 9th ed. / edited by Thomas M. Coffman
... [et al.].
p. ; cm.
Diseases of the kidney
Rev. ed. of: Diseases of the kidney & urinary tract. c2007.
Includes bibliographical references and index.
ISBN 978-1-4511-1075-3 (hardback : alk. paper) – ISBN 1-4511-1075-8
I. Coffman, Thomas M. II. Schrier, Robert W. III. Diseases of the kidney &
urinary tract. IV. Title: Diseases of the kidney.
[DNLM: 1. Kidney Diseases. WJ 300]

616.6’1--dc23
2012024363

Care has been taken to con rm the accuracy of the information presented and to describe generally
accepted practices. However, the authors, editors, and publisher are not responsible for errors or omissions
or for any consequences from application of the information in this book and make no warranty, expressed or
implied, with respect to the currency, completeness, or accuracy of the contents of the publication. Application
of the information in a particular situation remains the professional responsibility of the practitioner.
The authors, editors, and publisher have exerted every effort to ensure that drug selection and dosage
set forth in this text are in accordance with current recommendations and practice at the time of publication.
However, in view of ongoing research, changes in government regulations, and the constant ow of infor-
mation relating to drug therapy and drug reactions, the reader is urged to check the package insert for each
drug for any change in indications and dosage and for added warnings and precautions. This is particularly
important when the recommended agent is a new or infrequently employed drug.
Some drugs and medical devices presented in the publication have Food and Drug Administration
(FDA) clearance for limited use in restricted research settings. It is the responsibility of the health care
provider to ascertain the FDA status of each drug or device planned for use in their clinical practice.

To purchase additional copies of this book, call our customer service department at (800) 638-3030 or fax
orders to (301) 223-2320. International customers should call (301) 223-2300.

Visit Lippincott Williams & Wilkins on the Internet: at LWW.com. Lippincott Williams & Wilkins customer
service representatives are available from 8:30 a.m. to 6:00 p.m., EST.

10 9 8 7 6 5 4 3 2 1
CONTENTS ■

Contents vii 11 Computed Tomography and Magnetic


Preface xi Resonance Imaging 377
Contributors xiii Kyongtae T. Bae and Sonal Krishan

Volume I 12 Diagnostic Angiography and


Therapeutic Endovascular Intervention
of the Renal Circulation 401
SEC T I O N
Alex Perino, Rajan Gupta, and Ivan P. Casserly
I STRUCTURAL AND FUNCTIONAL
CORRELATIONS IN THE KIDNEY 1 13 Renal Biopsy: Indications and Evaluation 424
I. David Weiner, C. Craig Tisher, and Byron P. Croker
1 Structural–Functional Relationships
in the Kidney 1 SEC TI O N

2
Wilhelm Kriz, Astrid Weins, and Ruth Ellen Bulger

Clinical Importance of Nephron Mass 46


III CYSTIC AND TUBULAR
DISORDERS 458
Valerie A. Luyckx and Thomas F. Mueller
14 Introduction to Genetic Renal Disease 458
3 Renal Circulation and Glomerular Erum Hartung, Terry Watnick, and
Hemodynamics 74 Gregory G. Germino
William J. Arendshorst and L. Gabriel Navar
15 Nephronophthisis—Medullary Cystic
4 Regulation of Water Balance: Kidney Disease 501
Urine Concentration and Dilution 132 Friedhelm Hildebrandt, Rannar Airik,
Yumi Noda and Sei Sakaki and John A. Sayer
5 Tubular Sodium Transport 159 16 Autosomal Dominant Polycystic
Arohan R. Subramanya, Núria M. Pastor-Solar, Kidney Disease 519
W. Brian Reeves, and Kenneth R. Hallows Stefan Somlo and Arlene B. Chapman

6 Tubular Potassium Transport 194 17 Alport Syndrome, Fabry Disease, and


David B. Mount Nail-Patella Syndrome 564
Godela M. Brosnahan, Martin Gregory,
7 Renal Acid–Base Transport 214 Imed Halal, and Berenice Y. Reed
Thomas D. DuBose Jr. and Amanda K. Goode
18 Isolated Renal Tubular Disorders:
8 Hormones and the Kidney 230 Mechanisms and Clinical Expression 587
Sola Aoun Bahous, Maya Khairallah, Thomas D. DuBose Jr. and
Joumana T. Chaiban, and Kamal F. Badr Amanda K. Goode

19 Urinary Tract Obstruction and


SEC T I O N
Re ux Nephropathy 615
II CLINICAL EVALUATION 295
Ryan B. Pickens and Edward D. Kim

20 Nephrolithiasis 642
9 Laboratory Evaluation of Kidney Disease 295 Keith A. Hruska and Anne M. Beck
Lesley A. Inker, Richard A. Lafayette,
Ashish Upadhyay, and Andrew S. Levey

10 Ultrasonography and Nuclear Medicine 346


Frederic Rahbari-Oskoui, Andrew T. Taylor,
and William Charles O’Neill

vii
viii CONTENTS ■

SEC T I O N 33 Contrast-Induced Acute Kidney Injury 959

IV INFECTIONS OF THE URINARY TRACT


AND THE KIDNEY 673
Steven D. Weisbord and Paul M. Palevsky

34 Nephrotoxicity Secondary to Environmental


Agents, Heavy Metals, Drug Abuse,
21 Host–Pathogen Interactions and
and Lithium 981
Host Defense Mechanisms 673
Veronica Torres da Costa e Silva,
Keira Melican, Ferdinand X. Choong, and
Etienne Macedo, and Luis Yu
Agneta Richter-Dahlfors
35 Acute Tubulointerstitial Nephritis 994
22 Cystitis and Urethritis 689
Garabed Eknoyan and Rajeev Raghavan
Annelie Brauner and Milan Chromek
36 Acute Kidney Injury Associated with
23 Infections of the Upper Urinary Tract 699
Pigmenturia or Crystal Deposits 1018
Lindsay E. Nicollé
Abolfazl Zarjou and Anupam Agarwal
24 Renal and Perirenal Abscesses 725
37 Acute Kidney Injury Secondary to Tropical
Neha D. Nanda and Louise M. Dembry Hemorrhagic Viral Infections
25 Complicated Urinary Tract Infections 738 and Snakebites 1045
Dennis J. Mikolich* and Stephen H. Zinner Emmanuel A. Burdmann

26 Fungal Infections of the Urinary Tract 754 38 Acute Kidney Injury following
Hematopoietic Cell Transplantation
Carol A. Kauffman
and Severe Burns 1057
27 Urinary Tract Tuberculosis 764 Justin M. Belcher and Chirag R. Parikh
Mark S. Pasternack and Robert H. Rubin
SEC TI O N

SEC T I O N
VI HYPERTENSION 1086

V ACUTE KIDNEY INJURY 785

39 Role of the Kidneys in Hypertension 1086


28 Epidemiology, Diagnosis, and Therapy of Joey P. Granger and Eric M. George
Acute Kidney Injury 785
40 Hypertension in Chronic Kidney Disease 1109
Etienne Macedo and Ravindra L. Mehta
Peter N. Van Buren and Robert D. Toto
29 Pathophysiology of Ischemic Acute
41 Hypertension in End-Stage Renal Disease 1131
Kidney Injury 826
Rajiv Agarwal
Charles L. Edelstein and Robert W. Schrier
42 Renal Artery Stenosis, Renovascular
30 Pathophysiology of Nephrotoxic
Hypertension, and Ischemic Nephropathy 1153
Cell Injury 868
Stephen C. Textor
Brian S. Cummings and Rick G. Schnellmann
43 Hypertension due to Primary Aldosteronism
31 Antibiotic- and Immunosuppression-Related
and Related Disorders 1197
Renal Failure 901
Emmanuel L. Bravo, Muhammed A. Rafey,
Marc E. De Broe
and Surafel F. Gebreselassie
32 Nephrotoxicity of Nonsteroidal Anti-
44 Malignant Hypertension and
in ammatory Agents, Analgesics, and Inhibitors
Other Hypertensive Crises 1209
of the Renin-Angiotensin System 943
Charles R. Nolan and Stuart L. Linas
Biff F. Palmer
Index I-1
*Deceased.
PREFACE ■

T
he recent advances in many aspects of kidney diseases V Acute Kidney Injury is described in 11 chapters,
have mandated a new edition of Schrier’s Diseases of including the pathophysiology of renal cell ischemia
the Kidney. As in previous editions, a group of inter- and nephrotoxic injury, acute tubular necrosis, acute
national experts was assembled to present this information interstitial nephritis, and acute nephrotoxic renal disease.
in a comprehensive, authoritative, concise, and readily ac- VI Hypertension and its renal manifestations are cov-
cessible fashion. The chapters have been extensively revised ered in six chapters, which include pathophysiology,
and updated. renal vascular, and endocrine-related hypertension as
Nephrology is a discipline that combines the basic and well as hypertension in pregnancy and in diabetes.
clinical sciences. Successful integration of this knowledge is
VII Glomerular, Interstitial, and Vascular Renal
the goal of this Ninth Edition. The 11 sections of the two-
Diseases are discussed in 13 chapters, including
volume book are actually individual texts that can stand on
collagen vascular diseases, chronic interstitial nephritis,
their own.
primary glomerulonephritides, and vasculitides.
The rst section presents an overall view of the structural,
physiologic, and biochemical aspects of the kidney. This sec- VIII Systemic Diseases of the Kidney are covered in
tion incorporates the latest developments in cellular and mo- eight chapters, including diabetes, hepatorenal syn-
lecular biology, emphasizing the most current information drome, sickle cell disease, gout, myeloma/amyloido-
and concepts on cell signaling, receptors, and ion channels. sis, and tropical diseases.
The subsequent 10 sections are disease oriented, with each IX Disorders of Electrolyte, Water, and Acid Base
section beginning with a pathophysiology chapter. The goal are covered in nine chapters, including syndrome of
of Schrier’s Diseases of the Kidney is to publish the most com- inappropriate antidiuretic hormone secretion, central
prehensive material for practicing and academic physicians and nephrogenic diabetes insipidus, cardiac failure,
caring for patients with kidney disease and hypertension. The cirrhosis, and the nephrotic syndrome.
11 sections of the book cover 86 chapters and are summa- X Chronic Kidney Disease, a section of six chapters,
rized as follows: covers pathophysiology, anemia, osteodystrophy, the
I Structural and Functional Correlations in the nervous system, cardiovascular complications, and
Kidney includes structural, hemodynamic, hor- metabolic and endocrine dysfunctions.
monal, ion transport, and metabolic functions in XI Management of End-Stage Renal Disease by
eight chapters. transplantation, peritoneal dialysis and hemodialysis,
II Clinical Evaluation is covered in ve chapters on including complications, outcomes, and ethical
urinalysis, laboratory evaluation, urography, tomogra- considerations, is discussed in six chapters.
phy, angiography, with indications and interpretations As editors, we have substantial expertise in most of
for renal biopsy. these areas of nephrology and are very pleased with the
III Cystic and Tubular Disorders in seven chapters content of the Ninth Edition. Out of 86 chapters, 44 have
covers genetic mechanisms, medullary cystic and new authors. We would like to thank our authoritative and
sponge disorders, polycystic kidney disease, Alport remarkably talented contributing authors, whose dedication
syndrome, Fabry disease, and nail-patella syndrome, to nephrology is unmatched.
as well as isolated renal tubular disorders. Thomas Coffman, MD
IV Infections of the Urinary Tract and the Kidney are Ronald Falk, MD
contained in seven chapters, including host-defense Bruce Molitoris, MD
mechanisms; urinary bacterial infections, including Eric Neilson, MD
tuberculosis as well as fungal infections; renal Robert W. Schrier, MD
abscesses; and cystitis.

xi
SECTION I STRUCTURAL AND FUNCTIONAL
CORRELATIONS IN THE KIDNEY

C H A P T E R

1 Structural–Functional Relationships
in the Kidney
Wilhelm Kriz • Astrid Weins • Ruth Ellen Bulger

STRUCTURE–FUNCTION somewhat smaller. In both men and women, total kidney


mass best correlates with body surface area.
CORRELATIONS ALONG THE The concave medial margin has a slitlike aperture,
RENAL STRUCTURE called the renal hilum. Branches of the renal artery, vein,
The kidney functions as it does, in large part, because of nerves, lymphatics, and the expanded pelvis of the ureter
its architecture. In no instance is this more evident than in pass through the hilum. The hilum communicates with a
the urinary concentrating mechanism, where the complex attened space within the kidney called the renal sinus.
nephron and vascular interrelationships permit the coordi- Within this space, the renal pelvis branches into major and
nated function of different nephron and vascular elements minor calyces.
into countercurrent multiplication and exchange processes. Sections through the kidney reveal the cortex and me-
A recent proliferation of detailed structural, biochemical, dulla (Fig. 1.1). The human kidney is a multilobar organ
and functional information has led to an appreciation of containing 4 to 18 (average, 8) pyramids of medullary sub-
other structural–functional relationships that are relevant to stance1 and is situated so that their bases are adjacent to the
solute and water handling by the kidney. Moreover, it has cortex. The darker red cortical substance covers the base of
become evident that an understanding of the pathologic each medullary pyramid like the cap of an acorn. During fe-
developments in kidney diseases is only possible on the tal life, the kidney surface is demarcated by clefts that gradu-
basis of a thorough knowledge of kidney structure. ally disappear in the normal adult kidney. The apex of each
The purpose of this chapter is to review some of the re- medullary pyramid (called the papilla) extends into the renal
cent ndings, with special emphasis on structural–functional sinus and is capped by a funnel-shaped, minor calyx. The
relationships, to enhance our understanding of overall renal minor calyces receive the urine that is released from the kid-
function; therefore, this chapter is divided into two parts. ney into the extrarenal collecting system. A lobe of the kid-
The rst part considers the structural and functional inter- ney is composed of the conical medullary pyramid and the
relationships of each morphologic segment of the urinary surrounding cortical substance. During fetal development,
tubule, stressing the unique characteristics of each segment. some lobes may fuse and calyces are remodeled so that the
The second part discusses structure and function in terms of mature kidney has fewer calyces and papillae than the origi-
more general mechanisms used by several segments of the nal number of papilla anlagen 1; one calyx may drain a fused
renal tubule to accomplish speci c functions, such as ion or papilla developed from up to four anlagen, predominantly at
water transport. the kidney poles. Striated elements called medullary rays ex-
tend peripherally at intervals from the bases of the medullary
pyramids and penetrate into the cortex. These rays resemble
FORM OF THE HUMAN KIDNEY the medulla in structure and although they extend deeply
Human kidneys are paired, bean-shaped organs situated in into the cortex, they are part of the cortex. The rest of the
a retroperitoneal position on the posterior aspect of the ab- cortex is called the cortical labyrinth.
dominal cavity, on either side of the vertebral column against The medulla can be subdivided further either grossly or
the psoas major muscle. A brous capsule located within the microscopically (Fig. 1.2). The medulla has an outer zone
perirenal adipose tissue and surrounded by perirenal fascia that is adjacent to the cortex and an inner zone that includes
surrounds each kidney. The lateral border of each kidney the papilla. The outer zone is subdivided into an inner and
is convex. The kidneys of an adult man weigh approxi- outer stripe. This zonation is important because it represents
mately 120 to 170 g each and measure roughly 11 6 the location and orientation of the various segments of the
2.5 cm; those of an adult woman weigh slightly less and are renal tubules within the kidney.

1
2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

the functional kidney of the human. The metanephric kid-


ney is well suited to the human condition because of its ef-
cient ltering device and its complex tubule, which allows
for the production of not only dilute urine but also concen-
trated urine. This process occurs only in mammals and birds.
Although it is well suited for maintaining homeostasis, the
mammalian kidney is an inef cient organ for the elimination
of salt and water. In humans, 180 L of uid are ltered into
the tubular lumen every 24 hours, of which approximately
178 L must be returned to the blood.
Each human kidney contains approximately 1 million
functional units, called nephrons (with considerable inter-
individual variation).3 Each nephron is made up of a renal
corpuscle (glomerulus) and a complex tubular portion, which
drain into a unifying tubular system called the collecting duct
system. Both kinds of tubules represent the renal (or urinifer-
ous) tubules.
The nephrons are derived from the metanephric blas-
tema, the collecting ducts from the urethral bud. A connect-
ing tubule lies between the nephron and collecting ducts.
At present, there is controversy as to whether the connecting
tubule is derived from the metanephric blastema4–6 or the
ureteric bud.7 As is discussed in the next section, the con-
necting tubule has marked morphologic similarities to the
cortical collecting duct (CCD).
The segmentation of the renal tubule then includes the
following regions.8

The Nephron
FIGURE 1.1 Gross anatomic appearance of a human kidney. I. Renal corpuscle (most of which is called glomerulus)
A paraf n section through the whole kidney shows elements of A. Bowman’s capsule
the internal structure: C, cortex; M, medulla; P, papilla projecting B. Glomerular tuft
into a minor renal calyx; PE, pelvis; S, sinus; U, ureter. II. Proximal tubule
A. Convoluted part (pars convoluta) consists of P1 and
the rst part of P2 (PCT)
B. Straight part (pars recta) consists of the last part of
The relative volumes occupied by the cortex, outer me-
P2 and all of P3 (PST)
dulla, and inner medulla are 70%, 27%, and 3%, respectively,2
in humans. The relative thicknesses vary considerably among III. Thin limbs of the loop of Henle (intermediate tubule)
mammalian species. A. Thin descending part of short-looped nephrons
(SDTL)
B. Upper thin descending part of long-looped
RENAL (URINIFEROUS) TUBULES nephrons (LDTL up)
Human renal morphology resulted from a long evolution- C. Lower thin descending part of long-looped
ary process in which animals adapted to many changing nephrons (LDTL lp)
environmental conditions. The three sequential types of D. Ascending thin part of long-looped nephrons (ATL)
kidneys that evolved were the pronephros, mesonephros, IV. Distal tubule
and metanephros. The urogenital system of each human em- A. Straight part (pars recta)
bryo repeats this evolutionary process. The pronephros de- 1. Medullary thick ascending limb (MTAL), which
velops rst, but degenerates before attaining any functional includes regions located within the inner stripe
capacity. and outer stripe of the medulla
The mesonephric kidney functions for a short period 2. Cortical thick ascending limb (CTAL), which
in utero, but it also degenerates, with the notable exception includes the part ascending through the cortex,
of the part of the mesonephric tubules that form a portion the macula densa (MD), and the post macula
of the excurrent duct system of the male reproductive tract. densa segment
The metanephric kidney forms last and eventually becomes B. Convoluted part (pars convoluta) (DCT)
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 3

FIGURE 1.2 The relationship of nephron S UP ERFICIAL NEP HRON

segments to zones of the kidney.


Re na l corpus cle Dis ta l
convolute d
Conne cting tubule tubule
M
MIDCORTICAL E
NEP HRON P roxima l D
convolute d U
L
tubule L
A C
J UXTAMEDULLARY Colle cting duct
R
Y O
NEP HRON R
R
A T
Y E
Thick a s ce nding limb
X
Thick de s ce nding limb
Colle cting duct OU
S T TE
RI R
PE
O
U
T
Thick a s ce nding limb IN E
Thin de s ce nding limb S T NE R R
RI
Colle cting duct PE Z
O
N
E

M
E
D
Thin a s ce nding limb U
Thin de s ce nding limb L
Colle cting duct L
I A
N
N
E
R
Z
O
N
E

The Collecting Duct System The morphology of the nephron varies with the position
I. Connecting tubule (CNT) of the renal corpuscle in the cortex. Each nephron is classi-
II. Cortical collecting duct (or tubule) (CCD) ed as super cial, midcortical, or juxtamedullary, according
III. Outer medullary-collecting duct (or tubule) (OMCD) to the position of its renal corpuscle within the respective
regions of the cortex (Fig. 1.2) and the pattern of efferent
IV. Inner medullary-collecting duct (or tubule) including
vessel formation.9–11 A given segment tends to occupy a spe-
the papillary collecting ducts (also called the ducts of
ci c region of the kidney, which gives rise to the gross zona-
Bellini; IMCD)
tion referred to in the preceding text. In the human kidney,
Nephrons lie in characteristic positions (Fig. 1.2), with super cial nephrons empty singly into a terminal collecting
the renal corpuscles and proximal convoluted segments in duct, whereas several juxtamedullary nephrons empty into
the cortex (Figs. 1.3 and 1.4). The straight part of the prox- an arched tubular portion (arcade) that courses peripherally
imal tubule, the thin limb segments, and the straight part in the cortex before it turns to enter a medullary ray. Most
of the distal tubule form the loop of Henle, which enters a midcortical nephrons from humans empty individually as
medullary ray of the cortex and extends into the medulla, well.5,12 As known from the study of several species (rats and
where it bends, returning to the cortex by means of the rabbits), an arcade is established by the connecting tubule
same medullary ray. The loops of juxtamedullary nephrons epithelium (data from studies in humans are not available).
directly connect the outer stripe of the medulla without Nephrons also are classi ed as short or long looped ac-
ever being contained in a medullary ray. As the straight cording to the location of the position where their loops of
part of the distal tubule returns to the cortex, it passes by Henle turn within the kidney. Short-looped nephrons arise
the renal corpuscle from which the nephron originated, from renal corpuscles located in super cial and midcortical
forming the macula densa; then, after a short postmacula regions and have loops of Henle that turn within the outer
densa segment, it continues as the distal convoluted tubule medulla. In humans, some super cial nephrons may have
within the cortex. loops within the cortex itself. Short-looped nephrons have
4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 1.3 Scanning electron micrograph of the cortex. Con- FIGURE 1.4 Light micrograph of renal cortex (rat). Cortical ra-
voluted tubules are shown, along with renal corpuscles, some of dial vessels (A, artery; V, vein), glomeruli, and convoluted tubules
which contain a glomerular tuft (GT) and some from which the make up the cortical labyrinth. The straight tubular portions are
tuft is removed (arrow). A cortical radial artery (A) and vein (V) are found in the medullary rays of the cortex (one medullary ray is
also apparent. Note the thin wall of the vein. (Magni cation 140.) delineated by the hatched line). (Magni cation 80.)

short, thin limb segments that occur only along the descend- The renal corpuscle consists of Bowman’s capsule and
ing limb. Long-looped nephrons have loops of Henle that turn the glomerular tuft. The latter is made up of capillaries, de-
within the inner medulla and have thin limb segments in both rived from the afferent arteriole, their supporting cells, and an
descending and ascending limbs. Although most species have envelope consisting of the glomerular basement membrane
both long- and short-looped nephrons, some species, such as (GBM) and the visceral (podocyte) layer of Bowman’s capsule
dogs and cats, have only long ones,13 whereas other species, (Fig. 1.5). At the vascular pole, the visceral epithelium becomes
such as beavers, have only short ones.14,15 In human kidneys, the parietal epithelium, which then transforms into the proxi-
the ratio of short- to long-looped nephrons is 6:1 to 7:1. mal tubule epithelium at the urinary pole (Fig. 1.5). The space
between both layers is the urinary space (Bowman’s space).
The human renal corpuscle is roughly ovoid and
THE RENAL CORPUSCLE approximately 150 to 240 m in diameter. The term
The renal corpuscle ( rst segment of the nephron) is the site glomerulus is widely used to refer to the entire renal cor-
at which an ultra ltrate of the blood is produced (Fig. 1.5). puscle. The renal corpuscle without the parietal epithelial
The ltrate moves from the capillary lumen into Bowman’s cells is referred to as the glomerular tuft. The afferent arte-
space. This ow is in uenced by the following factors: re- riole enters the renal corpuscle at the vascular pole, where
nal blood ow; the oncotic and hydrostatic pressures in it divides into several primary branches that each ramify to
the capillaries and in Bowman’s space; the size, shape, and form a network of anastomosing capillaries, called a lobule.
charge of plasma molecules; and the various morphologic The lobule has a supporting region called the mesangium
components of the wall separating the capillary lumen from (Figs. 1.6 and 1.7). All lobules together establish the tuft;
Bowman’s space. The ltrate contains only barely detectable the converging mesangial regions are called the glomerular
quantities of plasma proteins.16 The ltration barrier increas- stalk, by which the tuft is connected to the extraglomerular
ingly restricts the passage of larger molecules, with very little mesangium (see Juxtaglomerular Apparatus). The capillar-
ltration of molecules larger than albumin (70 kDa).17 ies coalesce toward the center of the capillary tuft to form
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 5

FIGURE1.5 Schematic of a longitudinal section through a glom-


erulus and juxtaglomerular apparatus (JGA).The direction of blood
FIGURE 1.6 Schematic of a cross-section of a glomerular capillary
ow in the glomerular arterioles is indicated by arrows.The capil-
and its relationships to the mesangium.The capillary is made up of
lary network, together with the mesangium, is enclosed in a com-
a fenestrated epithelium.The peripheral part of the endothelium
mon compartment bounded by the glomerular basement mem-
tubule is surrounded by the glomerular basement membrane
brane (GBM) (shown in dark gray).The outer aspect of the GBM is
(GBM; shown in dark gray), which, at mesangial angles (arrows), de-
covered by the glomerular visceral epithelium (podocytes). Note
viates from a pericapillary course and covers the mesangium.The
that there is no basement membrane at the interface between the
outer aspect of the GBM is covered by the interdigitating pattern of
capillary endothelium and the mesangium. At the vascular pole,
podocyte foot processes. In the center, a mesangial cell is shown; its
the visceral epithelium, together with the GBM, is re ected into
many processes contain micro lament bundles and extend toward
the parietal epithelium of Bowman’s capsule, which, at the urinary
the GBM, to which they are connected.The mesangial matrix con-
pole, passes over into the epithelium of the proximal tubule.The
tains an interwoven network of micro brils. (From Venkatachalam
JGA consists of the macula densa of the distal tubule, the extraglo-
MA, Kriz W. Anatomy of the kidney. In: Heptinstall R, ed. Pathology of
merular mesangium (which is continuous between both arterioles
the Kidney, 4th ed. Boston: Little, Brown; 1991, with permission.)
and continues via the glomerular stalk into the intraglomerular
mesangium), and the granular cells within the afferent arterioles. All
cells that have been suggested to be of smooth muscle origin are
(podocytes), (c) the endothelial cells lining the capillaries,
shown in black. Note the sympathetic nerve terminals at the affer-
(d) the glomerular basement membrane (GBM), and (e) the
ent arteriole. (From Kriz W, Sakai T, Hosser H. Morphological aspects
intraglomerular mesangial cells and matrix. In the rat, the
of glomerular function. In: Davison AM, ed. Nephrology,Vol. 1. Pro-
ratio of the number of endothelial cells to mesangial cells to
ceedings of XInternational Congress of Nephrology, London, 1987.
visceral epithelial cells is 3:2:1.18
London: Bailliere Tindall; 1988:3, with permission.)
The Visceral Epithelium of Bowman’s Capsule
the efferent arteriole, which runs through the stalk and exits The visceral epithelial cells (nowadays generally called podo-
from the vascular pole. The efferent arteriole again breaks cytes) are octopus-shaped cells that reside in Bowman’s space
up to form a second capillary network, which surrounds the and attach to the GBM only by way of their processes (see be-
tubules and is called the peritubular capillary network. low). This shape was well described by Zimmermann19 and is
The renal corpuscle, therefore, consists of the following seen to advantage in scanning electron micrographs (SEM) (see
parts: (a) the parietal epithelium, (b) the visceral epithelium later, Fig. 1.10). The exact details of cell shape differ depending
6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

The cell bodies give rise to large primary processes that


may branch another time, nally splitting apart into ter-
minal processes, called foot processes (as seen from SEM
pictures, “ nger processes” would be a better word) or ped-
icels (Figs. 1.6, 1.8 to 1.11, and see Fig. 1.13). The foot
processes are anchored within the GBM to a depth of about

FIGURE 1.7 Transmission electron micrograph of a rat glomeru-


lar lobule. Glomerular capillaries and the glomerular mesangium
occupy a common compartment enclosed by the glomerular
basement membrane (GBM). The mesangial cell body (in the
center) gives rise to many processes that ll (together with the
mesangial matrix) radial arms that extend to the peripherally
located capillaries. The outer aspect of the GBM is covered by
podocytes. (Magni cation 3,500.)

on the species being studied.20,21 As a consequence of the high


degree of differentiation, podocytes, like neurons, are incapable A
of regenerative cell replication in the adult. Thus, podocytes that
were lost, for any reason, cannot be replaced by new ones.22–24
On the other hand, in glomerular diseases based on dedifferen-
tiation of podocytes (collapsing glomerulopathy), a vivid prolif-
eration of the dedifferentiated cells accompanies the disease.25
Recent observations in parietal epithelial cells question
this view. First, it has been proposed that a niche of glo-
merular epithelial stem cells resides within the parietal epi-
thelium at the transition to the proximal tubule.26,27 It is an
intriguing hypothesis that proliferating stem cells from this
locus may transform into podocytes and may reach the tuft
B
via the transitions of the epithelia at the glomerular vascular
pole. Migration of parietal cells onto the the vascular pole FIGURE 1.8 Transmission electron micrographs of glomerular
and subsequent transition into podocytes have been shown capillaries (C) and associated mesangium. A: A mesangial cell
to occur in the newborn mouse.28 However, evidence that body (M) gives rise to cell processes that extend to peripherally
such a process may be of any relevance in the adult has so located capillaries. Note that there is no basement membrane at
far not been presented.28 Moreover in the adult mouse, it the interface between the capillary endothelium and the mesan-
has been shown that proliferating parietal epithelial cells gium. (Magni cation 13,000.) B: Capillary mesangium interface.
may reach the glomerular tuft via tuft–capsule bridges; there Beneath the endothelium (E), tonguelike mesangial cell processes
they replace podocytes causing the collapse of the concerned run toward both opposing turning points of the GBM (arrows).
tuft area obviously by bringing the local cross talk between They contain micro lament bundles that obviously interconnect
podocytes and the endocapillary compartment to an end.29 the GBM of both mesangial angles. (Magni cation 24,000.)
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 7

FIGURE 1.9 Transmission electron micrograph showing the podocyte (P), pedicels (PC) near the basement membrane (BM), and the
endothelial cells lining the capillary (C). (Magni cation 34,000.)

40 to 60 nm. The foot processes interdigitate in a complicat- material (4 to 6 nm thick) called the ltration-slit membrane
ed manner with those from adjacent cells to form an elabo- (see Figs. 1.9 and 1.13). If tannic acid is added to the xative
rate layer of small processes along the glomerular basement solution, a highly ordered isoporous substructure is revealed
membrane. This interdigitation results in the formation of in en face views of the ltration-slit membrane.30 Staggered
an extensive series of narrow slits between the foot process- rodlike units project from the podocyte plasmalemma and
es, which provide a long extracellular path for ltration of connect to a central linear bar. These rodlike units delin-
water and solutes (Fig. 1.10). In transmission electron mi- eate rectangular pores 4 14 nm within the slit membrane
crographs, these slits are bridged by a layer of extracellular (i.e., which approximate the size of an albumin molecule).

FIGURE 1.10 Scanning electron


micrograph showing the elaborate
cell shape of rat podocytes.
(Magni cation 5,900.)
8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 1.11 Schematic drawing


of the molecular equipment of
podocyte foot processes. Cas,
p130Cas; Cat, catenins; CD, CD2-
associated protein; Ez, ezrin;
FAK, focal adhesion kinase; ILK,
integrin-linked kinase; M, myosin;
N, NHERF2; NSCC, nonselective
cation channel; PC, podocalyxin; S,
synaptopodin; TPV, talin-
paxilinvinculin; U, utrophin; Z,
ZO-1. See text for further expla-
nations. (From Endlich K, Kriz W,
Witzgall R. Update in podocyte
biology. Curr Opin Nephrol
Hypertens. 2001;10:331, with
permission.)

Generally, the slit diaphragm is considered as an ad- The cell body of podocytes contains a large nucleus
herenslike intercellular junction.31 Intensive research in that tends to be indented in the region of the large Golgi
recent years has uncovered several transmembrane pro- apparatus. Furthermore, it houses abundant rough-surface
teins that participate in the formation of the slit membrane endoplasmic reticulum; individual cisternae, generally ar-
(Fig. 1.11)—including P-cadherin,31 nephrin,32 Neph1,33 ranged in one complex per cell, are widened and lled with
and FAT.34 Other molecules, such as ZO1,35 Podocin,36 ne granular material of varying electrondensity—their
CD2AP,37 and catenins mediate the connection to the ac- relevance is unknown. In addition to the synthesis of mem-
tin cytoskeleton (see below). Nephrin is a member of the brane proteins necessary to supply the huge surface of their
immunoglobin superfamily (IgCAM); its gene, NPHS1, has processes, podocytes in the adult synthesize and secrete all
been identi ed as the gene whose mutations cause congeni- components of the GBM42 (see below). On the other hand,
tal nephrotic syndrome of the Finnish type.32 In addition to abundant multivesicular bodies (predominantly found in
its role as a structural component, nephrin acts as a signaling the large cell processes) demonstrate strong catabolic activ-
molecule that can activate MAP kinase cascades.38 Neph1 ity in podocytes.
is considered as a ligand for nephrin. Podocin belongs to Podocytes contain a well-developed cytoskeleton that
the raft-associated stomatin family, whose gene, NPHS2, is accounts for the unique shape of the cells and the main-
mutated in a subgroup of patients with autosomal recessive tenance of the processes. In the cell body and the primary
steroid-resistent nephrotic syndrome.36 These patients show processes, microtubules and intermediate laments, such as
disease onset in early childhood and rapid progression to vimentin and desmin, dominate, whereas micro laments are
end-stage renal failure. Podocin interacts with nephrin and densely accumulated in the foot processes. Here, they are
CD2AP.39 FAT is a novel member of the cadherin superfam- part of a complex contractile apparatus.43 The micro laments
ily with 34 tandem cadherinlike extracellular repeats and form loop-shaped bundles, with their limbs running along
a molecular weight of 516 KDa.40 Because FAT has a huge the longitudinal axis of the foot processes. The bends of these
extracellular domain, it is speculated that it dominates the loops are located centrally at the transition to the primary
molecular structure of the slit membrane34; the FAT mutant processes and are probably connected to the microtubules by
mouse fails to develop a slit membrane.41 P-cadherin 31 is the microtubule-associated protein .44 Peripherally, the actin
thought to mediate with its intracellular domain the linkage bundles appear to be anchored in the dense cytoplasm asso-
to - and -catenin, a complex which then connects to the ciated with the cell membrane of the soles of foot processes,
actin cytoskeleton via -catenin and -actinin. Taken to- and are dynamically linked to the slit diaphragm complex
gether, many components of the slit membrane are known, (discussed earlier). The importance of the podocyte actin cy-
but an integrative model of its substructure including all toskeleton is emphasized by the discovery of inherited forms
components is thus far lacking. of focal segmental glomerulosclerosis (FSGS) caused by
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 9

mutations in actin-binding proteins: -actinin-4 is a widely Endothelium


expressed homodimeric protein that bundles and crosslinks The endothelium consists of a simple squamous layer of
actin, and is highly expressed within podocyte foot processes. fenestrated (porous) cells with the cell nuclei generally lo-
It interacts with a variety of other adhesion and signalling cated near the axial region of the capillary loop (Fig. 1.6).
molecules. Several mutations in the ACTN4 gene encoding The fenestrated regions (Fig. 1.12), which compose roughly
this protein cause a late-onset, autosomal dominant form of 55% of the surface area,66 have a thin layer of cytoplasm
kidney failure.45,46 More recently, nine independent missense (about 50 nm thick) penetrated by numerous fenestrae of
mutations in INF2, which encodes a member of the formin round, oval, or irregular shape and varying sizes. The total
family of actin-regulating proteins, were shown to segregate area occupied by fenestrae accounts for 13% of the capillary
with FSGS in 11 unrelated families.47 surface. The fenestrated regions outline the pericapillary
Speci c transmembrane matrix receptors anchor the portions of the glomerular basement membrane, but also
podocyte foot processes to the GBM. Two systems are so far may be found adjacent to the mesangium (Fig. 1.8). The
known. The rst is a speci c integrin heterodimer consisting fenestrae have a diameter of 50 to 100 nm (thus they are
of 3 1 integrins. Within the GBM, these integrins bind to larger than endothelial fenestrae elsewhere in the body) and
collagen type IV, bronectin, and laminin 11.48,49 Second, are not bridged by diaphragms. Thus, this kind of a porous
a dystroglycan complex connects the intracellular molecule endothelium is unique for glomerular capillaries. Fenestrae
utrophin to laminin 11, agrin, and perlecan in the GBM.50,51 with diaphragms are found only in the out ow segment of
Both integrins and dystroglycans are coupled via adapter the efferent arteriole.67 Nonfenestrated regions are generally
molecules (paxillin, vinculin, -actinin, etc.) to the podocyte seen over nuclei and mesangial cell regions. Human glo-
cytoskeleton, allowing outside-in and inside-out signaling as merular endothelial cells also are fenestrated.68,69 A cell coat
well as transmission of mechanical force in both directions. that is rich with polyanionic glycoproteins, including podo-
In addition to the actin cytoskeleton within the foot calyxin, covers the endothelial surface70–72 and appears to
processes, a subplasmalemmal actin system is found in ll the fenstraelike “sieve plugs.”73 In addition, above this
podocytes.43 This actin network is connected to the trans- “classic” glycocalix a 200-nm thick endothelial surface lay-
membrane sialoprotein podocalyxin, which represents the er has been revealed consisting of loosely attached plasma
major protein of the negatively charged surface coat of podo- compounds.74
cytes.52,53 The cell coat has the characteristics of a glycocalyx
that contains sialic acid. A decrease in the content of sialic
acid is associated with a podocyte foot process effacement
and results in protein leakage through the lter, which has
been shown under a great variety of circumstances.54–58
A huge body of data has been accumulated in recent
years concerning the inventory of receptors and signaling
processes starting from them in podocytes. cGMP signaling
(stimulated by atrial natriuretic peptide [ANP], brain natri-
uretic peptide [BNP], and C-type natriuretic peptide [CNP],
as well as by nitric oxide [NO]), cAMP signaling (stimulated
by prostaglandin E2, dopamine, isoproterenol, parathyroid
hormone [PTH]/PTH-related peptide [PTHrP]), and Ca2
signaling (stimulated by a huge number of ligands, includ-
ing angiotensin II, acetylcholine, prostaglandin F2 (PGF2),
arginine vasopressin [AVP], adenosine triophosphate [ATP],
endothelin, histamine, etc.) have been identi ed.59 An ex-
ample of an ion channel of particular importance in podo-
cytes is transient receptor potential canonical 6 (TRPC6), a
nonselective cation channel, which is activated by diacyl-
glycerol in a protein kinase C-dependent manner.60 Muta-
tions in TRPC6 were found to cause autosomal dominant,
late adult-onset proteinuria, with a similar clinical phenotype
as seen in ACTN4-mediated FSGS.37 The major target of this
signaling orchestra is the cytoskeleton, the concrete effects
of which, however, are poorly understood. Other receptors,
such as for C3b,61 Heymann’s antigen,62 transforming growth
factor (TGF ),63,64 broblast growth factor 2 (FGF2),65 and FIGURE 1.12 Scanning electron micrograph of a sectioned
various other cytokines and chemokines have been shown to capillary loop showing the pores (arrows) of the endothelium.
be involved in the development of podocyte diseases.59 (Magni cation 50,400.)
10
10 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

As elsewhere in the body, glomerular endothelial cells noncollagenous globular domain called NC 1. At the amino-
are active participants in processes controlling coagulation, terminus the helix possesses a 60- m triple helical rod, the
in ammation, and immune processes. Renal endothelial 7S domain. Interactions between the 7S domain and the NC1
cells express surface antigens of the class 2 histocompatibility domain allow collagen type IV monomers to form tetramers
complex. Like platelets, glomerular endothelial cells contain that, by lateral association of triple helical strands, assemble
components of the coagulation pathway and are capable of into a three-dimensional network.100,101 Laminin forms a
binding factors IXa and Xa and synthesize, release, and bind second network that is superimposed to the collagenous net-
von Willebrand factor (factor VIII).69 Glomerular endothelial work. Laminin is a noncollagenous glycoprotein consisting
cells synthesize and release endothelin-1 and endothelium- of three polypeptide chains, two of which are glycosylated
derived relaxing factor (EDRF).75 Glomerular endothelial and cross-linked by disul de bridges.95,102,103 Laminin binds
cells have receptors for vascular endothelial growth fac- to speci c sites on the polymerized network of type IV col-
tor (VEGF) and angiopoetin I that are produced by podo- lagen as well as the basal endothelial and epithelial integrins
cytes.76,77 The signaling axis via VEGF appears to have a (see the preceding). The -5-, -2-, -1-laminin chains are
major relevance for the maintenance of the glomerular tuft. assembled to form the GBM-speci c heterotrimeric laminin
Glomerular endothelial cells, in turn, synthesize and secrete 11.48 This combined network of collagen type IV and lam-
platelet-derived growth factor (PDGF ) that acts on adja- inin provides mechanical stability to the basement mem-
cent mesangial cells.78 brane and serves as a basic structure on which other matrix
components attach.
Glomerular Basement Membrane The proteoglycans of the basement membrane consist
The GBM covers the capillary loops except in the axial re- of core proteins and covalently bound glycosaminoglycans,
gion, where it is re ected over the mesangium to the next which are concentrated in the laminae rarae internae and
capillary loop, accompanied by the layer of foot processes externae, where they have been referred to as anionic sites
(Figs. 1.6 to 1.8). The endothelial cells do not have a sepa- and can be localized with cationic probes.104 The major pro-
rate basement membrane; thus, the endothelial cells directly teoglycans of the GBM are of the heparan sulfate type—the
abut the mesangium toward axial regions. In adult humans, most prominent is agrin,96 but perlecan also has been shown
the basement membrane has a mean diameter of 320 to 340 to occur in the GBM.42 Digestion of these molecules with
m.79 It is thinner in young children and most laboratory heparinase leads to a dramatic increase in the permeability
animals.80 of the basement membrane to anionic native ferritin used as
The basement membrane is composed of three lay- a probe.70
ers: an outer, less dense subepithelial layer, the lamina rara
externa; a central, electron-dense layer, the lamina densa; Mesangium
and an inner subendothelial layer, the lamina rara interna, The mesangium consists of mesangial cells that are embed-
which is continuous with the mesangial matrix (see Figs. 1.9 ded in a mesangial matrix. The term mesangium was intro-
and 1.13). duced by Zimmermann in 192919 to describe the cells that
During glomerulogenesis, the GBM is generated as two form the stalk of the glomerulus and the axes of its lobules.
separate layers produced by glomerular endothelial and epi- Glomerular capillaries pursue a tortuous, highly anastomos-
thelial cells. The two sheets are fused together to form the ing course around the mesangial axes. Together with the
mature GBM.81–84 In the adult, the GBM is subject to a con- capillaries, the mesangium occupies the space inside the
tinuous turnover,85–88 but few details are known so far about GBM, frequently termed the “endocapillary region.” Topo-
these processes. Podocytes appear to be the dominant cell graphically, the mesangium can be subdivided into a juxta-
type to synthesize and probably to degrade the GBM. Podo- capillary region, where it abuts the capillary endothelium,
cytes are alone capable to synthesize all the components of and more centrally located axial regions, which are bound
the GBM42,89,90; glomerular endothelial cells and also mesan- by the perimesangial GBM (Figs. 1.5 to 1.7).105 The glo-
gial cells may contribute to the formation of the GBM.91 It merular mesangium is continuous with the extraglomerular
is less clear how the GBM degrades. In recent years, several mesangium (Polkissen or lacis cells) along the glomerular
extracellular matrix degrading enzymes have been described stalk (Fig. 1.5). Both intraglomerular and extraglomerular
being produced by podocytes and mesangial cells.92–94 The mesangial cells have many similarities.
relevance of these enzymes for the turnover of the GBM re- Mesangial cells are quite irregular in shape, with many
mains to be established. cytoplasmic processes extending from the cell body toward
The GBM is generally considered as a hydrated mesh- the GBM. They have structural characteristics similar to
work consisting of collagen type IV, laminin, entactin/nido- those of smooth muscle cells in that they contain many bun-
gen, and sulfated proteoglycans including agrin and per- dles of micro laments (especially within the cell processes)
lecan.42,95–99 Models of the ultramicroscopic structure of the and peripheral dense bodies. Actin, myosin, and -actin
basement membrane picture the GBM as a mat of collagen have been shown by immunocytochemistry to be contained
type IV. Monomers of type IV collagen consist of a 400- m in mesangial cells.106,107 The relevance of mesangial cell con-
triple helix that, at its carboxy-terminal end, has a large tractility is discussed in the following text.
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 111
1

The processes of mesangial cells extend toward the Supportive Functions of the Mesangium
GBM, to which they are attached either directly or by the in- and Podocytes
terposition of extracellular bundles of micro brils (Figs. 1.6
The glomerular tuft is constantly exposed to comparably
and 1.9). The GBM has to be considered as the effector struc-
high intraglomerular pressures within glomerular capillaries
ture of mesangial contractility.105,108 Connections between
and mesangium. The high intraglomerular pressures chal-
mesangial cells and the GBM are especially prominent in the
lenge not only the glomerular capillaries themselves but also
juxtacapillary region. At this site, tonguelike mesangial cell
the folding pattern of the glomerular tuft. Increased pres-
processes (packed with micro lament bundles) run under-
sures lead to the loss of the folding pattern and to dilation of
neath the capillary endothelium toward the turning points
the glomerular capillaries. Therefore, we have to ask: what
(mesangial angles) of the GBM, where they are anchored.
are the speci c structures and mechanisms that counteract
Generally, two of these processes interconnect the GBM
the expansile forces in the glomerular tuft? To answer this
from two opposing mesangial angles (Fig. 1.8). In the axial
question we have to distinguish between the structures and
mesangial region, contractile lament bundles are predomi-
mechanisms maintaining (1) the folding pattern of the glo-
nantly found within the numerous ngerlike projections of
merular tuft and those maintaining (2) the width of glomer-
mesangial cells. These microprojections also run toward the
ular capillaries.
GBM and are anchored to it. As in the juxtacapillary region,
The folding pattern of the glomerular tuft is primarily
these micro lament bundles interconnect opposing parts of
sustained by the mesangium.105,108,121 Mesangial cells are
the GBM.105
connected to the GBM by their contractile processes—they
The mesangial matrix lls the highly irregular spaces
maintain the infoldings of the GBM by centripetal contrac-
between mesangial cells and the perimesangial part of the
tions, thereby allowing for the capillaries to arrange in the
GBM. In immunocytochemical studies, collagen types IV
peripheral expansion of the GBM. This supporting role of
and V, heparan sulfate proteoglycan, bronectin, laminin,
mesangial cells is best illustrated under circumstances with
and entactin have been localized within the mesangial ma-
loss of mesangial cells, such as Thy-1 nephritis.122 Under
trix.99,109,110 Among these components, bronectin is the
those circumstances the folding pattern of the GBM is pro-
most abundant and has been shown to be associated with
gressively lost, nally resulting in mesangial aneurysms.
micro brils.109,111 Fibrillin 1 and other speci c elastic ber
Podocytes clearly contribute to maintenance of the folding
proteins have been detected in the glomerular mesangium
pattern by speci c cell processes that interconnect oppos-
and have been shown to be produced by mesangial cells.112,113
ing parts of the GBM from outside within the niches of the
In specimens prepared for transmission electron micros-
infoldings. This function is also best illustrated under cir-
copy according to routine methods, the mesangial matrix
cumstances with loss of mesangial support: podocytes are
appears as basement membranelike material, albeit more -
capable of maintaining a high degree of the GBM folding
brillar in character than the basement membrane proper.114
pattern for 2 to 4 days after which they obviously fail, and
In specimens prepared by a technique that avoids osmium
mesangial aneurysms become prominent.122
tetroxide and uses tannic acid for staining, the mesangial ma-
The width of glomerular capillaries, in the long run, is
trix is seen to contain a dense network of micro brils.105,115
probably controlled by growth processes accounting for dif-
Micro brils are noncollagenous, nonbranching, hollow struc-
ferently sized capillaries. The width of a given capillary, in an
tures of indeterminate length that are about 15 m thick.116
acute situation being exposed to changes in blood pressure,
Within the mesangium, micro brils form a three-dimension-
appears to be stabilized by the GBM, which is a strong elas-
al network that establishes a solid base of contact between
tic structure123 and, together with the mesangial cell bridges
mesangial cells and the GBM, fettering the GBM to mesangial
(see previous text), capable of developing wall tension.121,124
cells. Distinct bundles of micro brils may be regarded as “mi-
In addition, the tensile strength of the GBM is reinforced
crotendons” that allow the transmission of contractile force of
by podocytes. Podocytes are a kind of pericyte; their foot
mesangial cells to speci c sites of the GBM.105,115 The func-
processes represent a unique type of pericyte process which,
tional relevance of this system is discussed later.
like elsewhere in the body, counteract the dilation of the ves-
The relevance of mesangial cells in phagocytosis is well
sel. Podocyte processes are rmly attached to the underlying
documented. Mesangial cells are able to ingest particular
GBM (see previous text); their cytoskeletal tonus counteracts
tracers as well as macromolecules, such as thorotrast,117
the elastic extension of the GBM. In this function, podocytes
ferritin,114 and aggregated proteins, as well as immune
cannot be replaced by any other cell—failure in this function
complexes.118 An increased uptake of such materials by
will lead to capillary dilation.
mesangial cells has been noted in proteinuric states.114,119
It appears, however, that mesangial cells proper (i.e.,
mesangial cells that have contractile properties) are not pri- Glomerular Filtration Barrier
marily phagocytotic. A small subpopulation (3% to 7%) of The essential components of the glomerular ltration barrier
cells in this region has been recognized as bone marrow- are the endothelium (Fig. 1.13), which is perforated by large
derived—they represent macrophages that have taken up open pores, the extracellular matrix feltwork of the GBM
residence in the mesangium.120 membrane, and the slit diaphragms between the podocyte
12
12 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

On the basis of evidence of contractility of mesangial


cells exposed to vasoconstrictor stimuli in culture,129,130
of dimensional changes observed in intact glomeruli ex
vivo,131,132 and of changes in ultra ltration coef cient Kf
found in vivo in response to vasoactive substances,133,134
some researchers have concluded that mesangial cells con-
tract in situ and that this contraction alters glomerular ltra-
tion dynamics by decreasing ltration surface area.
Other considerations speak against this possibility. The
geometric arrangement of the mesangial contractile appara-
tus (Fig. 1.6), however, does not seem to be compatible with
the previously mentioned sequence of actions. Shortening
of the mesangial cell processes connecting opposing angles
would only bring the angles closer together, compressing
the mesangial capillary interface, but leaving peripheral
capillary wall area ( ltration area) unaltered. In addition,
with regard to the contractility of mesangial cells ex vivo
(in culture as well as preparations of whole glomeruli), it
should be remembered that mesangial contraction in these
cases is not opposed by intercapillary hydrostatic pressure
as it is in situ. These considerations, together with the ab-
sence of measurable changes in glomerular tuft dimensions
in morphometric studies,135,136 as well as in response to va-
soactive substances in vivo,137 have led to the suggestion 121
FIGURE 1.13 Transmission electron micrograph from a rat renal that the mechanical action of the mesangial cell contraction
corpuscle showing the endothelial lining (E), the basement is essentially static in nature, developing tension that serves
membrane (BM), and the pedicels (P). The ltration-slit membrane to counteract expansile forces on the tuft without induc-
(arrow) bridges the pedicels. (Magni cation 23,600.) ing signi cant changes in capillary dimension. If mesangial
contractility acutely alters the glomerular ultra ltration co-
ef cient Kf, it is, therefore, probably not because of an acute
foot processes. When compared with the barrier established change in ltration surface area.
in capillaries elsewhere in the body, a glomerular ltration The low permeability of the glomerular ltration barrier
barrier is quite different in two respects: its permeability to to plasma proteins is still poorly understood. Several points
water, small solutes, and ions is extremely high, whereas seem to be relevant. First, there is no vesicular transport of
its permeability to plasma proteins the size of albumin and proteins through this barrier as in most other capillaries else-
larger is very low. where in the body. The barrier function of the glomerular
The high hydraulic permeability is rooted in the fact lter for macromolecules is quite speci c and is selective for
that ltration occurs along extracellular routes. All compo- size, shape, and charge.138–140
nents of this route—the endothelial pores, the highly hy- In early extensive studies, Farquhar and associ-
drated GBM, and the slit membrane—can be expected to be ates,138,141,142 as well as Rennke and associates,143 used
quite permeable to water and small solutes. Drummond and tracers, such as ferritin and dextrans of different sizes
Deen 125 have calculated the hydraulic conductance of the and charge, to elucidate the role of the various layers in
individual layers. According to this calculation, the hydrau- determining the selectivity of this ltration barrier. When
lic resistance of the endothelium is negligible. The GBM and their results are summarized, it appears that the basement
the ltration slits each make up roughly one-half of the total membrane may be the major barrier to anionic substances,
hydraulic resistance of the ltration barrier. whereas the most restrictive part for uncharged and cationic
Any decrease in the length of the ltration slit, and thus substances may be the slit diaphragm. Uncharged macro-
in slit area, as in experimental and clinical glomerulopa- molecules up to an effective radius of 1.8 m pass freely
thies along with footprocess effacement, correlates with the through the lter. Larger compounds are more and more
decrease in the ultra ltration coef cient Kf.126,127 A model restricted (indicated by their fractional clearances, which
simulating those conditions showed, along with a decrease progressively decrease) and are totally restricted at effective
in slit area, its relevance in determining increases in ow radii of more than 4.0 m. The effective radius is an empiric
resistance. The decrease in ltration slit area caused the aver- value, measured in arti cial membranes, that takes into ac-
age path length for the ltrate, through the basement mem- count the shape of micromolecules and also attributes a
brane, to increase, thereby explaining the overall decreased radius to nonspherical molecules. Plasma albumin has an
hydraulic permeability.128 effective radius of 3.6 m; without the repulsion because
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 113
3

of the negative charge, plasma albumin would pass through


the lter in considerable amounts.144
Thus, since these early studies, the glomerular barrier
has been generally suggested to contain a size- and a charge-
restrictive element. Despite more than 40 years of intensive
research, the principles of the glomerular barrier function
are still controversial and poorly understood—a situation
that gives room even to hypotheses claiming that the glo-
merular barrier itself does not have any restrictive properties
to macromolecules, such as albumin.145–147 A more realistic
and elegant recent barrier hypothesis is based on the ob-
servation that ltration—the ow of ltrate through the
barrier—creates a potential difference of 0.02 to 0.05 mV
negative in Bowman’s space and that this potential differ-
ence is suf cient to hinder the negatively charged macro-
molecules like albumin from passing the lter.148
Summarizing the present status of knowledge, accu-
mulating evidence suggest that the GBM has little relevance
in size restriction, but the slit membrane is the decisive
size-restrictive element within the glomerular barrier.149
The current strong engagement in elucidating the molecu-
lar architecture of the slit membrane promises to enlighten FIGURE 1.14 Transmission electron micrograph of a proximal
the porous pattern of the slit membrane and, hopefully, tubule (P1 segment) of the rat. Note the apical brush border,
explain its size-restrictive property. Regarding the charge the vesicular zone of the apical cytoplasm, and the basal zone
selectivity, it appears that the proximal portion of the bar- of interdigitating cell processes lled with mitochondria.
rier, above all the endothelium, are most important.73,150–152 (Magni cation 2,300.)
Furthermore, prevention of albumin from entering the
lter is also dependant on normal hemodynamic condi-
tions in glomerular capillaries, as rst shown by Ryan and occupying the medullary ray; and S3, or P3, corresponds to
Karnovsky.153 the remaining part of the pars recta located primarily in the
outer stripe of the outer zone of the renal medulla. The tran-
sition from P1 to P2 is gradual, whereas the transition from P2
PROXIMAL TUBULE to P3 is generally abrupt, except in rabbits.158
At the urinary pole of the glomerulus, the at parietal epi- To confuse matters further, the morphology of the sub-
thelium of Bowman’s capsule transforms abruptly into the divisions differs not only from each other but also among
high epithelium of the proximal tubule. In some species species, including mice,159 rats,156 rhesus monkeys,160 rab-
(rabbits),154 a neck segment is found interposed between bits,158 and dogs.161 For a detailed description of a particular
the glomerulus and the proximal tubule—short neck seg- species, the reader should consult the original studies. Seg-
ments also may be seen in humans.80 In contrast, in mice, mentation of human proximal tubules has not been studied
the proximal tubule epithelium generally begins deep within as recently or thoroughly because of a lack of availability of
the Bowman’s capsule. well- xed normal human kidney and, therefore, has been
The proximal tubule is composed of segments that have divided only into convoluted and straight regions.162
differing morphology, functional relevance, and vulnerabil- In general, cells of the P1 region are tall, have a well-
ity to toxins (Figs. 1.14 to 1.18). The two segments most developed apical microvillus border, an elaborate cell shape
frequently identi ed are the proximal convoluted portion with well-developed lateral interdigitating processes con-
(pars convoluta) occupying the cortical labyrinth (Figs. 1.14 taining abundant, large mitochondria, and a well-developed
to 1.17), and the straight portion (pars recta) in the medul- endocytic apparatus (Fig. 1.16). P2 cells decrease in cell
lary rays of the cortex and the outer stripe of the outer me- height from those seen in P1, have a shorter microvillus bor-
dulla (Fig. 1.18). Further subdivision on structural criteria der, and are less elaborately shaped cells with smaller mi-
results in the three segments of P1, P2, and P3 according to tochondria (Fig. 1.17). The P3 cells are more cuboidal in
Jacobsen and Jorgensen,155 and S1, S2, and S3 according to shape (less elaborate) and their microvillus border is gen-
Maunsbach.156,157 S1, or P1, corresponds to the rst segment erally less elaborate. The cells of rats are an exception and
of the pars convoluta and lies exclusively in the cortical laby- have a well-developed brush border of very long microvilli
rinth; S2, or P2, corresponds to the remainder of the convo- (Fig. 1.18).156 This cytologic segmentation of the proximal
luted segment and the beginning of the pars recta, with the tubule is maintained in super cial, midcortical, and juxta-
rst part occupying the cortical labyrinth and the remainder medullary nephrons.
14
14 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 1.15 Transmission electron micrograph of a human proximal convoluted tubule showing the brush border (BB) and apical
condensing vacuoles containing small dense bodies (arrows). (Magni cation 7,800.)

FIGURE 1.16 Transmission electron micrograph of a P1 segment from a rat kidney. (Magni cation 8,000.)
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 115
5

FIGURE 1.17 Transmission electron micrograph of a P2 segment from a rat kidney. Note the large dense lysosomes (L) and peroxi-
somes (P). (Magni cation 10,000.)

FIGURE 1.18 Transmission electron micrograph of a P3 segment from a rat kidney showing the extensive microvillus border (BB) and
more simple cell shape of this segment. (Magni cation 8,000.)
16
16 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Proximal Convoluted Tubule (P1 and Part of P2 )


The proximal convoluted tubule is the longest and larg-
est segment of the mammalian nephron. The tubule is
lined by cells that have an elaborate cell shape based on
extensive basolateral interdigitations (Figs. 1.19 to 1.21),
a well-developed microvillus border, a prominent intracel-
lular digestive tract (endocytic apparatus and lysosomes
(Fig. 1.15), and numerous large peroxisomes (microbod-
ies). The single ovoid nucleus lies in the middle to basal
region of the cytoplasm.

Cell Shape and Mitochondria


The cells are characterized by an extensive system of lateral
cell processes that interdigitate with lateral processes from
adjacent cells (Figs. 1.19 to 1.21). These lateral processes
can extend the entire height of the epithelium, especially
in the P1 segment, but become more elaborate toward the
basal regions of the lateral surface (Fig. 1.16). The com- FIGURE 1.20 Transmission electron micrograph of the basal
plex shape of these cells serves to increase the lateral cell cytoplasm from a rat proximal tubular cell. (Magni cation
surface area manyfold, which provides a greater area for 20,600.)
transporters, above all of the Na -K -ATPase. These lateral
extensions usually contain one or two layers of mitochon-
dria (Fig. 1.20). The mitochondria are long, narrow rods that
branch and double back on themselves. They are oriented cell surface.163 The lateral extensions also establish a com-
perpendicular to the basement membrane and lie adjacent to plex and extensive surface labyrinth of lateral intercellular
the cell membrane, to which they presumably supply energy spaces (“basal labyrinth”).
for transport processes.
The lateral extensions and their mitochondria cause Cell Junctions
the pattern of basal striations that are typical for numerous The proximal convoluted tubular cells have an apical junc-
transporting cells. The lateral processes of proximal convo- tional complex that consists of a shallow, beltlike, tight
luted tubule cells in humans are not as elaborate as those junction next to the tubular lumen; a deeper, beltlike in-
seen in rats (compare Figs. 1.15 and 1.16). In rabbits, the termediate junction (the zonula adherens); and only small
lateral cell surface is increased 20 times over that of the basal and infrequently seen desmosomes. The proximal tubular
tight junctions are shallow and consist of only one or two
lines of fusion of the outer lea et of the cell membrane
(Fig. 1.22). Freeze-fracture studies of normal proximal tight
junctions, however, reveal focal discontinuities. During vol-
ume expansion, striking increases in the length of disconti-
nuities were found.164 Transmission micrographs also show
multiple areas of nonfusion of tight junctions associated
with renal venous constriction and increased ureteral pres-
sure.165 These sites of nonfusion may explain the increased
permeability seen in these situations. Proximal tubular cells
are electrically coupled by gap junctions (Fig. 1.22).166

Microvilli
A layer of slender (approximately 80 to 90 m), nger-
shaped processes extends into the tubular lumen, forming
the microvillus, or brush border. The length and number of
processes vary with the segment and species. The luminal
surface of the pars convoluta is increased 40 times in rats.157
The luminal surface is increased 36 times in the pars con-
FIGURE 1.19 Scanning electron micrograph of a rat proximal voluta and 15 times in the pars recta in rabbits.163 Each mi-
tubule epithelial cell showing the apical brush border (BB) and crovillus has a core of micro laments that extend into the
the lateral processes (LP). (Magni cation 14,700.) apical cytoplasm connecting them to the cytoskeleton.
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 117
7

FIGURE 1.21 Diagram of a proximal convoluted tubular cell showing the elaborate shape of these cells. Some interdigitating pro-
cesses extend the full height of the cells. The apical and basal cytoplasmic regions also have smaller, more elaborate interdigitating
processes. (From Bulger RE. The shape of rat kidney tubular cells. Am J Anal. 1965;116:237, with permission.)

Cytoplasm
The cells contain a very prominent vacuolar apparatus (see
later text), a Golgi apparatus, free ribosomes, and cisternae of
rough and smooth surface endoplasmic reticulum. The latter
forms specialized cisternae, called the perimembranous cis-
ternal system, near the lateral cell membranes.80 In addition,
the cells contain many peroxisomes that frequently have
eyecatching shapes.167 Peroxisomes are limited by a single
membrane and contain a dense matrix, frequently exhibit-
ing crystalloid nucleoids (Fig. 1.17); platelike inclusions
at the peroxisomal edge, which are called marginal plates,
are also frequently encountered. Peroxisomes are invariably
wrapped by elements of the smooth-surfaced endoplasmic
reticulum. In the kidney, large peroxisomes are exclusively
found in the proximal tubule, most frequently in the P3 seg-
ments167—all other nephron portions contain only micrope-
roxisomes. Peroxisomes contain enzymes from a primitive
respiratory chain in which oxidases produce hydrogen per-
oxide, which is, in turn, destroyed by the catalase, hence the
name peroxisome.168 Peroxisomes participate in the break-
down of very-long-chain fatty acids by lipid -oxidation;
in addition, they may play a protective role by destroying
hydrogen peroxide produced by free radicals.

Straight Part of the Proximal Tubule


(Last Part of P2 and All of P3 )
FIGURE 1.22 Freeze-fracture electron micrograph of the apical The straight part of the proximal tubule (pars recta) begins
part of proximal tubule cells (rabbit) showing the shallow tight in the medullary ray and penetrates into the outer stripe
junction (arrows) and a gap junction (arrowhead). (Magni cation of the outer zone of the medulla (Fig. 1.18). The proximal
37,000.) (From Kriz W, Kaissling B, Schiller A, et al. Morpholo- pars recta converts into the thin limb near the junction of
gische merkmale transportie-render epithelien. Klin Wochenschr. the inner and outer stripe. In rats, the pars recta includes
1979;57:967, with permission.) the nal region of both P2 and P3,169 and the transition
18
18 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

between the two segments occurs at various levels in the Megalin is a 600-kDa transmembrane protein be-
medullary ray. This region is marked by a sudden increase longing to the LDL-receptor family (see reviews).176,177
in microvillar length, a decrease in endocytic apparatus, The extracellular domain contains four clusters of cys-
and a decrease in interdigitation between adjacent cells. teine-rich, complement-type repeats, constituting the
The microvilli of the pars recta cells decrease in height and ligand-binding regions. Cubulin is a 416-kDa periph-
number in rabbits158 and humans162 but remain high in eral membrane protein identical to the intrinsic factor
rats (Fig. 1.18). vitamin B12 receptor, known from the small intestine.
In general, the P3 pars recta cells have been described It contains 27 CUB domains, which are responsible for
as lower in height with a less elaborate shape. In humans, the ability to interact with a great variety of ligands.
the pars recta cells have a convex apical surface, and some 2. Small apical vesicals pinch off from the tubular invagi-
lipid droplets are found in the basal cytoplasm. The mito- nation at the intermicrovillar areas to ferry the protein
chondria are fewer and no longer closely applied to the cell to the next component.
membrane. The lysosomes are smaller and the Golgi and en- 3. Large apical vacuoles located in the apical cytoplasm
docytic apparatuses are less well developed. Peroxisomes are are formed by fusion of small vesicles representing the
more numerous in the pars recta.80,162 The tight junctions early endocytotic compartment.
of P3 can be more complex in shape, consisting of several
4. Condensing vacuoles form in which the protein is
junctional strands in rats, dogs, and cats.170
condensed. These vacuoles move basally in the cell
and acquire hydrolytic enzymes by fusion either with
Structure–Function Correlations primary or secondary lysosomes. The proteins seques-
The functional relevance of the proximal tubule is manifold tered within lysosomes appear to be broken down into
and quantitatively enormous. It reabsorbs about 70% of amino acids that are reused by the cell.
ltered Na , Cl , and water; 95% of bicarbonate; 60% of 5. Already sequestered in the early endosomal compart-
Ca2 ; and sodium phosphate. Reabsorption of the ltered ment, the receptors are concentrated in dense, api-
glucose is complete, whereas reabsorption of amino acids is cal tubules by which they are returned to the apical
almost complete. This occurs either by transcellular trans- plasma membrane.
port via speci c channels and transporters in both mem-
branes (water, sodium, phosphate, glucose, amino acids, This process of megalin-mediated internalization ap-
and bicarbonate in a speci c enzyme-mediated mode) or pears to be of even much wider relevance. As mentioned
predominantly by paracellular transport through the leaky previously, the proximal tubule reabsorbs phosphate, repre-
tight junctions (Ca2 , Cl ). Furthermore, ltered macro- senting a key element in phosphate homeostasis. Depending
molecules undergo reuptake by a prominently developed on the amount of sodium phosphate cotransporters pres-
endocytotic apparatus. In addition, the proximal tubule ent in the brush border cell membrane, the reabsorption
(predominantly S3 segments) secretes organic cations and varies. Internalization of the phosphate transporter type II
anions, which constitute an extraordinarily diverse array of (e.g., PTH induced to downregulate reabsorption) occurs via
compounds of physiologic, pharmacologic, and toxicologic megalin-mediated endocytosis.178,179 The proximal tubule
importance. The transepithelial transport involves separate has also endocrine relevance. In response to stimulation by
entry steps at the basolateral membrane and exit steps at PTH, it produces, by adding a second hydroxyl group, the
the luminal membrane with speci c transporters at both active vitamin D3, calcitriol.
sites.171–174 Other potentially toxic xenobiotic compounds
are metabolized within the well-developed smooth endo- Thin Limbs of the Loop of Henle
plasmic reticulum of S3 segments.175 (Intermediate Tubule)
Unique to the proximal tubule is the reabsorption and
Thin limbs can be short, occurring only along the descend-
degradation of ltered protein and peptides (Figs. 1.15 and
ing limb, or they can be long, reaching varying distances
1.16). As discussed previously, all proteins smaller than al-
into the inner medulla. In the long-looped variety, thin limb
bumin leak through the glomerular lter; even albumin,
segments compose part of both the descending and ascend-
in small amounts, is always contained in the ltrate. All
ing limbs. Four types of thin limb segments are routinely
these diverse macromolecules are taken up by proximal tu-
identi ed: (1) descending thin limbs of short-looped neph-
bule cells via receptor-mediated endocytosis and degraded
rons (SDTL) (Figs. 1.23 to 1.25), (2) upper portions of de-
in lysosomes. In proteinuric states, this function is greatly
scending thin limbs of long-looped nephrons (LDTL up),
enhanced. The uptake and digestion include the following
(3) lower portions of descending thin limbs of long-looped
steps.176,177
nephrons (LDTL lp), and (4) ascending thin limbs of long-
1. Binding of the ltered protein to two multiligand looped nephrons (ATL) (Figs. 1.24 and 1.25). This pattern
receptors, megalin and cubulin, which are densely has been observed in rats,180,181 mice,182 syric hamsters,183
accumulated within clathrin-coated pits in the the desert rodent Psammomys obesus,184,185 and the rabbit.158
intermicrovillar areas of the apical membrane. Thin limbs have not been studied as carefully in humans as
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 119
9

pieces of functionally different cells are encountered within


an otherwise homogenous thin limb segment.189,190 Beyond
all of these heterogeneities, there are prominent differences
among species. This situation may explain the doggedly per-
sistent discussion about the integrated function of thin limbs
in the urine-concentrating process; a generally accepted con-
cept of how the nal concentration of the urine in the inner
medulla occurs is lacking.
The type-1 epithelium lining (SDTL) is composed of
at, noninterdigitating cells, joined by tight junctions that
consist of several anastomosing strands. Cell organelles
are exceedingly sparse. Functionally, this segment contains
aquaporin 1 (AQP1) and the urea transporter UT-A2 in its
FIGURE 1.23 Diagrammatic representation of thin limb struc- membranes.191–194 Thus, it is water and urea permeable.
ture in short loops of Henle showing the shapes of constituent However, these properties are unequally distributed; within
cells and the morphology of the occluding junctions (inset). the proximal part, the water permeability is high, whereas,
(From Schwartz MM, Venkatachalam MA. Structural differences within the distal part, the urea permeability is high.191,193,195
in thin limbs of Henle: physiologic implications. Kidney Int. In species with complex vascular bundles (rat, mouse, etc.),
1974;6:193, with permission.) the SDTLs lie within vascular bundles187; in these surround-
ings, the thin limbs are in an ideal position to recycle urea
from the ascending vasa recta into the short-loop nephrons.
in several other animal species.13,186,187 An additional sub- The descending limbs of long loops (LDTL) are generally
segment of thin limbs has been identi ed in chinchillas.188 much larger in diameter and have a thicker epithelium than
Surprisingly, as seen by light microscopy, these simple those of short loops (Fig. 1.25). Moreover, these thin limbs
looking epithelia are strikingly different from each other— segments are heterogeneous; those of the longest long loops
not only the ascending from the descending limbs but, most begin in the inner stripe as a much thicker tubule than those of
remarkably, the descending limbs of short from those of long shorter long loops. The character of the epithelium gradually
loops. Furthermore, within the descending segments, the changes as the limbs descend toward and into the inner me-
proximal portion, although structurally not different from dulla. The subdivision of the long descending thin limbs into
the distal portion, express a different pattern of transport- an upper part (type-2 epithelium) and a lower part (type-3 epi-
ers than the distal portion. Even islets or interposed limb thelium) is an approximation and re ects the gradual change

FIGURE 1.24 Diagrammatic representation of thin


limb structure in long loops of Henle, showing the
shapes of constituent cells and the morphology of the
occluding junctions (inset). (From Schwartz MM, Ven-
katachalam MA. Structural differences in thin limbs of
Henle: physiologic implications. Kidney Int. 1974;6:193,
with permission.)
20
20 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 1.25 Low power electron


micrograph of a cross-section through
the inner stripe (rabbit). A vascular
bundle with arterial (A) and venous
(V) vasa recta is surrounded by de-
scending thin limbs (DL; the small
pro les belong to short loops and the
large pro les to long loops), and thick
ascending limbs (AL) and collecting
ducts (CD); note the dense pattern of
capillaries between the tubules. (Mag-
ni cation 900.)

between the two epithelia. Moreover, this process of epithelial epithelium is much more simply organized. The prominent
transformation appears to be related to the length of each loop. paracellular pathway is lacking; the cells do not interdigitate
It occurs earlier and more quickly in short long loops and is and are joined by deep tight junctions. In other respects,
delayed in the longest long loops.158,159,181,182,187,189,196 however, the epithelia are similar in the two groups: numer-
Furthermore, considerable interspecies differences, ous luminal microvilli, many mitochondria, and a dense
particularly prominent in type 2 epithelia, complicate the assembly of intramembrane particles in luminal and basolat-
situation. Two patterns of type-2 epithelium may be distin- eral membranes are present in both groups. The high density
guished.187,197 In one group of species (mouse, rat, Psamomys, of intramembrane particles may, at least, be partially due to
etc.),159,184,185,198,199 the type-2 epithelium is characterized the high density of AQP1 channels in both membranes—
by an extremely high degree of cellular interdigitation and corresponding to the decrease of particle density along its
a shallow tight junction consisting of only one junctional descending course. The density of AQP1 channels also de-
strand pointing to prominent paracellular transports. In creases and nally terminates completely.195,204,205 Because
addition, the epithelium has numerous apical microvilli, structurally a clear cut border between the upper and the
considerable numbers of mitochondria, and exhibits a high lower segment of the LDTL cannot be de ned, it may be
expression of Na -K -ATPase.200,201 In a second group of reasonable to discriminate both segments by the absence of
species (rabbit, minipig, and, possibly, man),202,203 the type-2 AQP1 in the lower segments.189,195,204
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 221
1

Type-3 epithelium (found in LDTL lp) is comparably sim- cells is found in the wall of the CTAL in this region; this
ple; interspecies differences are no longer prominent. The epi- plaque of cells forms the macula densa ( JGA). In humans,
thelium consists of at, noninterdigitating cells, joined by tight the cells of the MTAL are not as elaborately shaped as those
junctions of intermediate apico-basal depth181; it lacks AQP1 described in the majority of laboratory animals that have
and may, accordingly, have a very low water permeability. Re- been studied.212
garding the permeability to urea and the distribution of the After a short post macula densa region of the CTAL,
urea transporter, UT-A, con icting data are published, especial- the distal tubule becomes more convoluted and it is now
ly when comparing data from different species.190,191,204,206–208 lined by epithelial cells that increase in height from the
The ascending thin limb (ATL) is present only in long- CTAL, which forms the distal convoluted tubule (DCT)
loop nephrons and is uniformly organized among mammals. (see Fig. 1.27).214,216 In rabbits, the DCT contains one type
Generally the transition from the type-3 epithelium of the of cell known as the distal convoluted tubular cell.158 In
DTL to the type-4 epithelium of the ATL occurs in a short, this species, the DCT is abruptly replaced by the con-
but fairly constant, distance before the bend (“prebend seg- necting tubule (CNT). In other species, such as rats and
ment”).180,185,189 Therefore, functionally, the entire bend should mice,217 humans,218 or minipigs,203 the transition is more
be regarded as part of the ATL. The type-4 epithelium is char- gradual with intermixing of cells so that in the late DCT of
acterized by very at but heavily interdigitating cells joined by these species connecting tubule cells and intercalated (IC)
shallow tight junctions, consisting of only one, but prominent, cells begin to dominate, although DCT cells and principal
junctional strand. This leaky organization of the paracellular cells of the collecting duct also can be identi ed. This seg-
pathways correspond with functional studies,209,210 which all ment is then de ned as the CNT. The CNT of super cial
demonstrate that the ATLs are highly permeable for ions. nephrons begin as unbranched segments, each emptying
The change from the type-3 epithelium to the type-4 into a collecting duct (some investigators call the rst part
epithelium coincides with the full disappearance of the urea of the collecting duct the initial collecting duct). In mid-
transporter UT-A and the abrupt beginning of the expression cortical and juxtamedullary nephrons, the change from
of the chloride channel ClC-K1189,204; aquaporins are also the DCT to the CNT occurs a few cells before the fusion
lacking. Thus, the ATL is water and urea impermeable, but of the two tubules.158 These CNT segments join to form
highly permeable for Cl and also Na . branching connecting tubule segments called arcades,
In humans,211,212 dogs,161 and minipigs,203 a gradual transi- which arch upward in the cortex and then empty into
tion is seen from the thin limb to the ascending thick limb; how- a CCD.7 The number of each type of CNT architecture
ever, an abrupt transition is seen in most other species.158,213,214 varies with the species.

Medullary Thick Ascending Limb


THE DISTAL TUBULE OVERVIEW The MTAL is lined by cells of one type that has extensive
The distal tubule of mammalian kidneys has been de ned basal interdigitating processes lling the basal three-fourths
in many and con icting ways. For morphologists, the distal of the cytoplasm (Fig. 1.26). The larger cell processes con-
tubule is divided into several serial segments with differ- tain large mitochondria that are elongated on an axis per-
ing locations in the kidney and of varying ultrastructural pendicular to the tubular basement membrane. In addition
patterns215 (Fig. 1.2). The rst morphologic segment of to their normal contents, the mitochondria contain promi-
the distal tubule for long-looped nephrons begins at the nent intramitochondrial granules and occasional lamentous
boundary between the inner zone of the medulla and the bodies.219 The epithelium lining the MTAL is approximately
inner stripe of the outer zone of the medulla. At this bound- 7 m in height.213,220
ary, the cells lining the ascending thin limbs of long-looped The apical surface of the MTAL cells does not have an
nephrons increase in height, forming the MTAL. The MTALs elaborate shape in rabbits158; it is more elaborate in rats.221
of short-looped nephrons are encompassed within the in- Tisher and associates,213,216,220 using scanning electron mi-
ner stripe of the outer zone in which the descending thin croscopy, have described two surface con gurations of the
limb of short-looped nephrons converts into a descending MTAL cells, with variations in apical cell outlines and in the
thick limb, which then makes a hairpin turn and ascends number of apical microplications. Some cells have smooth
as an MTAL. The MTAL traverses the inner stripe of the apical surfaces with a few microprojections, whereas others
outer zone of the medulla and continues toward the cortex have a rough surface with numerous microprojections. The
through the outer stripe of the outer zone of the medulla. MTAL cells have both extensive invaginations of the baso-
The MTAL then enters the cortex, ascending within the lateral plasma membrane, as well as a large lateral interdigi-
medullary ray as the CTAL. These two segments, the MTAL tating process of adjacent cells.220 Because a main function
and CTAL, are also called the straight part of the distal tu- of the TAL is the reabsorption of sodium and chloride from
bule. The CTAL then leaves the medullary ray and enters the tubular lumen to the interstitium, these basolateral pro-
the pars convoluta of the cortex, running between the affer- cesses have Na -K -ATPase activity.222 In addition, they
ent and efferent arteriole of the renal corpuscle from which have an apical bumetanide-sensitive sodium-potassium-2
that tubule was derived. A plaque of taller, but narrower, chloride cotransporter called NKCC2 located in the apical
22
22 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 1.26 Transmission


electron micrograph of the pars
recta of the distal tubule from a
rat showing the interdigitating
cellular processes. (Magni cation
15,000.)

membrane (see the discussion of the membrane ampli - or basal cell surface.228 These interdigitating processes are
cation principle later in this chapter). The MTAL demon- more prominent in a circumferential direction.228 Mitochon-
strates a low permeability for water so the active transport dria are still prominent in the basolateral cell processes, but
of the sodium ions out of the basolateral membranes of the somewhat smaller than those of the MTAL. The apical cell
cells in exchange for potassium ions participates in forming border becomes more tortuous in this segment, having more
the hypertonicity of the medullary interstitium. The api- prominent invaginations of the entire lateral cell margins,
cal cytoplasm contains a variable number of vesicles and including the apical region of the cell,158 and has more mi-
a prominent Golgi apparatus. Cisternae of rough-surfaced crovilli along its surface.213 Tamm–Horsfall glycoprotein has
endoplasmic reticulum can be seen throughout the cyto- been identi ed covering the plasma membrane in the TAL
plasm. The tight junction is of low to intermediate apical– of rat.229,230
basal depth, consisting of several parallel strands.223 These The MTAL and CTAL differ from each other physiologi-
tight junctions appear to be related to the relative imper- cally, especially in hormone responsiveness.231,232 The MTAL
meability to water (see discussion later in this chapter of has a high density of Na -K -ATPase.233 Hebert et al.222 have
tight junction structure and their role in transepithelial shown in mice that antidiuretic hormone (ADH) increases
solute and water transport). As the MTAL traverses the the transepithelial voltage and net chloride reabsorption in
outer stripe in rats, it decreases in cell height, but retains the medullary, but not the cortical, region of the ascending
its prominent lateral interdigitations. In rabbits, the apical thick segment. The ascending thick region functions in ow-
surface becomes more elaborate in this region.158 dependent absorption of NaCl mediated by the furosemide
The administration of vasopressin (antidiuretic hor- (bumetanide)-sensitive cotransport NKCC2.234,235 Nielsen
mone) to rats that have hereditary diabetes insipidus causes et al.236 and Obermuller et al.,237 using immunohistochemistry,
hypertrophy of the MTAL in Brattleboro rats.224 This effect have demonstrated bumetanide-sensitive Na-K-2Cl labeling in
can also be shown by water restriction in normal rats225 and the apical plasma membrane and the subapical intracellular
after high protein intake.226,227 vesicles of MTAL and CTAL in the rat and rabbit. The apical
plasma membrane of the macula densa region also had distinct
Cortical Thick Ascending Limb labeling consistent with a role in tubuloglomerular feedback.
The cells of the CTAL are lower in height than those of the The TAL in the inner stripe has a greater reabsorptive
MTAL in the rat, measuring about 5 m in height221 and capacity for NaCl than the cortical segment,238 but the corti-
2 m in the rabbit.2 The cells still have prominent basolat- cal segment can maintain a higher concentration gradient239
eral interdigitating cell projections, which increase the baso- (see the discussion of Na -K -ATPase and the basolateral
lateral cell membrane approximately tenfold over the apical membrane area later in this chapter). The sodium chloride
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 223
3

reabsorbed by the TAL contributes to the hypertonicity of the the CCD.241 In humans, small lipid droplets are seen in the
medullary interstitium. As the CTAL ascends toward the renal cytoplasm.212 The endocytic apparatus is not well developed,
corpuscle from which it was derived, there is an increase in but some vacuoles and numerous small vesicles are seen in
apical plasma membrane surface area, both by an increase in the apical region. Dorup 241 describes four types of vesicles
apical microvilli and by an increase in lateral cell margins.221 in these cells: intermediate vesicles (80 to 200 m), which
The macula densa region is formed by a plaque of cells are generally uncoated; small vesicles with a mean diame-
in the wall of the CTAL (see Fig. 1.35), where the ascend- ter of 50 m that are continuous with these intermediate
ing thick tubule runs between the afferent and efferent arteri- vesicles; large vesicles ( 200 m); and tubular pro les (these
oles of the renal corpuscle from which the tubule is derived. being less frequent in DCT cells). Some rough-surfaced en-
A group of extraglomerular mesangial cells (lacis cells, Polkis- doplasmic reticulum and lysosomes are also present in this
sen cells) lls the cone-shaped area formed by those three segment. The cell nuclei occupy an apical position, because
structures.240 These four elements—the afferent and efferent the basal two thirds of the cytoplasm is lled with extensive
arterioles, the extraglomerular mesangium, and the macula lateral interdigitating processes surrounded by basolateral
densa—constitute the juxtaglomerular apparatus, which will cell membranes. These basolateral processes are lled with
be discussed separately in more detail later in this chapter. large elongated mitochondria that have their longitudinal
axis perpendicular to the basement membrane. The presence
Distal Convoluted Tubule of the large mitochondria lying adjacent to the basolateral
The DCT (distal pars convoluta) is shorter than the proxi- cell membranes is consistent with the need for ATP for the
mal convolution, being about 1.2 mm in length in the rat.217 continued active reabsorption of solute that occurs in this tu-
Therefore, fewer pro les are seen in sections of the renal cor- bule (see Fig. 1.27). The volume of mitochondria in the DCT
tex. The DCT begins with a rather marked increase in the cells is larger than in the CNT cells or cortical collecting duct
height of the lining cells from those of the TAL, although the (CCT) cells.241 An increased delivery of sodium to the DCT
cells are similar to those lining the TAL. In the rabbit, DCT brings about an increase in the volume of the cell, in the mi-
cells are three to four times taller in the DCT than the cells tochondrial volume, and in the proliferation of the basolat-
lining the CTAL.214 The DCT tubule has a variable diameter eral membranes in the cells of the DCT, as well as in the CNT
and contains one type of cell with more nuclear pro les than cells and the principal cells of the collecting ducts.242–244
are seen in the proximal convoluted tubule (Fig. 1.27). The The apical membrane of the DCT cells has numerous
DCT extends from the CTAL to the CNT. The cells lining the small microprojections. The tight junctions between the
DCT have short, bulbous luminal microvilli, but no regu- cells of the distal tubule are elaborate and are composed of
lar brush border. The microvilli are more numerous than multiple lines of membrane fusion,245 a characteristic that
seen on the TAL cells, the CNT cells, or the principal cells of correlates with the ability of the cells to maintain a large

FIGURE 1.27 Transmission


electron micrograph from a rat
distal convoluted tubule showing
the numerous mitochondria
within interdigitating processes.
(Magni cation 2, 850.)
24
24 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

electrochemical gradient. The DCT reabsorbs NaCl against a


steep chemical gradient and, as expected for such a sodium-
absorbing epithelium, contains abundant Na -K -ATPase
on its basal lateral cell membrane.233,246,247 DCT prolifera-
tion and basolateral membrane ampli cation occur when
active sodium reabsorption increases.248,249 Kaissling et al.250
increased the NaCl load to this segment by administering
furosemide and demonstrated a marked adaptive increase in
the basolateral membrane in the DCT.
In rabbits, an abrupt transition is seen from the dis-
tal tubule to the connecting piece of the nephrons.158 The
situation is less clear in other species, in which the transi-
tion does not appear to be either as abrupt or as completely
studied. Recent studies by Biner et al.251 used immunohis-
tochemistry to localize the various transport systems along
the human cortical distal nephron. The DCT demonstrates
luminal thiazide-sensitive sodium-chloride cotransporter
(NCC). The NCC overlaps with epithelial sodium chloride
channels (ENaC) for a short region at the end of the DCT. IC
cells were interspersed among the DCT cells near the end of
the DCT. (For more detailed information, see recent reviews
from the groups of Kaissling251,252 and Bachmann.253)

The Connecting Tubule


The CNT forms the next region of the distal nephron and
lies between the DCT and the collecting duct system. At the
present time, the CNT may be best classi ed as part of the
DCT because it appears to be derived from the metanephric
blastema.4 Peter7 believed that the arcades arise from the
FIGURE 1.28 Light micrograph of the renal cortex (rabbit)
ureteric bud, whereas Oliver,5 Potter,254 and, more recently,
showing a connecting tubule composed of connecting tubule
Neiss,4 all believe that the metanephric blastema is the cor-
cells (1) and intercalated cells (2). (Magni cation 900.) (From
rect source. However, Howie et al.,255 using immunohisto-
Kaissling B, Zurich, with permission.)
logic methods with substances related to the ABO blood
groups, various cytokines, and Tamm–Horsfall protein,
found that the ureteric bud and the connecting piece ex-
press the same antigens. The morphology of the CNT cells (presumed to be type B intercalated cells), were observed
appears to have features of both the DCT cells, such as some seemingly being developed from two embryologically dis-
degree of lateral interdigitations that contain mitochondria, tinct parts of the kidney. Some of these IC cells in certain
but also has features of the CCD cells, such as the presence locations subsequently disappeared.256
of more true basal infoldings (Fig. 1.28). Because of the in- The CNT of super cial nephrons are short and drain
termixing of DCT cells, CNT cells, IC cells, and principal individually into the collecting duct (some classify the point
cells of the CCD in some species, such as rats and mice, of junction as the initial collecting duct). CNTs of juxtamed-
there is a gradual transition from the DCT to the CNT in ullary and some midcortical nephrons generally form arched
these species.213,217 Neiss4 demonstrated that IC cells (also collecting ducts in many species. The arched duct starts
called dark cells) in the CNT arose from the metanephric deep within the cortex with the conversion and con uence
blastema, whereas the IC cells found in the collecting duct of several DCT into the arcade and then ascends, while col-
arose from the ureteric bud. Kim et al.,256 using speci c an- lecting parts of other nephrons, before the duct turns and
tibodies to carbonic anhydrase II, H -ATPase, and band- enters a medullary ray. This arched duct results from an ear-
3 protein, demonstrated that IC appeared simultaneously ly embryonic type of nephron induction.4,7 The number of
in both the CNT and the medullary collecting duct. These nephrons that empty directly into the CCD, compared with
IC cells differentiated from separate foci: one in the neph- the number that enter rst into an arched tubule, varies with
ron (developing from the metanephric blastema) and one the species.5 Both types occur in humans.
in the collecting duct (developing from the ureteric bud). Two types of cells generally line the CNT, although in-
When rst identi ed, cells with distinct apical staining for termixing of these two cell types with cells of other cell
H -ATPase (presumed to be type A intercalated cells), as types, such as the DCT cells and the principal cells of the
well as cells with distinct basolateral H -ATPase labeling CD, are seen in some species. The two main types of cells
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 225
5

seen are CNT and IC cells (Fig. 1.28). The CNT cells appear tubule in that region. Electrophysiologic studies in rabbits
to be characterized mainly by extensive true infoldings of suggest that approximately 98% of the IC cells in the con-
the basal cell membrane, which can extend quite deeply necting tubule are the HCO3 -secreting B type.262
into the cytoplasm. However, basolateral interdigitations
have also been described, but are less pronounced than
seen in DCT cells.241 In rabbits, the plicated membranes THE COLLECTING DUCTS
can reach the apical cytoplasm.158 Stanton et al.244 demon- The collecting ducts extend from the CNT through the med-
strated a striking increase in basolateral membranes in the ullary rays as CCDs. They then cross the outer and inner
rat CNT and the initial CCD of potassium-adapted animals, stripe of the outer medulla as well as the inner medulla, to
indicating that potassium is secreted by the CNT cells, as empty their contents at the tip of the renal papillae. They
well as the principal cells of the (initial) collecting duct. In include the CCDs (including the initial collecting ducts), the
this study, no changes were seen in the IC cells. This sug- outer medullary collecting ducts (OMCDs), and the inner
gests that potassium is secreted by the CNT cell and the medullary collecting ducts (IMCDs).
principal cell of the initial collecting duct.244,257 The collecting ducts are the nal regulators of uid and
The basolateral membranes of CNT cells are partially electrolyte balance, playing roles in the handling of Na , Cl ,
separated by mitochondria, which are smaller, more ran- K , and acid and base. Although there has been great em-
domly distributed, and less numerous than those found in phasis on the role of the collecting ducts as the main control-
the DCT. The volume density of mitochondria in rat CNT lers of urinary sodium and potassium excretion, Meneton et
cells was signi cantly lower than in the DCT cells.241 This al.263 stress the pivotal role played by both the late part of the
arrangement differs from principal cells of the collecting DCT and the CNT, especially in situations that prevail in our
duct, in which the mitochondria are found mainly above current environment, in which the dietary sodium intake is
the basal infoldings, not among the basolateral membranes. high and the potassium intake is low. They propose that a
Microvilli on the apical surface tend to be slender and in- large proportion of the aldosterone-regulated sodium reab-
frequent. Apical vesicles were about as frequent as seen in sorption and potassium secretion occurs before the tubular
the DCT cells.241 Mitochondria, the nucleus, and other cell uid reaches the collecting duct. The difference between the
organelles ll the apical cytoplasm. The CNT cells appear function of the late DCT and the CNT compared to the col-
to exist in most species, including rats, mice,212,217 and lecting duct seems to be more quantitative than qualitative,
humans.212 with a large proportion of the aldosterone-regulated sodium
Biner et al.251 demonstrated an ENaC along the entire reabsorption and potassium secretion being done in the late
CNT in humans. The major part of the CNT also coexpresses DCT and CNT. The collecting duct would function mainly
aquaporin 2 with the ENaC. IC cells were identi ed inter- when the requirement for sodium and water conservation is
spersed among the CNT cells in the human. Lof ng and maximal and the upstream segments are overloaded by diet
Kaissling258 reviewed the transport pathways along several or some genetic defect.
mammalian distal nephrons and showed that ENaC was The collecting ducts are lined by two types of cells: col-
present along the CNT from the rabbit, rat, mouse, and hu- lecting duct principal (light) cells and intercalated (dark)
man. Frindt et al.,259 using patch clamp techniques in rat cells (Figs. 1.29 and 1.30). About 30% of CCD cells are IC
kidney tubule segments, demonstrated that the CNT could cells in rat.264 Kaissling and Kriz158 estimate that rabbits have
reabsorb sodium at a rate 10 times higher than that of the 33% IC cells in the CCD and 50% in the outer medullary
CCT. Using immunogold labeling and electron microscopy, collecting duct. About one-third of the cells lining the outer
aquaporin 2 (apical), 3, and 4 (basolateral) were all shown medullary collecting duct in the rat are IC cells.265 The num-
to be colocalized in CNT cells of rat.260 ber of IC cells decreases as the collecting duct descends into
The CNT cell displays an ampli cation of basal cell the medulla and are absent below the rst portion of the
membrane in rats244 and rabbits249 in situations in which IMCD. The principal cells of the collecting duct gradually
there is a low Na and high K intake. This effect is axial change in morphology as they descend toward the papilla.
along the tubule, being greatest at the early segment of the They increase in cell height and have more complex tight
CNT and decreasing along its length.249 The axial change junctions, whereas the amount of basal infoldings and the
in structure is paralleled by similar changes in Na -K - number of mitochondria decrease. Fusions of the collect-
ATPase.261 ing ducts occur in the inner renal medulla to form the large
The second cell type is the IC cells (or dark cells). Three papillary collecting ducts (ducts of Bellini). These large col-
types of IC cells have been described: type A involved with lecting ducts exit at the papillary tip in an area known as the
the secretion of protons into the lumen; type B involved area cribrosa.
with the secretion of bicarbonate into the tubular lumen; The principal cells of the collecting duct in the cor-
and non-A–non-B, which may be able to secrete both pro- tex are cuboidal in humans218 and low cuboidal in rats
tons and bicarbonate into the tubular lumen. IC cells will (see Fig. 1.29). They form the most numerous cell types.
be discussed under the collecting ducts, because these cells They have a simple cell shape, with fairly straight lateral
comprise such an important number of the cells lining the cell borders that have small interlocking projections. Their
26
26 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

they do not have mitochondria lying between them. The


mitochondria are located mainly above the infoldings and
in the apical cytoplasm, which contained few intermediate
and small vesicles, tubular pro les, and large vesicles.241 The
tight junctions are deep,223,266 and the apical surface has a
prominent glycocalyx.267
Conditions that increase potassium secretion, such as
potassium adaptation or high endogenous or exogenous
mineralocorticoid levels, bring about dramatic increases in
these basal cell membrane infoldings.249,262,268,269 For exam-
ple, striking increases in the basolateral membranes of the
principal cells of the initial segment of the collecting duct
have been demonstrated in potassium-adapted animals.244 In
addition, the principal cells in rats showed a 35% decrease in
the basolateral membranes in adrenalectomized animals that
was restored to control levels by the administration of physi-
ologic amounts of aldosterone, but not of glucocorticoids.270
In addition, increasing the dose of aldosterone over control
levels caused an increase in the basolateral membranes by
111% compared with controls. They did not note changes
in the luminal membranes of the principal cells.270 These

FIGURE 1.29 Transmission electron micrograph of the epithelium


of a cortical collecting duct (rat) showing a collecting duct cell
(principal cell) (above) and an intercalated cell (A-type) (below).
Note the basal infoldings in the collecting duct cell and the apical
vesicles in the intercalated cell. (Magni cation 5,000.)

pale-staining cytoplasm contains a few small, oval, ran-


domly oriented mitochondria and other organelles, and the
nucleus is situated in the middle to upper one-half of the
cell in the cortex. The luminal surface is generally smooth
with a few short microvilli and a single cilium. The basal
surface is characterized by true basal infoldings, with few lat- FIGURE 1.30 Scanning electron micrograph of a cortical col-
eral interdigitations. The ampli cation factor of basolateral lecting duct (rat) showing the apical aspect of collecting duct
membranes was signi cantly lower than in the CNT cells.241 cells (with an apical cilium) and intercalated cells (with apical
Because these infoldings are short and closely spaced, microfolds). (Magni cation 2,400.)
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 227
7

studies provide evidence that principal cells in the collecting The collecting duct cells undergo gradual, although
duct are involved with potassium secretion. considerable, changes from the cortex downstream to the
The collecting duct responds to ADH by an increase in upper one-third of the inner medulla.158,218 The cells of the
water permeability. The principal cells are vasopressin sensi- outer medulla include principal cells similar to those seen in
tive and show dramatic increases in water permeability of the cortex; however, the cells become taller with decreasing
the apical membrane. Sun et al.271 have demonstrated direct concentrations of several cellular organelles and basal infold-
evidence that AQP2 on principal cells is located in clathrin- ings. IC cells similar to type A are found in the outer medul-
coated pits that recycle between the plasma membrane and la. From the deeper cortical levels downward into the inner
intracellular vesicles in response to the availability of ADH. medullary region, the basal labyrinth of the principal cells
The mechanisms underlying these changes are discussed in continues to decrease gradually, with a steeper reduction
detail later in this chapter. within the outer stripe.158,215,218 The number of mitochon-
Biner et al.,251 using immunohistochemical localization dria also continues to decrease, whereas lysosomal elements
techniques on human kidney, demonstrated the presence of and apical-coated vesicles seem to increase. The density of
an amiloride-sensitive ENaC and AQP2 activity on collect- the cytoskeletal network lying under the apical cell mem-
ing duct principal cells. Lof ng et al.272 using immunohisto- brane becomes more prominent and the tight junctional belt
chemistry in rabbit kidney cortex, demonstrated that ENaC becomes deeper.158
is found in the CNT cells and the CCD cells. The ENaC From the second one-third of the inner medulla, col-
shifted from the apical membrane in the upstream CNT cells lecting duct cell size increases steeply. These tall IMCD cells
to a cytoplasmic location downstream in the CNT and CCD are distinct from collecting duct cells upstream according to
cells. In the rabbit, the AQP2 was seen only on the CCD several criteria213,220,274 (Fig. 1.31). Their luminal membrane
cells. The apical membranes of the collecting duct princi- is covered by numerous stubby microvilli and lacks the cen-
pal cells of humans contain ENaC and AQP2, whereas the tral cilium. The lateral intercellular spaces are more exten-
basal membranes contain Na -K -ATPase and aquaporins sively developed and are prominent by their dense assembly
3 (AQP3) and 4 (AQP4); hence, the CCD plays a critical role of microvilli and microfolds projecting from the lateral cell
in the concentration of urine. The CCD also responds to the membranes. A prominent feature of principal cells of the last
mineralocorticoid aldosterone.273 one-third of IMCDs is the expression of the ADH-sensitive

FIGURE 1.31 Transmission electron micrograph of the inner medullary collecting duct epithelium (rat) showing the high inner
medullary collecting duct cells with many stubby microvilli of the luminal membrane and prominent lateral intercellular spaces lled
with lateral microfolds. (Magni cation 15,500.)
28
28 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

CONNECTING TUBULE

P ROXIMAL, PARS CONVOLUTA (S 1 )

CORTICAL, COLLECTING DUCT

P ROXIMAL, PARS CONVOLUTA (S 2 )

DIS TAL, PARS CONVOLUTA

P ROXIMAL, PARS RECTA (S 3 )

DIS TAL, PARS RECTA

THIN LIMB,
DES C., UP P ER L.L.

THIN LIMB, AS C., L.L. MEDULLARY COLLECTING DUCT

THIN LIMB, LOWER, L.L.

FIGURE 1.32 Summary diagram, showing cells from the various regions of the urinary tubule.

urea transporter UT-A1.193,275 In most other respects, IMCD tubule in rabbits158,248 and humans,251 in the CCD (as listed
cells resemble the other collecting duct cells. The morphol- previously), in the outer medullary collecting ducts in rats,265
ogy of the various regions of the renal tubule is summarized and in the upper part of the inner medullary cells in rats and
in Figure 1.32. rabbits.158 IC cells constitute about 37% to 40% of the cells
in CCD in rats and rabbits.265,280,283 Kaissling and Kriz158 es-
Intercalated Cells timate that rabbits have 33% IC cells in the CCD and 50%
IC cells have long been identi ed in CCDs in a variety of spe- in the outer medullary collecting duct, whereas Hansen et
cies such as rats,213,217,264,276–279 mice,264,280 humans,218,251 al.265 estimate that 36% to 40% of IC cells are present in the
and rabbits158,248,256,281–283 (Figs. 1.29 and 1.33). It has be- outer medullary collecting ducts in rat. IC cells show a slow
come obvious that IC cells are not only found in the CCD decrease in the upper part of the IMCD in rats.283
but, depending on the species, in the latter region of the DCT IC cells (Fig. 1.33) are sometimes called dark cells
in humans218,251,278 and rat,217,241,265,278 in the connecting because their cytoplasm stains more densely. They exhibit
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 229
9

FIGURE 1.33 Transmission electron micro-


graphs of intercalated cells (rat). A: Type A
exhibiting apical microfolds and at vesicles
in the apical cytoplasm. (Magni cation
6,800.) B: Type B showing a rather smooth
apical surface, many small round vesicles, and
mitochondria located predominantly in the
lateral and basal parts of the cell. (Magni ca-
tion 6,800.)

signi cant structural heterogeneity even within a single Three types of IC cells have been de ned morphologically
segment of the collecting duct. IC cells can also be distin- and by immunohistochemical staining, including type A, type
guished from principal cells by differences in cell shape, B, and non-A–non-B.264,280 However, there is a striking differ-
cytochemical staining, and uptake of the pH-sensitive ence in the number and distribution of the various types of IC
uoroprobe, 2 ,7 -bis(carboxyethyl)—5,6-carboxy uores- cells from animal to animal and among the nephron segments of
cein (BCECF). IC cells have a more circular, rather than a particular species.264 Carbonic anhydrase II immunoreactivity
hexagonal, pro le that bulges into the tubule lumen when was seen in all IC cells, but type A stained more intensely than
observed in isolated perfused tubules by means of inter- type B. The immunostaining in type A cells was pronounced
ference-contrast optics. In rabbits, IC cells can be identi- in the apical cytoplasm and apical microprojections. In type B
ed by positive staining with peanut lectin 284,285 and by cells, the staining was more diffuse throughout the cytoplasm.
luminal uptake of acetoxymethyl BCECF.285 The apical In the non-A–non-B cells, the staining was also diffuse.286
surface of the IC cell is frequently adorned by luminal ex- Type A IC cells tend to have a more circular apical cell
tensions, which include microvilli and microridges (called pro le,278 a centrally placed nucleus, prominent apical micro-
microplicae). Basal membrane infoldings resemble those of villi and microplicae extending from the apical plasma mem-
the collecting duct cell. brane, and prominent apical cytoplasmic tubulovesicular
30
30 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

pro les (see Fig. 1.33A). Mitochondria are numerous and to an increased luminal membrane area with few apical ves-
can be located above the nucleus, as well as between the icles.268 It has, therefore, been postulated that IC cells func-
nucleus and the basal membrane infoldings. Using freeze- tion in potassium reabsorption.
fracture techniques, there are rod-shaped particles and studs A similar increase in the apical plasma membrane with
present on the cytoplasmic face of the apical plasmalemma a decrease in tubulovesicular pro les was seen in IC cells
and on the tubulovesicular pro les in the IC cells of the of the outer medulla in chronic metabolic acidosis298 and
CCD and the outer medullary collecting ducts.280,287,288 acute respiratory acidosis in rats.299 In respiratory acidosis, a
H -ATPase is expressed on the apical plasma membrane and marked increase in apical microprojections was seen as was
in the cytoplasmic tubulovesicular and vesicular pro les of an increase in the surface density of the apical membrane of
type A IC cells. (See review of renal vacuolar H -ATPase.289) type A cells. No changes were seen with type B cells in rat
Verlander et al.278 showed that IC cells in animals CCD.278 Madsen and Tisher213 postulated that hydrogen ion
with respiratory acidosis show a striking increase in apical pumps located in the apical vesicles had been inserted into
microprojections and tubulovesicular pro les as well as an the apical cell membrane by vesicle fusion with the apical
increase in surface density of the apical plasma membrane. membrane.
However, no changes were seen with the number of IC cells. Type B IC cells are present in the CCD and the CT of
When there was a stimulation of bicarbonate secretion in rats,276,278,286,300 mice,264,280 and rabbits.230,256,281,301 The type
rats, Verlander and associates288 documented a withdrawal B cell (Fig. 1.33B) has an angular outline278 and a relatively
of the marker for H -ATPase from the apical plasma mem- smooth apical plasma membrane with short sparse microvilli
brane with its storage in apical cytoplasmic vesicles in IC (without rod-shaped studs in the apical membrane).280 The
cells in both the CCD and the OMCD. H -ATPase activity apical membrane generally lacks studs, but the basolateral
appeared to be inserted into the basal plasma membrane of membrane exhibits studs.278 The nucleus lies in an eccentric
type B IC cells.288 position and the numerous small mitochondria are densely
Luminal and/or tubulovesicular membranes exhibit two packed and are concentrated at the basal part of the cyto-
speci c types of particles. Large club-shaped particles called plasm. The cytoplasm and the organelles stain more dense-
“studs” have been observed on the cytoplasmic surface of ly in light and electron microscopy. Cytoplasmic vesicles
the tubulovesicular structures on the cytoplasmic side of (mostly noncoated) are seen throughout the cell cytoplasm.
the apical plasma membrane; the rapid-freeze, deep-etch In the type B cells, the H -ATPase is expressed in the ba-
technique shows 10- m spherical structures composed of solateral plasma membrane and in the cytoplasmic vesicles
multiple subunits and arranged in paracrystalline hexagonal throughout the cells.277,280,291,300,302–305 Using freeze-fracture
arrays.290 Brown et al.291 have presented evidence that the techniques, rod-shaped rectangular particles are found on
studlike material coating the vesicles contains cytoplasmic the basal membranes in these cells and not on the apical
domains of the proton-pumping H -ATPase. On the basis of plasma membrane.277,300 The basal hydrogen ion secretion is
both morphologic characteristics and immunocytochemistry, thought to mediate HCO3 secretion into the luminal uid
the structures appear to be (or related to) the vacuolar-type by an apical Cl /HCO3 exchanger different from AE1.256,264
H -ATPase.277,291 In addition, freeze-fracture studies have One candidate for this anion exchanger is pendrin, a
shown the presence of rod-shaped particles in vesicles and Na -independent Cl /HCO3 exchanger. Quentin et al.306
cell surface membranes,292 which may form a component have shown that the Cl /HCO3 exchanger pendrin in rat
of this H -ATPase. Changes in membrane structure have kidney is speci cally regulated in response to chloride bal-
been shown to be related to proton secretion.248,261 When ance independent of sodium and acid/base balance. Using a
H secretion is stimulated, cytoplasmic vesicles bearing rod- mouse model, Wall et al.307 demonstrated that pendrin pro-
shaped intramembranous particles (IMPs) fuse with the api- tein was localized in the apical cytoplasmic vesicles in both
cal plasma membrane, inserting IMPs into the membrane. type B and non-A–non-B IC cells from a subset of cells in
This supports the idea that type A IC cells demon- the DCT, the type B cells in the CNT, and the CCD. Pendrin
strate net excretion of protons into the tubular lumen that mRNA was expressed mainly in the cortex. Pendrin immu-
is accomplished by the vacuolar H -ATPase located in the noreactivity was highest in the apical cytoplasmic vesicles,
apical plasma membrane and apical tubulovesicular pro- although there was little immunogold staining along the api-
les.264,277,280,288,293,294 For this process, it has been suggested cal plasma membrane of type B IC cells, but non-A–non-
that the hydrogen ions are produced by cytosolic carbonic B IC cells had intense pendrin immunoreactivity along the
anhydrase in CNT cells and CCD cells of the mouse and apical plasma membrane. Kim et al.,308 using immunoelec-
rat264,280,286,294,295 and human.251,296,297 The bicarbonate that tron microscopy, demonstrated pendrin in both the apical
is generated is released by a band 3-like Cl /HCO3 ex- plasma membrane and intracellular vesicles throughout the
changer AE1, located in the basolateral plasma membrane, cell. Kim et al.286 found the carbonic anhydrase activity more
into the interstitium at the base of the cell. diffuse in the cytoplasm of type B than type A cells.
Medullary IC cells from rats fed a diet with a high K The third type of IC cell is non-A–non-B cells that have
content had a small luminal membrane area and a cell apex been identi ed in the CT and the CCD of rats264,276,286,309
with numerous vesicles. The ingestion of a low K diet led and mice.264,276,280 Non-A–non-B cells have vacuolar
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 331
1

type H -ATPase in both apical plasma membranes and api- of cells in the wall of the CTAL of the distal nephron in the
cal vesicles, but do not have the basolateral band 3-like im- region of the vascular pole of a renal corpuscle, called the
munoreactivity of AE1.264 In rabbits and mice, most non- “macula densa”; (2) the termination of the afferent arteri-
A–non-B cells in the collecting duct have an electroneutral ole as it enters this renal corpuscle; (3) the initiation of the
Na-independent Cl /HCO3 exchanger in the apical mem- efferent arteriole as it exits from the same renal corpuscle;
brane.264 Pendrin immunoreactivity is intense along the api- and (4) a cone-shaped region of extraglomerular mesangial
cal membrane, as well as being present in apical vesicles in cells (also called the lacis, Polkissen, or Goomaghtigh cells)
some cells in the DCT, in the CNT, and CT.307 Because both lying in the space between the macula densa and the two
pendrin immunoreactivity and pendrin-mediated HCO3 arterioles. This area receives a rich supply of sympathetic
secretion are present in the apical plasma membrane and nerve endings.
apical intracellular vesicles in type B and non-A–non-B IC In the human kidney, 40% of the basal lamina region of
cells, Kim et al.308 suggest that HCO3 secretion could be the macula densa lies adjacent to the base of the cone-shaped
regulated by traf cking of pendrin between the two mem- extraglomerular mesangium, 10% of the macula densa basal
braneous compartments. In addition, since non-A–non B lamina is in contact with the afferent arteriole, and 5% is in
cells seem capable of both apical HCO3 and H secretion, contact with the efferent arteriole.308
the simultaneous secretion of both HCO3 in exchange for The MD consists of the cells in the wall of the CTAL
Cl and proton secretion mediated by electrogenic H - of the distal tubule, which lies adjacent to the glomerular
ATPase, would cause chloride reabsorption with no change vascular pole. The MD appears as a dense spot upon hema-
in acid/base status.308 toxylin and eosin staining because the cells are narrow and
the nuclei are close together. MD cells (Fig. 1.35) are not
interdigitated with each other by large lateral cell processes,
THE JUXTAGLOMERULAR APPARATUS as in other regions of the distal tubule. In contrast, the lateral
At the vascular pole of the renal corpuscle, the macula densa intercellular spaces between MD cells extend very straight
region of the CTAL comes into close proximity to the efferent in an apical–basal direction—their width appears to vary
and afferent arterioles and a group of cells called the extra- according to function.312 The mitochondria are shorter and
glomerular mesangium.310,311 The juxtaglomerular appara- more randomly arranged. The Golgi apparatus lies on the
tus consists of four parts (Figs. 1.5 and 1.34): (1) a plaque basal side of the nucleus. The basal aspect of the MD touches

FIGURE 1.34 Light micrograph of a renal corpuscle with both urinary and vascular poles in the section. The juxtaglomerular appa-
ratus contains the macula densa (MD), the two arterioles (A), and the extraglomerular mesangium (between the As). The urinary pole
(UP) is also apparent. (Magni cation 13,000.) (From Dobyan D, with permission.)
32
32 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

The extraglomerular mesangium 310 (Goormaghtigh


cells, polar cushion, Polkissen cells, and lacis cells) lls
the area between the afferent and efferent arterioles and
the MD (see Figs. 1.5 and 1.35). It is composed of non-
granulated cells that are continuous with granular cells
and smooth muscle cells of the arteriolar walls and with
the intraglomerular mesangial cells.310 The extraglomeru-
lar mesangial cells are “ atly pressed” cylinders with both
ends splitting into a group of parallel processes. These ex-
traglomerular mesangial cells are surrounded by abundant
extracellular matrix. Numerous gap junctions occur be-
tween the processes of the same cell, with adjacent smooth
muscle cells, with granular cells, and at the renal hilus with
intraglomerular mesangial cells.240,322,323
The initmate and systematic juxtaposition of tubular
and vascular cells within the JGA has given rise to early

FIGURE 1.35 Transmission electron micrograph of the juxtaglo-


merular apparatus (rat). The transition of the thick ascending limb
(TAL) into the macula densa (MD) is seen; the basal aspect of the
macula densa abuts the extraglomerular mesangium (EGM) and
also a granular cell (GC). G, glomerulus. (Magni cation 1,900.)

the extraglomerular mesangium. Additional, but variable,


contacts are found with the efferent as well as the afferent
arterioles.13 The most conspicuous difference of MD cells to
any other cells of the nephron is the occurrence of nitric ox-
ide synthase I (see Fig. 1.37C).313,314 A prominent feature of A
the MD is also the expression of cyclooxygenase-2.315
Modi ed smooth muscle cells in the wall of the afferent
arterioles, called granular cells (formerly also called juxtaglo-
merular cells), contain speci c membrane-bound granules
(Fig. 1.36). In situ hybridization316 and immunocytochemistry
317
have shown that these cells synthesize renin (Fig. 1.37A),
which is then stored in granular form (Figs. 1.36B and 1.37B).
The granules stain with the Bowie method and have a positive
periodic acid-Schiff reaction. Like other smooth muscle cells,
the juxtaglomerular cells also contain intracellular laments
and dense bodies, but they have more cisternae of rough-
surfaced endoplasmic reticulum, a large Golgi apparatus, and
mature and immature secretory granules in their cytoplasm,
consistent with the ability of these cells to synthesize small
B
peptides. The immature granules appear to have a paracrys-
talline structure.80,318 The secretory product is released by FIGURE 1.36 Transmission electron micrograph (rat).
exocytosis into the extracellular space within or surrounding A: An afferent arteriole containing in its wall a granular
the wall of the arteriole.319,320 Processes of the juxtaglomerular cell (GC). (Magni cation 2,100.) B: Part of a granular cell
cells contact the surrounding cells as well as the endothelial containing in its cytoplasm speci c membrane-bound granules.
cells by means of gap junctions.321 (Magni cation 14,500.)
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 333
3

speculations about a functional connection in which a signal


related to the composition of the tubular uid at the MD
affects glomerular vascular tone and the glomerular ltra-
tion rate.324 It has now become clear that the JGA serves
two different functions: it regulates the ow resistance of af-
ferent arterioles in the so-called tubuloglomerular feedback
mechanism and it participates in the control of renin synthe-
sis and release from granular cells in the afferent arteriole.325
Researchers originally assumed that the two responses might
be related to each other in that renin released from the
granular cells not only has systematic relevance, but locally
triggers the formation of angiotensin II and thus is respon-
A
sible for the afferent vasoconstriction as well; however, it
now appears that the nal activation of smooth muscle and
granular effector cells occurs through largely independent
pathways. Renin release from granular cells is the major
source of systemic angiotensin II and thus plays an essential
role in controlling extracellular volume and blood pressure,
whereas the vasoconstriction of the afferent arteriole locally
serves to modulate the ltration of this nephron.
For both mechanisms, it is well established that changes
in the chloride concentration of the tubular uid at the MD
cause graded releases of mediators that reach their target by
diffusion, thus acting in a paracrine fashion.326 Note that
the extraglomerular mesangium that mediates the contact
between the MD and the effector cells is not vascularized,
B
so that the buildup of any paracrine agent would not be
perturbed by blood ow.
With respect to renin release, the most likely para-
crine mediators of this process are prostaglandin E2 and
nitric oxide.314,327,328 With respect to the vasoconstrictor
response purinergic mediators,, either ATP or adenosine,
as rst suggested by Oswald and colleagues in 1980,329
appear to play the major role.325,330,331 For an up-to-date
discussion of the function of the JGA, see the reviews by
Schnermann and Levine,325 Persson and colleagues,332 and
Komlosi et al.333

RENAL BLOOD VESSELS


C The renal arteries arise from the lateral region of the abdomi-
nal aorta at the level of the rst and second lumbar verte-
FIGURE 1.37 Rat kidney containing granular cells in afferent brae. Each artery divides into an anterior and posterior divi-
arterioles (arrows) of two glomeruli. The two pictures show sion before traversing the renal hilus. These divisions usually
that renin is synthesized (A) and stored (B) in the same cells form a total of ve segmental branches. The anterior division
(same section). A: In situ hybridization using a 330-bp rat renin gives rise to the upper, middle, and lower segmental arter-
riboprobe (cRNA) labeled with digoxygenin (detection system: ies, whereas the posterior division becomes the posterior
alkaline phosphatase). B: Immunocytochemistry using a rab- segmental artery. The apical segmental artery can arise from
bit polyclonal antirat renin antibody (detection system: Texas either division. The segmental arteries give rise to interlobar
red coupled second antibody). (From Bachmann S, Heidelberg, arteries within the renal sinus. The interlobar arteries enter
with permission.) C: NADPH diaphorese reaction showing the renal columns adjacent to the renal pyramids.
positivity of exclusively macula densa cells re ecting activity The intrarenal microvaculature has been extensively
of nitric oxide synthase. (From Bosse H-M, Heidelberg, with studied by several groups10,11,158,334; a basic pattern is es-
permission.) tablished throughout the mammalian kidneys that may be
described as follows. At the corticomedullary junction, the
interlobar artery branches into several arcuate arteries that
34
34 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Glomerular capillaries are derived from the afferent ar-


teriole, which—strictly at the entrance level—divides into
several (two to ve) primary capillary branches.335–337
Each of these branches gives rise to an anastomosing
capillary network that runs toward the urinary pole and then
turns back toward the vascular pole. Thereby, the glomerular
tuft is subdivided into several lobules, each of which con-
tains an afferent and efferent capillary portion. The lobules
are not strictly separated from each other—some anastomo-
ses between lobules occur. The capillaries converge to form
the more centrally located efferent arteriole, which is already
established inside the glomerular tuft. Thus, the efferent ar-
teriole has a signi cant intraglomerular segment that runs
through the glomerular stalk (Fig. 1.5).337 At this site a me-
sangial layer surrounds the vessel. After leaving the glomeru-
lus, the efferent arteriole is reestablished as a proper arteriole.
The efferent arterioles of super cial (or subcapsular)
glomeruli perfuse convoluted tubules through long path-
ways extending to the kidney surface or through interme-
diate branches near the renal corpuscle. In the midcortex,
the efferents either branch near the glomerulus and perfuse
convoluted tubules in that region or extend directly to the
long meshed network of the medullary ray. Efferent arteri-
oles from juxtamedullary nephrons extend downward to
form the descending vasa recta (Fig. 1.38), and occasional
branches to regions between the bundles. The early divisions
give rise to capillaries located in the outer stripe of the outer
medulla. The descending vasa recta then descend within
the vascular bundles to the inner stripe and inner medulla
FIGURE 1.38 A basic pattern of renal microvasculature. C, cortex;
(Fig. 1.39).10 The medulla is drained by venous (ascending)
OS, outer stripe; IS, inner stripe; IM, inner medulla. The left panel
shows the arterial vessels and capillaries. An arcuate artery (arrow)
gives rise to cortical radial arteries from which the glomerular
afferent arterioles originate. Efferent arterioles of juxtamedullary
glomeruli descend into the medulla and divide into the descend-
ing vasa recta, which, together with ascending vasa recta, form
the vascular bundles. At intervals, descending vasa recta leave the
bundles to feed the adjacent capillaries. The right panel shows the
venous vessels. The cortical radial veins start within the super cial
cortex; in the human kidney, some of them start as stellate veins
on the surface of the kidney (shown on the right side). They all
drain into arcuate veins. The venous drainage of the medulla is
carried out by venous vasa recta; those from the inner medulla
all traverse the inner stripe within the vascular bundles, whereas
most of the venous vasa recta from the inner stripe ascend
outside the bundles. After traversing the outer stripe as wide
tortuous channels, the ascending vasa recta drain into arcuate or
cortical radial veins. (From Rollhäuser H, Kriz W. Das Gefäss-system
der Rattenniere. Z Zellforsch. 1964;64:381, with permission.)

arch across the base of the renal pyramid (Fig. 1.38). The
arcuate arteries give rise to interlobular arteries (cortical ra-
dial arteries) that course peripherally, between the medullary
rays and, thus, within the cortical labyrinth. The interlobular
arteries also branch, and the branches give rise to afferent FIGURE 1.39 Light micrograph of a vasa recta bundle, showing
arterioles that supply the renal corpuscles. descending (D) and ascending (A) vessels. (Magni cation 950.)
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 335
5

vasa recta, which lie adjacent to descending vasa recta, form- In functional studies, the interstitial volume of the kid-
ing the vascular bundles that function as a countercurrent ney has been estimated to amount to 13% of the total kidney
exchanger. Venous vasa recta that drain the inner medulla volume, whereas stereologically derived values for the cell-
remain in the vascular bundles through the inner and outer free interstitial space of the cortex and outer medulla of the
stripe region. The venous vessels that drain the region of rat range between 3% and 5%.344–346 Thus, the functional
the upper and middle inner stripe ascend within the inter- interstitium includes more than just the peritubular spaces;
bundle region to the outer stripe, there joining the vessels the prominent periarterial connective tissue sheaths (see the
leaving the bundles to form together the major part of the following) may, in fact, account for one-half of the entire
blood supply to this region by a network of venous vessels. interstitial volume.347
The density of capillaries that are derived directly from effer- The interstitium is differently developed within the
ent arterioles is debated.338 Finally, the ascending vasa recta various regions regions of the kidney.342,343 In the cortex,
empty into arcuate or interlobular veins (Fig. 1.38). the peritubular interstitium is distinguished from the peri-
The cortical venous system is subject to some varia- arterial connective tissue (Fig. 1.40). The peritubular inter-
tion. In the human, the venous drainage starts on the re- stitium consist of the narrow spaces between tubules and
nal surface beneath the capsule with small venules (stellate
veins) that converge to form the interlobular veins (cortical
radial veins); in most other species, the interlobular veins
begin deeply beneath the cortical surface and stellate veins
are lacking. Interlobular veins receive tributaries from the
cortical peritubular capillary network (Fig. 1.38) and nally
empty into arcuate veins that accompany the arcuate arteries.
The arcuate veins receive blood from the venous vasa recta,
as described in the preceding section. Interlobar veins form
by con uence of arcuate veins and the latter nally form
the renal vein. In contrast to the arcuate arteries, which are
terminal arteries, the arcuate veins form true anastomosing
arches at the corticomedullary border.
The morphology of the descending and ascending parts
of the vasa recta differs markedly (Fig. 1.39).187,339,340 The de-
scending vasa recta are lined by a continuous nonfenestrated
endothelium, with cells oriented longitudinally along the
cell axis, forming 10 to 20 cell pro les in a single cross-
section.187,340 The endothelium contains the urea transporter
UT-B1.193 Pericytes are seen encircling the descending vasa
recta, but they become less frequent as the vessels descend
into the inner medulla, where these vessels nally convert
into capillaries. As long as the descending vasa recta have
pericytes nerve terminals are seen in their neighborhood.
The capillaries and the ascending vasa recta are lined
by a thin fenestrated epithelium. The fenestrae are similar
to those seen in peritubular capillaries, as well as in most
regions of the body, being 40 to 70 m in diameter187 and
bridged by a thin diaphragm. Uniquely, fenestrated endothe-
lium can line quite large vessels in the kidney.
Lymphatic vessels are seen only in the cortex accompa-
nying the arteries within the periarterial tissue sheaths (see
below). The medulla has no lymphatic drainage.341

FIGURE 1.40 Low power electron micrograph of a cross-


INTERSTITIUM section through the renal cortex (rat). A cortical radial artery
The interstitium of the kidney comprises the extravascular (A) and vein (V), an afferent arteriole (AA), and several tubular
intertubular spaces of the renal parenchyma, with their at- pro les are seen. The arteries are surrounded by the loose con-
tendant cellular elements and extracellular substances.342,343 nective tissue sheath that contains the intrarenal lymphatics
It is bounded on all sides by tubular and vascular basement (Ly); the interstitial spaces of this sheath are continuous with the
membranes. The lymphatics are considered as part of the peritubular interstitium, which includes wide (stars) and narrow
interstitium. (arrows) portions. (Magni cation 1,000.)
36
36 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

around glomeruli. Sometimes, the peritubular capillaries


are considered as part of the peritubular interstitium. Cap-
illaries may abut directly to the the outer surface of tubules
accounting for 54% to 67%348 of the capillary surface,
whereas only 26% of the tubular surface is directly adjacent
to peritubular capillaries.349 Thus, most of the exchanges
among tubules and vessels have to pass through the inter- Inte rlobula r
stitial compartment. a rte ry + ve in

The periarterial connective tissue forms a uid-rich loose


connective tissue sheath that surrounds the intrarenal arter-
ies and contains the lymphatic vessels of the kidney (Figs.
1.40 and 1.41).341,347,350 The periarterial lymphatic sheath
extends along the intrarenal arteries as far as the afferent ar-
teriole, where it becomes quite attenuated. It is particularly
well developed around the arcuate and cortical radial arter-
ies. It contains the renal nerves.
The lymphatic capillaries begin within these sheaths;
lymphatics do not, in general, penetrate the renal paren-
chyma proper and are not found in the medulla.341,351 The
lymphatic vessels converge along with the intrarenal arteries Arcua te
a rte ry + ve in
to emerge at the renal hilus. The peritubular interstitium of
the cortex freely communicates with the periarterial tissue
sheaths. Within the sheaths, uid and solutes gradually may Va s cula r
enter the lymphatic vessels as they converge toward the hilus bundle
(Fig. 1.41). In addition to lymphatic drainage, the periarte- A
rial connective tissue sheaths probably participate in the dis-
tribution of vasoactive substances alongside of the intrarenal
arterioles and arteries.350 It also serves for the intrarenal dis-
tribution of in ammatory cells.
1
In the medulla, the interstitium is differently developed
within the three medullary regions342: outer and inner stripes A 2
and inner medulla. The relative interstitial volume exhibits a
3 V
pronounced axial gradient from cortex to the inner medulla.
The outer stripe has a very narrow, sparse interstitium, oc- Ly
cupying 3% to 5% of outer stripe volume.342,352 Also the vasa
recta within the bundles of the inner stripe are very narrowly
packed. The interstitial volume of the interbundle regions N
of the inner stripe is somewhat greater (10% in rats). The
B
most elaborate and distinctive type of regional interstitium
is that of the inner medulla. Here, the interstitium consti- FIGURE 1.41 Schematics showing (A) the distribution and
tutes a much larger part of the total tissue volume (30% to (B) the relationships of periarterial connective tissue sheath.
40%),345,352 and shows a particular arrangement of its bro- The periarterial sheath is schematically indicated as a wide
blasts (see later). “stocking” drawn over the intrarenal arteries. In reality, there is
no limiting membrane between the sheath and the surround-
The Cells of the Interstitium ing interstitium. The lymphatics (stippled area) originate and
The renal interstitium contains two types of cells: bro- travel within the periarterial sheath. The medullary rays are
blasts and dendritic cells. The broblasts are quite differently indicated by a broken line. The traverse section in (B) shows
developed in the cortex and medulla.342,353–356 Let us rst possible relationships between the sheath and the surround-
consider the cortex. Within the peritubular interstitium ing structures (double-headed arrows): (1) with the peritubular
roughly 50% of the cells normally present are broblasts interstitium, (2) with the accompanying vein (V), and (3) with
and the other 50% dendritic cells, which cannot be sepa- lymphatics (Ly). The single-headed arrows indicate the ow of
rated from each other by simple light microscopy. The - the respective uid. N, nerve. (From Kriz W. A periarterial path-
broblasts in this region are extensively branched, with long, way for intrarenal distribution of renin. Kidney Int. 987;31:551,
often sheetlike processes (Fig. 1.42).343,357,358 They contain with permission.)
abundant rough endoplasmic reticulum. Mitochondria,
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 337
7

In the inner medulla, the broblasts represent a particular


variety. They contain numerous homogeneous osmiophilic
lipid droplets. Hence, they are generally called lipid-laden
interstitial cells (Fig. 1.43).353,355,367 These star-shaped cells
interconnect loops of Henle and vasa recta, spanning these
axial structures like the rungs of a ladder.342,358 They in-
crease in number toward the tip of the papilla and have
an abundant rough endoplasmic reticulum, with cisternae
that are often dilated and lled with occulent material.
A cytoskeleton is especially well developed in their most
peripheral cell processes. These cells possess receptors for
angiotensin II and bradykinin.368,369 They are responsible for

FIGURE 1.42 Transmission electron micrograph of interstitial


cells in the cortical peritubular interstitium. Two types are seen:
broblasts (1) with many processes and a dendritic cell (2). C,
capillary. (Magni cation 5,800.)

Golgi complexes, lysosomes, and micro lament bundles


are regularly encountered. Fibroblasts perform a scaffold-
ing function interconnecting the nephrons and the peri-
tubular capillaries. They perform this function by focal
contacts of their cytoplasmic processes with neighboring
broblasts as well as with capillaries and tubules.343,356,357
In addition, they produce the brous matrix, which serves
together with these cells as a common scaffold throughout
the renal parenchyma. These broblasts can be unequivo-
cally detected by electron microscopy and uorescence mi-
croscopy by their strong expression of ecto-5 -nucleotidase
(CD73),343,356,357,359,360 as well as of the PDGF receptor beta FIGURE 1.43 Transmission electron micrograph of a longi-
(PDGFR ).361 tudinal section of the inner medulla (rat) showing lipid-laden
Fibroblasts of the renal cortex with their enzyme ecto- interstitial cells arranged like the rungs of a ladder between
5 -nucleotidase362,363 can generate adenosine within the cor- parallel running tubes or vessels. Note the numerous lipid drop-
tical interstitium. In addition, a subfraction of cortical inter- lets (arrowheads) and the prominent endoplasmic reticulum
stitial cells synthesize erythropoietin.364–366 (arrows). (Magni cation 3,300.)
38
38 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

the production of extracellular bers and ground substance, developed into a complex structural system that accounts
including the abundant glycosaminoglycans and hyaluronic for this function. Whereas the mechanisms to produce
acid of the inner medulla.370 a concentrated urine up to a concentration of about 600
The lipid-laden interstitial cells participate in produc- mOsmol/L in the outer medulla are fairly well known, the
ing the medullary prostaglandins. The lipid droplets of mechanisms underlying the nal urine concentration in the
these cells contain polyunsaturated fatty acids that appear inner medulla are still poorly understood.
to be precursors of prostaglandins and other lipid-derived Within the outer medulla the reabsorption of NaCl from
hormones.368,369 The cells produce vasodepressing lipids, in the MTAL represents the driving force to produce a corti-
particular, prostaglandin E2.371,372 comedullary osmotic gradient that provokes the osmotic
The second abundant cell type in the renal interstitium water withdrawal from the collecting duct passing through
is the dendritic cell (Fig. 1.42). They originate from the bone this region. ADH initiates the insertion of AQP2 chan-
marrow and, as in other organs, are subject to a vivid turn- nels into the apical plasma membrane of collecting duct
over.373 In the kidney, dendritic cells are found in the intersti- principal cells. Together with the constitutive AQP3 and
tium throughout the cortex and the outer medulla.343,356,357 AQP4 channels in the basolateral membrane of these cells,
They enter the interstitium from the blood, reside for some this allows osmotic water withdrawal into the hypertonic
days in interstitial spaces, and leave the interstitium with the interstitium of the outer medulla.192 The reabsorbed water
lymph ow. is brought back into the systemic circulation by venous
The extracellular components of the interstitium form a vasa recta.2
matrix that may be thought of as a hydrated gel of ground The nal step of the urine concentrating process in the
substance within a brillar reticulum.342 Several bers inner medulla is basically identical insofar as water is reab-
make up the interstitial reticulum. Collagen bers (types sorbed by osmosis through ADH-stimulated water-permeable
I, III, and VI) are present in the matrix, both in isolation collecting ducts into a hypertonic interstitium. However, the
and in bundles. Type I collagen forms typical cross-banded mechanisms for creating the osmotic gradient in the inner
bers, generally more than 30 m in diameter. Type III medulla up to 1200 mOsmol/L in humans and much higher
bers (10 to 40 m in diameter) and type VI bers (6 to in many rodent species are much more complex than in the
10 m in diameter) are often seen associated with type outer medulla and are insuf ciently understood. A driving
I bers. In addition, unbanded micro brils with a diam- force like the sodium pump in MTALs in the outer medulla is
eter of 15 to 30 m and an electron-lucent core have been lacking. This has led to several “passive” models,379,380 which
described.342,354 take into account the speci c transport properties of thin
Myo broblasts are absent from healthy kidneys. They limbs, collecting ducts, and vasa recta to elucidate, by math-
differ from broblasts by their high production of extra- ematical modeling, how part of the osmotic energy produced
cellular matrix, expression of vimentin, and -smooth by the function of MTALs in the outer medulla may be trans-
muscle actin ( SMA).343,374 The accumulation of myo - ferred into the inner medulla, leading there to an osmotic
broblasts in a diseased kidney is generally suggested to oc- gradient toward the tip of the papilla. A review of these mod-
cur by transformation of broblasts into myo broblasts in els is far beyond the scope of this chapter. From a structural
response to stimuli from locally produced cytokines.374,375 point of view, two features appear most relevant.
An alternative hypothesis postulates that myo broblasts
may develop by “epithelialmesenchymal transition” (EMT) 1. The shape of the inner medulla. The inner medulla
from tubular cells376; however, the evidence for this origin has a particular shape, tapering from a broad basis to a
is not conclusive.377 tiny papilla. The mass of the inner medulla is, there-
Macrophages (histiocytes) are also rarely found in the fore, unevenly distributed along the longitudal axis.
healthy kidney, except for the periarterial tissue sheaths356 A reconstruction study in the rat2,381,382 has shown that
(Fig. 1.42). These round cells demonstrate primary and the inner medulla is shaped like a mushroom, consist-
secondary lysosomes and characteristic surface folds. Along ing of a broad head and a thin stalk. Calculations in
with interstitial affections, the number of macrophages may the model have shown that the rst one-half of the in-
dramatically rise. ner medulla comprises roughly 80% of the total inner
medullary volume and, consequently, only 20% are left
for the second, papillary, half. Thus, the decrease of the
STRUCTURE–FUNCTION mass in the inner medulla along the longitudal axis is
almost exponential. This shape perfectly re ects what
RELATIONSHIPS WITHIN THE happens with the structures within the inner medulla:
RENAL MEDULLA loops of Henle, collecting ducts, and vasa recta all
During phylogeny, the renal medulla has developed in re- decrease rapidly in number from the base to the tip of
sponse to the necessity to conserve water by excreting a the papilla.381,382 It has been calculated that of an esti-
concentrated urine.378 Loops of Henle, collecting ducts, mated 10,000 long loops entering the inner medulla at
and a speci c blood supply through vascular bundles have its base, only about 1,500 reach the papillary one-half
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 339
9

of the inner medulla.2 Thus, the majority of thin limbs urea delivery in the papilla (i.e., at the “ultimate bend” of the
(the “short” long thin limbs) turn back shortly after complex countercurrent exchange system of the inner me-
entering the inner medulla; a smaller, but still substan- dulla) is optimal because (1) by countercurrent exchange in
tial, number of thin limbs reach the middle part of the vasa recta and thin limbs, urea is largely trapped and distrib-
inner medulla; and only a small population of “long” uted in a longitudinal gradient within the inner medulla and
long loops really reach the papilla. (2) the fraction of urea that escapes the inner medulla is sub-
This has led to the proposal383 and, later, math- ject to recycling via short loops back into the inner medulla.
ematical models204,384 that this arrangement might Thus, this latter fraction of urea is available another time
account for a cascadelike transport of solutes toward within the papillary interstitium ready to withdraw and/or to
the tip of the papilla. The large fraction of “short” long balance water reabsorption from the collecting duct. Recy-
loops, by some way or another, manages the transport cling is not simply trapping because this fraction of urea on
of solutes into the rst one-third of the inner medulla, its nephron way back into the inner medulla is concentrated
the intermediate fraction of long loops brings a propor- a second time, essentially by the work of the sodium pump
tion of these solutes further down into the middle in the MTALs. In essence, urea recycling by short loops (not
part, and nally the small fraction of “long” long loops by long loops) represents an effective transport of osmotic
completes the transport of a still much smaller solute energy from the outer into the inner medulla.
fraction down into the papilla. Whether these mechanisms are suf cient to build an
2. Structural arrangements in the renal medulla that osmotic gradient up to several osmoles in desert rodents,
allow the recycling of urea into the inner medulla or whether there are additional sources complementing the
via short loops of Henle. The descending thin limbs solute gradients (e.g., continuous production of osmotic ac-
of short loops in the outer medulla are arranged in a tive substances), is presently unknown. Even if this question
pattern that allows the shifting of urea from the venous does not belong to the most urgent problems in medicine, it
blood coming up with the venous vasa recta from the represents a highly challenging biologic enigma.
inner medulla into tubular uid of descending thin
limbs of short loops. This possibility is perfectly de-
veloped in the complex vascular bundles in the outer REFERENCES
medulla of rodents with high urine-concentrating 1. Inke G. G. The
The Protolobar
Protolobar Structure
Structure of
o the
the Human
Human Kidney.
Kidney. New York: Alan R.
Liss, Inc.; 1988.
abilities.159,187,197,198 Within these vascular bundles, 2. Jamison R, R,Kriz
KrizW.WUrinary
Urinary Concentrating
Concentrating Mechanism.
Mechanism. Structure
Structure andand Func-
Function.
the SDTLs are arranged in a countercurrent fashion tion.
NewNew York:York: Oxford
Oxford University
University Press;Press;
1982.1982.
with ascending vasa recta coming up from the inner 3. Amann K, Plank C, Dotsch J. Low nephron number - a new cardiovascular
risk factor in children? Pediatr Nephrol. 2004;19:1319–1323.
medulla. Thus urea, by countercurrent exchange, may http://www.ncbi.nlm.nih.gov/pubmed/15490250
4. Neiss W. Morphogenesis and histogenesis of the connecting tubule in the
directly enter the descending thin limbs through the rat4.kidney.
NeissAnat
W Morphogenesis and histogenesis of the connecting tubule in the
Embryol. 1982;165:81.
urea transporter UT-A2193,275 (the fenestrated endo- rat 5.
kidney.
OliverAnat Embryol 1982;165:81.
J. Nephrons and Kidneys. New York/Evanston/London: Harper &
http://www.ncbi.nlm.nih.gov/pubmed/7149277
Row, Hoeber Medical; 1968.
thelium of venous vasa recta may readily be expected 5. Oliver J. Nephrons
6. Osathanondh and E.
V, Potter Kidneys. New York/Evanston/London:
Development of human kidney as shown Harper by&
to be highly permeable for urea). In a renal medulla Row, Hoeber Medical;
microdissection. 1968.
III. Formation and interrelationship of collecting tubules and
with “simple” vascular bundles (as they are found in 6. Osathanondh
nephrons. Arch Pathol. V 1963;76:290–302.
Potter E. Development of human kidney as shown by
microdissection. III. Formationüber
7. Peter K. Untersuchungen andBau
interrelationship
und Entwicklung of der
collecting tubules
Niere. Jena: and
Gustav
most species), countercurrent exchange of urea rst nephrons. Arch Pathol. 1963;76:290–302.
Fischer; 1909.
occurs from ascending to descending vasa recta (which http://www.ncbi.nlm.nih.gov/pubmed/14058156
8. Kriz W, Bankir L. A standard nomenclature for structure of the kidney. The
contain the urea transporter UT-B1), which, afterward, 7. Peter
Renal K. Untersuchungen
Commission über Bau Union
of the International und Entwicklung der Niere.
of Physiological Jena: (IUPS).
Sciences Gustav
Fischer; 1909.1988;411:113–120.
P ugers Arch.
will deliver their blood to the capillary plexus of the 8. Kriz W, Bankir
9. Beeuwkes L. A standard
R. Efferent vascularnomenclature for structure
patterns and early of the kidney.
vascular-tubular The
relations
interbundle region surrounding the thin limbs of short Renal
in the Commission
dog kidney. Am of the International
J Physiol. Union of Physiological Sciences (IUPS).
1971;221:1361–1374.
loops on their descending direction. Thus, even if Pf10.
ugers Arch. 1988;411:113–120.
Beeuwkes R. The vascular organization of the kidney. Annu Rev Physiol.
http://www.ncbi.nlm.nih.gov/pubmed/3353210
1980;42:531–542.
probably less effectively, urea from the inner medulla 9. Rollhäuser
11. Beeuwkes R. H,Efferent
Kriz W, vascular
Heinke W.patterns and early vascular-tubular
Das Gefässsystem der Rattenniere.relations
Z Zell-
has access to the SDTLs.193,197,385,386 in the Mikrosk
forsch dog kidney.
Anat.Am J Physiol. 1971;221:1361–1374.
1964;64:381–403.
http://www.ncbi.nlm.nih.gov/pubmed/5124281
12. Sperber J. Studies of the mammalian kidney. Zool Bidrag. 1944;22:249–431.
Starting with the MTAL, thus abruptly at the end of the 10. Beeuwkes
13. Kriz R. TheB.vascular
W, Kaissling organization
Structural organizationof of
thethekidney. Annu Rev
mammalian Physiol.
kidney. In:
ATL, the uriniferous tubule is impermeable to urea until the 1980;42:531–542.
Seldin D, Giebisch G, eds. The Kidney: Physiology and Pathophysiology. New York:
terminal collecting duct in the inner medulla, which, start- http://www.ncbi.nlm.nih.gov/pubmed/6996596
Raven Press; 1992:707–777.
11. Rollhäuser
14. Pfeiffer H, Kriz W Heinke
E. Comparative W Dasobservations
anatomical Gefässsystemofder theRattenniere.
mammalianZrenalZell-
ing abruptly, expresses the ADH-dependent urea transporter orsch Mikrosk
pelvis Anat. 1964;64:381–403.
and medulla. J Anat. 1968;102:321–331.
UT-A1.193,275 Because of water reabsorption upstream from http://www.ncbi.nlm.nih.gov/pubmed/14320463
15. Schmidt-Nielsen B, Pfeiffer E. Urea and urinary concentrating ability in the
this segment, urea becomes concentrated within the tubular 12. Sperber
mountain beaverJ. Aplodontia
Studies ofrufa. the Ammammalian kidney. Zool Bidrag. 1944;22:
J Physiol. 1970;218:1370–1375.
249–431.
16. van Liew J, Buentig W, Stolte H, et al. Protein excretion: Micropuncture
urine to levels always higher than the adjacent interstitium. 13. of
study Kriz
ratW, Kaissling
capsular andB.proximal
Structural organization
tubule uid. Am of the mammalian
J Physiol. kidney. In:
1970;219:299–305.
In the terminal CD segment, a major part of this urea diffuses Seldin D, Giebisch G,J.eds.
17. Pappenheimer The Kidney:
Passage Physiology
of molecules and capillary
through Pathophysiology. New YRev.
walls. Physiol ork:
into the papillary interstitium and where it mixes with other Raven Press; 1992:707–777.
1953;33:387.
14. Pfeiffer E. Comparative
18. Helmchen U. Die Zahl der anatomical observations
Mesangiumzellen of thenormalen
in einem mammalian renal
Glomeru-
solutes (NaCl), contributing to the osmotic driving force pelvis
lum derand medulla. JEine
Rattenniere: Anat.dreidimensionale
1968;102:321–331. elektronenoptische Analyse. Tuebingen,
that reclaims water from the collecting duct. The locus of http://www.ncbi.nlm.nih.gov/pubmed/5643845
Germany: Inaugural dissertation; 1980.
40
40 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

15. Zimmermann
19. Schmidt-Nielsen B, P eiden
K. Ueber er E.
BauUreadesand urinary concentrating
Glomerulus der menschlichen abilityNiere.
in the 49. Schwarz
39. Adler K, Simons
S. Integrin M, Reiser
receptors in J,
theet glomerulus:
al. Podocin, potential
a ra t-associated
role in compo nent
glomerular
Zmountain
Mikrosk Anatbeaver Aplodontia
Forsch. ru a. Am J Physiol. 1970;218:1370–1375.
1929;18:520–552. o the glomerular
injury. Am J Physiol. slit1992;262:F697–F704.
diaphragm, interacts with CD2AP and nephrin. J Clin Invest.
16. Arakawa
20. van LiewM. J, ABuentig
scanning W electron
Stolte H,microscopy
et al. Protein excretion:
of the glomerulusMicropuncture
of normal 2001;108:1621–1629.
50. Raats C, van den Born J, Bakker M, et al. Expression of agrin, dystroglycan,
study
and o rat capsular
nephrotic rats. LabandInvest.
proximal tubule uid. Am J Physiol. 1970;219:299–305.
1970;23:489–496. and40.utrophin
Dunne in J, Hanby
normalA,renal Poulsom
tissueR, andet in
al. experimental
Molecular cloning and tissue expres-
glomerulopathies. Am J
http://www.ncbi.nlm.nih.gov/pubmed/5448057
21. Spinelli F. Structure and development of the renal glomerulus as revealed sion o 2000;156(5):1749–1765.
Pathol. FAT, the human homologue o the Drosophila at gene that is located on
by17. Pappenheimer
scanning J. Passage o Int
electron microscopy. molecules
Rev Cytol. through capillary walls. Physiol Rev.
1974;39:345–381. chromosome
51. Regele H, 4q34-q35
Fillipovicand encodes
E, Langer a putative
B, et adhesion
al. Glomerular molecule.
expression Genomics.
of dystrogly-
1953;33:387.
22. Fries J, Sandstrom D, Meyer T, et al. Glomerular hypertrophy and epi- 1995;30:207–223.
cans is reduced in minimal change nephrosis but not in focal segmental glomeru-
http://www.ncbi.nlm.nih.gov/pubmed/13088295
thelial cell injury modulate progressive glomerulosclerosis in the rat. Lab Invest. http://www.ncbi.nlm.nih.gov/pubmed/8586420
losclerosis. J Am Soc Nephrol. 2000;11(3):403–412.
18. Helmchen U. Die Zahl der Mesangiumzellen in einem normalen Glomerulum
1989;60:205–218. 41. Kerjaschki
52. Ciani L, Patel A, AllenD, N,
D, Sharkey et al. Mice
Farquhar lacking
M. Identi the and
cation giantcharacterization
protocadherin
der
23.Rattenniere:
Sterzel R, Eine Pabst dreidimensionale
R. The temporal elektronenoptische
relationship betweenAnalyse. Tuebingen,
glomerular Ger- cell mFAT1
of exhibit renalmajor
podocalyxin-the slit junction
sialoproteinabnormalities
of the renal and aglomerular
partially penetrant
epithelialcyclo-
cell.
many: Inaugural
proliferation anddissertation;
monocyte 1980. in ltration in experimental glomerulonephritis. Jpia and
Cell anophthalmia
Biol. 1984;98:1591. phenotype. Mol Cell Biol. 2003;23:3575–3582.
19. Zimmermann
Virchows Arch B Cell Pathol K. Ueber Incl den
Mol Bau
Pathol.des1982;38:337–350.
Glomerulus der menschlichen Niere. http://www.ncbi.nlm.nih.gov/pubmed/12724416
53. Orlando R, Takeda T, Zak B, et al. The glomerular epithelial cell anti-
Z24.
Mikrosk
Kriz Anat Forsch.N,1929;18:520–552.
W, Gretz Lemley K. Progression of glomerular diseases: Is the 42. Miner
adhesion J. Renal associates
podocalyxin basement with membrane
the actincomponents.
cytoskeleton KidneythroughInt. 1999;56:
interactions
20. Arakawa
podocyte M. A scanning
the culprit? Kidney Int.electron microscopy o the glomerulus o normal
1998;54:687–697. 2016–2024.
with ezrin. J Am Soc Nephrol. 2001;12:1589–1598.
and
25.nephrotic
Kriz W, rats. Le Hir LabM. Invest.
Pathways1970;23:489–496.
to nephron loss starting from glomerular http://www.ncbi.nlm.nih.gov/pubmed/10594777
54. Michael A, Blau E, Vernier R. Glomerular polyanion. Alterations in amino-
http://www.ncbi.nlm.nih.gov/pubmed/5483882
diseases - Insights from animal models. Kidney Int. 2005;67:404–419. 43. Drenckhahn
nucleoside nephrosis. D, Lab
Franke R. 1970;23:649–657.
Invest. Ultrastructural organization o contractile and
21. Sagrinati
26. Spinelli FC, . Structure
Netti G, and development
Mazzinghi B, et al.o Isolation
the renaland glomerulus as revealed
characterization of cytoskeletal
55. Andrews proteins in glomerular
P. Glomerular epithelial podocytes
alterations o resulting
chicken, from rat, sialic
and man. Lab
acid sur-
by scanning progenitor
multipotent electron microscopy.
cells from the Int Rev
Bowman’Cytol.s 1974;39:345–381.
capsule of adult human kidneys. Invest.
face 1988;59:673–682.
coat removal. Kidney Int. 1979;15:376–385.
Jhttp://www.ncbi.nlm.nih.gov/pubmed/4611944
Am Soc Nephrol. 2006;17:2443–2456. http://www.ncbi.nlm.nih.gov/pubmed/3141719
56. Seiler M, Rennke H, Venkatachalam M, et al. Pathogenesis of polycation-
22. Ronconi
27. Fries J, Sandstrom
E, SagrinatiD,C,Meyer T, et M,
Angelotti al. et
Glomerular hypertrophy
al. Regeneration and epi-
of glomerular 44. Sanden
induced alteration W, (fusion)
Elger M, Mundel P,epithelium.
of glomerular et al. TheLab architecture o podocyte
Invest. 1977;36:48–61.
thelial cell by
podocytes injury
human modulate progressive Jglomerulosclerosis
renal progenitors. in the rat. Lab Invest.
Am Soc Nephrol. 2009;20:322–332. cytoskeleton
57. Kerjaschki suggests a role
D, Vernillo in glomerular
A, Farquhar M. Reduced f ltration dynamics.
sialylation Ann Anat.
of podocalyxin-
1989;60:205–218.
28. Appel D, Kershaw D, Smeets B, et al. Recruitment of podocytes from 1995;177:44–45.
the major sialoprotein of the rat kidney glomerulusin aminonucleotide neph-
http://www.ncbi.nlm.nih.gov/pubmed/2915515
glomerular parietal epithelial cells. J Am Soc Nephrol. 2008;20:333–343. 45. Am
rons. Kaplan J, Kim
J Pathol. S, North K, et al. Mutations in ACTN4, encoding a -actinin-4.
1985;118:343.
23. Smeets
29. Sterzel B, R, Uhlig
Pabst S, R.Fuss
The A, temporal relationship
et al. Tracing the originbetween glomerular
of glomerular cell pro-
extracapil- cause amilialT,ocal
58. Takeda segmental
McQuistan glomerulosclerosis.
T, Orlando R, et al. Loss Natof Genet. 2000;24:251–256.
glomerular foot processes
li eration
lary lesionsand frommonocyte
parietalinf ltration cells.
epithelial in experimental glomerulonephritis.
J Am Soc Nephrol. 2009;20:2604–2615.Virchows http://www.ncbi.nlm.nih.gov/pubmed/10700177
is associated with uncoupling of podocalyxin from the actin cytoskeleton. J Clin
Arch
30. BRodewald
Cell PatholR,Incl Mol Pathol.M.1982;38:337–350.
Karnovsky Porous substructure of the glomerular slit 46. Weins
Invest. A, Kenlan P, Herbert S, et al. Mutational and biological analysis
2001;108:289–301.
http://www.ncbi.nlm.nih.gov/pubmed/6121416
diaphragm in the rat and mouse. J Cell Biol. 1974;60:423–433. o59.alpha-actinin-4
Pavenstadt H, in Krizocal segmental
W, Kretzler glomerulosclerosis.
M. Cell biology of the glomerularJ Am Socpodocyte.
Nephrol.
24. Reiser
31. Kriz WJ, Gretz
Kriz W,N,Kretzler
LemleyM, K. etProgression o glomerular
al. The glomerular diseases: isIsathe
slit diaphragm modi-po- 2005;16:3694–3701.
Physiol Rev. 2003;83:253–307.
docyte the culprit?
ed adherens Kidney
junction. J Am Int.Soc1998;54:687–697.
Nephrol. 2000;11:1–8. http://www.ncbi.nlm.nih.gov/pubmed/16251236
60. Reiser J, Polu K, Moller C, et al. TRPC6 is a glomerular slit diaphragm-asso-
http://www.ncbi.nlm.nih.gov/pubmed/9734594
32. Ruotsalainen V, Ljungberg P, Wartiovaara J, et al. Nephrin is speci cally 47. channel
ciated Braun E,requiredSchlondor D, Becker
for normal D, et al. Nat
renal function. Mutations in the ormin gene
Genet. 2005;37:739–774.
25. Kriz
located W, slit
at the Le Hir M. Pathways
diaphragm to nephron
of glomerular loss starting
podocytes. Procrom
Natlglomerular
Acad Sci USA. dis- INF2
61. cause ocal segmental
Kazatchkine M, Fearonglomeruloscerlosis.
D, Appay M, et al.Nat Genet. 2010;42:72–76.
Immunohistochemical study of
eases - Insights rom animal models. Kidney Int. 2005;67:404–419.
1999;96(14):7962–7967. http://www.ncbi.nlm.nih.gov/pubmed/20023659
the human glomerular C3b receptor in normal kidney and in seventy- ve cases
http://www.ncbi.nlm.nih.gov/pubmed/15673288
33. Donoviel D, Freed D, Vogel H, et al. Proteinuria and perinatal lethality in of48.
renalKreidberg
diseases. J,J Clin Symons
Invest.J. 1982;69:900–912.
Integrins in kidney development, unction, and
26. lacking
mice Sagrinati C, Netti
NEPH1, G, Mazzinghi
a novel protein with B, et al. Isolation
homology and characterization
to nephrin. Mol Cell Biol. disease. Am J Physiol
62. Kerjaschki Renal Physiol.
D, Farquhar 2000;279(2):F233–F242.
M. Immunocytochemical localization of the Hey-
o multipotent progenitor cells rom the Bowman’s capsule o adult human
2001;21:4829–4836. 49. antigen
mann Adler S. (gp Integrin
330) receptors
in glomerular in the glomerulus:
epithelial cells potential
of normalrole in glomerular
Lewis rats. J Exp
kidneys.J
34. Inoue AmT,Soc Nephrol.
Yaoita 2006;17:2443–2456.
E, Kurihara H, et al. Fat is a component of glomerular slit injury.1983;157:667–686.
Med. Am J Physiol. 1992;262:F697–F704.
http://www.ncbi.nlm.nih.gov/pubmed/16885410
diaphragms. Kidney Int. 2001;59:1003–1012. 50. Schiffer
63. Raats C,M, vanSchiffer
den Born J, Bakker
L, Gupta M,al.
A, et et Inhibitory
al. Expression smadso agrin, dystroglycan,
and TGF- signal-
27. Schnabel
35. Ronconi E, E, Sagrinati
AndersonC,J,AngelottiFarquharM,M. et al.
TheRegeneration
tight junction o glomerular
protein ZO-1 po- and in
ling utrophin
glomerular in normal
cells. Jrenal
Am Soctissue and in2002;13:2657–2666.
Nephrol. experimental glomerulopathies. Am J
docytes
is by human
concentrated alongrenalslitprogenitors.
diaphragmsJ of Amthe Socglomerular
Nephrol. 2009;20:322–332.
epithelium. J Cell Biol. Pathol. 2000;156(5):1749–1765.
64. Wogensen L, Nielsen C, Hjorth P. Under control of the Ren-1c promoter,
http://www.ncbi.nlm.nih.gov/pubmed/19092120
1990;111:1255–1263. http://www.ncbi.nlm.nih.gov/pubmed/10793086
locally produced transforming growth factor-beta1 induced accumulation of glo-
28. Boute
36. AppelN, D, Gribouval
Kershaw D,O,Smeets RoselliB,S,etetal.al.Recruitment
NPHS2, encoding o podocytes rom glo-
the glomerular 51. Regele
merular H, Fillipovic
extracellular matrix E, in
Langer B, et al.
transgenic Glomerular
mice. Diabetes.expression o dystrogly-
1999;48:182–192.
merular podocin,
protein parietal epithelial
is mutated cells.
in Jautosomal
Am Soc Nephrol.recessive2008;20:333–343.
steroid-resistant nephrotic cans
65. isKriz
reduced in minimal
W, Haehnel changeS,nephrosis
B, Rosener but not in
et al. Long-term ocal segmental
treatment glomeru-
of rats with FGF-
http://www.ncbi.nlm.nih.gov/pubmed/19092119
syndrome. Nat Genet. 2000;24:349–354. 2losclerosis.
results in Jfocal
Am Soc Nephrol.glomerulosclerosis.
segmental 2000;11(3):403–412. Kidney Int. 1995;48:1435–1450.
29. Li
37. Smeets B, Uhlig S, V
C, Ruotsalainen Fuss A, et al. Tracing
, Tryggvason K, et al.the originisoexpressed
CD2AP glomerular withextracapil-
nephrin 52. Bulger
66. Kerjaschki D, Sharkey
R, Eknoyan D, Farquhar
G, Purcell D, et M.al. Identif
Endothelialcationcharacteristics
and characterization
of glo-
lary
in lesions rom
developing parietal and
podocytes epithelial
is foundcells.widely
J Am Soc Nephrol.kidney
in mature 2009;20:2604–2615.
and elsewhere. o podocalyxin-the
merular capillaries major sialoprotein
in normal, mercuric o the renal glomerularand
chloride-induced, epithelial cell. J
gentamicin-
Am 30.J Physiol
Rodewald RenalR,Physiol.
Karnovsky M. Porous substructure o the glomerular slit dia-
2000;279(4):F785–F792. Cell Biol. acute
induced 1984;98:1591.
renal failure in the rat. J Clin Invest. 1983;72:128–141.
phragm
38. Huber in the T, rat and mouse.
Kottgen J Cell Biol.
M, Schilling B, et1974;60:423–433.
al. Interaction with podocin facilitates http://www.ncbi.nlm.nih.gov/pubmed/6371025
67. Elger M, Sakai T, Kriz W. The vascular pole of the renal glomerulus of rat.
http://www.ncbi.nlm.nih.gov/pubmed/4204974
nephrin signaling. J Biol Chem. 2001;276:41543–41546. Adv53.Anat
Orlando
Embryol R, Cell
TakedaBiol.T,1998;139:1–98.
Zak B, et al. The glomerular epithelial cell anti-adhe-
31. Schwarz
39. Reiser J, Kriz W, Kretzler
K, Simons M, et al.
M, Reiser J, etTheal.glomerular
Podocin, aslit diaphragm iscompo-
raft-associated a modi- sion
68. podocalyxin
Dobyan D, Eknoyan associatesG, with the L,
Magill actin cytoskeleton
et al. A quantitative through interactions
evaluation of the with
glo-
f ed adherens
nent junction.slit
of the glomerular J Am Soc Nephrol.
diaphragm, 2000;11:1–8.
interacts with CD2AP and nephrin. J Clin ezrin. J Am
merular Soc Nephrol.
capillary 2001;12:1589–1598.
endothelium in rat and human kidneys: Utilization of vascular
http://www.ncbi.nlm.nih.gov/pubmed/10616834
Invest. 2001;108:1621–1629. http://www.ncbi.nlm.nih.gov/pubmed/11461930
perfusion, freeze-cracking of tissue, and scanning electron microscopy. J Electron
32. Dunne
40. Ruotsalainen
J, Hanby V, A,
Ljungberg
PoulsomP,R,Wartiovaara
et al. Molecular J, et cloning
al. Nephrin is specif
and tissue cally
expres- 54. Michael
Microsc A, Blau E, Vernier R. Glomerular polyanion. Alterations in amino-
Tech. 1984;1:185.
located
sion at the
of FAT, theslit diaphragm
human homologue o glomerular podocytes.
of the Drosophila fatProc
geneNatl
thatAcad Sci USA.
is located on nucleoside
69. Savage nephrosis.
C. The biology Lab Invest.of 1970;23:649–657.
the glomerulus: Endothelial cells. Kidney Int.
1999;96(14):7962–7967.
chromosome 4q34-q35 and encodes a putative adhesion molecule. Genomics. http://www.ncbi.nlm.nih.gov/pubmed/4098608
1994;45:314–319.
http://www.ncbi.nlm.nih.gov/pubmed/10393930
1995;30:207–223. 55. Kanwar
70. AndrewsY,PLinker . Glomerular epithelial
A, Farquhar alterationspermeability
M. Increased resulting rom sialic
of the acid sur-
glomerular
33. Ciani
41. Donoviel D, Freed
L, Patel D, Vogel
A, Allen N, et H, al.
et al.
MiceProteinuria
lacking the and giant
perinatal lethality in
protocadherin ace coat removal.
basement membrane Kidney Int. 1979;15:376–385.
to ferritin after removal of glycosaminoglycans (heparan
mice lacking
mFAT1 exhibitNEPH1,
renal slit a novel
junction protein with homology
abnormalities to nephrin.
and a partially Mol Cell
penetrant Biol.
cyclo- http://www.ncbi.nlm.nih.gov/pubmed/513496
sulfate) by enzyme digestion. J Cell Biol. 1980;86:688–693.
2001;21:4829–4836.
pia and anophthalmia phenotype. Mol Cell Biol. 2003;23:3575–3582. 56. Latta
71. SeilerH,M,Johnston
Rennke W, H, Stanley
Venkatachalam M, et al. Pathogenesis
T. Sialoglycoproteins and ltration o polycation-
barriers in
34. Miner
42. Inoue T, J. Y aoita E,
Renal Kuriharamembrane
basement H, et al. Fat is a component
components. Kidneyo glomerular
Int. 1999;56: slit induced
the alteration
glomerular ( usion)
capillary o glomerular
wall. J Ultrastructepithelium. Lab Invest. 1977;36:48–61.
Res. 1975;51:354–376.
diaphragms. Kidney Int. 2001;59:1003–1012.
2016–2024. http://www.ncbi.nlm.nih.gov/pubmed/63647
72. Mohos S, Skoza L. Histochemical demonstration and localization of sialo-
http://www.ncbi.nlm.nih.gov/pubmed/11231355
43. Drenckhahn D, Franke R. Ultrastructural organization of contractile and 57. Kerjaschki
proteins D, VernilloExp
in the glomerulus. A, Farquhar
Mol Pathol. M.1970;12:316–323.
Reduced sialylation o podocalyxin-
35. Schnabel
cytoskeletal E, Anderson
proteins in glomerular J, Farquhar
podocytesM. The tight junction
of chicken, rat, andprotein
man.ZO-1Lab the
73.major sialoprotein
Rostgaard J, Qvortrup o theK.rat Sievekidney
plugsglomerulusin
in fenestrae of aminonucleotide neph-
glomerular capillaries
is concentrated
Invest. along slit diaphragms o the glomerular epithelium. J Cell Biol.
1988;59:673–682. -rons. Amthe
site of J Pathol.
ltration 1985;118:343.
barrier? Cells Tissues Organs. 2002;170:132–138.
1990;111:1255–1263.
44. Sanden W, Elger M, Mundel P, et al. The architecture of podocyte cytoskel- http://www.ncbi.nlm.nih.gov/pubmed/3976840
74. Hjalmarsson C, Johansson B, Haraldsson B. Electron microscopic evaluation of
http://www.ncbi.nlm.nih.gov/pubmed/2202736
eton suggests a role in glomerular ltration dynamics. Ann Anat. 1995;177:44–45. the58.endothelial
Takeda T,surfaceMcQuistanlayer ofT,glomerular
Orlando R,capillaries.
et al. LossMicrovasc
o glomerular oot processes
Res. 2004;67:9–17.
36. Kaplan
45. Boute N, Gribouval
J, Kim S, NorthO,K,Roselli S, et al. NPHS2,
et al. Mutations in ACTN4, encoding
encoding the glomerular
-actinin-4, is75.
associated
Ballermannwith B, uncoupling
Marsden Po. Endothelium-derived
podocalyxin rom thevasoactive actin cytoskeleton.
mediatorsJ andClin
proteinfamilial
cause podocin, focalissegmental
mutated in autosomal recessive
glomerulosclerosis. Nat steroid-resistant
Genet. 2000;24:251–256. nephrotic Invest.glomerular
renal 2001;108:289–301.function. J Clin Invest. 1991;14:508–517.
syndrome.
46. WeinsNat A, Genet.
Kenlan2000;24:349–354.
P, Herbert S, et al. Mutational and biological analysis 59. Eremina
76. Pavenstadt H, Kriz W,
V, Quaggin S. Kretzler
The role M. Cell biology
of VEGF-A o the glomerular
in glomerular podocyte.
development and
http://www.ncbi.nlm.nih.gov/pubmed/10742096
of alpha-actinin-4 in focal segmental glomerulosclerosis. J Am Soc Nephrol. Physiol Rev.
function. 2003;83:253–307.
Curr Opin Nephrol Hypertens. 2004;13:9–15.
37. Li C, Ruotsalainen V, Tryggvason K, et al. CD2AP is expressed with ne-
2005;16:3694–3701. http://www.ncbi.nlm.nih.gov/pubmed/12506131
77. Satchell S, Anderson K, Mathieson P. Angiopoietin 1 and vascular endo-
phrin in developing
47. Braun E, Schlondorffpodocytes and isD,ound
D, Becker widely in mature
et al. Mutations kidneygene
in the formin andINF2
else- 60. Reiser
thelial growthJ,factorPolu modulate
K, Moller humanC, et al.glomerular
TRPC6 iscell a glomerular slit diaphragm-
barrier properties. Am Soc
where.focal
cause Am segmental
J Physiol Renal Physiol. 2000;279(4):F785–F792.
glomeruloscerlosis. Nat Genet. 2010;42:72–76. associated
Nephrol. channel required or normal renal unction. Nat Genet. 2005;37:
2004;15:566–574.
38. Kreidberg
48. Huber T, KottgenJ, Symons M, Schilling
J. Integrins B, etinal.kidney
Interaction with podocin
development, acilitates
function, and 739–774.
78. Grandaliano G, Choudhury G, Poptic E, et al. Thrombin regulates PDGF ex-
nephrin Am
disease. signaling.
J Physiol J Biol
Renal Chem.
Physiol.2001;276:41543–41546.
2000;279(2):F233–F242. http://www.ncbi.nlm.nih.gov/pubmed/15924139
pression in bovine glomerular endothelilal cells. J Am Soc Nephrol. 1998;9:583–589.
http://www.ncbi.nlm.nih.gov/pubmed/11562357
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 441
1

61. Jorgensen
79. Kazatchkine M, Fearon
F, Bentzon M. D, TheAppay M, et al. Immunohistochemical
ultrastructure of the normal human glomeru- study o 84. Schwartz
111. St John P,E,Abrahamson
Gold scher D. Glomerular endothelial
S, Coltoff-Schiller B, et al. cells and podocytes
Extracellular matrix
the human
lus: Thicknessglomerular
of glomerular C3b receptor
basementinmembrane.
normal kidney and in 1968;18:42–48.
Lab Invest. seventy-f ve cases jointly
micro synthesize laminin-1ofand
brils are composed core-11 chains.coated
proteins KidneywithInt. 2001;60:1037–1046.
bronectin. J Histochem
o80.renal diseases.
Trump J Clin R.
B, Bulger Invest. 1982;69:900–912.
Morphology of the kidney. In: Becker E, ed. Structural 85. Abrahamson
Cytochem. D. Origin o the glomerular basement membrane visu-
1985;33:268–274.
http://www.ncbi.nlm.nih.gov/pubmed/7042757
Basis of Renal Disease. New York: Harper&Row; 1968:1–92. alized a ter inM,
112. Gibson vivo labeling o J,laminin
Kumaratilake Cleary inE. newborn
The protein rat kidneys.
components J Cellof Biol.
the
62. Abrahamson
81. Kerjaschki D,D. Farquhar
Structure M. and
Immunocytochemical
development of the localization
glomerular o capillary
the Hey- 1985;100:1988–2000.
12–nanometer micro brils of elastic and nonelastic tissues. J Biol Chem.
mannand
wall antigen
basement(gp 330) in glomerular
membrane. epithelial
Am J Physiol. cells o normal Lewis rats. J Exp
1987;253:F783–F794. 86. Daha M, Blok A, de Grae J, et al. Synthesis and degradation o glomeru-
1989;264:4590–4598.
Med.
82. 1983;157:667–686.
Miner J. Developmental biology of glomerular basement membrane com- lar
113.basement
Sterzel R, membrane
Hartner in A, rats with nephrotoxicU,nephritis.
Schlotzer-Schrehard Nephron.
et al. Elastic ber 1978;22:
proteins
http://www.ncbi.nlm.nih.gov/pubmed/6337231
ponents. Curr Opin Nephrol Hypertens. 1998;7:13–19. 522–528.
in the glomerular mesangium in vivo and in cell culture. Kidney Int. 2000;58:
63. Sariola
83. Schi erH,M,Timpl Schi R, er von
L, Gupta A, etK,
der Mark al.etInhibitory smadsofand
al. Dual origin TGF-b signal-
glomerular base- http://www.ncbi.nlm.nih.gov/pubmed/740113
1588–1602.
ling inmembrane.
ment glomerularDev cells. J Am
Biol. Soc Nephrol. 2002;13:2657–2666.
1984;101:86–96. 87. Farquhar
114. Cohen M,M, Surma
Palade M,G.Wu V. In vivoevidence
Functional biosynthesisfor the andexistence
turnoverofoa glomeru-
third cell
http://www.ncbi.nlm.nih.gov/pubmed/12397035
84. St John P, Abrahamson D. Glomerular endothelial cells and podocytes lar
typebasement
in the renalmembrane
glomerulus.in diabetic rats. AmofJ Physiol.
Phagocytosis ltration1982;242:F385–F389.
residues by a distinctive
64. Wogensen
jointly synthesizeL, Nielsen C,
laminin-1 andHjorth P. Under
-11 chains. control
Kidney o the Ren-1c promoter,
Int. 2001;60:1037–1046. 88. Reddi
“third” cell. JA.
CellMetabolism o glomerular basement membrane in normal, hy-
Biol. 1962;13:55–87.
locally produced trans
85. Abrahamson orming
D. Origin of growth actor-beta1
the glomerular induced
basement accumulation
membrane visualized o after
glo- pophysectomized,
115. Mundel P, Elger and M, growth-hormone-treated
Sakai T, et al. Micro diabetic brils are rats. Exp component
a major Mol Pathol.
merular
in extracellular
vivo labeling of laminin matrix in transgenic
in newborn mice. Diabetes.
rat kidneys. J Cell Biol.1999;48:182–192.
1985;100:1988–2000. 1985;43:196–208.
of the mesangial matrix in the glomerulus of the rat kidney. Cell Tissue Res.
http://www.ncbi.nlm.nih.gov/pubmed/9892241
86. Daha M, Blok A, de Graeff J, et al. Synthesis and degradation of glomerular 89. Bernstein J, Cheng F, Roszka B. Glomerular di erentiation in metanephric
1988;254:183–187.
65. Krizmembrane
basement W, Haehnel B, Rosener
in rats S, et al. Long-term
with nephrotoxic nephritis. treatment o rats with FGF-
Nephron. 1978;22:522–528. culture.
116. Inoue Lab S,Invest.
Leblond1981;45:183–190.
C. The micro brils of connective tissue: I. Ultrastructure.
287.
results in ocal
Cohen segmental
M, Surma M, Wu glomerulosclerosis.
V. In vivo biosynthesis Kidneyand Int.turnover
1995;48:1435–1450.
of glomerular http://www.ncbi.nlm.nih.gov/pubmed/7265915
Am J Anat. 1986;176:121–138.
http://www.ncbi.nlm.nih.gov/pubmed/8544400
basement membrane in diabetic rats. Am J Physiol. 1982;242:F385–F389. 90. Latta
117. Bonadio J, Sage H, Cheng
H, Maunsbach F, et al.S.Localization
A, Madden The centrolobularo collagen types
region of IV
theand
renal V,
66. Reddi
88. Bulger A. R, Eknoyan
Metabolism G, Purcell D, et al. basement
of glomerular Endothelialmembranecharacteristics o glo-
in normal, laminin,
glomerulus andstudied
heparanbysul ate proteoglycan
electron microscopy. to Jthe basal lamina
Ultrastruct o kidney epithelial
Res. 1960;4:455–472.
merular capillaries inand
hypophysectomized, normal, mercuric chloride-induced,
growth-hormone-treated diabetic rats.and Expgentamicin-in-
Mol Pathol. cells
118. inSchlondorff
transf lter metanephric
D. The glomerular culture. mesangial
Am J Pathol.cell: 1984;116:289–296.
an expanding role for a
duced acute renal ailure in the rat. J Clin Invest. 1983;72:128–141.
1985;43:196–208. http://www.ncbi.nlm.nih.gov/pubmed/6235751
specialized pericyte. FASEB J. 1987;1:272–281.
http://www.ncbi.nlm.nih.gov/pubmed/6223941
89. Bernstein J, Cheng F, Roszka B. Glomerular differentiation in metanephric 91. Mauer
119. Lee L, Pollock
S, Fish A, Lovett Blau E,D.etAsymmetric origin o mesangium.
al. The glomerular the mature glomerular
I. Kinetic
67. Elger
culture. Lab M, Sakai
Invest. T, Kriz W. The vascular pole o the renal glomerulus o rat.
1981;45:183–190. basement
studies ofmembrane.
macromolecular J Cell Physiol.
uptake 1993;157:169–177.
in normal and nephrotic rats. J Clin Invest.
Adv
90.Anat Embryol
Bonadio Cell H,
J, Sage Biol.Cheng
1998;139:1–98.
F, et al. Localization of collagen types IV and V, http://www.ncbi.nlm.nih.gov/pubmed/8408235
1972;51:1092–1101.
http://www.ncbi.nlm.nih.gov/pubmed/9386305
laminin, and heparan sulfate proteoglycan to the basal lamina of kidney epithe- 92. Schreiner
120. Davies M, G, Thomas
Unanue G, Shrewing
E. OriginL,ofetthe al. Mesangial
rat mesangial cell proteoglycans:
phagocyte andsyn- its
68.cells
lial Dobyan
in trans D, lter
Eknoyan G, Magill
metanephric L, et al.Am
culture. A quantitative evaluation o the glo-
J Pathol. 1984;116:289–296. thesis and metabolism.
expression of the leukocyte J Amcommon
Soc Nephrol. 1992;2(10
antigen. Suppl):S88–94.
Lab Invest. 1984;51:515–523.
merular
91. Leecapillary
L, Pollock endothelium
A, Lovett in D.rat and humanorigin
Asymmetric kidneys:of theUtilization o vascular
mature glomerular 93. Kriz
121. Martin W,J, Elger
Steadman R, Knowlden
M, Mundel P, et J,al.et Structure-stabilizing
al. Di erential regulation forceso in
matrix
the
per usion,membrane.
basement reeze-cracking J CelloPhysiol.
tissue, 1993;157:169–177.
and scanning electron microscopy. J Electron melalloproteinases
glomerular tuft. J Am asnSoc their inhibitors
Nephrol. in human glomerular epithelial cells in
1995;5:1731–1739.
Microsc Tech. 1984;1:185.
92. Davies M, Thomas G, Shrewing L, et al. Mesangial cell proteoglycans: vitro.
122. JKriz
Am Soc W, Nephrol.
Haehnel 1998;9:1629–1637.
B, Hosser H, et al. Pathways to recovery and loss of
69. Savage
synthesis C. The biology
and metabolism. J Amo Soc theNephrol.
glomerulus: Endothelial
1992;2(10 cells. Kidney Int.
Suppl):S88–94. 94. Martin
nephrons J, Eynstonenephritis.
in anti-Thy-1 L, DaviesJ Am M, etSocal.Nephrol.
Induction o metalloproteinases by
2003;14:1904–1926.
1994;45:314–319.
93. Martin J, Steadman R, Knowlden J, et al. Differential regulation of matrix glomerular
123. Welling mesangial
L, Zupka cellsM,stimulated
Welling by D. proteins
Mechanical o the extracellular
properties matrix. J
of basement
http://www.ncbi.nlm.nih.gov/pubmed/8164414
melalloproteinases asn their inhibitors in human glomerular epithelial cells in Am Soc Nephrol.
membrane. News2001;12:88–96.
Physiol Sci. 1995;10 (1):30–35.
70. JKanwar
vitro. Am Soc Y , Linker 1998;9:1629–1637.
Nephrol. A, Farquhar M. Increased permeability o the glomerular http://www.ncbi.nlm.nih.gov/pubmed/12374147
124. Kriz W, Mundel P, Elger M. The contractile apparatus of podocytes is
basement
94. Martin membrane
J, Eynstone to L, erritin
Davies a ter
M, removal o glycosaminoglycans
et al. Induction of metalloproteinases(heparanby 95. Timpl
arranged R, Brown J.GBM
to counteract Supramolecular
expansion. Contrib assembly o basement
Nephrol. membranes. Bi-
1994;107:1–9.
sul ate) by enzyme
glomerular mesangial digestion. J Cell Biol.by
cells stimulated 1980;86:688–693.
proteins of the extracellular matrix. oEssays. 1996;18 M,
125. Drumond (2):123–132.
Deen W. Structural determinants of glomerular hydraulic
Jhttp://www.ncbi.nlm.nih.gov/pubmed/6447156
Am Soc Nephrol. 2001;12:88–96. http://www.ncbi.nlm.nih.gov/pubmed/8851045
permeability. Am J Physiol. 1994;266:F1–F12.
71. Timpl
95. Latta H,R,Johnston
Brown J.WSupramolecular
Stanley T. Sialoglycoproteins
assembly of and f ltration
basement barriers in
membranes. 96. Kiberd
126. Gro enB.A,The Ruegg M, Dijkman
functional H, et al.
and structural Agrin of
changes is the
a major heparan
glomerulus sul ate
through-
the glomerular
BioEssays. 1996;18 capillary wall. J Ultrastruct Res. 1975;51:354–376.
(2):123–132. proteoglycan
out the courseinofthe human
murine glomerular
lupus nephritis.basement
J Am Soc membrane. J Histochem Cyto-
Nephrol. 1992;3:930–939.
http://www.ncbi.nlm.nih.gov/pubmed/48557
96. Groffen A, Ruegg M, Dijkman H, et al. Agrin is a major heparan sulfate chem.
127. 1998;46:19–27.
Guasch A, Myers B. Determinants of glomerular hypo ltration in
72. Mohos S,inSkoza
proteoglycan L. Histochemical
the human glomerular demonstration and localization
basement membrane. o sialo-
J Histochem http://www.ncbi.nlm.nih.gov/pubmed/9405491
nephrotic patients with minimal change nephropathy. J Am Soc Nephrol. 1994;4:
proteins in1998;46:19–27.
Cytochem. the glomerulus. Exp Mol Pathol. 1970;12:316–323. 97. Hudson B, Reeders S, Tryggvason K. Type IV collagen: structure, gene or-
1571–1581.
73. Hudson
97. RostgaardB,J,ReedersQvortrup S, K. Sieve plugsK.inType
Tryggvason enestrae o glomerular
IV collagen: capillaries
structure, gene ganization
128. Drumond and roleM, in human
Kristal diseases.
B, Myers B, etJ Biol Chem. 1993;268:26033–26036.
al. Structural basis for reduced glomer-
- site o the f and
organization ltration
rolebarrier?
in human Cells Tissues JOrgans.
diseases. Biol Chem.2002;170:132–138.
1993;268:26033–26036. http://www.ncbi.nlm.nih.gov/pubmed/8253711
ular ltration capacity in nephrotic humans. J Clin Invest. 1994;94:1187–1195.
74. Inoue
98. Hjalmarsson C, Johanssonarchitecture
S. Ultrastructural B, Haraldsson B. Electronmembranes.
of basement microscopicContrib
evalu- 98. Barnes
129. Inoue S. Ultrastructural
J, Reznicek M, Radnikarchitecture
R, et al.o Anionization
basement membranes.of an antigen Contrib
pro-
ation o 1994;107:21–28.
Nephrol. the endothelial sur ace layer o glomerular capillaries. Microvasc Res. Nephrol. 1994;107:21–28.
motes glomerular binding and immune complex formation. Kidney Int. 1988;34:
2004;67:9–17.
99. Madri J, Roll F, Furthmayr H, et al. Ultrastructural localization of bro- http://www.ncbi.nlm.nih.gov/pubmed/8004970
156–163.
75. Ballermann
nectin and lamininB,inMarsden the basement P. Endothelium-derived
membranes of the murine vasoactive mediators
kidney. and
J Cell Biol. 99. Simonson
130. Madri J, Roll M, FDunn
, Furthmayr H, et al. Ultrastructural
M. Leukotriene C4 and D4 contract localization o f bro-
rat glomerular
renal glomerular unction. J Clin Invest. 1991;14:508–517.
1980;86:G82–G87. nectin
mesangialand cells.
laminin in the
Kidney Int.basement membranes o the murine kidney. J Cell Biol.
1986;30:524–531.
http://www.ncbi.nlm.nih.gov/pubmed/1794204
100. Timpl R, Dziadek M. Structure, development, and molecular pathology of 1980;86:G82–G87.
131. Savin V. In vitro effects of angiotensin II on glomerular function. Am J
76. Eremina
basement V, Quaggin
membranes. S. The
Int Rev Exprole o VEGF-A
Pathol. in glomerular development and
1986;29:1–112. 100.
Physiol.Timpl R, Dziadek M. Structure, development, and molecular pathology o
1986;251:F627–F634.
unction.
101. Curr Opin
Yurchenco Nephrol Hypertens.
P, Schittny J. Molecular 2004;13:9–15.
architecture of basement membranes. basement membranes.
132. Scharschmidt L, Int Rev Exp
Douglas J, Pathol.
Dunn M. 1986;29:1–112.
Angiotensin II and eicosanoids in
http://www.ncbi.nlm.nih.gov/pubmed/15090854
FASEB J. 1990;4:1577–1590. http://www.ncbi.nlm.nih.gov/pubmed/3536789
the control of glomerular size in the rat and human. Am J Physiol. 1986;250:
77. Colognato
102. Satchell S, H, Anderson
Yurchenco K, Mathieson
P. Form and P. Angiopoietin
function: the 1 and vascular
laminin familyendo-
of 101. Yurchenco P, Schittny J. Molecular architecture o basement membranes.
F348–F356.
thelial growth Dev
heterotrimers. actorDyn.modulate human glomerular cell barrier properties. Am Soc
2000;218:213–234. FASEB J. 1990;4:1577–1590.
133. Blantz R, Konnen K, Tucker B. Angiotensin II effects upon the glo-
Nephrol.
103. 2004;15:566–574.
Miner J, Yurchenco P. Laminin functions in tissue morphogenesis. Annu http://www.ncbi.nlm.nih.gov/pubmed/2180767
merular microcirculation and ultra ltration coef cient of the rat. J Clin Invest.
http://www.ncbi.nlm.nih.gov/pubmed/14978158
Rev Cell Dev Biol. 2004;20:255–284. 102. Colognato H, Yurchenco P. Form and unction: the laminin amily o het-
1976;57:419–434.
78. Kanwar
104. Grandaliano G, Choudhury
Y, Farquhar G, Popticsites
M. Anionic E, etinal. the
Thrombin regulates
glomerular PDGF
basement erotrimers.
134. Dworkin Dev L,Dyn. 2000;218:213–234.
Ichikawa I, Brenner B. Hormonal modulation of glomerular
expression in
membrane: In bovine
vivo andglomerular endothelilal
in vitro localization cells. J Am
to laminae raraeSocbyNephrol.
cationic 1998;9:
probes. http://www.ncbi.nlm.nih.gov/pubmed/10842354
function. Am J Physiol. 1983;244:F95–F104.
J583–589.
Cell Biol. 1979;81:137–153. 103. Miner D,
135. Haley J, YSarra
urchenco an M, P. Laminin
Bulger R,unctions in tissue and
et al. Structural morphogenesis. Annu
functional corre-
http://www.ncbi.nlm.nih.gov/pubmed/9555660
105. Sakai T, Kriz W. The structural relationship between mesangial cells and Rev
latesCell Dev Biol.
of effects of 2004;20:255–284.
angiotensin-induced changes in rat glomerulus. Am J Physiol.
79. Jorgensen
basement membrane F, Bentzon M. The
of the renal ultrastructure
glomerulus. AnatoEmbryol.
the normal human glomeru-
1987;176:373–386. 104. Kanwar Y, Farquhar M. Anionic sites in the glomerular basement mem-
1987;253:F111–F119.
lus: Thickness
106. Drenckhahn o glomerular
D, Schnittler basement membrane.
H, Nobiling R, etLab al.Invest. 1968;18:42–48.
Ultrastructural organi- brane: In vivo G,
136. Olivetti andGiacomelli
in vitro localization
F, Wiener J.toMorphometry
laminae raraeofbysuper cationic
cialprobes. J Cell
glomeruli in
http://www.ncbi.nlm.nih.gov/pubmed/5643970
zation of contractile proteins in rat glomerular mesangial cells. Am J Pathol. Biol.
acute1979;81:137–153.
hypertension in the rat. Kidney Int. 1985;27:31–38.
80. Trump B, Bulger R. Morphology o the kidney. In: Becker E, ed. Structural
1990;137:1343–1351. http://www.ncbi.nlm.nih.gov/pubmed/90048
137. Steinhausen M, Endlich K, Wiegman D. Glomerular blood ow. Kidney Int.
Basis of
107. Renal Disease.
Becker New York: of
C. Demonstration Harper&Row;
actomyosin 1968:1–92.
in mesangial cells of the renal 105. Sakai T, Kriz W. The structural relationship between mesangial cells and
1990;38:769–784.
http://www.ncbi.nlm.nih.gov/pubmed/5702212
glomerulus. Am J Pathol. 1972;66:97–110. basement
138. Farquhar membraneM. The o the renal glomerulus.
glomerular basement Anat Embryol.a 1987;176:373–386.
membrane: selective macromo-
81. Kriz
108. Abrahamson
W, Elger M, D. Lemley
Structure K, and
et al.development
Structure of the o the glomerular
glomerular capillary
mesangium: http://www.ncbi.nlm.nih.gov/pubmed/3631536
lecular lter. In: Hay E, ed. Cell Biology of Extracellular Matrix. New York: Plenum
Awall and basement
biomechanical membrane. Am
interpretation. J Physiol.
Kidney 1987;253:F783–F794.
Int. 1990;38:S2–S9. 106.
Press; Drenckhahn
1991:365–418. D, Schnittler H, Nobiling R, et al. Ultrastructural organi-
82. Courtoy
109. Miner J. Developmental
P, Timpl R, Farquhar biologyM. o glomerular
Comparative basement membrane
distribution com-
of laminin, zation o contractile
139. Daniels B. The roleproteins
of theinglomerular
rat glomerular mesangial
epithelial cells.
cell in the Am J Pathol.
maintenance of
ponents.
type Curr Opinand
IV collagen, Nephrol Hypertens.
bronectin 1998;7:13–19.
in the rat glomerulus. J Histochem Cytochem. 1990;137:1343–1351.
the glomerular ltration barrier. Am J Nephrol. 1993;13:318–323.
http://www.ncbi.nlm.nih.gov/pubmed/9442357
1982;30:874. http://www.ncbi.nlm.nih.gov/pubmed/2260624
140. Daniels B, Deen W, Mayer G, et al. Glomerular permeability barrier in the
83. Martinez-Hernandez
110. Sariola H, Timpl R, von der Mark
A, Chung A. K,
Theet al. Dual origin olocalization
ultrastructural glomerularofbase-two 107. Becker
rat. J Clin C. 1993;92:929–936.
Invest. Demonstration o actomyosin in mesangial cells o the renal
ment membrane.
basement membrane Devcomponents
Biol. 1984;101:86–96.
entactin and laminin - in rat tissue. J Histochem glomerulus.
141. Farquhar AmM, J Pathol.
Wissig1972;66:97–110.
S, Palade G. Glomerular permeability: I.Ferritim trans-
http://www.ncbi.nlm.nih.gov/pubmed/6420214
Cytochem. 1984;32:289. http://www.ncbi.nlm.nih.gov/pubmed/4621555
fer across the normal glomerular capillary wall. J Exp Med. 1961;113:47–66.
42
42 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

108. Chang
142. Kriz W,R,ElgerDeenM, W,Lemley
Robertson K, et
C,al. Structure
et al. o the glomerular
Permselectivity mesangium:
of the glomerular cap- 132. Burckhardt
172. Scharschmidt B, L,Burckhardt
Douglas J, G. Dunn M. Angiotensin
Transport of organic II and
anionseicosanoids
across thein
A biomechanical
illary interpretation.
wall: III. Restricted transportKidney Int. 1990;38:S2–S9.
of polyanions. Kidney Int. 1975;8:212–218. the control membrane
basolateral o glomerular size in the
of proximal rat and
tubule cells.human.
Rev PhysiolAm JBiochem
Physiol. Pharmacol.
1986;250:
109. Rennke
143. CourtoyH, P, Timpl
CotranR,R,Farquhar
VenkatachalamM. Comparative
M. Role of distribution
molecular ochargelaminin,in F348–F356.
2003;146:95–158.
type IV collagen,
glomerular and f bronectin
permeability. in the rat
Tracer studies withglomerulus.
cationized Jferritins.
HistochemJ Cytochem.
Cell Biol. 133. Lee
173. Blantz R, Konnen
W, Kim K, Tucker
R. Transporters and B. renalAngiotensin II e ects
drug elimination. Annuupon the glo-
Rev Pharma-
1982;30:874.
1975;67:638–646. merular
col Toxicol. microcirculation
2004;44:137–166. and ultraf ltration coe f cient o the rat. J Clin Invest.
http://www.ncbi.nlm.nih.gov/pubmed/7130672
144. Deen W, Bohrer M, Brenner B. Macromolecule transport across glomerular 1976;57:419–434.
174. Wright S, Dantzler W. Molecular and cellular physiology of renal organic
110. Martinez-Hernandez
capillaries: Application of pore A, Chung
theory. A. The Int.
Kidney ultrastructural localization o two
1979;16:353–365. http://www.ncbi.nlm.nih.gov/pubmed/1254727
cation and anion transport. Physiol Rev. 2004;84:987–1049.
basement
145. Burne membrane
M, Osicka components
T, Comperentactin and laminin
W. Fractional - in ratoftissue.
clearance high Jmolecular
Histochem 134. Lock
175. Dworkin E, ReedL, Ichikawa I, Brenner
C. Xenobiotic B. Hormonal
metabolizing modulation
enzymes o glomerular
of the kidney. Toxicol
Cytochem.
weight 1984;32:289.
proteins in conscious rats using a continuous infusion method. Kidney unction.
Pathol. Am J Physiol. 1983;244:F95–F104.
1998;26:18–25.
http://www.ncbi.nlm.nih.gov/pubmed/6198358
Int. 1999;55:261–270. 135. Christensen
176. Haley D, Sarraf an M, Bulger
E, Gburek R, et reabsorption
J. Protein al. Structuralinand renalunctional
proximal corre-
tu-
111. Greive
146. Schwartz K, E, Goldf scher S, D,
Nikolic-Paterson Colto -SchillerM,B, etet al.
Guimaraes al. Glomerular
Extracellularpermse-
matrix lates o- function
bule e ects o and angiotensin-induced
dysfunction in kidney changes in rat glomerulus.
pathophysiology. Am JNephrol.
Pediatr Physiol.
microf brils
lectivity are composed
factors o core proteins
are not responsible for thecoated
increasewithin ffractional
bronectin.clearance
J Histochem of 1987;253:F111–F119.
2004;19:714–721.
Cytochem.in1985;33:268–274.
albumin rat glomerulonephritis. Am J Pathol. 2001;159:1159–1170. 136. V
177. Olivetti
erroust G, Giacomelli F,E.Wiener
P, Christensen Megalin J. Morphometry
and cubilin - the o superf
story of cial
twoglomeruli
multipur-in
http://www.ncbi.nlm.nih.gov/pubmed/3980980
147. Smithies O. Why the kidney glomerulus does not clog: a gel permeation/diffu- acutereceptors
pose hypertension in the
unfolds. rat. Kidney
Nephrol Int. 1985;27:31–38.
Dial Transplant. 2002;17:1867–1871.
112.hypothesis
sion Gibson M, Kumaratilake
of renal function. Proc J, Cleary
Natl Acad E. Sci
The protein
USA. components o the
2003;100:4108–4113. http://www.ncbi.nlm.nih.gov/pubmed/3981871
178. Bacic D, Wagner C, Hernando N, et al. Novel aspects in regulated expres-
12-nanometer
148. Hausmannmicrof brils C,
R, Kuppe o Egger
elasticH,and et al.nonelastic
Electrical tissues. J Biol Chem.
forces determine glo- 137. ofSteinhausen
sion the renal type M, IIa
Endlich K, Wiegman D. Glomerular
Na/Pi-contransporter. Kidney Int. blood ow. Kidney Int.
2004;91:S5–S12.
1989;264:4590–4598.
merular permeability. J Am Soc Nephrol. 2010;21:2053–2058. 1990;38:769–784.
179. Bachmann S, Schlichting U, Geist B, et al. Kidney-speci c inactivation
http://www.ncbi.nlm.nih.gov/pubmed/2647740
149. Tryggvason K, Pettersson E. Causes and consequences of proteinuria: http://www.ncbi.nlm.nih.gov/pubmed/2266660
of the megalin gene impairs traf cking of renal inorganic sodium phosphate
113.kidney
the Sterzelltration
R, HartnerbarrierA, Schlotzer-Schrehard
and progressive renalU,failure. et al. Elastic
J Internf ber
Med.proteins
2003; 138. Farquhar
cotransporter M. The glomerular
(NaPi-IIa). J Am Soc Nephrol basement membrane: a selective macromo-
. 2004;15:892–900.
in the glomerular mesangium in vivo and in cell culture. Kidney Int. 2000;58:
254:216–224. lecularKriz
180. f lter.
W,In: Hay E, ed. Cell
Schnermann Biology ofH.Extracellular
J, Dieterich Differences Matrix. New York: Plenum
in the morphology of de-
1588–1602.
150. Bolton G, Deen W, Daniels B. Assessment of the charge selectivity of Press; 1991:365–418.
scending limbs of short and long loops of Henle in the rat kidney. In: Wirz H,
http://www.ncbi.nlm.nih.gov/pubmed/11012893
glomerular basement membrane using Ficoll sulfate. Am J Physiol. 1998;274: 139. Daniels
Spinelli F, eds. B. The Advances
Recent role o the in glomerular
Renal Physiology. epithelial
Basel:cell in the1972:140–144.
Karger; maintenance o
114. Farquhar M, Palade G. Functional evidence or the existence o a third cell
F889–F896. the glomerular
181. Schwartz fM, ltration barrier. Am M.
Venkatachalam J Nephrol.
Structural1993;13:318–323.
differences in thin limbs of
type inSorensson
151. the renal J, glomerulus.
Ohlson M,Phagocytosis
Haraldssono B.f ltration residues analysis
A quantitative by a distinctive
of the http://www.ncbi.nlm.nih.gov/pubmed/8116684
Henle: Physiological implications. Kidney Int. 1974;6:193–208.
“third” cell. Jcharge
glomerular Cell Biol. 1962;13:55–87.
barrier in the rat. Am J Physiol Renal Physiol. 2001;280: 140. Dieterich
182. Daniels B,H, Deen W, Mayer
Barrett J, KrizG,W,et etal.al.Glomerular permeability
The ultrastructure barrier
of the thininloop
the
http://www.ncbi.nlm.nih.gov/pubmed/19866600
F646–F656. rat. J Clin
limbs of the Invest.
mouse 1993;92:929–936.
kidney. Anat Embryol. 1975;147:1–18.
115. Ciarimboli
152. Mundel P, G, Elger M, Sakai C,
Hjalmarsson T, Bokenkamp
et al. MicrofA,brils et al.are a majoralterations
Dynamic component of http://www.ncbi.nlm.nih.gov/pubmed/7688767
183. Bachmann S, Kriz W. Histotopography an ultrastructure of the thin limbs
o the mesangial
glomerular charge matrix
densityinin the xedglomerulus
rat kidneyso suggest
the ratinvolvement
kidney. CellofTissue
endothe-Res. 141.
of Farquhar
the loop of HenleM, in Wissig S, Palade
the hamster. CellG. Glomerular
Tissue permeability: I.Ferritim
Res. 1982;225:111–127.
1988;254:183–187.
lial coat. Am J Physiol Renal Physiol. 2003;285:F722–F730. trans er
184. acrossJ,the
Barrett Kriz normal glomerular
W, Kaissling B, etcapillary wall. J Exp Med.of1961;113:47–66.
al. The ultrastructure the nephrons of
http://www.ncbi.nlm.nih.gov/pubmed/2461802
153. Ryan G, Karnovsky M. Distribution of endogenous albumin in the rat http://www.ncbi.nlm.nih.gov/pubmed/13698249
the desert rodent (Psammomys obesus) kidney. I. thin limbs of Henle of short
116. Inoue Role
glomerulus. S, Leblond C. The microf
of hemodynamic brilsino glomerular
factors connectivebarrier
tissue: function.
I. Ultrastructure.
Kidney 142. Chang
looped nephrons. R, DeenAm JW, Robertson
Anat. C, et al. Permselectivity o the glomerular
1978;151:487–498.
Am 1976;9:36–45.
Int. J Anat. 1986;176:121–138. cap illary
185. wall:J,III.
Barrett KrizRestricted transport
W, Kaissling B, etoal.polyanions. Kidney Int.
The ultrastructure of1975;8:212–218.
the nephrons of
http://www.ncbi.nlm.nih.gov/pubmed/2943153
154. Schonheyder H, Maunsbach A. Ultrastructure of a specialized neck region http://www.ncbi.nlm.nih.gov/pubmed/1202253
the desert rodent (Psammomys obesus) kidney. II. Thin limbs of Henle of long-
117.
in Latta H,
the rabbit Maunsbach
nephron. Kidney A,Int.
Madden S. The centrolobular region o the renal
1975;7:145–153. 143. Rennke
looped nephrons. H, Am Cotran
J Anat.R, 1978;151:499–514.
Venkatachalam M. Role o molecular charge in
glomerulus
155. Jacobsen studied by electron
N, Jorgensen F. microscopy.
Further enzyme J Ultrastruct Res. 1960;4:455–472.
histochemical observations on glomerular
186. Dobyan permeability.
D, Jamison Tracer studies
R. Structure andwith cationized
function of the erritins. J CellSemin
renal papilla. Biol.
http://www.ncbi.nlm.nih.gov/pubmed/13759385
the segmentation of the proximal tubules in the kidney of the male rat. Histoche- 1975;67:638–646.
Nephrol. 1984;4:5.
118.1973;34:11–32.
mie. Schlondor D. The glomerular mesangial cell: an expanding role or a spe- http://www.ncbi.nlm.nih.gov/pubmed/1202017
187. Kriz W. Structural organization of the renal medulla: Comparative and
cialized
156. pericyte. FASEB
Maunsbach J. 1987;1:272–281.
A. Observations on the segmentation of the proximal tubule in 144. Deenaspects.
functional W, Bohrer AmM, Brenner
J Physiol. B. Macromolecule transport across glomerular
1981;241:R3–R16.
http://www.ncbi.nlm.nih.gov/pubmed/3308611
the rat kidney. J Ultrastruct Res. 1966;16:239–258. capillaries:
188. ChouApplication
C, Nielsen S, o Knepper
pore theory. M. Kidney Int. 1979;16:353–365.
Structural-functional correlation in chin-
119. Maunsbach
157. Mauer S, Fish A, Blau E, etofal.theThe
A. Ultrastructure glomerular
proximal tubule. mesangium.
In: Orloff J,I.Berliner
Kinetic http://www.ncbi.nlm.nih.gov/pubmed/529682
chilla long loop of Henle thin limbs: a novel papillary subsegment. Am J Physiol.
studies
R, eds. oHandbook
macromolecular uptakevol.8.
of Physiology, in normal and nephrotic
Washington: American rats. Physiological
J Clin Invest. 145. Burne M, Osicka T, Comper W. Fractional clearance o high molecular
1993;265:F863–F874.
1972;51:1092–1101.
Society; 1973:31–79. weightPannabecker
189. proteins in conscious
T, Abbott ratsD,using a continuous
Dantzler in usion method. functional
W. Three-dimensional Kidney Int.
120. Kaissling
158. Schreiner B, G, Kriz
Unanue E. Origin oanalysis
W. Structural the rat of mesangial
the rabbit phagocyte
kidney. andAdvitsAnat
ex- 1999;55:261–270.
reconstruction of inner medullary thin limbs of Henle’s loop. Am J Physiol Renal
pression Cell
Embryol o the leukocyte
Biol. common antigen. Lab Invest. 1984;51:515–523.
1979;56:1–123. http://www.ncbi.nlm.nih.gov/pubmed/9893135
Physiol. 2004;286:F38–F45.
http://www.ncbi.nlm.nih.gov/pubmed/6238206
159. Kriz W, Koepsell H. The structural organization of the mouse kidney. 146. Pannabecker
190. Greive K, Nikolic-Paterson
T, Dahmann A, D, Brokl
Guimaraes O, etM,al.etMixed
al. Glomerular
descending- permse-
and
121.
Z AnatKriz W, Elger M, Mundel
Entwicklungsgesch. P, et al. Structure-stabilizing orces in the glo-
1974;144:137–163. lectivity actors thin
ascending-type are not responsible
limbs of Henle’sorloop the increase
in mammalianin ractional
renal clearance o al-
inner medulla.
merular
160. tu t.C.
Tisher J Am Soc Nephrol.
Relationship 1995;5:1731–1739.
between renal structure and concentrating ability in bumin
Am in ratRenal
J Physiol glomerulonephritis. Am J Pathol. 2001;159:1159–1170.
Physiol. 2000;278:F202–F208.
http://www.ncbi.nlm.nih.gov/pubmed/7787139
the rhesus monkey. Am J Physiol. 1971;220:1100–1106. 147. Kim
191. Smithies
Y, KimO.D,Why Han the
K, etkidney glomerulus
al. Expression of ureadoes not clog: ina the
transporters gel develop-
permea-
122. Bulger
161. Kriz W,R,Haehnel
Cronin R, B, Hosser
Dobyan H, D. etSurvey
al. Pathways to recovery and
of the morphology of thelossdogo tion/di
ing u sion hypothesis
rat kidney. Am J PhysioloRenal renalPhysiol.
unction. Proc Natl Acad Sci USA. 2003;100:
2002;282:F530–F540.
nephrons
kidney. Anatin anti-Thy-1 nephritis. J Am Soc Nephrol. 2003;14:1904–1926.
Rec. 1979;194:41–66. 4108–4113.
192. Nielsen S. Renal aquaporins: an overview. BJU International. 2002;90:1–6.
http://www.ncbi.nlm.nih.gov/pubmed/12819253
162. Tisher C, Bulger R, Trump B. Human renal ultrastructure. I. Proximal http://www.ncbi.nlm.nih.gov/pubmed/12655073
193. Bankir L, Trinh-Trang-Tan M. Renal urea transporters. Direct and indirect
123. Welling
tubule of healthy L, Zupka M, Welling
individuals. D. Mechanical
Lab Invest. properties o basement mem-
1966;15:1357–1394. 148. Hausmann
regulation R, Kuppe
by vasopressin. Exp C,Physiol.
Egger H, et al. Electrical orces determine glo-
2000;85:243S-252S.
brane.Welling
163. News Physiol Sci. 1995;10
L, Welling D. Surface (1):30–35.
areas of brush border and lateral cell walls in merular
194. permeability.
Nielsen S, KwonJ Am Soc Nephrol.B,2010;21:2053–2058.
T, Christensen et al. Physiology and pathophysiology
124.rabbit
the Krizproximal
W, Mundel P, ElgerKidney
nephron. M. The Int.contractile apparatus o podocytes is ar-
1975;8:343–348. http://www.ncbi.nlm.nih.gov/pubmed/20947631
of renal aquaporins. J Am Soc Nephrol. 1999;10:647–663.
rangedHarmanci
164. to counteractM, WadeGBMJ,expansion.
DiScala V. Contrib
Altered Nephrol.
structure1994;107:1–9.
of tight junctions in rat 149. Nielsen
195. Tryggvason K, Pettersson
S, Pallone T, Smith E. Causes
B, et al.and consequences
Aquaporin-1 o proteinuria:
water channels in theshort
kid-
125. Drumond
proximal tubules M, withDeenvolume W. expansion.
Structural determinants o glomerular hydraulic
Kidney Int. 1975;8:478. ney flong
and ltration
loopbarrier and progressive
descending thin limbs renal
and ailure. J Intern Med.
in descending vasa2003;
recta 254:216–224.
in rat kidney.
permeability.
165. Bulger R, AmLorentz
J Physiol. W1994;266:F1–F12.
Jr, Colindres R, et al. Morphological changes in rat http://www.ncbi.nlm.nih.gov/pubmed/12930230
Am J Physiol Renal Physiol. 1995;268:1023–1037.
126. proximal
renal Kiberd B. tubules
The unctional
and their and structural changes
tight junctions o the
with glomerulus
increased through-
intraluminal 150. Kriz
196. Bolton W,G, Deen W,
Schiller A, Daniels
TaugnerB.R. Assessment
Freeze-fractureo the charge
studiesselectivity
on the thin o glomeru-
limbs of
out the course
pressure. o murine
Lab Invest. lupus nephritis. J Am Soc Nephrol. 1992;3:930–939.
1974;30:136–144. lar basement
Henle’ s loop in membrane
Psammomys usingobesus.
Ficoll sulAmate. Am J1981;162:23–34.
J Anat. Physiol. 1998;274: F889–F896.
http://www.ncbi.nlm.nih.gov/pubmed/1450369
166. Silverblatt F, Bulger R. Gap junctions occur in vertebrate renal proximal 151. Kriz
197. Sorensson J, Ohlson
W. Structural M, Haraldsson
organization of the B. renal
A quantitative
medullaryanalysis
countero ow the system.
glomer-
127. Guasch
tubule A, Biol.
cells. J Cell Myers B. Determinants o glomerular hypof ltration in ne-
1970;47:513. ular Proc.
Fed charge barrier in the rat. Am J Physiol Renal Physiol. 2001;280: F646–F656.
1983;42:2379–2385.
phrotic
167. patients
Barrett with minimal
J, Heidger change nephropathy.
P. Microbodies of the ratJ Am renalSocproximal
Nephrol. 1994;4:
tubule: http://www.ncbi.nlm.nih.gov/pubmed/11249856
198. Kaissling B, de Rouf gnac C, Barrett J, et al. The structural organization of the
1571–1581.
Ultrastructural and cytochemical investigations. Cell Tissue Res. 1975;157: 152. Ciarimboli
kidney of the desertG,rodentHjalmarsson
Psammomys C, Bokenkamp
obesus. Anat A, Embryol.
et al. Dynamic alterations o
1975;148:121–143.
128. Drumond M, Kristal B, Myers B, et al. Structural basis or reduced glomer-
283–305. glomerular
199. Schwartz chargeM, density
Karnovsky in f xed rat kidneys suggest
M, Venkatachalam involvement
M. Regional o endothe-
membrane spe-
ular f DeDuve
168. ltration capacity
C, Baudhuinin nephrotic humans.(microbodies
P. Peroxisome J Clin Invest. 1994;94:1187–1195.
and related particles). lial coat. Am
cialization in Jthe
Physiol
thinRenal
limbsPhysiol.
of Henle’2003;285:F722–F730.
s loops as seen by freeze-fracture electron
http://www.ncbi.nlm.nih.gov/pubmed/8083359
Physiol Rev. 1966;46:323–357. 153. Ryan G,
microscopy. Karnovsky
Kidney M. Distribution o endogenous albumin in the rat glo-
Int. 1979;16:577–589.
129. Jacobsen
169. Barnes J,N, Reznicek
Jorgensen M, FRadnik R, et al. observations
. Ultrastructural Anionization on o the
an antigen pro-
pars decen- merulus.
200. MajackRoleR, o Paull
hemodynamic
W, Barrett actors
J. Thein glomerular barrier
ultrastructural unction.
localization Kidney Int.
of membrane
motesofglomerular
dens the proximal binding
tubule andinimmune
the kidney complex
of the ormation.
male rat. Kidney Int. 1988;34:
Z Zellforsch Mikrosk 1976;9:36–45.
ATPase in rat thin limbs of the loop of Henle. Histochemie. 1979;63:23–33.
156–163.
Anat. 1973;136:479–499. http://www.ncbi.nlm.nih.gov/pubmed/940256
201. Ernst S, Schreiber J. Ultrastructural localization of Na , K -ATPase in rat
http://www.ncbi.nlm.nih.gov/pubmed/2460660
170. Roesinger B, Schiller A, Taugner R. A freeze-fracture study of tight 154.rabbit
and Schonheyder H, Maunsbach
kidney medulla. A. Ultrastructure
J Cell Biol. 1981;91:803–813. o a specialized neck region
130. Simonson
junctions M, Dunn
in the pars convolutaM. Leukotriene
and pars recta C4ofandthe D4
renal contract
proximal rattubule.
glomerular
Cell in the Schiller
202. rabbit nephron.
A, Taugner Kidney
R, Kriz Int.W.
1975;7:145–153.
The thin limbs of Henle’s loop in the rabbit:
mesangial
Tissue Res. cells. Kidney Int. 1986;30:524–531.
1978;186:121–133. http://www.ncbi.nlm.nih.gov/pubmed/1123890
A freeze-fracture study. Cell Tissue Res. 1980;207:249–265.
http://www.ncbi.nlm.nih.gov/pubmed/3465969
171. Koepsell H, Schmitt B, Gorboulev V. Organic cation transporters. Rev 155. Dobyan
203. JacobsenD,N,Bulger Jorgensen F. Further of
R. Morphology enzyme histochemical
the minipig kidney. J observations
Electron Microsc on
131. Savin
Physiol Biochem V. Pharmacol.
In vitro e ects o angiotensin II on glomerular unction. Am J
2003;150:36–90. the segmentation
Tech. 1988;9:213–234. o the proximal tubules in the kidney o the male rat. Histoche-
Physiol. 1986;251:F627–F634. mie. 1973;34:11–32.
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 443
3

156. Layton
204. Maunsbach A. Observations
A, Pannabecker on the segmentation
T, Dantzler W, et al. Two modes o the proximal tubule in
for concentrating 180. Kriz W,R,Schnermann
235. Greger Schlatter E, Lang J, Dieterich
F. EvidenceH. Di for erences in thesodium
electroneutral morphology chloride o
the ratinkidney.
urine rat inner J Ultrastruct
medulla. Am Res.J1966;16:239–258.
Physiol Renal Physiol. 2004;287:F816–F839. de scending in
cotransport limbs o shortthick
the cortical and long loopslimb
ascending o Henle in the
of Henle’ rat kidney.
s loop of rabbitIn: Wirz
kidney.
http://www.ncbi.nlm.nih.gov/pubmed/5333381
205. Verbavatz J, Brown D, Sabolic I, et al. Tetrameric assembly of CHIP28 H, Spinelli
P ugers Arch.F,1983;396:308–314.
eds. Recent Advances in Renal Physiology. Basel: Karger; 1972:
157. channels
water Maunsbach in A. Ultrastructure
liposomes and cell o membranes:
the proximalatubule. In: Orlostudy.
freeze-fracture J, Berliner
J Cell 140–144.
236. Nielsen S, Maunsbach A, Ecelbarger C, et al. Ultrastructural localization of
R, eds.
Biol. Handbook of Physiology, vol.8. Washington: American Physiological Soci-
1993;123:605–618. 181.
Na-K-2ClSchwartz M, Venkatachalam
cotransporter in thick ascendingM. Structural
limb anddimacula erences in thin
densa of ratlimbs
kidney.o
ety; 1973:31–79.
206. Hediger M, Knepper M. Introduction: recent insights into the urinary Henle: Physiological
Am J Physiol implications.
Renal Physiol. Kidney Int. 1974;6:193–208.
1998;275:F885–F893.
158. Kaisslingmechanism:
concentrating B, Kriz W. Structural
from cDNA analysis
cloning o to thedodelin
rabbit kidney. Adv AnatAm
renal function. Em-J http://www.ncbi.nlm.nih.gov/pubmed/4427417
237. Obermüller N, Kunchaparty S, Ellison D, et al. Expression of the Na-K-2Cl
bryol Cell
Physiol. Biol. 1979;56:1–123.
1998;275:F317. 182. DieterichbyH,
cotransporter Barrettdensa
macula J, KrizandW,thick
et al.ascending
The ultrastructure
limb cells of o rat
theandthinrabbit
loop
http://www.ncbi.nlm.nih.gov/pubmed/443090
207. Trinh-Trang-Tan M, Bankir L. Integrated function of urea transporters in limbs
nephron.o the mouse
J Clin kidney.
Invest. Anat Embryol. 1975;147:1–18.
1996;98:635–640.
159.mammalian
the Kriz W, Koepsellkidney. Exp H. The structural
Nephrol. organization o the mouse kidney. Z
1998;6:471–479. http://www.ncbi.nlm.nih.gov/pubmed/1200392
238. Burg M, Green N. Function of the thick ascending limb of Henle’s loop. Am
Anat Entwicklungsgesch.
208. Shayakul C, Knepper 1974;144:137–163.
M, Smith C, et al. Segmental localization of urea 183. Bachmann
J Physiol. S, Kriz W. Histotopography an ultrastructure o the thin limbs
1973;224:659–668.
http://www.ncbi.nlm.nih.gov/pubmed/4472393
transporter mRNAa in rat kidney. Am J Physiol. 1997;272:F654–F660. o239.
the Burg
loop M. o Henle
Thick in the hamster.
ascending limbCell Tissues Res.
of Henle’ loop.1982;225:111–127.
Kidney Int. 1982;22:454.
160. Imai
209. Tisher M.C.Function
Relationship of thebetween renal structure
thin ascending limb ofand Henleconcentrating ability in
of rats and hamsters http://www.ncbi.nlm.nih.gov/pubmed/6889464
240. Spanidis A, Wunsch H, Kaissling B, et al. Three-dimensional shape of a
the rhesusinmonkey.
perfused vitro. Am Am J Physiol.
J Physiol. 1971;220:1100–1106.
1977;232:F201–F209. 184. Barrett J,cell
Goormaghtigh KrizandW, itsKaissling
contactB,with et al. The ultrastructure
a granular o the kidney.
cell in the rabbit nephrons o
Anat
161. Jamison
210. Bulger R,R.Cronin R, Dobyan
Micropuncture D. Survey
study o theof
of segments morphology
thin loops of o Henle
the dog inkid-
the the desert1982;165:239–252.
Embryol. rodent (Psammomys obesus) kidney. I. thin limbs o Henle o short
ney. Am
rat. AnatJ Physiol.
Rec. 1979;194:41–66.
1968;215:236–242. looped
241. Dorupnephrons. Am J Anat. 1978;151:487–498.
J. Ultrastructure of distal nephron cells in rat renal cortex. J Ultra-
http://www.ncbi.nlm.nih.gov/pubmed/443563
211. Bulger R, Tisher C, Myers C, et al. Human renal ultrastructure. II. The http://www.ncbi.nlm.nih.gov/pubmed/645614
struct Res. 1985;92:101–118.
162. limb
thin Tisher C, Bulger
of Henle’ s loop R, and
Trump the B. Human renal
interstitium ultrastructure.
in healthy individuals.I. Proximal
Lab Invest. tu- 185. Barrett J, B,
242. Kaissling Kriz W, Kaissling
Stanton B, et al.
B. Adaptation of The
distalultrastructure o the nephrons
tubule and collecting duct to in- o
bule o healthy individuals. Lab Invest. 1966;15:1357–1394.
1967;16:124–141. the desert
creased rodent
sodium (Psammomys
delivery. obesus) kidney.
I. Ultrastructure. II. Thin
Am J Physiol. limbs o Henle o long-
1988;255:F1256–F1268.
http://www.ncbi.nlm.nih.gov/pubmed/5920363
212. Tisher C, Bulger R, Trump B. Human renal ultrastructure. III. The distal looped nephrons.
243. Stanton B, Am J Anat. B.
Kaissling 1978;151:499–514.
Adaptation of distal tubule and and collect-
163. Welling
tubule in healthy L, Welling D. Sur
individuals. Lab ace areas1968;18:655–668.
Invest. o brush border and lateral cell walls in http://www.ncbi.nlm.nih.gov/pubmed/645615
ing duct to increased Na delivery. II. Na and K transport. Am J Physiol.
the rabbit
213. Madsenproximal nephron.
K, Tisher Kidney Int. 1975;8:343–348.
C. Structural-functional relationships along the distal 186. Dobyan D, Jamison R. Structure and unction o the renal papilla. Semin
1988;255:F1269–F1275.
http://www.ncbi.nlm.nih.gov/pubmed/1206850
nephron. Am J Physiol. 1986;250:F1–F15. Nephrol.
244. Stanton1984;4:5. B, Biemesderfer D, Wade J, et al. Structural and functional study
164. Kaissling
214. HarmanciB,M,Peter Wade S, J, DiScala
Kriz W. The V. Altered
transition structure o tight
of the thick junctions
ascending in rat
limb of 187.
of the Kriz W. Structural
rat distal organization
nephron: Effects o the renal
of potassium medulla:
adaptation and Comparative
depletion. Kidney and
proximal
Henle’ tubules
s loop into with volume
the distal expansion.
convoluted Kidney
tubule in Int.
the 1975;8:478.
nephron of thr rat kidney. unctional
Int. aspects. Am J Physiol. 1981;241:R3–R16.
1981;19:36–48.
165.Tissue
Cell Bulger Res.R,1977;182:111–118.
Lorentz W Jr, Colindres R, et al. Morphological changes in rat 188. Chou C, Nielsen
245. Farquhar M, Palade S, Knepper
G. JunctionalM. Structural-
complexes unctional
in variouscorrelation
epithelia. in Jchin-
Cell
renal proximal
215. Kaisslingtubules
B, Kriz and their tight junctions
W. Morphology of the loop with increased
of Henle, intraluminal
distal tubulepres- and chilla long loop o Henle thin limbs: a novel papillary subsegment. Am J Physiol.
Biol. 1963;17:375.
sure. Lab Invest.
collecting duct. 1974;30:136–144.
In: Windhager E, ed. Handbook of Physiology: Section on Renal 1993;265:F863–F874.
246. Schmidt U, Dubach U. Activity of (Na K )-stimulated adenosine tri-
http://www.ncbi.nlm.nih.gov/pubmed/4591678
Physiology. New York, NY: Oxford University Press; 1992:109–167. 189. Pannabecker
phosphatase in the rat T, Abbott
nephron. D,PDantzler
ugers Arch. W. 1969;306:219–226.
Three-dimensional unctional re-
166. Allen
216. Silverblatt
F, TisherF, Bulger R. Gap junctions
C. Morphology occur inthick
of the ascending vertebrate
limb ofrenal
Henle. proximal
Kidney construction
247. Schmidto U,inner Dubachmedullary thin limbs oadenosinetriphosphatase:
U. NaK-stimulated Henle’s loop. Am J Physiol Renal
Intracel-
tubule
Int. cells. J Cell Biol. 1970;47:513.
1976;9:8–22. Physiol. 2004;286:F38–F45.
lular localisation within the proximal tubule of the rat nephron. P ugers Arch.
http://www.ncbi.nlm.nih.gov/pubmed/19866748
217. Crayen M, Thoenes W. Architecture and cell structures in the distal neph- 190. Pannabecker T, Dahmann A, Brokl O, et al. Mixed descending- and as-
1971;330:265–270.
167.of Barrett
ron J, Heidger
the rat kidney. P. Microbodies
Cytobiol. o the rat renal proximal tubule: Ultra-
1978;17:197–211. cending-type
248. Kaissling thin B. limbs o Henle’
Structural s loopofin adaptive
aspects mammalian renal inner
changes medulla.
in renal Am J
electrolyte
structural
218. Myers and C,cytochemical
Bulger R, Tisher investigations.
C, et al. Human Cell Tissue
renal Res. 1975;157:IV
ultrastructure. 283–305.
. Collect- Physiol
excretion.Renal AmPhysiol.
J Physiol.2000;278:F202–F208.
1982;243:F211–F226.
http://www.ncbi.nlm.nih.gov/pubmed/1122543
ing duct of healthy individuals. Lab Invest. 1966;15:1921–1950. 191. Kim Y, Kim
249. Kaissling B, D,
LeHanHir K,M.etDistal
al. Expression o urea transporters
tubular segments in the rabbit in the develop-
kidney after
168. Suzuki
219. DeDuveT,C,Mosto Baudhuin P. Peroxisome (microbodies
F. Intramitochondrial lamentous and related
bodies in particles).
the thick ing rat kidney.
adaptation Am J Physiol
to altered Na- and RenalK-intake.
Physiol. 2002;282:F530–F540.
I. Structural changes. Cell Tissue Res.
Physiol
limb ofRev. 1966;46:323–357.
Henle of the rat kidney. J Cell Biol. 1967;33:605–623. 192. Nielsen S. Renal aquaporins: an overview. BJU International. 2002;90:1–6.
1982;224:469–492.
http://www.ncbi.nlm.nih.gov/pubmed/5325972
220. Madsen K, Verlander J, Tisher C. Relationship between structure and func- http://www.ncbi.nlm.nih.gov/pubmed/12445090
250. Kaissling B, Bachmann S, Kriz W. Structural adaptation of the distal con-
169.inJacobsen
tion distal tubuleN, Jorgensen
and collecting F. Ultrastructural
duct. J Electronobservations
Microsc Tech.on the pars decen-
1988;9:187–208. 193.
volutedBankirtubule L, Trinh-Trang-Tan
to prolonged furosemide M. Renal urea transporters.
treatment. Direct and
Am J Physiol. indirect
1985;248:
dens oKone
221. the B,proximal
Madsentubule K, Tisher in the kidney o the of
C. Ultrastructure male
the rat.
thickZ ascending
Zellforsch Mikrosk
limb of regulation
F374–F381. by vasopressin. Exp Physiol. 2000;85:243S-252S.
Anat. 1973;136:479–499.
Henle in the rat kidney. Am J Anat. 1984;171:217–226. http://www.ncbi.nlm.nih.gov/pubmed/10795928
251. Biner H, Arpin-Bott M, Lof ng J, et al. Human cortical distal nephron;
http://www.ncbi.nlm.nih.gov/pubmed/4346669
222. Hebert S, Culpepper R, Andreoli T. ADH-stimulated NaCl transport in 194. NielsenofS, electrolyte
Distribution Kwon T, Christensen
and water B, et al. Physiology
transport pathways.and pathophysiology
J Am Soc Nephrol.
170. Roesinger
mouse medullaryB,thick Schiller A, Taugner
ascending limbs. R. I.AEvidence
reeze- racture study o tight junc-
for ADH-stimulated neu- o2002;13:836–847.
renal aquaporins. J Am Soc Nephrol. 1999;10:647–663.
tionsNaCl
tral in theco-transport
pars convoluta andandfunctional
pars recta nephron
o the renal proximal tubule.
heterogeneity. Am Cell Tissue
J Physiol. http://www.ncbi.nlm.nih.gov/pubmed/10073616
252. Lof ng J, Cueni-Lof n D, Valderrabano V, et al. Distribution of transcel-
Res. 1978;186:121–133.
1981;241:F412–F431. 195. Nielsenand
lular calcium S, Pallone
sodiumT,transport
Smith B,pathways
et al. Aquaporin-1
along mouse water channels
distal nephron. in short
Am J
http://www.ncbi.nlm.nih.gov/pubmed/627008
223. Schiller A, Taugner R, Roesinger B. Vergleichende Morphologie der Zonu- and long
Physiol loopPhysiol.
Renal descending thin limbs and in descending vasa recta in rat kidney.
2001;281:F1019–F1020.
171.
lae Koepsell H,
occludentes amSchmitt B, Gorboulev
Nierentubulus. V. Organic
Verh Anat cation transporters. Rev Phys-
Ges. 1978;72:229–234. Am
253.J Physiol
Bachmann RenalS,Physiol. 1995;268:1023–1037.
Bostanjoglo M, Schmitt R, et al. Sodium transport-related
iol Biochem
224. BoubyPharmacol.
N, Bankir2003;150:36–90.
L, Trinh-Trang-Tan M, et al. Selective ADH-induced hy- 196.
proteinsKriz in W,theSchiller
mammalianA, TaugnerdistalR.nephron
Freeze- -racture studies ontogeny
distribution, on the thinand limbs
func-o
http://www.ncbi.nlm.nih.gov/pubmed/12827517
pertrophy of the medullary thick ascending limb in Brattleboro rats. Kidney Int. Henle’
tional saspects.
loop in Anat Psammomys
Embryol. obesus. Am J Anat. 1981;162:23–34.
1999;200:447–468.
172. Burckhardt B, Burckhardt G. Transport o organic anions across the
1985;28:456–466. http://www.ncbi.nlm.nih.gov/pubmed/7030046
254. Potter E. Normal and Abnormal Development of the Kidney. Chicago: Year
basolateral
225. Bankirmembrane
L, Fischer oC, proximal
Fischer S,tubule cells. Rev Physiol
et al. Adaptation Biochem
of the rat kidneyPharmacol.
to altered 197.
Book; Kriz
1972.W Structural organization o the renal medullary counter ow system.
2003;146:95–158.
water intake and urine concentration. P ugers Arch. 1988;412:42–53. Fed
255.Proc.
Howie 1983;42:2379–2385.
A, Schmithson N, Rollason T. Reconsideration of the development
http://www.ncbi.nlm.nih.gov/pubmed/12605306
226. Bouby N, Trinh-Trang-Tan M, Laouari D, et al. Role of the urinary concentrat- 198.
of the Kaissling
distal tubule B, de Rouhuman
of the f gnackidney.
C, BarrettJ Anat.J, 1993;183:141–147.
et al. The structural organiza-
173.process
ing Lee W, in Kim R. Transporters
the renal effects of high andprotein
renal drug
intake. elimination.
Kidney Int. Annu Rev Pharma-
1988;34:4–12. tion
256. oKim theJ,kidney
Tisher oC, the MadsendesertK.rodent Psammomys
Differentiation obesus. Anat
of intercalated cellsEmbryol.
in de-
col Toxicol.
227. Bankir 2004;44:137–166.
L, Kriz W. Adaptation of the kidney to protein intake and to urine 1975;148:121–143.
veloping rat kidney: An immunohistochemical study. Am J Physiol. 1994;266:
174. Wright S,activity:
concentrating DantzlerSimilar
W. Molecular and cellular
consequences in healthphysiology
and CFR. o renal
Kidneyorganic
Int. http://www.ncbi.nlm.nih.gov/pubmed/1211658
F977–F990.
cation and anion transport. Physiol Rev. 2004;84:987–1049.
1995;47:7–24. 199. SchwartzB.M,
257. Stanton Karnovsky
Renal potassium M,transport:
Venkatachalam M. Regional
morphological membraneadap-
and functional spe-
http://www.ncbi.nlm.nih.gov/pubmed/15269342
228. Welling L, Welling D, Hill J. Shape of cells and intercellular channals in cialization
tations. AminJ Physiol.
the thin1989;257:R989–R997.
limbs o Henle’s loops as seen by reeze- racture electron
175. Lock
rabbit thick E, Reed C.limb
ascending Xenobiotic
of Henle. metabolizing enzymes o the kidney. Toxicol
Kidney Int. 1978;13:144–151. microscopy.
258. Lof ngKidney Int. 1979;16:577–589.
J, Kaissling B. Sodium and calcium transport pathways along the
Pathol.Hoyer
229. 1998;26:18–25.
J, Sisson S, Vernier R. Tamm-Horsfall glycoprotein. Ultrastructural http://www.ncbi.nlm.nih.gov/pubmed/548602
mammalian distal nephron from rabbit to human. Am J Physiol Renal Physiol.
http://www.ncbi.nlm.nih.gov/pubmed/9502382
immunoperoxidase localization in rat kidney. Lab Invest. 1979;41:168–173. 200. Majack R, Paull W, Barrett J. The ultrastructural localization o membrane
2003;284:F628–F643.
176. Sikri
230. Christensen
K, FosterE,C,Gburek MacHugh J. N,
Protein reabsorptionofinTamm-Horsfall
et al. Localization renal proximal tu-
glyco- ATPase
259. Fridtin rat G,thin
Palmerlimbs L. oNathe loop o in
channels Henle.
the rat Histochemie.
connecting1979;63:23–33.
tubule. Am J Physiol
bule - in
protein unction
the human and dys
kidney unction
usinginimmuno-
kidney pathophysiology. Pediatr Nephrol.
uorescence and immuno-electron http://www.ncbi.nlm.nih.gov/pubmed/159881
Renal Physiol. 2004;286:F669–F674.
2004;19:714–721.
microscopical techniques. J Anat. 1981;132:597–605. 201. Ernst S, Schreiber
260. Coleman R, Wu D, J. Ultrastructural
Liu J, Wade J. localization
Expression oofNa + , K + -ATPase
aquaporin in the in rat
renal
http://www.ncbi.nlm.nih.gov/pubmed/15146321
231. Chabardes D, Imbert-Teboul M, Gagnan-Brunette M, et al. Different hor- and rabbit kidney
connecting tubule.medulla.
Am J PhysiolJ CellRenal
Biol. Physiol.
1981;91:803–813.
2000;279:F874–F883.
177. Verroust
monal target sites P, Christensen
along the mouse E. Megalin
and rabbitand cubilin
nephrons. - theCurr
story o two
Probl Clinmultipur-
Biochem. http://www.ncbi.nlm.nih.gov/pubmed/6276410
261. Le Hir M, Kaissling B, Dubach U. Distal tubular segments of the rabbit
pose receptors un olds. Nephrol Dial Transplant. 2002;17:1867–1871.
1977;8:447–454. 202.
kidneySchiller A, Taugner
after adaptation to R, Kriz W.
altered Na-Theandthin limbs oII.Henle’
K-intake. Changes s loop in the rabbit:
in Na-K-ATPase
http://www.ncbi.nlm.nih.gov/pubmed/12401836
232. Morel F, Imbert-Teboul M, Chabardes D. Receptors to vasopressin and A reeze-Cell
activity. racture
Tissue study. Cell Tissue Res. 1980;207:249–265.
Res. 1982;224:493–504.
178. hormones
other Bacic D, Wagner C, Hernandokidney.
in the mammalian N, et al. Novel
Kidney Int.aspects in regulated expres-
1987;31:512–520. http://www.ncbi.nlm.nih.gov/pubmed/7388918
262. Muto S, Yasoshima K, Yoshitomi K, et al. Electrophysiological identi ca-
sion
233. oEl-Mernissi
the renal type IIa Na/Pi-contransporter.
G, Doucet A. Quantitation of [3H]ouabain Kidney Int. 2004;91:S5–S12.
binding and turnover 203.
tion ofDobyan- and D,-intercalated
Bulger R. Morphology
cells and theiro the minipig kidney.
distribution alongJtheElectron
rabbitMicrosc
distal
179.
of Na Bachmann
-K-ATPase along S, Schlichting
the rabbit U, Geist B,
nephron. AmetJ Physiol.
al. Kidney-specif c inactivation
1984;247:F158–F167. Tech.
nephron1988;9:213–234.
segments. J Clin Invest. 1990;86:1829–1839.
o theGreger
234. megalin R, gene impairs
Schlatter tra f cking
E. Presence of oluminal
renal K inorganic sodium phosphate
, a prerequisite for active http://www.ncbi.nlm.nih.gov/pubmed/3199239
263. Meneton P, Lof ng J, Warnock D. Sodium and potassium handling by the
cotransporter
NaCl transport(NaPi-IIa). J Am Soc
in the cortical Nephrol
thick . 2004;15:892–900.
ascending limb of Henle’s loop of rabbit 204. Layton A, Pannabecker
aldosterone-sensitive T, Dantzler
distal nephron: W, et al.
the pivotal roleTwo modes
of the distalorandconcentrating
connecting
http://www.ncbi.nlm.nih.gov/pubmed/15034091
kidney. P ugers Arch. 1981;392:92–94. urine
tubule.inAm rat Jinner
Physiol.medulla. Am J Physiol Renal Physiol. 2004;287:F816–F839.
2004;287:F593–F601.
44
44 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

205. Kim
264. Verbavatz
J, KimJ,YBrown, Cha J,D,etSabolic I, et al. Tetrameric
al. Intercalated cell subtypes assembly o CHIP28
in connecting wa-
tubule 229. Brown
295. Hoyer J,D,Sisson S, VernierT,R. Roth
Kumpulainen Tamm-Hors
J, et al.allImmunohistochemical
glycoprotein. Ultrastructurallocal-
ter channels
and in liposomes
cortical collecting ductand celland
of rat membranes:
mouse. J Ama reeze- racture 1999;10:1–12.
Soc Nephrol. study. J Cell Biol. immunoperoxidase
ization of carbonic localization
anhydrase inin postnatal
rat kidney.and Labadult
Invest.rat1979;41:168–173.
kidney. Am J Physiol.
1993;123:605–618.
265. Hansen G, Tisher C, Robinson R. Response of the collecting duct to distur- http://www.ncbi.nlm.nih.gov/pubmed/379435
1983;245:F110–F118.
http://www.ncbi.nlm.nih.gov/pubmed/7693713
bances of acid-base and potassium balance. Kidney Int. 1980;17:326–337. 230. Dobyan
296. Sikri K, D, Foster
MagillC, L,
MacHugh
Friedman N,Pet, etal.al.Localization o Tamm-Hors
Carbonic anhydrase all glyco-
histochemistry
206. Pricam
266. HedigerC,M, Knepper
Humbert M. Introduction:
F, Perrelet recent insights
A, et al. A freeze-etch study ofintothethe
tighturinary
junc- protein
in rabbitinand themouse
humankidneys.
kidney Anat
usingRec.immuno uorescence and immuno-electron
1982;204:185–197.
concentrating
tions of the rat mechanism:
kidney tubules. romLab cDNA
Invest.cloning to dodelin renal unction. Am J
1974;30:286–291. microscopical
297. Dobyan techniques.
D, Bulger R. J Anat.
Renal1981;132:597–605.
carbonic anhydrase. Am J Physiol. 1982;
Physiol.
267. Grif1998;275:F317.
th L, Bulger R, Trump B. Structure and staining of mucosubstance http://www.ncbi.nlm.nih.gov/pubmed/7028707
243:F311–F324.
207.
on Trinh-Trang-Tan
“intercalated cells” fromM, Bankir
the ratL.distal
Integrated
tubule unction o ureaduct.
and collecting transporters
Anat Rec. in 231. Madsen
298. Chabardes D, Imbert-Teboul
K, Tisher C. ResponseM,ofGagnan-Brunette
intercalated cells M, et al.
of rat Di medullary
outer erent hor-
the mammalian kidney. Exp Nephrol. 1998;6:471–479.
1968;160:643–662. monal target
collecting duct sites along themetabolic
to chronic mouse and rabbit Lab
acidosis. nephrons.
Invest. Curr Probl Clin Biochem.
1984;51:268–276.
http://www.ncbi.nlm.nih.gov/pubmed/9807016
268. Rastegar A, Biemesderfer D, Kashgarian M, et al. Changes in membrane sur- 1977;8:447–454.
299. Madsen K, Tisher C. Cellular response to acute respiratory acidosis in rat
208. ofShayakul
faces collectingC, ductKnepper M, Smith adaptation.
cells in potassium C, et al. Segmental
Kidney Int.localization o urea
1980;18:293–301. http://www.ncbi.nlm.nih.gov/pubmed/616378
medullary collecting duct. Am J Physiol. 1983;14:F670–F679.
transporter
269. Wade mRNAaJ, O’NeilinR,rat kidney.
Pryor J, et Am J Physiol. 1997;272:F654–F660.
al. Modulation of cell membrane area in renal 232. Brown
300. Morel FD, , Imbert-Teboul
Orci L. Junctional M, Chabardes
complexesD.and Receptors to vasopressin
cell polarity in the urinary and
209. Imaitubules
collecting M. Function o the thin ascending
by corticosteroid hormones.limb J Cello Biol.
Henle o rats and hamsters
1979;81:439–445. other hormones
tubule. J Electronin the mammalian
Microsc kidney. Kidney Int. 1987;31:512–520.
Tech. 1988;9:145–170.
per used
270. in vitro.
Stanton Am J Physiol.
B, Janzen 1977;232:F201–F209.
A, Klein-Robbenhaar G, et al. Ultastucture of rat ini- http://www.ncbi.nlm.nih.gov/pubmed/3031358
301. Weiner I, Hamm L. Regulation of intercellular pH in the rabbit cortical
210.collecting
tial Jamison tubule.
R. Micropuncture
Effect of adrenalstudy o corticosteroid
segments o thin loops o JHenle
treatment. in the
Clin Invest. 233. El-Mernissi
collecting tubule. J G, ClinDoucet A. Quantitation o [3H]ouabain binding and
Invest. 1990;85:274–281.
rat. Am J Physiol. 1968;215:236–242.
1985;75:1327–1334. turnover
302. o Na+-K-ATPase
Verlander J, Madsen along K, Stonethe rabbit
D, et nephron. Am J Physiol.
al. Ultrastructural 1984;247:
localization of
http://www.ncbi.nlm.nih.gov/pubmed/5659337
271. Sun T, Hoek W, Huang Y, et al. Aquaporin-2 localization in clathrin-coated HF158–F167.
ATPase in rabbit cortical collecting duct. J Am Soc Nephrol. 1994;44:
211. inhibition
pits: Bulger R, of Tisher C, MyersbyC,dominant-negative
endocytosis et al. Human renal ultrastructure.
dynamin. II. The
Am J Physiol. 234. Greger R, Schlatter E. Presence o luminal K+, a prerequisite or active
1546–1557.
thin limb o Henle’s loop and the interstitium in healthy individuals. Lab Invest.
2002;282:F998–F1011. NaCl Emmons
303. transport C, in the cortical
Kurtz thick ascending
I. Functional limb o Henle’
characterization s loop
of three o rabbit
intercalated
1967;16:124–141.
272. Lof ng J, Cueni-Lof n D, Macher A, et al. Localization of epithelial sodium kidney.
cell Pf ugers
subtypes in Arch. 1981;392:92–94.
the rabbit outer cortical collecting duct. J Clin Invest. 1994;93:
http://www.ncbi.nlm.nih.gov/pubmed/4164459
channel and aquaporin-2 in rabbit cortex. Am J Physiol Renal Physiol. 2000;272:530. http://www.ncbi.nlm.nih.gov/pubmed/7322839
417–423.
212. Marver
273. Tisher C, D.Bulger
Evidence R, Trump B. Human renal
of corticosteroid actionultrastructure.
along the nephron.III. The Am distalJ 235. Milton
304. Greger A, R, Schlatter
Weiner I.E,Regulation
Lang F. Evidenceof B-type or electroneutral
intercalated cell sodium
apical chloride
anion
tubule in1984;246:F111–F123.
Physiol. healthy individuals. Lab Invest. 1968;18:655–668. cotransport
exchange in theby
activity cortical thick ascending
CO2/HCO3. Am J Physiol. limb1998;275:F1086–F1094.
o Henle’s loop o rabbit kidney.
http://www.ncbi.nlm.nih.gov/pubmed/5667863
274. Madsen K, Clapp W, Verlander J. Structure and function of the inner med- Pf ugers
305. Arch. 1983;396:308–314.
Schwartz G, Al-Awqati Q. Carbon dioxide causes exocytosis of vesicles
213. Madsen K,duct.
ullary-collecting Tisher C. Structural-
Kidney unctional relationships along the distal
Int. 1988;34:441–454. 236. Nielsen
containing H S, pumps
Maunsbach A, Ecelbarger
in isolated perfused C, etproximal
al. Ultrastructural localization
and collecting tubules.o
nephron.
275. Bankir AmL, J Physiol. 1986;250:F1–F15.
Trinh-Trang-Tan M. Urea and the kidney. In: Brenner B, ed. The JNa-K-2Cl
Clin Invest. cotransporter in thick ascending limb and macula densa o rat kidney.
1985;75:1638–1644.
214. Kaissling
Kidney. Philadelphia:B, Peter S, Kriz 2000:637–679.
Saunders; W. The transition o the thick ascending limb o Am J Physiol
306. Quentin Renal Physiol. 1998;275:F885–F893.
F, Chambrey R, Trinh-Trang-Tan M, et al. The Cl/HCO3 exchang-
Henle’Alper
276. s loopS,into Natalethe J,distal
Gluck convoluted tubule in
S, et al. Subtypes of the nephroncells
intercalated o thr in rat kidney
kidney. 237.
er Obermüller
pendrin N, Kunchaparty
in rat kidney is regulatedS,inEllison
response D, ettoal. Expression
chronic o theinNa-K-2Cl
alteration chloride
Cell Tissueduct
collecting Res. de1977;182:111–118.
ned by antibodies against erythroid band 3 and renal vascular cotransporter
balance. by macula
Am J Physiol densa
Renal and2004;287:1179–1188.
Physiol. thick ascending limb cells o rat and rabbit
Hhttp://www.ncbi.nlm.nih.gov/pubmed/884723
-ATPase. Proc Natl Acad Sci USA. 1989;80:5429–5433. nephron.
307. WallJ S,Clin Invest.K,1996;98:635–640.
Hassell Royaux I, et al. Localization of pendrin in mouse kidney.
215. Brown
277. Kaissling B, Kriz S,
D, Hirsch W.Gluck
Morphology o the loop
S. Localization o Henle, distal tubule
of a proton-pumping ATPaseand in 238.J Physiol
Am Burg M, Green
Renal N. Function
Physiol. o the thick ascending limb o Henle’s loop. Am
2003;284:F229–F241.
collecting
rat kidney. duct.
J Clin In: Windhager
Invest. E, ed. Handbook o Physiology: Section on Renal
1988;82:2114–2126. J Physiol.
308. Kim1973;224:659–668.
Y, Kwon T, Frische S, et al. Immunocytochemical localization of pen-
Physiology.
278. New J,
Verlander York,
MadsenNY: K, OxTisher
ord University
C. Effect ofPress;
acute1992:109–167.
respiratory acidosis on two http://www.ncbi.nlm.nih.gov/pubmed/4691283
drin in intercalated cell subtypes in rat and mouse kidney. Am J Physiol Renal
216. Allen Fof
populations , Tisher C. Morphology
intercalated cells in rato cortical
the ascending
collectingthick limb
duct. Am o JHenle.
PhysiolKidney
Renal 239. Burg
Physiol. M. Thick ascending limb o Henle’s loop. Kidney Int. 1982;22:454.
2002;283:F744–F754.
Int. 1976;9:8–22.
Fluid Electrolyte Physiol. 1987;253:F1142–F1156. http://www.ncbi.nlm.nih.gov/pubmed/6296522
309. Madsen K, Verlander J, Kim J, Tisher C. Morphological adaptation of the
http://www.ncbi.nlm.nih.gov/pubmed/181632
279. Verlander J, Madsen K, Tisher C. Two populations of intercalated cells exist 240. Spanidis
collecting duct toA,acid-base
Wunsch disturbances.
H, Kaissling KidneyB, et al. Int.Three-dimensional shape o a
1991;40, Suppl.33:S57–S63.
217.
in Crayen
cortical M, Thoenes
collecting ducts W. Architecture
in the and1985;33:501A.
rat. Clin Res. cell structures in the distal neph- Goormaghtigh
310. Barajas L.cell Theandultrastructure
its contact with of athegranular cell in the rabbit
juxtaglomerular kidney.
apparatus as Anat
dis-
ron o Teng-Umnuay
280. the rat kidney.P,Cytobiol.
Verlander 1978;17:197–211.
J, Yuan W, et al. Identi cation of distinct sub- Embryol.
closed by1982;165:239–252.
three-dimensional reconstrucion from serial sections: The anatomi-
http://www.ncbi.nlm.nih.gov/pubmed/689251
populations of intercalated cells in the mouse collecting duct. J Am Soc Nephrol. http://www.ncbi.nlm.nih.gov/pubmed/7158812
cal relationship between the tubular and vascular components. J Ultrastruct Res.
218. Myers C, Bulger R, Tisher C, et al. Human renal ultrastructure. IV. Collect-
1996;7:260–274. 241. Dorup J. Ultrastructure o distal nephron cells in rat renal cortex. J Ultra-
1970;33:116–147.
ing duct
281. o healthy
Schuster individuals.
V, Bonsib S, JenningsLab Invest.
M. Two 1966;15:1921–1950.
types of collecting duct mitochon- struct Res.
311. Barajas1985;92:101–118.
L, Latta H. Structure of the juxtaglomerular apparatus. Circ Res.
http://www.ncbi.nlm.nih.gov/pubmed/5335544
dria-rich (intercalated) cells: Lectin and band 3 cytochemistry. Am J Physiol Cell 242. Kaissling
1967;20–21 Suppl.B, 2:15.
Stanton B. Adaptation o distal tubule and collecting
219. Suzuki
Physiol. T, Mostof F. Intramitochondrial f lamentous bodies in the thick
1986;251:C347–C355. duct to
312. in creased
Kaissling sodium
B, Kriz delivery. of
W. Variability I. intercellular
Ultrastructure. Ambetween
spaces J Physiol.macula
1988;255:
den-
limb oVerlander
282. Henle o J,the rat kidney.
Madsen J Cell Biol.
K, Tisher 1967;33:605–623.
C. Axial distribution of band 3–positive F1256–F1268.
sa cells: A transmission electron microscopic study in rabbits and rats. Kidney Int.
http://www.ncbi.nlm.nih.gov/pubmed/6036524
intercalated cells in the collecting duct of control and ammonium chloride-loaded 243. Stanton B, Kaissling B. Adaptation o distal tubule and and collecting
1982;22:9–17.
220. Madsen
rabbits. Kidney Int. K, Verlander J, Tisher C. Relationship between structure and
1996;57:S137–S147. duct toBachmann
313. increased S,NaMundel delivery.
P. II. Na+oxide
Nitric and K+ transport.
in the kidney:Am J Physiol.localization
synthesis, 1988;255:
unction
283. in distal
LaFurgey A, tubule
Tisher C. andMorphology
collecting duct. J Electron
of rabbit Microsc
collecting duct.Tech.
Am 1988;9:
J Anat. F1269–F1275.
and function. Am J Kidney Dis. 1994;24:112–129.
187–208.
1979;155:111–124. 244. Wilcox
314. StantonC,B, WelchBiemesder er D, Wade
W, Murad F, et al.J, Nitric
et al. Structural
oxide synthase and unctional
in maculastudy den-
http://www.ncbi.nlm.nih.gov/pubmed/3058890
284. O’Neil R, Hayhurst R. Functional differentiation of cell types of cortical o the
sa rat distal
regulates nephron:capillary
glomerular E ects o pressure.
potassium adaptation
Proc Natl Acad andScidepletion. Kidney
USA. 1992;89:
221. Kone
collecting B, Madsen
duct. K, Tisher
Am J Physiol. C. Ultrastructure o the thick ascending limb o
1985;248:F449–F453. Int. 1981;19:36–48.
11993–11997.
HenleWeiner
285. in the rat kidney.L.Am
I, Hamm Use J Anat. 1984;171:217–226.
of uorescent dye BCECF to measure intracellular http://www.ncbi.nlm.nih.gov/pubmed/7218667
315. Harris R, McKanna J, Akai Y, et al. Cyclooxygenase-2 is associated with
http://www.ncbi.nlm.nih.gov/pubmed/6496376
pH in cortical collecting tubule. Am J Physiol. 1989;256:F957–F964. 245.macula
the FarquhardensaM,ofPalade G. Junctional
rat kidney and increases complexeswith salt in restriction.
various epithelia. J Cell
J Clin Invest.
222. Kim
286. Hebert S, Culpepper
J, Tisher C, Linser R, PAndreoli T. ADH-stimulated
, et al. Ultrastructural NaCl transport
localization of carbonic in Biol. 1963;17:375.
1994;94:2504–2510.
mouse medullary
anhydrase thick ascending
ll in subpopulations limbs. I. Evidence
of intercalated cells of ortheADH-stimulated
rat kidney. J Amneu- Soc http://www.ncbi.nlm.nih.gov/pubmed/13944428
316. Bachmann S, Peters J, Engler E, et al. Transgenic rats carrying the mouse
tral NaCl
Nephrol. co-transport and unctional nephron heterogeneity. Am J Physiol.
1990;1:245–256. 246. gene
renin Schmidt U, Dubach characterization
- Morphological U. Activity o (Na+ K+)-stimulated
of a low-renin adenosine
hypertension model.tri-
1981;241:F412–F431.
287. Humbert F, Pricam C, Perrelet A, et al. Speci c plasma membrane dif- phosphatase
Kidney in the rat nephron. Pf ugers Arch. 1969;306:219–226.
Int. 1992;41:24.
223. SchillerinA, the
ferentiations Taugner
cells R,ofRoesinger
the kidney B. Vergleichende
collecting tubule.Morphologie der Zonu-
J Ultrastruct Res. http://www.ncbi.nlm.nih.gov/pubmed/4238475
317. Taugner R, Hackenthal E, Rix E, et al. Immunocytochemistry of the renin-
lae occludentes am Nierentubulus. Verh Anat Ges. 1978;72:229–234.
1975;52:13–20. 247. Schmidt
angiotensin U, Dubach
system: Renin,U.angiotensinogen,
NaK-stimulated adenosinetriphosphatase:
angiotensin I, angiotensinIntracel- II, and
http://www.ncbi.nlm.nih.gov/pubmed/746705
288. Verlander J, Madsen K, Galla J, et al. Response of intercalated cells to chlo- lular localisation
converting enzyme within
in thethekidneys
proximal of tubule
mice, rats, o the and rattree
nephron.
shrews.Pf Kidney
ugers Arch.
Int.
224.depletion
ride Bouby N, Bankir L,alkalosis.
metabolic Trinh-Trang-Tan M, et1992;262:F309–F319.
Am J Physiol. al. Selective ADH-induced hy- 1971;330:265–270.
1982;22 (Suppl 12):S33–S43.
pertrophy
289. Wagner o the medullary
C, Finberg K, thick
Bretonascending limb in
S, et al. Renal Brattleboro
vacuolar rats. Kidney
H -ATPase. Int.
Physiol http://www.ncbi.nlm.nih.gov/pubmed/4258313
318. Barajas L. The development and ultrastructure of the juxtaglomerular cell
1985;28:456–466.
Rev. 2004;84:1263–1314. 248. Kaissling
granule. B. Structural
J Ultrastruct aspects o adaptive changes in renal electrolyte ex-
Res. 1966;15:400–413.
http://www.ncbi.nlm.nih.gov/pubmed/4068480
290. Brown D, Orci L. The “coat” of kidney intercalated cell tubulovesicles does cretion.
319. Am S.
Peter J Physiol. 1982;243:F211–F226.
Ultrastructural studies on the secretory process in the epitheloid
225.contain
not Bankirclathrin.
L, Fischer AmC, Fischer 1986;250:C605–C608.
J Physiol. S, et al. Adaptation o the rat kidney to altered 249. ofKaissling
cells B, Le Hir M.apparatus.
the juxtaglomerular Distal tubular segments
Cell Tissue in the rabbit kidney a ter
Res. 1976;168:45–53.
water Brown
291. intake and urine concentration.
D, Gluck S, Hartwig J. Structure Pf ugers Arch.
of the1988;412:42–53.
novel membrane-coating adaptation
320. Ryan to G, altered
Alcorn Na- and K-intake.
D, Coghlan J, et al.I.Ultrastructural
Structural changes. Cell Tissue
morphology Res.
of gran-
http://www.ncbi.nlm.nih.gov/pubmed/3174386
material in proton-secreting epithelial cells and identi cation as an H ATPase. 1982;224:469–492.
ule release from juxtaglomerular myoepithelioid and peripolar cells. Kidney Int.
J226. Bouby
Cell Biol. N, Trinh-Trang-Tan M, Laouari D, et al. Role o the urinary
1987;105:1637–1648. http://www.ncbi.nlm.nih.gov/pubmed/7116409
1982;22 (Suppl.12):3–8.
concentrat
292. Stetson ingD, process
Wade J, in Giebisch
the renalG.eMorphologic
ects o highalterations
protein intake.
in the ratKidney
medul-Int. 250. Gorgas
321. Kaissling K. B,Structure
Bachmann andS,innervation
Kriz W. Structural adaptation o the
of the juxtaglomerular distal con-
apparatus of
1988;34:4–12.
lary collecting duct following potassium depletion. Kidney Int. 1980;17:45–56. voluted
the tubule
rat. Adv Anatto prolonged
Embryol urosemide
Cell Biol. 1978;54:5–84. treatment. Am J Physiol. 1985;248:
227. Brown
293. Bankir D, L, Kriz
Sabolic W. Adaptation
I. Endosomal o the kidneyfor
pathways to protein intake and
water channel and toproton
urine F374–F381.
322. Biava C, West M. Fine structures of normal human juxtaglomerular
concentrating
pump recyclingactivity:
in kidney Similar consequences
epithelial cells. J Cell in
Sci.health and CFR.
1993;Suppl. Kidney Int.
17:49–59. http://www.ncbi.nlm.nih.gov/pubmed/3976898
cells. I. General structure and intercellular relationship. Am J Pathol. 1966;49:
1995;47:7–24.
294. Kriz W, Kaissling B. Structural organization of the mammalian kidney. 251. Biner H, Arpin-Bott M, Lo f ng J, et al. Human cortical distal nephron;
697–721.
228.
In: Welling
Seldin L, Welling
D, Giebisch G, D, eds.HillTheJ.Kidney.
Shape Philadelphia:
o cells and intercellular channals &
Lippincott Williams in Distribution
323. TaugneroR,electrolyte
Schiller A, and waterB,transport
Kaissling et al. Gappathways.
junctionalJcoupling
Am Soc between
Nephrol.
rabbit thick
Wilkins; ascending limb o Henle. Kidney Int. 1978;13:144–151.
2000:587–654. 2002;13:836–847.
the JGA and the glomerular tuft. Cell Tissue Res. 1978;186:279–285.
http://www.ncbi.nlm.nih.gov/pubmed/362035 http://www.ncbi.nlm.nih.gov/pubmed/11912242
CHAPTER 1 STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 445
5

252. Goormaghtigh
324. Lo f ng J, Cueni-Lo N. Facts f n D,in Valderrabano
favour of an V et al. Distribution
endocrine function ofo the transcel-
renal 277. Brown D, Hirsch S, H.
358. Takahashi-Iwanaga GluckTheS.three-dimensional
Localization o a proton-pumping
spaces of the kidney.ATPaseCell in
lular calcium
arterioles. and sodium
J Pathol Bacteriol.transport pathways along mouse distal nephron. Am J
1945;57:392. rat kidney.
Tissue J Clin Invest. 1988;82:2114–2126.
Res. 1991;264:269–281.
PhysiolSchnermann
325. Renal Physiol.J, 2001;281:F1019–F1020.
Levine D. Paracrine factors in tubuloglomerular feedback: 278. VerlanderT,J,Gandhi
359. Dawson MadsenR, K,Le Tisher
Hir C.M,Eet ectal. oEcto-5
acute -nucleotidase:
respiratory acidosis on two
Localization
253. Bachmann
adenosine, ATP, and S, nitric
Bostanjoglo
oxide. M, Annu Schmitt R, et 2003;65:501–529.
Rev Physiol. al. Sodium transport-related populations
in rat kidneyobyintercalated cells inhistochemical
light microscopic rat cortical collecting
methods. duct. Am J Physiol
J Histochem Renal
Cytochem.
proteins
326. in the
Kurtz mammalian
A. Renin release:distal
sites, nephron
mechanisms, - distribution,
and control. ontogeny
Annu Rev andPhysiol.
unc- Fluid Electrolyte Physiol. 1987;253:F1142–F1156.
1989;37:39–47.
tional aspects. Anat Embryol. 1999;200:447–468.
2011;73:377–399. 279. Verlander J, Madsen
360. Humphreys B, Lin SL,K, Tisher
KobayashiC. Two A, populations o intercalated
et al. Fate tracing reveals cells exist
the peri-
http://www.ncbi.nlm.nih.gov/pubmed/10526014
327. Peti-Peterdi J, Komlosi P, Fuson A, et al. Luminal NaCl delivery regulates in
cytecortical
and not collecting
epithelial ducts in the
origin rat. Clin
of myo Res. 1985;33:501A.
broblasts in kidney brosis. Am J Pathol.
254. PotterPGE
basolateral E. 2Normal
release andfromAbnormal
macla densa Development
cells. J Clin of Invest.
the Kidney. Chicago: Year
2003;112:76–82. 280. Teng-Umnuay P, Verlander J, Yuan W et al. Identif cation o distinct sub-
2010;176:85–97.
Book; Schweda
328. 1972. F, Kurtz A. Cellular mechanism of renin release. Acta Physiol populations
361. Lin SL,oKisselevaintercalated cells inD,
T, Brenner theetmouse collecting
al. Pericytes and duct. J Am Socbroblasts
perivascular Nephrol.
255. Howie
Scand. A, Schmithson N, Rollason T. Reconsideration o the development
2004;181:383–390. 1996;7:260–274.
are the primary source of collagen-producing cells in obstructive brosis of the
o the Osswald
329. distal tubule o the human
H, Nabakowski G, kidney.
HermesJH. Anat. 1993;183:141–147.
Adenosine as a possible mediator of http://www.ncbi.nlm.nih.gov/pubmed/8785396
kidney. Am J Pathol. 2009;173:1617–1627.
http://www.ncbi.nlm.nih.gov/pubmed/7505779
metabolic control of glomerular ltration rate. Int J Biochem. 1980;12:263–267. 281. Schuster R,
362. Gandhi V, Bonsib
Le HirS, M, Jennings M. Two
Kaissling B. types o collecting ductofmitochon-
Immunolocalization ecto-5 -
256. Castrop
330. Kim J, Tisher
H, Huang C, Madsen
Y, HashimotoK. DiS,erentiation
et al. Impairmento intercalated cells in de-
of tubuloglomerular dria-rich
nucleotidase (intercalated)
in the kidney cells: byLectin and band 3antibody.
a monoclonal cytochemistry. Am J Physiol
Histochemie. 1990;95:Cell
veloping regulation
feedback rat kidney:ofAn GFR immunohistochemical study. Am J Physiol.
in ecto-5 -nucleotidase/CD73–de 1994;266:
cient mice. J Clin Physiol.
165–174. 1986;251:C347–C355.
F977–F990.
Invest. 2004;114:634–642. 282.
363. Le Verlander J, MadsenB.K,Distribution
Hir M, Kaissling Tisher C. Axial of 5 distribution
-nucleotidaseo inband 3-positive
the renal in-
intersti-
257. Thomson
331. Stanton B.S,Renal Bao potassium
D, Deng A,transport: morphological
et al. Adenosine formedand by 5unctional adap-
-nucleotidase tercalated
tium of thecells rat. in theTissue
Cell collecting duct o control and ammonium chloride-loaded
Res. 1989;258:177–182.
tations. Am
mediates J Physiol. 1989;257:R989–R997.
tubuloglomerular feedback. J Clin Invest. 2000;106:289–298. rabbits.
364. Koury Kidney S, Int. 1996;57:S137–S147.
Koury M, Bondurant M, et al. Quantitation of erythropoietin-
258. Persson
332. Lo f ng J,A,Kaissling
Ollerstam B. A,
Sodium
Liu R,and calcium
et al. transport
Mechanism forpathways
macula densaalong cell
the 283.
producingLaFurgey
cells inA,kidneys
Tisher C. Morphology
of mice by in situo hybridization:
rabbit collecting duct. Amwith
Correlation J Anat.
he-
mammalian
release of renin.distal
Actanephron rom rabbit
Physiol Scand. to human. Am J Physiol Renal Physiol.
2004;181:471–474. 1979;155:111–124.
matocrit, renal erythropoietin mRNA, and serum erythropoietin concentration.
2003;284:F628–F643.
333. Komlosi P, Fintha A, Bell P. Current mechanisms of macula densa cell sig- http://www.ncbi.nlm.nih.gov/pubmed/463786
Blood. 1989;74:645–651.
259. Fridt
naling. Acta G, Palmer
Physiol L. Na
Scand. channels in the rat connecting tubule. Am J Physiol
2004;181:463–469. 284. O’Neil R, C,
365. Lacombe Hayhurst
Da Silva R. J,Functional
Bruneval di P, eterentiation o cellcells
al. Peritubular typesare
o the
cortical
site
Renal Physiol.
334. Moffat D, 2004;286:F669–F674.
Fourman J. The vascular pattern of the rat kidney. J Anat. collecting duct. Amsynthesis
of erythropoietin J Physiol.in1985;248:F449–F453.
the murine hypoxic kidney. J Clin Invest. 1988;
260. Coleman R, Wu D, Liu J, Wade J. Expression o aquaporin in the renal
1963;97:543–553. 285. Weiner I, Hamm L. Use o uorescent dye BCECF to measure intracellular
81:620–623.
connecting
335. Aikenstubule. Am J Physiol
B, Eenboom A, BohleRenal A.Physiol. 2000;279:F874–F883.
Untersuchungen zur Struktur des Glom- pH
366.in Bachmann
cortical collecting
S, Le Hirtubule.
M, EckardtAm J K.Physiol. 1989;256:F957–F964.
Co-localization of erythropoietin mRNA
261. LeVirchows
erulum. Hir M, Kaissling
Arch A Pathol B, Dubach U. Distal 1979;381:283–293.
Anat Histopathol. tubular segments o the rabbit 286. Kim J,
and ecto-5 Tisher C, Linser
-nucleotidase P, et al. Ultrastructural
immunoreactivity in peritubular localization
cells of rato renal
carbonic
cor-
kidneyMurakami
336. a ter adaptation to alteredvessels
T. Glomerular Na- and ofK-intake. II. Changes
the rat kidney. in Na-K-ATPase
A scanning electron anhydrase
tex indicates ll inthat
subpopulations
broblasts produceo intercalated cells o the
erythropoietin. rat kidney.Cytochem.
J Histochem J Am Soc
activity. Cell study
microscope TissueofRes. 1982;224:493–504.
corrosion casts. Arch Histol Jpn. 1972;34:87. Nephrol. 1990;1:245–256.
1993;41:335–341.
http://www.ncbi.nlm.nih.gov/pubmed/6288247
337. Winkler D, Elger M, Sakai T, et al. Branching and con uence pattern of http://www.ncbi.nlm.nih.gov/pubmed/2129509
367. Bulger R, Grif th L, Trump B. Endoplasmic reticulum in rat renal inter-
262. Mutoarterioles
glomerular S, Yasoshimain theK,rat. Yoshitomi
Kidney Int.K, 1991;39:S-2–S-8.
et al. Electrophysiological identif ca- 287.
stitial Humbert F, Pricam
cells: Molecular C, Perrelet A,after
rearrangement et al. Specif
water c plasma membrane
deprivation. Science. 1966;di -
tion oYamamoto
338. a - and b -intercalated
K, Wilson D, cells Baumal andR.their
Blood distribution
supply andalong the rabbit
drainage distal
of the outer erentiations in the cells o the kidney collecting tubule. J Ultrastruct Res.
151:83–86.
nephron of
medulla segments. J Clin Invest.
the rat kidney. Scanning 1990;86:1829–1839.
electron microscopy of microvascular casts. 1975;52:13–20.
368. Brown C, Zusman R, Haber E. Identi cation of an angiotensin receptor
263. Rec.
Anat Meneton P, Lo f ng J, Warnock D. Sodium and potassium handling by the
1984;210:273. http://www.ncbi.nlm.nih.gov/pubmed/1152104
in rabbit renomedullary interstitial cells in tissue culture. Circ Res. 1980;46:
aldosterone-sensitive
339. Bulger R, Trump distal
B. nephron: the pivotal
Fine structure of therolerato renal
the distal and Am
papilla. connecting
J Anat. 288.
802–807. Verlander J, Madsen K, Galla J, et al. Response o intercalated cells to chlo-
tubule. Am J Physiol. 2004;287:F593–F601.
1966;118:685–722. ride
369.depletion
Kuroda M, metabolic
Ueno H, alkalosis.
SakatoAm S, JetPhysiol.
al. A 1992;262:F309–F319.
unique af nity and adaptation
264. Schwartz
340. Kim J, Kim M,YKarnovsky
, Cha J, et al. M,Intercalated
Venkatachalam cell subtypes in connecting
M. Ultrastructural tubule
differences 289. Wagner C, Finberg
of renomedullary K, Breton
interstitial cells S,for
et al. Renal vacuolar
hypertonic medium.H+ -ATPase. Physiol
Prostaglandins.
and cortical
between rat collecting duct o descending
inner medullary rat and mouse. andJascending
Am Soc Nephrol. 1999;10:1–12.
vasa recta. Lab Invest. Rev. 2004;84:1263–1314.
1979;18:209–220.
http://www.ncbi.nlm.nih.gov/pubmed/9890303
1976;35:161–170. 290. Brown D,
370. Pitcock Orci L.H,The
J, Lyons “coat”
Brown o al.
P, et kidney intercalated cell of
Glycosaminoglycans tubulovesicles
the rat renomed-does
265. Kriz
341. Hansen G, Tisher H.
W, Dieterich C, Robinson
Das LymphgefäR. Response
systemo der the Niere
collecting duct to Säuge-
bei einigen distur- not contain
ullary clathrin.Ultrastructural
interstitium: Am J Physiol. 1986;250:C605–C608.
and biochemical observations. Exp Mol Pathol.
bances Licht-
tieren. o acid-base and potassium balance. Kidney
und elektronenmikroskopische Int. 1980;17:326–337.
Untersuchungen. Z Anat Entwick- 291. Brown D, Gluck S, Hartwig J. Structure o the novel membrane-coating
1988;49:373–387.
http://www.ncbi.nlm.nih.gov/pubmed/7401452
lungsgesch. 1970;131:111–147. material in proton-secreting
371. Muirhead epithelial cells
E. The renomedullary systemand of identif
bloodcation as ancontrol.
pressure H+ATPase. Am J
266. Lemley
342. Pricam C, K, Humbert F, Perrelet of
Kriz W. Anatomy A, the
et al.renal
A reeze-etch
interstitium.studyKidney
o the tight junc-
Int. 1991; Cell
MedBiol. 1987;105:1637–1648.
Sci. 1988;29531:231–251.
tions o the rat kidney tubules. Lab Invest. 1974;30:286–291.
39:370–381. 292. Stetson D,G.
372. Gothberg Wade J, Giebisch
Physiology of the G. renomedullary
Morphologic alterationsdepressorinsystem.
the ratJmedul-
Hyper-
http://www.ncbi.nlm.nih.gov/pubmed/4817823
343. Kaissling B, Le Hir M. The renal cortical interstitium: morphological and lary
tens.collecting duct ollowing potassium depletion. Kidney Int. 1980;17:45–56.
1994;12:S57–S64.
267. Gri faspects.
functional th L, Bulger
HistochemR, Trump B. Structure
Cell Biol. and staining o mucosubstance
2008;130:247–262. http://www.ncbi.nlm.nih.gov/pubmed/7374020
373. Steinman R. The dendritic cell system and its role in immunogenicity. Annu
on “intercalated
344. Larson M, cells” Sjoquist romM,theWolgast
rat distalM. tubule
Renal and collecting
interstitial volumeduct.ofAnat
the Rec.
rat 293. Brown D,
Rev Immunol. Sabolic I. Endosomal pathways or water channel and proton
1991;9:271–296.
1968;160:643–662.
kidney. Acta Physiol Scand. 1984;120:297–304. pump recycling in
374. Diamond kidney
J, van Goorepithelial
H, Ding cells.
G, JetCell
al. Sci.
Myo1993;Suppl.
broblasts in 17:49–59.
experimental
http://www.ncbi.nlm.nih.gov/pubmed/4174122
345. Pfaller W, Rittinger M. Quantitative morphology of the rat kidney. Int J http://www.ncbi.nlm.nih.gov/pubmed/8144705
hydronephrosis. Am J Pathol. 1995;146:121–129.
268. Rastegar
Biochem. A, Biemesder er D, Kashgarian M, et al. Changes in mem-
1980;12:17–22. 294. Kriz W, Kaissling
375. Desmouliere B. Structural
A, Darby I, Gabbiani organization
G. Normaloand thepathologic
mammalian softkidney.
tissue
brane Pinter
346. sur aces o collecting
G, Gärtner duct cellscapillary,
K. Peritubular in potassium adaptation.
interstitium, and lymphKidney Int.
of the In: Seldin D,role
remodeling: Giebisch G, eds.
of the myo The Kidney.
broblast, with Philadelphia:
special emphasis Lippincott
on liver Williams
and kidney &
1980;18:293–301.
renal cortex. Rev Physiol Biochem Pharmacol. 1984;99:184–202. Wilkins;
brosis. Lab2000:587–654.
Invest. 2003;83:1689–1707.
http://www.ncbi.nlm.nih.gov/pubmed/6257963
347. Swann H, Norman R. The periarterial spaces of the kidney. Tex Rep Biol 295. Brown M,
376. Iwano D, Neilson
Kumpulainen T, Roth of
E. Mechanisms J, tubulointerstitial
et al. Immunohistochemicalbrosis. Curr local-
Opin
269. 1970;28:317–334.
Med. Wade J, O’Neil R, Pryor J, et al. Modulation o cell membrane area in renal ization
NephroloHypertens.
carbonic2004;13:279–284.
anhydrase in postnatal and adult rat kidney. Am J Physiol.
collecting
348. Pedersen tubules by corticosteroid
J, Persson A, Maunsbach hormones. J Cell Biol.and
A. Ultrastructure 1979;81:439–445.
quantitative charac- 1983;245:F110–F118.
377. Barnes J, Glass W. Renal interstitial brosis: A critical evaluation of the
http://www.ncbi.nlm.nih.gov/pubmed/468913
terization of the cortical interstitium in the rat kidney. In: Maunsbach A, Olsen T, 296.
origin Dobyan
of myo D, Magill L,Contrib
broblasts. Friedman P, et 2011;169:97–123.
Nephrol. al. Carbonic anhydrase histochemistry
270. Stanton
Christensen B, Janzen
E, eds. FunctionalA, Klein-Robbenhaar
Ultrastructure of theG, et al.London:
Kidney. Ultastucture
Academico rat ini-
Press; in
378.rabbit
Smithand H.mouse
The kidneys. Anat Rec.and
Kidney: Structure 1982;204:185–197.
Function in Health and Disease. New
tial collecting tubule. E ect o adrenal corticosteroid treatment. J Clin Invest.
1980:443–457. http://www.ncbi.nlm.nih.gov/pubmed/6818876
York: Oxford University Press; 1951.
1985;75:1327–1334.
349. Kriz W, Napiwotzky P. Structural and functional aspects of the renal inter- 297. Dobyan D, J.Bulger
379. Stephenson R. Renalofcarbonic
Concentration urine in aanhydrase.
central core Am J Physiol.
model of the1982;
renal
http://www.ncbi.nlm.nih.gov/pubmed/2985657
stitium. Contr. Nephrol. 1979;16:104–108. 243:F311–F324.
counter ow system. Kidney Int. 1972;2:85–94.
271. Kriz
350. Sun W. T, Hoek W, Huang
A periarterial Y, et al.for
pathway Aquaporin-2 localization in
intrarenal distribution of clathrin-coat-
renin. Kidney 298. MadsenJ,K,
380. Kokko Tisher
Rector C. Countercurrent
F Jr. Response o intercalated multiplicationcells osystem
rat outer medullary
without active
ed pits:
Int. inhibition o endocytosis by dominant-negative dynamin. Am J Physiol.
1987;31:S-51–S-56. collecting
transport in duct to chronic
inner medulla.metabolic
Kidney Int. acidosis. Lab Invest. 1984;51:268–276.
1972;2:214–223.
2002;282:F998–F1011.
351. Albertine K, O’Morchoe C. Distribution and density of the canine renal http://www.ncbi.nlm.nih.gov/pubmed/6471808
381. Becker B. Quantitative Beschreibung der Innenzone der Rattenniere. Muenster:
272. Lolymphatic
cortical f ng J, Cueni-Lo f n D, Macher
system. Kidney A, et al. Localization o epithelial so-
Int. 1979;16:470–480. 299.
InauguralMadsen K, Tisher1978.
dissertation; C. Cellular response to acute respiratory acidosis in rat
dium Knepper
352. channel and aquaporin-2
M, Danielson in rabbit
R, Saidel G, etcortex. Am J Physiol
al. Quantitative Renal ofPhysiol.
analysis renal medullary
382. Han collecting
J, Thompson duct.K,Am J Physiol.
Chou C, et1983;14:F670–F679.
al. Experimental tests of three-dimen-
2000;272:530.
medullary anatomy in rats and rabbits. Kidney Int. 1977;12:313–323. 300.
sional Brown
model D, of Orci
urinaryL. Junctional
concentrating complexes
mechanism. and cell
J Ampolarity in the 1992;2:
Soc Nephrol. urinary
273. Bohman
353. Marver D.S.Evidence o corticosteroid
The ultrastructure of the action
rat renalalong the nephron.
medulla Am J Phys-
as observed after tubule.
1677–1688. J Electron Microsc Tech. 1988;9:145–170.
iol. 1984;246:F111–F123.
improved xation methods. J Ultrastruct Res. 1974;47:329–360. http://www.ncbi.nlm.nih.gov/pubmed/3058889
383. Kriz W. Structural organization of renal medullary circulation. Nephron.
274. Bulger
354. MadsenR,K,Nagle ClappR.W, Verlander J. Structure
Ultrastructure and unction
of the interstitium o the
in the inner
rabbit med-
kidney. 301. Weiner I, Hamm L. Regulation o intercellular pH in the rabbit cortical
1982;31:290–295.
ullary-collecting
Am duct. Kidney Int. 1988;34:441–454.
J Anat. 1973;136:183–204. collecting
384. Layton tubule. J Clin Invest.of
H. Distribution 1990;85:274–281.
Henle’s loops may enhance urine concentrating
http://www.ncbi.nlm.nih.gov/pubmed/3059025
355. Osvaldo L, Latta H. Interstitial cells of the renal medulla. J Ultrastruct Res. http://www.ncbi.nlm.nih.gov/pubmed/2153152
capability. Biophys J. 1986;49:1033–1040.
275. Bankir L, Trinh-Trang-Tan M. Urea and the kidney. In: Brenner B, ed. The
1966;15:589–613. 302. Verlander
385. Lemley K, J,Kriz
Madsen K, Stone
W. Cycles andD,separations:
et al. Ultrastructural localization of
The histotopography o the
H+
Kidney.Kaissling
356. Philadelphia:
B, HegyiSaunders;
I, Lof ng 2000:637–679.
J, et al. Morphology of interstitial cells in the ATPase
urinary in rabbit cortical
concentrating collecting
process. Kidneyduct.Int.J Am Soc Nephrol. 1994;44: 1546–1557.
1987;31:538–548.
276. Alper
healthy kidney.S, Natale J, Gluck 1996;193:303–318.
Anat Embryol. S, et al. Subtypes o intercalated cells in rat kidney http://www.ncbi.nlm.nih.gov/pubmed/8025228
386. Trinh-Trang-Tan M, Lasbennes F, Gane P, et al. UT-B1 proteins in rat: tissue
collecting
357. duct def
Kaissling B, LenedHirbyM. antibodies against erythroid
Characterization band 3 and
and distribution renal vascular
of interstitial cell 303. Emmons
distribution and C, Kurtz I.byFunctional
regulation antidiureticcharacterization
hormone in kidney. o three intercalated
Am J Physiol Renal
H+ -ATPase.
types Proc Natl
in the renal cortex Acad Sci USA.
of rat. Kidney 1989;80:5429–5433.
Int. 1994;45:709–720. cell subtypes
Physiol. in the rabbit outer cortical collecting duct. J Clin Invest. 1994;93:
2002;283:F912–F922.
http://www.ncbi.nlm.nih.gov/pubmed/2526338 417–423.
(contd.)
45
46
44 SECTION I ■ STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

304. Kim
264. Milton A, Weiner
J, Kim Y, ChaI.J,Regulation o B-type
et al. Intercalated cellintercalated
subtypes incell apical anion
connecting ex-
tubule 328. Schweda
295. Brown D, F,Kumpulainen
Kurtz A. Cellular T, Rothmechanism
J, et al. oImmunohistochemical
renin release. Acta Physiollocal-
change
and activity
cortical by CO2/HCO3.
collecting duct of ratAm andJ Physiol.
mouse.1998;275:F1086–F1094.
J Am Soc Nephrol. 1999;10:1–12. Scand.
ization2004;181:383–390.
of carbonic anhydrase in postnatal and adult rat kidney. Am J Physiol.
305. Hansen
265. Schwartz G,G, Al-Awqati
Tisher Q. Carbon
C, Robinson dioxideofcauses
R. Response exocytosis
the collecting ductotovesicles
distur- http://www.ncbi.nlm.nih.gov/pubmed/15283749
1983;245:F110–F118.
containing
bances H+ pumps
of acid-base andinpotassium
isolated perbalance.
used proximal
Kidney Intand collecting tubules. J Clin
. 1980;17:326–337. 329. OsswaldD,
296. Dobyan H,Magill
Nabakowski G, Hermes
L, Friedman H. Carbonic
P, et al. Adenosineanhydrase
as a possible mediator o
histochemistry
Invest.Pricam
266. 1985;75:1638–1644.
C, Humbert F, Perrelet A, et al. A freeze-etch study of the tight junc- metabolic
in rabbit and control
mouseo glomerular
kidneys. Anat f ltration rate. Int J Biochem. 1980;12:263–267.
Rec. 1982;204:185–197.
306. of
tions Quentin F, Chambrey
the rat kidney tubules.R, Trinh-Trang-Tan M, et al. The Cl/HCO3 exchang-
Lab Invest. 1974;30:286–291. http://www.ncbi.nlm.nih.gov/pubmed/7399032
297. Dobyan D, Bulger R. Renal carbonic anhydrase. Am J Physiol. 1982;
er pendrin
267. Grif in th rat
L, kidney
Bulger R, is regulated
Trump B.inStructure
response andto chronic
staining alteration in chloride
of mucosubstance 330. Castrop H, Huang Y, Hashimoto S, et al. Impairment o tubuloglomerular
243:F311–F324.
balance.
on Am J Physiol
“intercalated cells”Renal
fromPhysiol.
the rat2004;287:1179–1188.
distal tubule and collecting duct. Anat Rec. eedback
298. regulation
Madsen K, Tishero GFR in ecto-5’-nucleotidase/CD73-def
C. Response of intercalated cells of ratcient outermice. J Clin
medullary
307. Wall S, Hassell K, Royaux I, et al. Localization o pendrin in mouse kidney.
1968;160:643–662. Invest. 2004;114:634–642.
collecting duct to chronic metabolic acidosis. Lab Invest. 1984;51:268–276.
Am
268.J Physiol
Rastegar Renal Physiol. 2003;284:F229–F241.
A, Biemesderfer D, Kashgarian M, et al. Changes in membrane sur- http://www.ncbi.nlm.nih.gov/pubmed/15343381
299. Madsen K, Tisher C. Cellular response to acute respiratory acidosis in rat
308. ofKim
faces Y, Kwon
collecting ductT, cells
Frische S, et al. Immunocytochemical
in potassium adaptation. Kidney Int.localization
1980;18:293–301.o pen- 331. Thomson
medullary S, Bao
collecting D, Am
duct. Deng A, et .al.
J Physiol Adenosine ormed by 5’-nucleotidase
1983;14:F670–F679.
drin inWade
269. intercalated
J, O’Neilcell subtypes
R, Pryor in Modulation
J, et al. rat and mouse kidney.
of cell Am J Physiol
membrane area in Renal
renal mediates
300. Brown tubuloglomerular
D, Orci L. Junctionaleedback.complexes
J Clin Invest. and2000;106:289–298.
cell polarity in the urinary
Physiol. 2002;283:F744–F754.
collecting tubules by corticosteroid hormones. J Cell Biol. 1979;81:439–445. http://www.ncbi.nlm.nih.gov/pubmed/10903345
tubule. J Electron Microsc Tech. 1988;9:145–170.
309. Stanton
270. Madsen B, K,Janzen
Verlander J, Kim J, Tisher C.
A, Klein-Robbenhaar G, etMorphological
al. Ultastucture adaptation
of rat ini-o 332. Persson I,A,Hamm
301. Weiner Ollerstam A, Liu R, of
L. Regulation et intercellular
al. Mechanism pH or macula
in the densa
rabbit cell
cortical
the collecting tubule.
tial duct to Effect
acid-base disturbances.
of adrenal Kidney treatment.
corticosteroid Int. 1991;40, Suppl.33:
J Clin Invest. release
collectingo renin.
tubule.Acta Physiol
J Clin InvestScand. 2004;181:471–474.
. 1990;85:274–281.
S57–S63.
1985;75:1327–1334. 333. Komlosi P, J,Fintha
302. Verlander Madsen A, Bell
K, PStone
. CurrentD, mechanisms o maculalocalization
et al. Ultrastructural densa cell sig-
of
310. Sun
271. Barajas L. The
T, Hoek ultrastructure
W, Huang o the juxtaglomerular
Y, et al. Aquaporin-2 localization apparatus as dis-
in clathrin-coated naling.
H ATPase Acta Physiol
in rabbit Scand. 2004;181:463–469.
cortical collecting duct. J Am Soc Nephrol. 1994;44:
closedinhibition
pits: by three-dimensional
of endocytosis reconstrucion rom serialdynamin.
by dominant-negative sections: TheAm Janatomi-
Physiol. http://www.ncbi.nlm.nih.gov/pubmed/15283759
1546–1557.
cal relationship between the tubular and vascular components. J Ultrastruct Res.
2002;282:F998–F1011. 334. Mo at D,C,Fourman
303. Emmons Kurtz I.J. Functional
The vascular pattern o theofrat
characterization kidney.
three J Anat.
intercalated
1970;33:116–147.
272. Lof ng J, Cueni-Lof n D, Macher A, et al. Localization of epithelial sodium 1963;97:543–553.
cell subtypes in the rabbit outer cortical collecting duct. J Clin Invest. 1994;93:
http://www.ncbi.nlm.nih.gov/pubmed/4922486
channel and aquaporin-2 in rabbit cortex. Am J Physiol Renal Physiol. 2000;272:530. http://www.ncbi.nlm.nih.gov/pubmed/14064096
417–423.
311. Marver
273. Barajas L, D. Latta H. Structure
Evidence o the juxtaglomerular
of corticosteroid action along the apparatus.
nephron. CircAm Res.J 335. Aikens A,
304. Milton B, Eenboom
Weiner I. A,Regulation
Bohle A. Untersuchungen
of B-type intercalated zur Struktur des Glom-
cell apical anion
1967;20–21
Physiol Suppl. 2:15.
. 1984;246:F111–F123. erulum.
exchangeVirchows
activity Arch A Pathol Anat
by CO2/HCO3. AmHistopathol. 1979;381:283–293.
J Physiol. 1998;275:F1086–F1094.
http://www.ncbi.nlm.nih.gov/pubmed/4970997
274. Madsen K, Clapp W, Verlander J. Structure and function of the inner med- http://www.ncbi.nlm.nih.gov/pubmed/155927
305. Schwartz G, Al-Awqati Q. Carbon dioxide causes exocytosis of vesicles
312. Kaissling B,duct.
ullary-collecting KrizKidney
W. Variability o intercellular spaces between macula den-
Int. 1988;34:441–454. 336. Murakami
containing H pumps T. Glomerular
in isolated vessels o theproximal
perfused rat kidney. andAcollecting
scanning tubules.
electron
sa cells:
275. A transmission
Bankir electron microscopic
L, Trinh-Trang-Tan M. Urea andstudy in rabbits
the kidney. In:and rats. Kidney
Brenner Int.
B, ed. The microscope
J Clin Invest.study o corrosion casts. Arch Histol Jpn. 1972;34:87.
1985;75:1638–1644.
1982;22:9–17.
Kidney. Philadelphia: Saunders; 2000:637–679. http://www.ncbi.nlm.nih.gov/pubmed/5062282
306. Quentin F, Chambrey R, Trinh-Trang-Tan M, et al. The Cl/HCO3 exchang-
http://www.ncbi.nlm.nih.gov/pubmed/6957682
276. Alper S, Natale J, Gluck S, et al. Subtypes of intercalated cells in rat kidney 337. Winkler
er pendrin in ratD,kidney
Elger is M,regulated
Sakai T, in et response
al. Branching and con
to chronic uenceinpattern
alteration chlorideo
313. Bachmann
collecting duct deS,ned Mundel P. Nitric against
by antibodies oxide inerythroid
the kidney: bandsynthesis,
3 and renal localization
vascular glomerular
balance. Amarterioles
J Physiol in the Physiol
Renal rat. Kidney Int. 1991;39:S-2–S-8.
. 2004;287:1179–1188.
Hand -ATPase.
unction.Proc Am Natl
J Kidney
AcadDis. Sci 1994;24:112–129.
USA. 1989;80:5429–5433. 338. Yamamoto
307. Wall S, HassellK, Wilson
K, Royaux D, Baumal
I, et al.R. Blood supply
Localization and drainage
of pendrin o the
in mouse outer
kidney.
http://www.ncbi.nlm.nih.gov/pubmed/7517625
277. Brown D, Hirsch S, Gluck S. Localization of a proton-pumping ATPase in medulla
Am J Physiolo the rat Physiol
Renal kidney.. 2003;284:F229–F241.
Scanning electron microscopy o microvascular casts.
314.
rat Wilcox
kidney. C, Invest
J Clin Welch W, Murad F, et al. Nitric oxide synthase in macula
. 1988;82:2114–2126. Anat
308. Rec.
Kim1984;210:273.
Y, Kwon T, Frische S, et al. Immunocytochemical localization of pen-
densa Verlander
278. regulates J, glomerular
Madsen K,capillary
Tisher C.pressure.
Effect ofProc
acuteNatl Acad Sciacidosis
respiratory USA. 1992;89:
on two http://www.ncbi.nlm.nih.gov/pubmed/6507893
drin in intercalated cell subtypes in rat and mouse kidney. Am J Physiol Renal
11993–11997.
populations of intercalated cells in rat cortical collecting duct. Am J Physiol Renal 339.
PhysiolBulger R, Trump B. Fine structure o the rat renal papilla. Am J Anat.
. 2002;283:F744–F754.
http://www.ncbi.nlm.nih.gov/pubmed/1281548
Fluid Electrolyte Physiol. 1987;253:F1142–F1156. 1966;118:685–722.
309. Madsen K, Verlander J, Kim J, Tisher C. Morphological adaptation of the
315. Verlander
279. Harris R, J,McKanna Madsen K, J, Akai
TisherY,C.etTwoal. Cyclooxygenase-2
populations of intercalatedis associated with
cells exist http://www.ncbi.nlm.nih.gov/pubmed/5956103
collecting duct to acid-base disturbances. Kidney Int. 1991;40, Suppl.33:S57–S63.
thecortical
in maculacollecting
densa o ducts rat kidney
in theand rat. increases with salt restriction. J Clin Invest.
Clin Res. 1985;33:501A. 340. SchwartzL.M,
310. Barajas TheKarnovsky M, Venkatachalam
ultrastructure M. Ultrastructural
of the juxtaglomerular apparatus di erences
as dis-
1994;94:2504–2510.
280. Teng-Umnuay P, Verlander J, Yuan W, et al. Identi cation of distinct sub- between
closed byratthree-dimensional
inner medullary reconstrucion
descending and fromascending vasa recta.
serial sections: TheLab Invest.
anatomi-
316. Bachmann
populations S, Peters J,cells
of intercalated Engler
in theE, mouse
et al. Transgenic
collecting rats duct.carrying
J Am Soc theNephrol
mouse. 1976;35:161–170.
cal relationship between the tubular and vascular components. J Ultrastruct Res.
renin gene - Morphological characterization o a low-renin hypertension model.
1996;7:260–274. http://www.ncbi.nlm.nih.gov/pubmed/957604
1970;33:116–147.
KidneySchuster
281. Int. 1992;41:24.
V, Bonsib S, Jennings M. Two types of collecting duct mitochon- 341. Kriz W, L,
311. Barajas Dieterich
Latta H.H.Structure
Das Lymphge of theäbjuxtaglomerular
system der Niereapparatus.
bei einigen Säuge-
Circ Res.
http://www.ncbi.nlm.nih.gov/pubmed/1593860
dria-rich (intercalated) cells: Lectin and band 3 cytochemistry. Am J Physiol Cell tieren.
1967;20–21Licht-Suppl.
und elektronenmikroskopische
2:15. Untersuchungen. Z Anat Entwick-
317. Taugner
Physiol R, Hackenthal E, Rix E, et al. Immunocytochemistry o the renin-
. 1986;251:C347–C355. lungsgesch.
312. Kaissling 1970;131:111–147.
B, Kriz W. Variability of intercellular spaces between macula den-
angiotensin
282. Verlander system: Renin,K, angiotensinogen,
J, Madsen angiotensin ofI, band
Tisher C. Axial distribution angiotensin
3–positive II, http://www.ncbi.nlm.nih.gov/pubmed/5507220
sa cells: A transmission electron microscopic study in rabbits and rats. Kidney Int.
and converting
intercalated cells enzyme in the kidneys
in the collecting o mice,
duct of control andrats, and tree chloride-
ammonium shrews. loaded
Kidney 342. Lemley K, Kriz W. Anatomy o the renal interstitium. Kidney Int. 1991;
1982;22:9–17.
Int.1982;22
rabbits. Kidney (Suppl 12):S33–S43.
Int. 1996;57:S137–S147. 39:370–381.
313. Bachmann S, Mundel P. Nitric oxide in the kidney: synthesis, localization
318. LaFurgey
283. Barajas L. A, The development
Tisher C. Morphologyand ultrastructure o the juxtaglomerular
of rabbit collecting duct. Am J Anat cell. http://www.ncbi.nlm.nih.gov/pubmed/2062030
and function. Am J Kidney Dis. 1994;24:112–129.
granule. J Ultrastruct Res. 1966;15:400–413.
1979;155:111–124. 343. Kaissling
314. Wilcox C,B,Welch
Le HirW,M. The renal
Murad cortical
F, et al. Nitricinterstitium:
oxide synthase morphological
in macula den-and
http://www.ncbi.nlm.nih.gov/pubmed/4957359
284. O’Neil R, Hayhurst R. Functional differentiation of cell types of cortical unctional
sa regulates aspects. Histochem
glomerular Cell Biol.
capillary 2008;130:247–262.
pressure. Proc Natl Acad Sci USA. 1992;89:
319. Peterduct.
collecting S. Ultrastructural studies on the secretory process in the epitheloid
Am J Physiol. 1985;248:F449–F453. http://www.ncbi.nlm.nih.gov/pubmed/18575881
11993–11997.
cells oWeiner
285. the juxtaglomerular
I, Hamm L. Use apparatus.
of uorescent Cell Tissue Res. 1976;168:45–53.
dye BCECF to measure intracellular 344. Larson R,
315. Harris M,McKanna
Sjoquist M, Wolgast
J, Akai Y, etM.al.Renal interstitial volume
Cyclooxygenase-2 o the rat with
is associated kid-
http://www.ncbi.nlm.nih.gov/pubmed/1268928
pH in cortical collecting tubule. Am J Physiol. 1989;256:F957–F964. ney. Acta Physiol
the macula densaScand.
of rat1984;120:297–304.
kidney and increases with salt restriction. J Clin Invest.
320. Kim
286. RyanJ,G,Tisher AlcornC,D,LinserCoghlan P, etJ,al.
et Ultrastructural
al. Ultrastructural morphology
localization o gran-
of carbonic http://www.ncbi.nlm.nih.gov/pubmed/6369886
1994;94:2504–2510.
ule release llrom
anhydrase juxtaglomerularofmyoepithelioid
in subpopulations intercalated cellsandofperipolar cells. Kidney
the rat kidney. Int.
J Am Soc 345. P aller W Rittinger
316. Bachmann S, Peters M. Quantitative
J, Engler E, et al.morphology
Transgenic rats o the rat kidney.
carrying Jnt J
the mouse
1982;22. 1990;1:245–256.
Nephrol (Suppl.12):3–8. Biochem.
renin gene 1980;12:17–22.
- Morphological characterization of a low-renin hypertension model.
http://www.ncbi.nlm.nih.gov/pubmed/6752537
287. Humbert F, Pricam C, Perrelet A, et al. Speci c plasma membrane dif- 346.
KidneyPinter G, Gärtner K. Peritubular capillary, interstitium, and lymph o the
Int. 1992;41:24.
321. Gorgas in
ferentiations K. the
Structure
cells of andthe innervation o the juxtaglomerular
kidney collecting apparatus
tubule. J Ultrastruct Reso. renal
317. cortex.
TaugnerRev R, Physiol
Hackenthal Biochem Pharmacol.
E, Rix E, et al. 1984;99:184–202.
Immunocytochemistry of the renin-
the rat. Adv Anat Embryol Cell Biol. 1978;54:5–84.
1975;52:13–20. http://www.ncbi.nlm.nih.gov/pubmed/6695128
angiotensin system: Renin, angiotensinogen, angiotensin I, angiotensin II, and
http://www.ncbi.nlm.nih.gov/pubmed/358773
288. Verlander J, Madsen K, Galla J, et al. Response of intercalated cells to chlo- 347. Swannenzyme
converting H, Norman in theR.kidneys
The periarterial
of mice, rats, spaces ando tree
the kidney.
shrews.Tex Rep Biol
Kidney Int.
322.depletion
ride Biava C, metabolic
West M. Fine structures
alkalosis. Am JoPhysiol
normal human juxtaglomerular cells. I.
. 1992;262:F309–F319. Med.
1982;221970;28:317–334.
(Suppl 12):S33–S43.
General
289. structure
Wagner and intercellular
C, Finberg K, Bretonrelationship.
S, et al. Renal Amvacuolar
J Pathol.H1966;49:-ATPase. 697–721.
Physiol http://www.ncbi.nlm.nih.gov/pubmed/5521884
318. Barajas L. The development and ultrastructure of the juxtaglomerular cell
323.
Rev Taugner R, Schiller A, Kaissling B, et al. Gap junctional coupling between
. 2004;84:1263–1314. 348.
granule.Pedersen J, Persson
J Ultrastruct A, Maunsbach A. Ultrastructure and quantitative
Res. 1966;15:400–413.
the JGA
290. Brownand the glomerular
D, Orci tu t. Cell
L. The “coat” Tissue Res.
of kidney 1978;186:279–285.
intercalated cell tubulovesicles does charac terization
319. Peter o the corticalstudies
S. Ultrastructural interstitium
on theinsecretory
the rat kidney.
processIn: in Maunsbach
the epitheloidA,
http://www.ncbi.nlm.nih.gov/pubmed/627020
not contain clathrin. Am J Physiol. 1986;250:C605–C608. Olsen
cells ofT,the
Christensen
juxtaglomerularE, eds.apparatus.
Functional Cell Ultrastructure
Tissue Res.of1976;168:45–53.
the Kidney. London: Aca-
324. Brown
291. Goormaghtigh D, GluckN.S,Facts Hartwig in avour o an of
J. Structure endocrine
the novelunction o the renal
membrane-coating demic
320. RyanPress;G,1980:443–457.
Alcorn D, Coghlan J, et al. Ultrastructural morphology of gran-
arterioles.inJ proton-secreting
material Pathol Bacteriol. 1945;57:392.
epithelial cells and identi cation as an H ATPase. 349. Kriz W,
ule release from Napiwotzky
juxtaglomerularP. Structural and unctional
myoepithelioid aspects ocells.
and peripolar the renal
Kidneyinter-
Int.
J325. Schnermann
Cell Biol J, Levine D. Paracrine actors in tubuloglomerular eedback:
. 1987;105:1637–1648. stitium.
1982;22Contr. Nephrol. 1979;16:104–108.
(Suppl.12):3–8.
adenosine,
292. Stetson ATP , and
D, Wade nitric oxide. Annu
J, Giebisch Rev Physiol. 2003;65:501–529.
G. Morphologic alterations in the rat medul- 350. Kriz W. K.
321. Gorgas A periarterial
Structure and pathway or intrarenal
innervation distribution o renin.
of the juxtaglomerular Kidney
apparatus of
http://www.ncbi.nlm.nih.gov/pubmed/12208992
lary collecting duct following potassium depletion. Kidney Int. 1980;17:45–56. Int.
the 1987;31:S-51–S-56.
rat. Adv Anat Embryol Cell Biol. 1978;54:5–84.
326. Brown
293. Kurtz A. D,Renin
Sabolic release: sites, mechanisms,
I. Endosomal pathways for andwater
control. Annu and
channel Rev Physiol.
proton 351. Albertine
322. Biava K, O’Morchoe
C, West M. Fine C. Distribution
structures and density
of normal human o the canine renal
juxtaglomerular
2011;73:377–399.
pump recycling in kidney epithelial cells. J Cell Sci. 1993;Suppl. 17:49–59. cortical
cells. I. lymphatic system. Kidney
General structure Int. 1979;16:470–480.
and intercellular relationship. Am J Pathol. 1966;49:
http://www.ncbi.nlm.nih.gov/pubmed/20936939
294. Kriz W, Kaissling B. Structural organization of the mammalian kidney. http://www.ncbi.nlm.nih.gov/pubmed/548593
697–721.
327.Seldin
In: Peti-Peterdi
D, Giebisch J, Komlosi
G, eds.P,The Fuson A, etPhiladelphia:
Kidney. al. Luminal NaCl delivery
Lippincott regulates
Williams & 352. KnepperR,M,Schiller
323. Taugner Danielson R, Saidel
A, Kaissling B, G, et Gap
et al. al. Quantitative analysis between
junctional coupling o renal
basolateral
Wilkins; PGE2 release rom macla densa cells. J Clin Invest. 2003;112:76–82.
2000:587–654. medullary
the JGA and anatomy in rats and
the glomerular tuft.rabbits. Kidney
Cell Tissue ResInt. 1977;12:313–323.
. 1978;186:279–285.
http://www.ncbi.nlm.nih.gov/pubmed/12840061 http://www.ncbi.nlm.nih.gov/pubmed/604620
(contd.)
CHAPTER 1 ■ 47
45
STRUCTURAL–FUNCTIONAL RELATIONSHIPS IN THE KIDNEY 4 3

353. BohmanA,S.Pannabecker
204. Layton The ultrastructure
T, Dantzlero theW,rat renal
et al. Twomedulla
modes for as observed
concentratinga ter 369. KurodaR,M,
235. Greger Ueno E,
Schlatter H, Lang
Sakato S, et al.forAelectroneutral
F. Evidence unique a f nity and chloride
sodium adapta-
improved
urine in ratf xation methods.Am
inner medulla. J Ultrastruct Res. 1974;47:329–360.
J Physiol Renal Physiol. 2004;287:F816–F839. tion o renomedullary
cotransport in the cortical interstitial cells or limb
thick ascending hypertonic
of Henle’ medium.
s loop ofProstaglandins.
rabbit kidney.
354. Bulger R, Nagle
205. Verbavatz J, Brown R. Ultrastructure
D, Sabolic I, oet the al. interstitium in the rabbit
Tetrameric assembly kidney.
of CHIP28 1979;18:209–220.
P ugers Arch. 1983;396:308–314.
Am
waterJ Anat. 1973;136:183–204.
channels in liposomes and cell membranes: a freeze-fracture study. J Cell http://www.ncbi.nlm.nih.gov/pubmed/523679
236. Nielsen S, Maunsbach A, Ecelbarger C, et al. Ultrastructural localization of
http://www.ncbi.nlm.nih.gov/pubmed/4734341
Biol. 1993;123:605–618. 370. Pitcock
Na-K-2Cl J, Lyons H,inBrown
cotransporter thick Pascending
, et al. Glycosaminoglycans
limb and macula densa o the rat renomed-
of rat kidney.
355. Osvaldo M,
206. Hediger L, Latta
KnepperH. Interstitial cells o therecent
M. Introduction: renal insights
medulla.into J Ultrastruct
the urinaryRes. ullary interstitium:
Am J Physiol Ultrastructural
Renal Physiol and biochemical observations. Exp Mol Pathol.
. 1998;275:F885–F893.
1966;15:589–613.
concentrating mechanism: from cDNA cloning to dodelin renal function. Am J 1988;49:373–387.
237. Obermüller N, Kunchaparty S, Ellison D, et al. Expression of the Na-K-2Cl
http://www.ncbi.nlm.nih.gov/pubmed/5911654
Physiol. 1998;275:F317. http://www.ncbi.nlm.nih.gov/pubmed/2461872
cotransporter by macula densa and thick ascending limb cells of rat and rabbit
356. Kaissling B, HegyiM,I, Bankir
207. Trinh-Trang-Tan Lo f ngL.J, Integrated
et al. Morphology
functiono ofinterstitial cells in the
urea transporters in 371. Muirhead
nephron. E. The
J Clin Invest renomedullary system o blood pressure control. Am J
. 1996;98:635–640.
healthy kidney. Anat
the mammalian kidney.Embryol. 1996;193:303–318.
Exp Nephrol . 1998;6:471–479. Med
238.Sci.
Burg1988;29531:231–251.
M, Green N. Function of the thick ascending limb of Henle’s loop. Am
http://www.ncbi.nlm.nih.gov/pubmed/8694267
208. Shayakul C, Knepper M, Smith C, et al. Segmental localization of urea http://www.ncbi.nlm.nih.gov/pubmed/3284345
J Physiol. 1973;224:659–668.
357. Kaissling
transporter mRNAa B, LeinHir rat M. Characterization
kidney. Am J Physiol.and distribution o interstitial cell
1997;272:F654–F660. 372. Gothberg
239. Burg G. Physiology
M. Thick ascendingolimb theofrenomedullary depressor
Henle’s loop. Kidney Int. system. J Hyper-
1982;22:454.
types
209. in ImaitheM.renal cortexof
Function o the
rat.thin
Kidney Int. 1994;45:709–720.
ascending limb of Henle of rats and hamsters tens.
240.1994;12:S57–S64.
Spanidis A, Wunsch H, Kaissling B, et al. Three-dimensional shape of a
http://www.ncbi.nlm.nih.gov/pubmed/8196273
perfused in vitro. Am J Physiol. 1977;232:F201–F209. 373. Steinmancell
Goormaghtigh R. and
The its dendritic cell system
contact with andcell
a granular its inrole
theinrabbit
immunogenicity.
kidney. Anat
358. Takahashi-Iwanaga
210. Jamison R. Micropuncture H. Thestudythree-dimensional
of segments ofspacesthin loopso the kidney.in Cell
of Henle the Annu
EmbryolRev. 1982;165:239–252.
Immunol. 1991;9:271–296.
Tissue
rat. AmRes. 1991;264:269–281.
J Physiol . 1968;215:236–242. http://www.ncbi.nlm.nih.gov/pubmed/1910679
241. Dorup J. Ultrastructure of distal nephron cells in rat renal cortex. J Ultra-
http://www.ncbi.nlm.nih.gov/pubmed/1878946
211. Bulger R, Tisher C, Myers C, et al. Human renal ultrastructure. II. The 374. Diamond
struct Res J, van Goor H, Ding G, et al. Myof broblasts in experimental
. 1985;92:101–118.
359. Dawson
thin limb T, Gandhi
of Henle’ s loop and R, LetheHir M, et al. Ecto-5’-nucleotidase:
interstitium in healthy individuals.Localization
Lab Invest. hydronephrosis.
242. Kaissling B,Am J Pathol.
Stanton 1995;146:121–129.
B. Adaptation of distal tubule and collecting duct to in-
in rat kidney by light microscopic histochemical methods. J Histochem Cytochem.
1967;16:124–141. http://www.ncbi.nlm.nih.gov/pubmed/7856721
creased sodium delivery. I. Ultrastructure. Am J Physiol. 1988;255:F1256–F1268.
1989;37:39–47.
212. Tisher C, Bulger R, Trump B. Human renal ultrastructure. III. The distal 375. Desmouliere
243. Stanton A, DarbyB.I, Gabbiani
B, Kaissling AdaptationG. of Normal
distal andtubulepathologic
and and so tcollect-
tissue
http://www.ncbi.nlm.nih.gov/pubmed/2535703
tubule in healthy individuals. Lab Invest. 1968;18:655–668. remodeling:
ing duct to role o the myof
increased broblast,II.with
Na delivery. Na special
and emphasis
K transport. on liverAm and kidney.
J Physiol
360. Humphreys
213. Madsen B, Lin C.
K, Tisher SL,Structural-functional
Kobayashi A, et al. relationships
Fate tracing reveals
along the the distal
peri- f1988;255:F1269–F1275.
brosis. Lab Invest. 2003;83:1689–1707.
cyte and not
nephron. Am epithelial origin o myof broblasts in kidney f brosis. Am J Pathol.
J Physiol. 1986;250:F1–F15. 376. Iwano M,
244. Stanton B, Neilson
Biemesderfer E. Mechanisms
D, Wade J,o ettubulointerstitial
al. Structural and f brosis. Currstudy
functional Opin
2010;176:85–97.
214. Kaissling B, Peter S, Kriz W. The transition of the thick ascending limb of Nephrol
of the ratHypertens. 2004;13:279–284.
distal nephron: Effects of potassium adaptation and depletion. Kidney
http://www.ncbi.nlm.nih.gov/pubmed/20008127
Henle’s loop into the distal convoluted tubule in the nephron of thr rat kidney. http://www.ncbi.nlm.nih.gov/pubmed/15073485
Int. 1981;19:36–48.
361. Lin SL,
Cell Tissue Res.Kisseleva T, Brenner D, et al. Pericytes and perivascular f broblasts
1977;182:111–118. 377. Barnes J, M,
245. Farquhar Glass W Renal
Palade interstitialcomplexes
G. Junctional f brosis: Aincritical
variousevaluation
epithelia. oJ Cell
the
are
215.theKaissling
primary B, source
Kriz oW. collagen-producing
Morphology of the cells loopinofobstructive
Henle, distal f brosis
tubuleo and
the origin o myof broblasts. Contrib Nephrol. 2011;169:97–123.
Biol. 1963;17:375.
kidney.
collectingAmduct.
J Pathol.In: 2009;173:1617–1627.
Windhager E, ed. Handbook of Physiology: Section on Renal 378. Smith H.U,
246. Schmidt The Kidney:U.Structure
Dubach Activityand Function
of (Na K in Health andadenosine
)-stimulated Disease. Newtri-
362. Gandhi
Physiology. NewR,YLe ork,Hir
NY M, Kaissling
: Oxford B. Immunolocalization
University Press; 1992:109–167. o ecto-5’ -nucleoti- Y ork: Ox ord in
phosphatase University Press; 1951.
the rat nephron. P ugers Arch. 1969;306:219–226.
dase
216. inAllen
the kidney
F, TisherbyC.a Morphology
monoclonal antibody. Histochemie.
of the ascending thick 1990;95:
limb of Henle.165–174.
Kidney 379. Stephenson
247. Schmidt J. Concentration
U, Dubach o urine inadenosinetriphosphatase:
U. NaK-stimulated a central core model o the renal
Intracel-
http://www.ncbi.nlm.nih.gov/pubmed/2081690
Int. 1976;9:8–22. counter ow system.
lular localisation Kidney
within theInt. 1972;2:85–94.
proximal tubule of the rat nephron. P ugers Arch.
363. Le Hir M,
217. Crayen M, Kaissling
Thoenes B. W. Distribution
Architectureoand 5’-nucleotidase
cell structuresininthe therenal
distalintersti-
neph- http://www.ncbi.nlm.nih.gov/pubmed/4671532
1971;330:265–270.
tium
ron ofo the
theratrat.kidney.
Cell Tissue Res.. 1978;17:197–211.
Cytobiol 1989;258:177–182. 380. Kokko J, Rector
248. Kaissling F Jr. Countercurrent
B. Structural multiplication
aspects of adaptive changessystem without
in renal active
electrolyte
http://www.ncbi.nlm.nih.gov/pubmed/2553262
218. Myers C, Bulger R, Tisher C, et al. Human renal ultrastructure. IV. Collect- transport
excretion.inAm inner medulla.
J Physiol Kidney Int. 1972;2:214–223.
. 1982;243:F211–F226.
364.
ing ductKoury S, Koury
of healthy M, Bondurant
individuals. M, .et1966;15:1921–1950.
Lab Invest al. Quantitation o erythropoietin- 381. Becker B. B,
249. Kaissling Quantitative
Le Hir M.Beschreibung
Distal tubularder segments
Innenzone in derthe
Rattenniere. Muenster:
rabbit kidney after
producing
219. Suzuki cellsT, inMosto
kidneysF. oIntramitochondrial
mice by in situ hybridization:
lamentous Correlation
bodies in the withthick
he- Inaugural
adaptationdissertation;
to altered Na- 1978. and K-intake. I. Structural changes. Cell Tissue Res.
matocrit,
limb of Henle renaloferythropoietin
the rat kidney.mRNA, and
J Cell Biol serum erythropoietin concentration.
. 1967;33:605–623. 382. Han J, Thompson K, Chou C, et al. Experimental tests o three-dimen-
1982;224:469–492.
Blood. 1989;74:645–651.
220. Madsen K, Verlander J, Tisher C. Relationship between structure and func- sional model o B,urinary
250. Kaissling Bachmann concentrating
S, Kriz W. mechanism. J Am Soc of
Structural adaptation Nephrol. 1992;2:
the distal con-
http://www.ncbi.nlm.nih.gov/pubmed/2752138
tion in distal tubule and collecting duct. J Electron Microsc Tech. 1988;9:187–208. 1677–1688.
voluted tubule to prolonged furosemide treatment. Am J Physiol. 1985;248:
365. Lacombe
221. Kone C, Da K,
B, Madsen Silva J, Bruneval
Tisher P, et al. of
C. Ultrastructure Peritubular cells are the
the thick ascending limbsite
of 383. Kriz W. Structural organization o renal medullary circulation. Nephron.
F374–F381.
oHenle
erythropoietin
in the rat kidney.synthesisAm JinAnatthe. 1984;171:217–226.
murine hypoxic kidney. J Clin Invest. 1988; 1982;31:290–295.
251. Biner H, Arpin-Bott M, Lof ng J, et al. Human cortical distal nephron;
81:620–623.
222. Hebert S, Culpepper R, Andreoli T. ADH-stimulated NaCl transport in http://www.ncbi.nlm.nih.gov/pubmed/7177264
Distribution of electrolyte and water transport pathways. J Am Soc Nephrol.
http://www.ncbi.nlm.nih.gov/pubmed/3339134
mouse medullary thick ascending limbs. I. Evidence for ADH-stimulated neu- 384. Layton H. Distribution o Henle’s loops may enhance urine concentrating
2002;13:836–847.
366. Bachmann
tral NaCl S, Le and
co-transport Hir M, Eckardt nephron
functional K. Co-localization
heterogeneity. o erythropoietin
Am J Physiol. capability.
252. Lof Biophys J. 1986;49:1033–1040.
ng J, Cueni-Lof n D, Valderrabano V, et al. Distribution of transcel-
mRNA and ecto-5’ -nucleotidase immunoreactivity in peritubular cells o rat
1981;241:F412–F431. http://www.ncbi.nlm.nih.gov/pubmed/3708088
lular calcium and sodium transport pathways along mouse distal nephron. Am J
renal
223. cor tex indicates
Schiller A, Taugner thatR,f broblasts
Roesingerproduce erythropoietin.
B. Vergleichende J Histochem
Morphologie Cyto-
der Zonu- 385.
PhysiolLemley K, Kriz
Renal Physiol W. Cycles and separations: The histotopography o the
. 2001;281:F1019–F1020.
chem. 1993;41:335–341.
lae occludentes am Nierentubulus. Verh Anat Ges. 1978;72:229–234. urinary concentrating
253. Bachmann process. Kidney
S, Bostanjoglo Int. 1987;31:538–548.
M, Schmitt R, et al. Sodium transport-related
http://www.ncbi.nlm.nih.gov/pubmed/8429197
224. Bouby N, Bankir L, Trinh-Trang-Tan M, et al. Selective ADH-induced hy- http://www.ncbi.nlm.nih.gov/pubmed/3550222
proteins in the mammalian distal nephron - distribution, ontogeny and func-
367. Bulger
pertrophy of R,
theGri f th L, Trump
medullary thickB.ascending
Endoplasmic limbreticulum in rat rats.
in Brattleboro renalKidney
interstitial
Int. 386.
tional Trinh-Trang-Tan
aspects. Anat Embryol M, Lasbennes F, Gane P, et al. UT-B1 proteins in rat: tissue
. 1999;200:447–468.
cells: Molecular rearrangement a ter water deprivation. Science. 1966; 151:83–86.
1985;28:456–466. distribution
254. Potter and regulation
E. Normal andby antidiuretic
Abnormal hormone of
Development in the
kidney. Am. Chicago:
Kidney J Physiol Renal
Year
http://www.ncbi.nlm.nih.gov/pubmed/5908968
225. Bankir L, Fischer C, Fischer S, et al. Adaptation of the rat kidney to altered Physiol. 2002;283:F912–F922.
Book; 1972.
368. Brown and
water intake C, Zusman R, Haber E. Identif
urine concentration. P ugers cation
Arch.o1988;412:42–53.
an angiotensin receptor in 255. Howie A, Schmithson N, Rollason T. Reconsideration of the development
rabbit renomedullary
226. Bouby interstitial cells
N, Trinh-Trang-Tan in tissue
M, Laouari D,culture. CircofRes.
et al. Role the 1980;46: 802–807.
urinary concentrat- of the distal tubule of the human kidney. J Anat. 1993;183:141–147.
http://www.ncbi.nlm.nih.gov/pubmed/6247080
ing process in the renal effects of high protein intake. Kidney Int. 1988;34:4–12. 256. Kim J, Tisher C, Madsen K. Differentiation of intercalated cells in de-
227. Bankir L, Kriz W. Adaptation of the kidney to protein intake and to urine veloping rat kidney: An immunohistochemical study. Am J Physiol. 1994;266:
concentrating activity: Similar consequences in health and CFR. Kidney Int. F977–F990.
1995;47:7–24. 257. Stanton B. Renal potassium transport: morphological and functional adap-
228. Welling L, Welling D, Hill J. Shape of cells and intercellular channals in tations. Am J Physiol. 1989;257:R989–R997.
rabbit thick ascending limb of Henle. Kidney Int. 1978;13:144–151. 258. Lof ng J, Kaissling B. Sodium and calcium transport pathways along the
229. Hoyer J, Sisson S, Vernier R. Tamm-Horsfall glycoprotein. Ultrastructural mammalian distal nephron from rabbit to human. Am J Physiol Renal Physiol.
immunoperoxidase localization in rat kidney. Lab Invest. 1979;41:168–173. 2003;284:F628–F643.
230. Sikri K, Foster C, MacHugh N, et al. Localization of Tamm-Horsfall glyco- 259. Fridt G, Palmer L. Na channels in the rat connecting tubule. Am J Physiol
protein in the human kidney using immuno- uorescence and immuno-electron Renal Physiol. 2004;286:F669–F674.
microscopical techniques. J Anat. 1981;132:597–605. 260. Coleman R, Wu D, Liu J, Wade J. Expression of aquaporin in the renal
231. Chabardes D, Imbert-Teboul M, Gagnan-Brunette M, et al. Different hor- connecting tubule. Am J Physiol Renal Physiol. 2000;279:F874–F883.
monal target sites along the mouse and rabbit nephrons. Curr Probl Clin Biochem. 261. Le Hir M, Kaissling B, Dubach U. Distal tubular segments of the rabbit
1977;8:447–454. kidney after adaptation to altered Na- and K-intake. II. Changes in Na-K-ATPase
232. Morel F, Imbert-Teboul M, Chabardes D. Receptors to vasopressin and activity. Cell Tissue Res. 1982;224:493–504.
other hormones in the mammalian kidney. Kidney Int. 1987;31:512–520. 262. Muto S, Yasoshima K, Yoshitomi K, et al. Electrophysiological identi ca-
233. El-Mernissi G, Doucet A. Quantitation of [3H]ouabain binding and turnover tion of - and -intercalated cells and their distribution along the rabbit distal
of Na -K-ATPase along the rabbit nephron. Am J Physiol. 1984;247:F158–F167. nephron segments. J Clin Invest. 1990;86:1829–1839.
234. Greger R, Schlatter E. Presence of luminal K , a prerequisite for active 263. Meneton P, Lof ng J, Warnock D. Sodium and potassium handling by the
NaCl transport in the cortical thick ascending limb of Henle’s loop of rabbit aldosterone-sensitive distal nephron: the pivotal role of the distal and connecting
kidney. P ugers Arch. 1981;392:92–94. tubule. Am J Physiol. 2004;287:F593–F601.
C H A P T E R

2 Clinical Importance of Nephron Mass


Valerie A. Luyckx • Thomas F. Mueller

being re ected in their present, and affecting their future


INTRODUCTION risk of hypertension and renal disease. Although tradition-
The relationship between renal salt handling, intravascular ally it has been thought that all kidneys have about 1 million
volume homeostasis, and hypertension is well established, nephrons, recent studies have found that total glomerular
which points to the kidney as the central organ in the de- number varies up to 13-fold in human kidneys, much more,
velopment of hypertension.1 Based on the concept of devel- for example, than height or weight (Table 2.1).13 The terms
opmental programming, where an environmental stimulus “nephron endowment,” implying number of nephrons pres-
experienced during a critical period of early development ent upon completion of nephrogenesis; “nephron number,”
can induce long-term structural and functional adaptive implying the number of intact nephrons at the time of mea-
changes in the developing organism, Brenner et al. pro- surement; and “glomerular number,” including the number
posed that a low nephron number, acquired during fetal of tubular and atubular glomeruli, have all been used in-
life, may predispose an individual to hypertension and renal terchangeably.13 In this chapter we use the term “nephron
disease.2,3 This hypothesis was attractive because low birth number” more generally to describe the total number of
weight, a marker of an adverse intrauterine environment, if nephrons in a kidney at the time of discussion. The term
associated with a congenital reduction in nephron number, nephron mass is used more broadly as a clinical term to in-
could potentially explain the variability in hypertension and corporate nephron number, kidney weight, kidney size, and
renal disease prevalence observed among populations of dif- kidney volume. A discussion of the importance of acquired
ferent ethnicity where those with lower birth weights tend to reduction in nephron mass in later life is beyond the scope
have a higher burden of renal disease.4–7 The initial hypothe- of this chapter.
sis suggested that a kidney with fewer nephrons would have
a reduced ltration surface area with a limited capacity to
excrete sodium, thereby contributing to the development of DEVELOPMENTAL DETERMINANTS OF
hypertension. Although this “nephron number” hypothesis NEPHRON NUMBER
was initially quite controversial, and with time has proved
to be not entirely this straightforward, the association be- Low Nephron Number
tween nephron number and predisposition to hypertension Kidney development in humans proceeds from the 9th to the
and renal disease has been borne out in many animal experi- 36th week of gestation.9,14 Accurate determination of neph-
ments and human studies.8–12 In this chapter we put forward ron number is dif cult because nephron number cannot be
the existing evidence comprising animal and human studies, determined in humans in vivo. The unbiased fractionator-
which link nephron mass, birth weight, and other clinical sampling/dissector method is thought to be the most objec-
variables with clinical outcomes. Extrapolation from ani- tive nephron counting method, and is currently utilized in
mals to humans has many limitations and, therefore, where most human studies.15,16 This method, however, requires
possible we have included human studies to corroborate or postmortem kidney samples and is very labor intensive. An
refute animal ndings. Although this eld has grown signi - in vivo glomerular counting method comparing the fraction-
cantly within the last decade and many questions remain un- ator technique with a combined renal biopsy/magnetic reso-
answered, a clearer and consistent picture is emerging that nance imaging (MRI) method in explanted canine kidneys
shows nephron mass does have clinical importance. has been attempted.17 This study found a good correlation
An individual’s nephron mass is determined by a com- of glomerular number on average between the two meth-
plex interplay between genetics and environment, evolving ods, but, within kidneys, there was a 36% variance, calling
throughout their lifetime, bearing the imprint of their past, individual applicability into question. Large-scale human

46
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 447
7

TA B L E

2.1 Nephron Numbers in Humans

Reference Population Sample Mean Range Fold


Size

Nyengaard and Bendtsen Danish 37 617,000 331,000–1,424,000 4.3

Merlet-Benichou et al.a French 28 1,107,000 655,000–1,554,000 2.4

Keller et al. German 20 1,074,414 531,140–1,959,914 3.7


Hypertensive 10 702,379 531,104–954,893 1.8
Normotensive 10 1,429,200 884,458–1,959,914 2.2

Douglas-Denton et al. African Americans 105 884,938 210,332–2,026,541 9.6


White Americans 84 843,106 227,327–1,660,232 7.3

Hoy et al. Australian non- 21 861,541 380,517–1,493,665 3.9


Aborigines
Australian Aborigines 19 713,209 346,161–1,129,223 3.1

McNamara et al.b Senegalese 47 992,353 536,171–1,764,241 3.3

Hoy et al. African and white 420 901,902 210,332–2,702,079 12.8


Americans, Australian
Aborigines, and non-
Aborigines and
Senegalese

a
Used acid maceration technique. All other studies used unbiased stereology.
b
Values for 47 participants were combined from two publications.
Reprinted with permission from Puelles VG, Hoy WE, Hughson MD, et al. Glomerular number and size variability and risk for kidney disease. Curr Opin
Nephrol Hypertens. 2011;20:7–15. See original manuscript for detailed references.

studies of nephron number and association with phenotype beyond the scope of this chapter and are outlined in detail
are therefore not easily feasible. elsewhere.8,21,22 Extrapolating from the animal studies, from
Average nephron number has been reported to range a clinical point of view, the factors associated with develop-
from 617,000 (range 331,000–1,424,000) to 1,429,200 ment of low nephron number can be divided into two groups:
(range 884,485–1,959,914) per kidney among normal adult modi able and nonmodi able, as outlined in Table 2.2.
Caucasian Europeans.10,18 Other studies including subjects
of multiple ethnic origins from the United States, Africa, Modi able Factors
and Australia showed somewhat similar results, with a mean Modi able factors associated with low nephron number in-
number around the mid 800,000 glomeruli per kidney, with clude prenatal events—factors occurring during gestation
a very wide range, from 210,332 to 2,702,079 as shown in and postnatal events occurring in the neonate.
Table 2.1.13 The range appears widest in kidneys from sub-
jects of African origin.13,19 In general, nephron numbers are Prenatal Factors. Maternal diets de cient in protein, to-
lower in older subjects, attributed to age-related glomerulo- tal calories, or iron have all been shown to reduce nephron
sclerosis and obsolescence.18,20 Whether the high variability numbers in offspring of experimental animals, most of-
in nephron number across populations re ects true differ- ten in association with low birth weight.12,23–26 Figure 2.1
ences or is confounded by small sample sizes or limitations shows a reduction in nephron numbers in low birth weight
of counting methods will become clearer with time as more rats that were subjected to maternal low protein diets dur-
studies accumulate or as better techniques evolve. ing gestation. Maternal dietary de ciencies are common in
Various animal models have been used to study the pregnant mothers in developing countries and therefore
impact of developmental programming on nephrogene- likely clinically relevant in a large proportion of the world.27
sis. The details and pathophysiology of these models, and Maternal vitamin A de cient diets are associated with a
mechanisms whereby nephron numbers are reduced, are dose-dependent reduction in nephron number in animals.28
48
48 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

TA B L E

2.2 Factors Associated with Charges in Nephron Number and Kidney Size

REDUCED NEPHRON NUMBERS OR KIDNEY SIZE

Timing Condition Source of Effect on Nephron Number References


Data (NNx)/Kidney Size

Prenatal, Maternal low protein diet or Animal ↓ NNx, 16%–40% 12, 246
modi able total calorie restriction
Maternal vitamin A Animal ↓ NNx, in proportion to 28, 29
restriction reduction in vitamin A
Human Small infant kidney size
Maternal iron restriction Animal ↓ NNx, 22% 25
Gestational glucocorticoid Animal ↓ NNx, 20%–38% 37, 180
exposure
Uterine artery ligation/ Animal ↓ NNx, 20%–30% 81
embolization
Maternal diabetes/ Animal ↓ NNx, 10%–35% 50, 51, 247
hyperglycemia
Gestational drug exposure Animal
Gentamicin ↓ NNx, 10%–20% 40–45, 248
lactams ↓ NNx, 5%–10%
Cyclosporine ↓ NNx, 25%–33%
Ethanol ↓ NNx, 10%–20%
COX2 inhibitors ↓ NNx
Indomethacin ↓ NNx

Prenatal, Genetics
nonmodi able RET(1476A) Human 10% ↓ newborn kidney 63, 64
polymorphism volume
PAX2 AAA haplotype Human 10% ↓ newborn kidney
volume
Prematurity Human NNx ↓ with gestational age, 54, 69, 132
limited post natal
nephrogenesis
Reduced kidney size in growth
restricted children

Postnatal Nutrition Animal NNx ↓ with postnatal nutrient 32


restriction alone
Renal failure Human ? cause or consequence 54
of NNx ↓

NORMALIZATION OR INCREASE IN NEPHRON NUMBER

Source of Effect on Nephron Number


Timing Condition Data (NNx)/Kidney Size References

Prenatal Maternal vitamin A Animal Normalization of NNx in 82


supplementation LPD model
Maternal amino acid Animal Normalization of NNx in 24
supplementation LPD model
(continued)
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 449
9

TA B L E

2.2 Factors Associated with Charges in Nephron Number and Kidney Size (continued)

Source of Effect on Nephron Number


Timing Condition Data (NNx)/Kidney Size References

Ouabain administration Animal Normalization of NNx in 84


LPD model
Maternal uninephrectomy Animal NNx ↑ 88, 89
Genetics
ALDH1A2rs7169289(G) Human 22% ↑ newborn kidney size 83
allele

Postnatal Reinstitution of good Animal Catch-up of NNx in LPD model 81


nutrition
Overfeeding Animal NNx ↑ in normal birth 152
weight rats

↑, increase; ↓, decrease; ?, unknown.


Adapted from Luyckx VA, Brenner BM. The clinical importance of nephron mass. J Am Soc Nephrol. 2010;21:898–910.

Vitamin A de ciency was examined in a cohort of Indian kinase receptor critical for kidney development.30 Interest-
compared to Canadian mothers and found to be associat- ingly, vitamin A levels are reduced by smoking and alcohol
ed with signi cantly smaller newborn renal volume, which intake, both known to reduce birth weight.31 Uteroplacental
the authors suggest likely re ects lower nephron number.29 insuf ciency, induced by uterine artery ligation late in gesta-
Retinoic acid, the active metabolite of vitamin A, functions as tion, also results in low offspring birth weight and low neph-
a transcription factor regulating expression of Ret, a tyrosine ron number.32,33 This model may share some similarities

8,000

h
Δ S EM
t
r
i
b
t
Me a n
a
6,000
e
r
u
t
c
u
r
t
s
e
k

i
4,000
l
-
*
s
u
r
e
m
‡ Δ
o
l
g
f
2,000
o

r
e
*
b
m
u
N
0
I M I M I M I M
Ma le Fe ma le Ma le Fe ma le
(e ) NBW (NP) LBW (LP)

FIGURE 2.1 Relationship between glomerulogenesis, nephron number, and birth weight in rats subjected to maternal normal (NP) or
low (LP) protein diets. Renal cortex at days 0 (d0) and 10 (d10) in rat offspring of NP-fed dams (normal birth weight, NBW) and LP-fed
dams (low birth weight, LBW). (a) Normal glomerulogenesis in NBW offspring at d0, with comma-shape structure (immature renal cor-
puscle, white arrow) and inner vascularized structure (mature renal corpuscle, black arrow); (b) LBW d0 with fewer corpuscular structures
and moderately dilated tubules; (c) NBW d10 showing only mature renal corpuscles; (d) LBW d10 showing immature and mature renal
corpuscles; and (e) number of glomerulus-like structures (immature [I] and mature [M]) measured in NBW and LBW offspring (n 5 per
group). Symbols indicate group comparisons, P 0.05. (Reprinted with permission from Villar-Martini VC, Carvalho JJ, Neves MF, et al.
Hypertension and kidney alterations in rat offspring from low protein pregnancies. J Hypertens Suppl. 2009;27:S47–51.)
50
50 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

with preeclampsia in humans in terms of the reduction of Postnatal Factors. Although nephrogenesis is thought to
uterine blood ow and the restriction of fetal nutrient supply. be complete at birth in humans, this may not be the case
Increased fetal glucocorticoid exposure is a likely mech- for babies born prematurely, and therefore a window in
anism whereby maternal low protein diet reduces nephron which nephrogenesis may still be vulnerable likely exists
number, via reduced activity of placental 11 -hydroxy- soon after birth in these infants.54 Consistent with this pos-
steroid dehydrogenase activity, shown in both animals and sibility, early postnatal growth restriction alone in normal
humans.34,35 Similarly, administration of glucocorticoids birth weight rats was associated with a reduction in neph-
during gestation in rats and sheep leads to reduced nephro- ron number, demonstrating the importance of early post-
genesis, although this effect was not seen in the Marmoset natal nutrition on nephrogenesis.32 The relevance of these
monkey.36–38 Glucocorticoids are thought to reduce nephron ndings to the human, however, is questionable because
number by impacting ureteric bud invasion of the meta- nephrogenesis normally proceeds for 10 days after birth in
nephric mesenchyme, thereby limiting branching morpho- rodents and therefore this period is analogous to late gesta-
genesis.8 The impact of maternal glucocorticoid utilization tion in humans. These data may, however, have relevance to
during pregnancy on human nephrogenesis is not known. humans born prematurely. Indeed, in a cohort of children
Ingestion of other medications during pregnancy may also born either very low birth weight ( 1,000 g) or prema-
impact nephrogenesis in many ways.39 Gestational adminis- ture ( 30 weeks gestation), extrauterine growth restriction
tration of aminoglycosides, beta lactams, cyclosporine, cyclo- was associated with signi cantly lower glomerular ltration
oxygenase inhibitors, and nonsteroidal anti-in ammatory rates at a mean of 7.6 years of age, suggesting an impact of
drugs have all been associated with reduced nephron num- postnatal nutrition on renal development.55 Another study
ber in experimental models.39–43 Similarly, chronic and acute of postmortem kidneys from premature infants who died
gestational exposure to alcohol impairs embryonic ureteric after 40 days of life found glomerular number to be sig-
bud branching, resulting in fewer nephrons in offspring.44,45 ni cantly lower in those who developed renal failure com-
In humans one abstract suggested an impact of maternal pared to those who did not. These ndings may suggest
alcohol consumption on kidney development in Australian that renal failure itself inhibits glomerulogenesis; however,
Aboriginal children.9 it is also possible that fewer glomeruli made these extremely
Conceivably, therefore, all of these prenatal experimental ill infants more susceptible to renal failure. In another co-
conditions may impact human nephrogenesis and minimi- hort of critically ill premature infants, renal failure was a
zation of these exposures prior to and during pregnancy signi cant complication and associated with a high mortal-
would optimize fetal nephrogenesis. The timing of an insult ity, although not associated with birth weight.56 In contrast,
during gestation is also relevant to its impact on nephrogen- another study did nd neonatal acute kidney injury to be an
esis, with the greatest effect in animals generally seen with independent predictor of mortality in very low birth weight
interventions in the latter half of gestation.8 infants.57 Prematurity itself is a recognized risk for renal fail-
Maternal factors also impact fetal development during ure in infants, and has been shown to be associated with
gestation. Low birth weight is associated with multiple ma- increased risk of subsequent hypertension and chronic kid-
ternal factors although nephron number has not speci cally ney disease (CKD).58 Taking these human studies together,
been examined in most cases.46,47 Manalich et al. found a postnatal events do impact renal development in prema-
strong correlation between low birth weight and low neph- ture infants and may have potentially adverse short- and
ron number in a cohort of Cuban newborns.48 Maternal hy- long-term consequences.
pertension and maternal smoking were correlated with low
birth weight, although direct correlation with nephron num- Nonmodi able Factors
ber was not reported. In experimental animals, maternal Nonmodi able factors also impact nephrogenesis, and may
diabetes or hyperglycemia has been shown to result in occur in isolation or together with other potentially modi -
approximately 30% lower offspring nephron number in able factors described previously (Table 2.2).
some, but not all, studies, although differences in methods
of nephron number counting may account for some of the Genetics. Rare congenital and genetic abnormalities associ-
variability.49–51 In other studies, maternal diabetes was as- ated with abnormal kidney development manifest with renal
sociated with smaller kidneys, higher blood pressures, mi- dysfunction, often presenting very early in life.11,59 Approxi-
croalbuminuria, and reduced glomerular ltration rates in mately 40% to 60% of childhood end-stage renal disease
rat offspring.51,52 In young adults, renal functional reserve (ESRD) results from some form of congenital renal hypopla-
was found to be reduced in those who had been exposed to sia.60 More subtle renal developmental abnormalities—which
maternal diabetes during gestation, compared to those with may not manifest as overt syndromes but, rather, with later life
paternal diabetes (i.e., excluding a genetic component), or renal dysfunction—may well be the result of gene polymor-
those with nondiabetic parents.53 The reduced renal func- phisms impacting nephron number. Renal hypoplasia and re-
tional reserve was interpreted by the authors as a possible duced nephron number have been described with full or partial
surrogate for a reduced nephron number acquired in utero deletion of over 25 genes in mice, which are reviewed in detail
in the offspring of diabetic mothers. elsewhere.13,21,61 The important steps in kidney development
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 551
1

include speci cation of the metanephric blastema from the been reported previously.54,66 In addition, they found evi-
intermediate mesoderm, formation of the ureteric bud and its dence of active glomerulogenesis (indicated by the presence
outgrowth from the wolf an duct, and ureteric bud branch- of basophilic S-shaped bodies under the renal capsule in kid-
ing. Genes participating in speci cation of the metanephric neys) in premature infants up to, but not beyond, 40 days
blastema from the intermediate mesoderm include Odd-1, Eya of life.54 This was the rst study to demonstrate ongoing
1 Pax 2, Wt-1, Six 1, Gdnf, and Sall 1, of which Odd-1 and Eya -1 nephrogenesis in humans postnatally. Similarly, in preterm
are critical.21,60 Genes regulating formation of the ureteric bud baboons, nephrogenesis was found to continue after birth
and its outgrowth from the wolf an duct include Pax2, Lim1, and nephron number was within the normal range; however,
Bmp4, and Gdnf. 21,60 Gdnf (glial cell-derived neurotrophic fac- there was a greater proportion of abnormal glomeruli in the
tor) signals through the Gfr 1 receptor and the c-Ret receptor super cial cortex compared to full-term controls, suggest-
tyrosine kinase and, during branching, morphogenesis is only ing compromised nephrogenesis after premature birth.67 In
expressed on the tips of ureteric branches, selectively induc- contrast, Hinchliffe et al. did not nd an increase in nephron
ing branching at this location.21 Among the most important number in growth restricted infants who died as stillbirths
pathways impacting nephrogenesis, therefore, are Gdnf/Ret at varying gestations, or at 1 year of age, suggesting a lack of
and Pax2. In mice, deletion of Gdnf and c-Ret leads to re- nephrogenesis after birth.66,68 Gestational age was found to
nal agenesis or severe hypoplasia.21,60 Deletion of Pax2, the correlate with nephron number, which reached a maximum
“master organizer” of renal development, is incompatible with around 36 weeks.69
life.60 The impact of genetic polymorphisms in these pivotal
genes has been studied in humans. Haploinsuf ciency of the Gender. Gender likely plays a complicated role in develop-
PAX2 gene causes the autosomal dominant renal coloboma mental programming. In the largest series of kidneys analyzed
syndrome, associated with signi cant reduction in nephron to date, glomerular number in adult females was found to be
number and “oligomeganephronia.”60,62,63 Taking this nding reduced by up to 12% compared to males.13,70 In a cohort of
further, looking for a more subtle impact in the wider popula- Cuban newborns, however, nephron number was not affected
tion, Quinlan et al. found that the common AAA haplotype of by gender.48 In experimental models, reviewed in detail else-
PAX2, present in 18.5% of newborns in a Canadian cohort, where, males generally tend to be more severely affected than
was associated with reduced allele-speci c mRNA expression females in terms of reduction in nephron number, as well as
in vitro, and a 10% reduction in newborn kidney volume, subsequent manifestation of hypertension and renal dysfunc-
compared with the GGG haplotype.63 Similarly, a polymor- tion.71,72 These differences may in part result from differences
phic variant of RET, RET(1476A), was also associated with re- in postnatal growth rates between males and females,
duced mRNA synthesis, an almost 10% reduction in kidney gender-speci c differences in adaptation to adverse events,
volume, and higher levels of the renal function marker cystatin and gender-speci c regulation of genes and pathways im-
C at birth compared with the RET(1476G) variant in Cauca- pacting renal development, function, and hypertension.33,72
sian newborns.64 These authors found that newborn kidney Similarly, a large study in humans found an association of
volume is proportional to nephron number, therefore PAX2 CKD with low birth weight in adult males, but not in females,
and RET polymorphisms are likely associated with reduced suggesting a possible impact of gender on subsequent disease
nephron number in humans.64 Among 15% of Caucasians in- expression, although mechanisms are not yet clear.73
heriting both alleles, newborn kidney sizes were 23% small-
er.65 Surprisingly, however, none of 19 common GDNF gene Ethnicity. Hoy and colleagues have shown a reduction in
variants or three single nucleotide polymorphisms related to nephron number among Aboriginal compared with non-
a putative GDNF-PAX binding site were associated with small Aboriginal Australians (Table 2.1).70 Among African Ameri-
kidney size among 163 Caucasians newborns.65 One rare cod- cans and Caucasian Americans, nephron number was not
ing GDNF variant (R93W) was not found in any subject and signi cantly different in both groups and correlated with
therefore, the clinical impact of this potential mutation is not birth weight, although the distribution appeared to be
known.65 These early and small studies suggest that genetic more bimodal in the African American cohort.74 No low
polymorphisms in genes that are critical in nephrogenesis birth weight subjects were included in this study, but low
may contribute to the wide spectrum of nephron number birth weight is more prevalent among African Americans;
found in the general population. therefore, in the general U.S. population, a greater propor-
tion of African Americans may have lower nephron num-
Prematurity. Unlike in rodents, postnatal nephrogenesis ber. This remains to be studied. Nephron number among
does not occur in humans, except in extremely premature Senegalese Africans and African Americans was similar.75
infants; therefore, nephron number is predominantly de- Among Cuban neonates, nephron number was again not
termined in utero. Rodriguez et al. examined kidneys from different between black compared with white subjects.48 To
56 extremely premature infants compared with 10 full-term our knowledge kidneys of subjects from other ethnic groups
infants at autopsy.54 Radial glomerular counts were lower have not been studied. Ethnicity, therefore, may have an
in premature compared with full-term infant kidneys and impact on nephron number, although it is dif cult to dis-
glomerular number correlated with gestational age, as has sect out an impact independent of its association with birth
52
52 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

weight, socioeconomic factors, genetic polymorphisms, and females at birth, led to restoration of nephron number and
many other potential confounders. prevented the development of subsequent hypertension com-
pared to pups with continued growth restriction.81
Intergenerational Factors. Among both white and African
American women, mothers who had been of low birth weight Vitamin A Supplementation
had a signi cantly increased risk of having low birth weight Because vitamin A de ciency is associated with a nephron
offspring, independent of economic environment, suggesting a de cit, administration of a single dose of retinoic acid during
cross-generational effect of maternal low birth weight.76 Simi- early nephrogenesis restored nephron number to control
larly maternal, but not paternal birth weights, were associated levels in rat pups exposed to low protein diet in utero.82
with offspring birth weight, arguing for an intergenerational Postnatal administration of retinoic acid to preterm baboons,
programming effect of the maternal environment.77 Interest- however, was not able to stimulate nephrogenesis compared
ingly, in a large population-based study, mothers experienc- to preterm controls, suggesting a more proximal window for
ing preeclampsia, especially when associated with premature the effect of vitamin A on nephrogenesis, although these re-
birth and low birth weight in the offspring, are at increased sults may have been confounded by routine antibiotics given
risk of subsequent need for renal biopsy and/or ESRD.47,78 to all animals, which may have negatively impacted nephro-
A reduced maternal glomerular ltration rate (GFR) 90 mL genesis, confounding a potentially small vitamin A effect.30
per minute and hypertension are signi cant risk factors for
preeclampsia, small for gestational age infants, and premature Genetics
delivery.79 The question arises why the mother herself may
have been predisposed to these adverse pregnancy-related In a cohort of Caucasian newborns, a common variant of
and renal outcomes. It is conceivable that a vicious cycle may the ALHD1A2 gene involved in retinoic acid metabolism,
occur where a low birth weight mother would be predisposed ALHD1A2rs7169289(G), was associated with a 22% increase
to programmed adverse pregnancy outcomes, in turn impact- in newborn kidney size, and higher cord blood retinoic acid
ing fetal nephrogenesis and thereby future pregnancy out- levels, compared to the wild-type ALDH1A2 rs7169289(A)
comes and renal health of the subsequent generations. To our allele.83 These authors suggest this gene polymorphism
knowledge this speci c association has not been studied in could be protective for nephrogenesis in the setting of vita-
humans. In rats, the rst generation offspring of mothers fed min A de ciency.
low protein diets during gestation had low birth weights, low
nephron number, and developed spontaneous hypertension Prevention of Low Nephron Number
at 8 weeks of age. Offspring of these rst generation females, The ubiquitous plasma membrane protein Na /K -ATPase
although maintained on normal diets throughout gestation, functions as an ion pump as well as a signal transducer.
also exhibited low nephron number and hypertension, dem- Ouabain is a highly speci c Na /K -ATPase ligand that
onstrating intergenerational programming.80 Interestingly the triggers the release of calcium waves, which are important
effect was lost by the third generation, suggesting that the in- regulators of early development.84 Interestingly, erythrocyte
tergenerational cycle can be interrupted by optimization of membrane Na /K -ATPase activity was found to be reduced
risk factors such as maternal nutrition. in a cohort of low birth weight males at age 20, making this
a potentially relevant pathway.85 The impact of ouabain ad-
Strategies for Augmentation of Nephron ministration was studied experimentally as a modulator of
Number nephrogenesis under protein-de cient conditions in vitro and
in vivo.86 Ouabain was found to abrogate the effect of serum
Although total ltration surface area in individuals with fewer starvation on ureteric bud branching in cultured metaneph-
nephrons may not be reduced, as a result of compensatory roi, and to prevent reduction in nephron number in offspring
hypertrophy of the existing nephrons (see later), low nephron of low protein diet-fed dams.84 The ouabain was adminis-
number is still associated with an increased risk of hyperten- tered throughout pregnancy in this study and, therefore, the
sion and renal dysfunction in later life. Strategies to optimize potential of ouabain to rescue or restore nephron number
nephron number may therefore have an important impact on once an adverse event is already established has not been
clinical disease (Table 2.2). Interventions would likely need to be studied. Similarly, supplementation of maternal diet during
applied during gestation to have an optimal effect. Ideally, opti- gestation with glycine, urea, or alanine prevented the reduc-
mization of all modi able risk factors prior to pregnancy would tion in nephron number induced by maternal low protein
appear the simplest and most widely applicable intervention. diet in all offspring, but blood pressure was only normalized
Clinically feasible interventions are being studied to potentially in those receiving glycine.24 Interestingly, nephron number in
“rescue” nephron number and reduce subsequent hypertension. the offspring of mothers subjected to water restriction during
gestation was increased, but also did not abrogate develop-
Postnatal Nutrition ment of subsequent hypertension—again suggesting possible
Provision of adequate postnatal nutrition in low birth weight divergent programming mechanisms for nephron number
rat pups, achieved by cross-fostering onto normal lactating and blood pressure in some models.87
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 553
3

Maternal Nephrectomy Anthropomorphic Features


Uninephrectomy in rat mothers prior to pregnancy has been Birth Weight
associated with an increase in offspring nephron number Low birth weight is de ned by the World Health Organiza-
at birth; however, at 6 weeks, nephron numbers were not tion as a birth weight under 2,500 g. Very low birth weight
different from offspring of nonnephrectomized dams.88,89 is usually de ned a below 1,500 g. Low birth weight could
These authors suggest a possible circulating renotrophic result from prematurity itself (i.e., birth before the 37th week
factor in response to maternal uninephrectomy, possibly in- of gestation with an appropriate weight for gestational age),
ducing hypertrophy of the contralateral kidney, which may or from intrauterine growth restriction (IUGR) at any gesta-
accelerate nephrogenesis in the fetus but may not affect ulti- tion.46 A small for gestational age infant is de ned as having
mate nephron number. These observations may be relevant a birth weight below the 10th percentile of normal for that
in human cases such as maternal renal transplantation or gestational age.46 Full-term IUGR is the most strongly associ-
maternal kidney donation, although timing of pregnancy in ated with adult disease.91 Risk factors for low birth weight are
relation to nephrectomy may be an important variable. This diverse and, in poorer countries, maternal malnutrition, poor
area deserves more investigation. prenatal care, and infections are common, whereas in the de-
veloped world, factors such as high risk pregnancies, assisted
reproduction, multiple gestations, and advanced maternal age
CLINICAL SURROGATES FOR are becoming more frequent.46,92 High birth weight is de ned
NEPHRON NUMBER variably as a birth weight 4,000 g or 4,500 g, and is as-
In vivo, nephron number can only be grossly estimated by sociated with maternal obesity, maternal diabetes, prolonged
MRI or kidney biopsy.18,70,90 Associations of nephron num- gestation, and reduced maternal smoking.93 High birth weight
ber with readily available clinical variables have been de- has also been associated with adverse renal outcomes in the
scribed and are outlined in Table 2.3. offspring, especially as a consequence of maternal diabetes.94,95

TA B L E

2.3 Clinical Surrogates for Low Nephron Number

Clinical Feature Association with Nephron Number Population Reference

Low birth weight ↑ of 257,426 glomeruli per kg increase in birth U.S. white and black, 19
weight children and adults

Prematurity ↓ glomerular number in premature compared U.S. premature and full term 54, 68
to term infants neonates

Gender Nephron number is 12% lower in females U.S. white and black 70
Aboriginal Australian
Age ↓ 3,676 glomeruli per kidney per year of U.S. white and black 70
age 18 years Aboriginal Australian

Adult height ↑ 28,000 glomeruli per centimeter increase in Australian Aboriginal 10, 70
height German, white

Kidney mass ↑ 23,459 glomeruli per gram of kidney tissue Infants 3 months of age 64

Glomerular volume Inverse correlation between glomerular volume U.S. white and black 10, 13, 48
and nephron number Aboriginal Australian
German adults,
Cuban infants
Ethnicity ↓ Aboriginal Australians compared to U.S. white U.S. white and black 70
and black Aboriginal Australian

↑, increase; ↓, decrease.
54
54 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Low birth weight is the strongest current clinical surro- Other anthropomorphic correlates that have been associated
gate for nephron number. Nephron number has consistently with nephron number are highlighted in Table 2.3.18,20,70,98
been shown to correlate strongly with birth weight in hu-
mans, with an extrapolated increase of 257,426 glomeruli Kidney Size
per kilogram increase in birth weight.19,48,54,70 The relation-
Renal Mass
ship of birth weight to nephron number is preserved among
Australian Aboriginals, African Americans, and Whites and From autopsy studies, nephron number has been found to
therefore may be generalizable to other populations.19,70 The correlate directly with kidney weight in both adult and in-
speci c relationship between nephron number and low birth fant cohorts.18,64 Zhang et al. calculated a predicted increase
weight has only been examined in infants. Low birth weight of 23,459 glomeruli per gram of kidney mass in infants un-
was associated with lower nephron number than normal der 3 months of age.64 In living subjects, kidney weight is
birth weight, and was similar among black and white sub- not routinely obtainable, but donor kidney mass measured
jects.48,68 In experimental animals, however, not all low birth prior to transplantation, as a measure of nephron “dose,” has
weight animals have been found to have reduced nephron been shown to have clinical relevance (see later).
number and, conversely, low nephron number has been re-
ported in the absence of low birth weight.96,97 Birth weight Kidney Volume
alone, therefore, is not a universal surrogate for nephron Kidney volume can be measured in vivo, making it an
number. To our knowledge, nephron number has not been attractive potential surrogate for nephron number. Renal vol-
speci cally studied in high birth weight humans or animals. ume is dependent on nephron number, but is also strongly

TABLE

2.4 Differences in Nephron Number, Glomerular Volume, and Total Glomerular Surface Area in
the Right Kidney, U.S./Australian Adults (18 years), Means (SD)

No Hypertension
All No hypertension Hypertension versus Hypertension
US whites Nglom 855 183 (295 247) 894 339 (275 956) 747 727 (271 155) P 0.026
Nglom adja 866 722 (289 896) 912 876 (283 744) 779 808 (288 510) P 0.046
US blacks Nglom 921 708 (318 089) 931 463 (290 529) 912 480 (350 329) P 0.776
Nglom adja 946 379 (322 516) 949 934 (288 768) 952 441 (353 197) P 0.97
Aborigines Nglom 733 484 (217 763) 843 423 (199 384) 631 321 (105 298) P 0.04
Nglom adja 776 422 (253 631) 912 539 (218 432) 653 241 (178 609) P 0.110
US whites Mean Vglom, 7.1 (6.6–7.6) 6.87 (6.3–7.5) 7.82 (6.9–8.9) P 0.096
gmean
US blacks Mean Vglom, 7.7 (7.2–8.3) 6.92 (6.3–7.6) 8.64 (7.8–9.5) P 0.0012
gmean
Aborigines Mean Vglom, 7.7 (6.5–9.0) 6.88 (5.4–8.7) 8.0 (5.3–11.9) P 0.426
gmean
US whites Vglomtot, cm3 5.68 (5.3–6.1) 5.79 (5.2–6.5) 5.34 (4.7–6.3) P 0.484
US blacks Vglomtot, cm3 6.74 (6.3–7.3) 6.16 (5.6–6.8) 7.34 (6.6–8.2) P 0.020
Aborigines Vglomtot, cm3 5.40 (4.6–6.3) 5.89 (4.4–7.4) 4.96 (3.9–6.3) P 0.365

Nglom, Nglom adj, and Vglomtot: arithmetic means (SD). Vglomtot is the combined volume of all glomeruli in the kidney. Vglom in m3 106,
geometric mean (95% con dence interval). Nglom for US whites versus US blacks, P 0.133. Nglom adj for US whites versus US blacks, P 0.079.
Nglom for Aboriginal versus other, P 0.047.
a
Nglom adj: adjusted for proportions of sclerosed glomeruli seen on light microscopy.
Reprinted with permission from Hoy WE, Bertram JF, Denton RD, et al. Nephron number, glomerular volume, renal disease and hypertension. Curr Opin
Nephrol Hypertens. 2008;17:258–265.
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 555
5

correlated with current body size.18 In fetuses and at birth, others found no difference in kidney volume, adjusted for
kidney volume is presumed to be directly proportional to body surface area and gender, between individuals who had
nephron number; however, once normal kidney growth and been appropriate weight for gestational age at term, small for
adaptation occurs after a few months of life, impacted by gestational age at term, or preterm at age 9 to 12 years.106
body surface area, age, gender, glomerular hypertrophy, or Among young adults born premature (either appropriate or
nephron loss through injury, the relationship likely becomes small for gestational age) compared with term age-matched
less linear.63 Despite this caveat, several authors have inves- controls, prematurity was associated with smaller kidneys at
tigated utility of renal volume in relation to birth weight. age 20 years, whereas IUGR had only a small, nonsigni cant
Evaluation of fetal renal function by ultrasound in growth effect.107 Kidney volume may therefore be less reliable as a
restricted fetuses in utero found reduced hourly urine out- surrogate for nephron endowment as subjects age. In addi-
put, greater oligohydramnios, reduced renal perfusion, tion, as a note of caution, renal volumes were not compa-
and smaller kidney volume compared to normally growing rable between ultrasound and MRI in a neonatal population,
fetuses.99–101 Although these ndings could simply re ect suggesting that the same imaging modality should be used if
globally reduced renal perfusion, abnormal renal develop- measurements are to be compared.108
ment cannot be excluded. Another study utilizing serial
ultrasounds in a cohort of small for gestational age compared Histologic Features
to appropriate for gestational age fetuses found that kidneys
were smaller in the small for gestational age cohort, although Glomerular Volume
kidney length was relatively preserved compared to width Glomerular number has been found to vary up to 13-fold
and circumference after 26 weeks of gestation.102 Follow- among all the cohorts studied, and glomerular volume has
up of kidney size and growth postnatally in 178 premature been found to vary up to 6.7-fold (Table 2.1).13 Because
or small for gestational age children, compared with 717 nephron number is xed at birth, glomerular size is likely
term appropriate for gestational age controls, at 0, 3, and the major variable determining adaptation of ltration
18 months found that weight for gestational age correlated capacity to match the body’s demands.19,20,48,98,109,110 In-
with kidney volume at all three time points.103 Slight catch- deed, calculated total ltration surface area in kidneys with
up in kidney growth was observed in the growth-restricted, varying nephron number tends to be very similar, suggesting
but not the premature infants. Among a cohort of low birth compensatory hypertrophy in those with fewer nephrons as
weight Australian Aboriginal children aged 5 to 18, renal shown in Table 2.4.9 Consistent with this, mean glomeru-
volumes were found to be lower when adjusted for body size lar volumes vary directly with body size, and inversely with
compared to normal birth weight children.104 These authors nephron number and birth weight in all populations stud-
also found that the reduction in kidney volume was driven ied (Fig. 2.3).10,48,70,111 Most studies, however, report mean
more by a shorter depth than length of the kidney. Kidney glomerular volumes for a whole kidney. Further investiga-
length and volume were also both found to be smaller in a tion into glomerular heterogeneity has found signi cant in-
cohort of low birth weight children aged 10 to 12 years, and ter- and intraindividual variability in glomerular size.13,112
correlated weakly with lower GFR (Fig. 2.2).105 In contrast, Individual glomerular volume was found to vary up to eight-
fold within a single subject.13 Glomerular size tended to be
larger and more variable in the outer cortex compared to
those deeper within the kidney.13 In multiple analyses, hy-
pertension, obesity, age, and low nephron number emerged
as major predictors of larger glomeruli and greater het-
erogeneity.13,70,75,111–113 Interestingly, consistently, African
American and African subjects have higher mean glomeru-
lar volumes and greater heterogeneity of glomerular volume
compared to white subjects, independent of nephron num-
ber (Fig. 2.4).13,75 Furthermore, between Senegalese and
African American subjects, glomerular volumes were higher
in the U.S. cohort after controlling for body size.75 Increased
glomerular volume and heterogeneity therefore may result
FIGURE 2.2 Correlation between growth restriction and kidney from different mechanisms among African origin popula-
length. Kidney length (expressed as percent expected from the tions compared to Caucasians. Whether this may have posed
literature) is signi cantly shorter in Caucasian children aged an evolutionary survival advantage which may be maladap-
11.3 2.1 years who were born small weight for gestational tive in the current environmental circumstances given the
age (SGA) compared with appropriate weight for gestational greater burden of renal disease among African Americans,
age (AGA) at birth. (Reprinted with permission from Simonetti or whether this may re ect differences in glomerular perfu-
GD, Raio L, Surbek D, et al. Salt sensitivity of children with low sion, circulating glucose concentrations, or chronic in am-
birth weight. Hypertension. 2008;52:625–630.) mation, remains to be elucidated.75,114 It is possible that the
56
56 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

diabetes) will eventually become maladaptive and contrib-


(a )
ute to ongoing nephron loss. A kidney starting with fewer
Vglom × 10 6 (m 3 ) 14
(95% Cl) nephrons would therefore reach a critical de cit within a
12
shorter time. Consistent with this, evaluation of glomerular
10
size in donor kidney biopsies again found higher maximal
8 planar area of glomeruli to be a predictor of poorer trans-
6 plant function and, again, glomerular size was higher in
4 African American subjects.115 From a clinical point of view,
2 therefore, an otherwise unexplained increased in glomerular
0 volume should raise the suspicion of a coexisting reduction
<7 0 7 40 0 <8 76 3 0 0 <11 0 0 4 0 0 <2 0 2 5 5 00 in nephron number, although this relationship is less clear in
Ne phron numbe r
African origin populations.
(b)
Vglom × 10 6 (m 3 ) 14
(95% Cl)
12 Pathologic Changes
10
Most renal biopsy results in subjects with lower nephron
8 number have commented on glomerular size. In general
6 the degree of glomerulosclerosis present in kidneys with
4 low nephron number has been found to increase with age
2 and hypertension, but has not been a prominent feature in
0 most studies.116 The pattern of glomerulosclerosis has been
<3 .0 0 <3.5 0 described as a global ischemic collapse rather than classical
Birth we ight
focal and segmental glomerulosclerosis that might have been
(c)
expected with hyper ltration causing cumulative glomerular
Vglom × 10 6 (m 3 ) 14
(95% Cl)
injury.74 A recent case series of six very low birth weight sub-
12
jects, aged 15 to 52 years, who had been born prematurely,
10 however, did nd evidence of secondary focal and segmental
8 glomerulosclerosis, associated with glomerulomegaly in all
6 biopsies.117 Although all biopsies were performed because
4 of a clinical indication, and therefore may not be a gener-
2 alizable sample, the authors suggest that low birth weight
0 was a common denominator predisposing to hyper ltra-
Obe s e Le a n tion and glomerulosclerosis. Analyses of histologic changes
Body ma s s index in human biopsies may be confounded by multiple factors.
In kidneys of rats exposed to gestational low protein diet,
FIGURE 2.3 Relationship of glomerular volume (Vglom ) to neph- fewer and more immature glomeruli were present on day 10
ron number, birth weight, and body size. A: Inverse association at the end of nephrogenesis, exhibiting a markedly thick-
of mean glomerular volume (Vglom ) with nephron number, ened glomerular basement membrane and abnormal podo-
n 252; (B) inverse association of mean glomerular volume cyte structure (Fig. 2.1).118 Similarly, in prehypertensive
(Vglom ) with birth weight, n 58; and (C) direct association of Gdnf heterozygous mice, which have a 30% reduction in
mean glomerular volume (Vglom ) with body mass index (BMI; nephron number, glomerular enlargement was observed, as-
obese 30 kg per m 2, n 95; lean 25 kg per m 2, n 78). sociated with an increase in cellular proliferation, thickened
Subjects were U.S. whites and African Americans. * P 0.05. glomerular basement membrane, reduced podocyte density,
(Reprinted with permission from Puelles VG, Hoy WE, Hughson and a mild expansion of the tubulointerstitium.119 In rats
MD, et al. Glomerular number and size variability and risk for rendered diabetic at 12 weeks, and followed until 40 weeks,
kidney disease. Curr Opin Nephrol Hypertens. 2011;20:7–15.) glomerular changes associated with diabetes were similar in
those that had been of low birth weight compared to normal
birth weight. However, again, in addition to lower nephron
greater average adult height among African American sub- number, podocyte density was reduced and the area covered
jects may augment nal nephron number, diminishing the by each podocyte was greater in the low birth weight diabetic
relationship with birth weight, but may not compensate for, animals.120 Interestingly, proteinuria tended to be higher in
or may exacerbate, other potentially programmed changes the low birth weight group, likely suggesting a greater de-
such as glomerular size. Compensatory hyper ltration may gree of hyper ltration. Taken together, these authors pos-
be adequate in the short term to augment glomerular ltra- tulate that early subtle structural abnormalities in kidneys
tion, but sustained hyper ltration, especially in the setting with reduced nephron number may enhance susceptibility
of additional renal stresses (e.g., rapid growth, hypertension, to subsequent renal injury.
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 557
7

FIGURE 2.4 Relationship between mean glomerular Me a n glome rula r 12


volume and glomerular number (Nglom) in U.S. whites volume (µm 3 ×10 6 )
me a ns (95% Cl)
and African Americans. Mean glomerular volume by quar-
10
tiles of glomerular number. Light bars are U.S. whites and
dark bars are African American adults. U.S. whites, P for
trend 0.0001; African Americans P for trend 0.0008. 8
(Reprinted with permission from Hoy WE, Bertram JF,
Denton RD, et al. Nephron number, glomerular volume, re-
6
nal disease and hypertension. Curr Opin Nephrol Hypertens.
2008;17:258–265.)
4
Ng lo m ≥227 327 ≥669 856 ≥854 419 ≥1 101 498

of lower birth weight, therefore this programming effect is


CLINICAL IMPACT OF NEPRHON MASS evident early and tracks through to adulthood (Fig. 2.6).127
Blood Pressure Importantly, low birth weight children tend to have higher
blood pressures, although not in the hypertensive range,
Birth Weight and Blood Pressure
compared to normal birth weight children. With increas-
Many studies in humans and animal models have sup- ing age, however, blood pressure differences between low
ported the observation that low birth weight is associated birth weight and normal birth weight subjects become am-
with higher blood pressures in later life (Fig. 2.5).12,121–127 pli ed and do reach hypertensive levels with time.128,129
Higher blood pressures have been reported in newborns Prematurity and gestational age have also been associated
with higher blood pressures in young adults; however, low
birth weight for gestational age was a more signi cant pre-
dictor of blood pressure at birth and 18 years of age than
170

low birth weight of prematurity.107,130–133 This observa-
tion suggests that ongoing intrauterine stress may have a
160 † greater impact than premature birth. Consistent with this
S EM Δ
possibility, abnormal placental morphology, a marker of

150 Me a n adverse intrauterine conditions, has been associated with
)
higher blood pressures in children at 7 years of age.134 The
g
H
140
m
importance of the intrauterine environment has also been
m

(
† highlighted in both monozygotic and dizygotic twin stud-
e
r
130
ies, where the lower birth weight twin has been found to
u
s

s
Δ have a greater increase in blood pressure in infancy and
e
r
120
p
higher blood pressure in adulthood.135,136 These data have
d
o
o
been interpreted to suggest that genetic factors play a
l
B
110
smaller role than fetal growth in developmental program-
100 ming. Although this may be the case, a signi cant modula-
10 tion of the relationship between birth weight and blood
5
0
pressure has been found by genotype of beta adrenergic
d90 d180 d90 d180 d90 d180 d90 d180 receptors, suggesting possible developmental interaction
Ma le Fe ma le Ma le Fe ma le with gene expression as well.137 The relationship between
NBW (NP)
birth weight and blood pressure has not been universally
LBW (LP)
found, however, and in particular appears to be weaker, but
FIGURE 2.5 Relationship between birth weight, nephron num- not always absent, in African American children.128,138–142
ber, and hypertension in rats subjected to maternal normal (NP) However, interestingly, the association between low birth
or low (LP) protein diet. Systolic blood pressure in 90-day-old weight and higher blood pressures does appear preserved
(d90) and 180-day-old (d180) male and female rats with normal in African and Caribbean black children, suggesting that
(NBW) or low (LBW) birth weights. Symbols indicate group com- early childhood growth rates, genetic, and/or other envi-
parisons, P 0.05. (Reprinted with permission from Villar-Martini ronmental factors are also important.143–145 Current body
VC, Carvalho JJ, Neves MF, et al. Hypertension and kidney altera- mass index (BMI) is a frequent confounder in these stud-
tions in rat offspring from low protein pregnancies. J Hypertens ies, and may play a greater role among black compared to
Suppl. 2009;27:S47–51.) white subjects.137
58
58 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Cha nge in s ys tolic blood pre s s ure (mmHg) pe r kg incre a s e in birth we ight
(95% Cl)

1

0


0
5
1
1
1
0
5
0
Ne wborn
3 ye a rs
3 ye a rs
3–4
ye a rs
3–12 ye a rs
5–7 ye a rs
5 ye a rs
6–16 ye a rs
6.5 ye a rs
6–9 ye a rs
7 ye a rs
7–11 ye a rs
8–9 ye a rs
8–10 ye a rs
8–11 ye a rs , M
8–11 ye a rs , F
10–12 ye a rs
11–16 ye a rs
11 ye a rs
12–14 ye a rs , M
12–14 ye a rs , F
15 ye a rs , M
15 ye a rs , F
17 ye a rs
17 ye a rs
17–24 ye a rs
18 ye a rs , M
18–23 ye a rs , M
18–23 ye a rs , F
16–26 ye a rs
18 ye a rs
18–25 ye a rs , M
18–25 ye a rs , F
20–44 ye a rs
36 ye a rs , M
36 ye a rs , F
36.5 ye a rs , F
40 ye a rs
40–75 ye a rs , M
41–47 ye a rs , M
41–54 ye a rs
42–47 ye a rs
46–50 ye a rs , M
46–50 ye a rs , F
50 ye a rs
47–56 ye a rs
50 ye a rs , M FIGURE 2.6 Studies reporting multiple
50–53 ye a rs
regression analyses in children, adoles-
51–54 ye a rs , M
51–54 ye a rs , F cents, and adults. Higher blood pressures
58 ye a rs , F are generally associated with lower birth
59–63 ye a rs , M weights across all ages and in both genders.
59–63 ye a rs , F CI, con dence interval. See original source
64–71 ye a rs , M
for detailed references. (Reprinted with
64–71 ye a rs , F
permission from Huxley RR, Shiell AW, Law
CM. The role of size at birth and postnatal
catch-up growth in determining systolic
Highe r blood pre s s ure , low blood pressure: a systematic review of the
birth we ight literature. J Hypertens. 2000;18:815–831.)
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 559
9

Nephron Number and Blood Pressure these populations.70,113 An important caveat is that these
The association between low birth weight and later life studies are all performed on postmortem kidney samples
hypertension has been well documented in various animal mostly from adults of varying ages, and therefore may not
models utilizing gestational interventions such as maternal re ect nephron endowment at birth. Much larger sample
dietary protein restriction, dexamethasone administration, or sizes would be required to control for all possible confound-
uterine artery ligation, as described previously.12,28,37,146–150 ing variables between subjects, which are not easily feasible.
The link between low birth weight and subsequent sponta- In addition, associations have been well described between
neous hypertension in these models appears to be, at least in birth weight and nephron number, nephron number and hy-
part, attributable to an inborn nephron de cit.12,37,148 Most pertension, and birth weight and hypertension but, to date,
interventions result in a 20% to 30% reduction in glomeru- to our knowledge, no study has analyzed nephron number,
lar number in offspring, and hypertension emerges by early birth weight, and hypertension in individual subjects.
adulthood.12,151 Conversely, optimization of postnatal nutri- Timing of nephron loss appears to impact subsequent
tion after growth restriction has been found to normalize risk of hypertension and renal disease. In humans, congenital
nephron number and abrogate the development of hyper- conditions associated with signi cant reductions in nephron
tension in rats, suggesting that nephron number per se is mass (e.g. unilateral renal agenesis or bilateral renal hypopla-
an important contributor to the pathogenesis of hyperten- sia) result in worsening proteinuria, hypertension, and renal
sion.24,81,152 In contrast, however, augmentation of nephron dysfunction with time.11 In contrast, nephrectomy later in life,
number does not always protect against high blood pres- resulting in a comparable loss of nephron mass, does not nec-
sure. Restoration of nephron number by supplementation essarily result in progressive renal functional decline.153 Simi-
of maternal low protein diet with glycine, urea, or alanine larly, nephrectomy in adult animals in varying experimental
only normalized blood pressure in the offspring supple- settings does not invariably lead to hypertension and renal
mented with glycine.24 Similarly, a 20% increase in nephron dysfunction.154 Removal of a kidney on postnatal day 1 in
number, induced by postnatal overfeeding in normal birth rats, or fetal uninephrectomy in sheep (i.e., loss of nephrons
weight rats, did not prevent hypertension and glomerulo- during active nephrogenesis), however, does lead to adult hy-
sclerosis with age, although concomitant obesity may have pertension in the absence of evidence of renal injury.155–157
been a confounder in this study.152 Based on these studies, Taken together these animal and human observations suggest
therefore, developmental programming of hypertension is that loss of nephrons during renal development, as opposed
dependent on more than a reduction in nephron number, to after nephrogenesis is completed, may have a more critical
although under certain circumstances nephron number does impact on the long-term risk of hypertension.
appear to be the predominant predisposing factor. In support of this hypothesis, glomerular numbers in
Evidence in humans suggests a similar association adult rats were similar after uninephrectomy at day 3 or
between nephron number and risk of hypertension. In a day 120 of age; however, a greater proportion of immature
cohort 35- to 59-year-old European Caucasians who died glomeruli were present in kidneys of rats having undergone
in accidents, mean nephron number was signi cantly lower, removal of the contralateral kidney at day 3.158 In addition,
and glomerular volume signi cantly higher in the 10 sub- mean glomerular volume compared to controls was 59%
jects with a history of essential hypertension, compared to higher in rats undergoing neonatal nephrectomy compared
10 normotensive matched controls.10 There was no evi- with 20% higher in rats nephrectomized as adults, suggest-
dence of disproportionate glomerulosclerosis or renal injury, ing more vigorous compensatory hypertrophy and hyper-
leading the authors to suggest that an intrinsic de cit in function in response to neonatal nephrectomy, which may
nephron number was the most likely factor associated with become maladaptive over the long term. Developmentally
development of essential hypertension. Birth weights were acquired low nephron mass may, therefore, be considered
not available in this study, therefore potential associations along the broader continuum of renal hypoplasia and associ-
with nephron number could not be speculated. A possible ated with long-term consequences.13
limitation of this study is the high mean glomerular num-
ber in the nonhypertensive group compared to that reported Glomerular Volume and Blood Pressure
in other Caucasian populations; however, mean nephron Glomerular volume varies inversely with nephron number
number in the hypertensive group was similar to that in a and, in U.S. White, Black, and Australian Aboriginal subjects,
hypertensive U.S. Caucasian cohort. Similarly, lower neph- is associated with increased risk for high blood pressure
ron numbers have been associated with higher blood pres- (Fig. 2.8).70 In addition, among the U.S. Black and Australian
sures among Caucasians and Australian Aboriginal subjects, Aboriginal populations, large glomeruli on renal biopsy are as-
although the relationship is not as consistent among subjects sociated with poorer renal outcomes in native and transplanted
of African origin (Fig. 2.7).10,70,98 Conversely, the prevalence kidneys.98,115,159,160 Among Black subjects, glomerular size ap-
of hypertension was found to be lower among Caucasians pears to be an independent predictor of higher blood pressure,
and Australian Aboriginals with higher nephron numbers, whereas in White and Australian Aboriginal subjects, the rela-
suggesting a protective effect of higher nephron numbers in tionship appears also dependent on low nephron number.113
60
60 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

(a ) (b)

P roba bility of 1 P roba bility of 1


hype rte ns ion hype rte ns ion
(95% Cl) 0.8 (95% Cl) 0.8

0.6 0.6

0.4 0.4

0.2 0.2

0 0
Nglom ≥227 327 ≥657 561 ≥833 711 ≥1 021 701 Nglom ≥387 550 ≥693 771 ≥873 436 ≥1 162 345
2 2
Nglom/m ≥94 409 ≥333 414 ≥441 347 ≥532 777 Nglom/m ≥53 718 ≥344 305 ≥454 688 ≥550 531

(c)

P roba bility of 1
hype rte ns ion
(95% Cl) 0.8

0.6

0.4

0.2
*
0
Nglom ≥491 163 ≥625 930 ≥785 584 ≥976 244
2
Nglom/m ≥310 863 ≥378 873 ≥476 217 ≥618 548

FIGURE 2.7 Probability of hypertension by glomerular number (Nglom) and ethnicity. Probability of hypertension in (A) U.S. white
adults, by Nglom (light bars), P .097; adjusted for sex, P .042. By Nglom per m 2 body surface area (BAS), Nglom per m 2 (dark bars),
P .0012; adjusted for sex, P .0006. B: African American adults, by Nglom, P .625; adjusted for sex, P .71. By Nglom per m 2,
P .246; adjusted for sex, P .245. C: Australian Aboriginal adults, by Nglom, P .167; adjusted for sex, P .173. By Nglom per m 2,
P .167; adjusted for sex, P .109. *Among subjects with Nglom in this category none had hypertension. CI, con dence interval. (Re-
printed with permission from Hoy WE, Bertram JF, Denton RD, et al. Nephron number, glomerular volume, renal disease and hyperten-
sion. Curr Opin Nephrol Hypertens. 2008;17:258–265.)

The observation that in some animal models normaliza-


tion of nephron number did not prevent development of sub-
sequent hypertension, argues against nephron number be-
ing the sole programmed link between early developmental
stress and higher blood pressures.161,162 Given that the ltra-
tion surface area in kidneys with low nephron numbers has
not been found to be signi cantly lower than in kidneys with
a higher number of nephrons, and may even be increased in
subjects with hypertension (Table 2.4), other nonglomerular
contributors to sodium avidity in low birth weight kidneys
have been investigated.8,10,70 Blood pressure is dependent on
renal, neuroendocrine, and vascular factors, all of which may
FIGURE 2.8 Probability of hypertension by glomerular volume be subject to simultaneous developmental programming.8
(Vglom ) in U.S. white and African American adults. Increasing Programmable vascular factors which have been studied in-
probability of hypertension with increasing (Vglom ) in U.S. white clude altered structure and function of large vessels, impaired
(light bars) and African American (dark bars) subjects aged vascular reactivity, and endothelial dysfunction, which are
18 years or older (n 252). (Reprinted with permission from reviewed in detail elsewhere.163–165 Neuroendocrine factors
Puelles VG, Hoy WE, Hughson MD, et al. Glomerular number and include altered stress responsiveness, cortisol levels, insulin
size variability and risk for kidney disease. Curr Opin Nephrol resistance, and sympathetic nervous system activity.71,166,167
Hypertens. 2011;20:7–15.) Within the kidney, in addition to programming of nephron
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 661
1

number, alterations in sodium transport and modulation of malnutrition in utero, therefore a low sodium diet during
the renin-angiotensin system have also been well described. gestation, which also may have modulated their renal so-
dium handling. Interestingly, manipulation of sodium intake
Renal Programming of Blood Pressure postnatally was found to impact long-term blood pressure in
young low birth weight rats.174 Low birth weight rats were
Salt Sensitivity and Birth Weight placed on either low, normal, or high salt diets from weaning
The original programming hypothesis postulated a reduc- to 6 weeks of age, followed by normal diets thereafter. Later
tion in ltration surface area and limitation in sodium excre- life hypertension was abrogated by early low salt diet and
tory capacity in a kidney with fewer nephrons.2 Consistent worsened by early high salt diet. In addition, salt sensitivity
with this, prenatal dexamethasone administration in rats was after 40 weeks was lost in the rats subjected to early low salt
associated with lower GFR, higher urine albumin excretion, diets.174 Early low salt diet therefore appears to be able to
reduced urinary sodium excretion, and higher tissue sodium “re-program” the kidney and prevent hypertension in low
content compared to controls.37 Interestingly, in a study of birth weight rats.
Caucasian men aged 20 years, those who had been of low Filtration surface area, blood pressure, and response to
birth weight had an increase in systolic blood pressure and sodium loading was assessed in GNDF heterozygous mice in
no change in GFR, but an increase in fractional excretion of which nephron numbers are reduced, and 20% of animals
sodium compared to normal birth weight controls.85 have unilateral renal agenesis.175 Total nephron number was
Salt-sensitive hypertension has subsequently been reduced by 25% in mice with two kidneys (HET2K) and
shown in low birth weight rats (induced by uterine artery 65% in mice with single kidneys (HET1K) compared to the
ligation), and in adult rats that had been exposed to ma- wild type. The degree of glomerular hypertrophy was simi-
ternal gestational diabetes.52,168,169 In contrast, however, lar in HET1K and HET2K, resulting in normalization of glo-
blood pressures did not increase more in low birth weight merular surface area in HET2K but a persistent reduction in
rats (induced by maternal protein restriction) compared to the HET1K. In this model, reduced nephron number alone
normal birth weight rats on a high salt diet.170 Timing of the was not associated with increased blood pressures, but both
dietary sodium challenge may have an impact, however.171 groups of HET mice became signi cantly hypertensive on a
Blood pressure rises more consistently in response to a high high sodium diet, with blood pressures being highest in the
salt diet in older rats, suggesting loss of potential adaptive HET1K mice.175 Interestingly, at baseline urine sodium excre-
mechanism with age, or greater susceptibility to salt sensi- tion was progressively higher in HET2K and HET1K mice,
tivity as nephron numbers decline with age.172 Plasma vol- suggesting augmented natriuresis that maintained normal
ume and blood pressures were found to be higher in low blood pressures. In the same model, tubule sodium trans-
birth weight compared to normal birth weight juvenile rats porters were not found to be increased, in contrast to other
on normal diets, however, suggesting positive sodium bal- programming models. This suggested a different adaptation
ance at baseline.173 Interestingly, although a further increase in baseline sodium handling in this genetic model, which
in sodium intake did not increase blood pressures or plasma may be overwhelmed in the face of a high sodium diet.119,175
volume expansion more than in normal birth weight rats, In healthy human adults, salt sensitivity was found to
GFR was signi cantly increased in the low birth weight correlate inversely with birth weight, independent of GFR
rats on a high sodium diet, suggesting a shift in the pres- (Fig. 2.9).176 Similarly, in low birth weight children, the prev-
sure natriuresis curve.173 These rats were subjected to global alence of salt sensitivity was found to be high and to correlate
(a ) (b)
5
R = –0.60 P = 0.002
)
)
4
g
g
9
H
H
m
m
3
6
m
m
s a lt s e ns itive (n=9)
(
(
2
y
P
3
t
A
vi
M
1
i
t
i
s
h
0
n
0
4
e
2
s
–3
a
t
–1
l
t
l
a
e
s a lt re s is ta nt (n=15)
S
D
–2 –6
2.5 3.0 3.5 4.0 4.5 88 90 92 94 96 98
Birth We ight (kg) Tota l kidney le ngth (%)

FIGURE 2.9 Salt-sensitivity in humans relative to birth weight and kidney length. A: Correlation between birth weight and salt sensi-
tivity in 27 normotensive subjects aged 37.2 14.5 years. B: Correlation between salt sensitivity and percent expected kidney length
in children aged 11.3 2.1 years. Kidney lengths were smaller in children who had been small weight for gestational age (SGA) com-
pared to appropriate weight for gestational age (AGA) children at birth, suggesting a correlation of salt sensitivity with birth weight.
(A, reprinted with permission from de Boer MP, Ijzerman RG, de Jongh RT, et al. Birth weight relates to salt sensitivity of blood pressure
in healthy adults. Hypertension 2008;51:928–932. B, reprinted with permission from Simonetti GD, Raio L, Surbek D, et al. Salt sensitiv-
ity of children with low birth weight. Hypertension. 2008;52:625–630.)
62
62 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

inversely with kidney size, again independent of GFR, sug- balance, or is an independent simultaneously programmed
gesting that renal function itself was not a confounder change in the renal tubules is not yet clear.
(Fig. 2.9).105 Developmental programming of blood pressure The intrarenal renin-angiotensin system is important
in most animal models and humans does, therefore, appear for nephrogenesis as well as blood pressure regulation.8,72
to be associated with altered sodium handling by the kidney. Administration of inhibitors of the renin-angiotensin system
during renal development results in abnormal kidneys and
Renal Sodium Transport reduced nephron numbers.8 Maternal low protein diet, pre-
The expression of renal sodium transporters has been in- natal dexamethasone, and uterine artery ligation all induce
vestigated in several animal models of developmental pro- intrauterine growth restriction and have been associated with
gramming as a contributor to salt sensitivity. In offspring of altered expression of components of the renin-angiotensin
mothers fed a low protein diet or given dexamethasone dur- system.8,181–185 The observed alterations in renal renin,
ing gestation, chloride transport in the thick ascending limb angiotensinogen, angiotensin converting enzyme activity,
(mTAL) and the lumen positive transepithelial potential dif- angiotensin II, and angiotensin receptor subtype 1 and 2
ference were signi cantly higher compared to control rats, levels all appear to be modulated at different times of devel-
demonstrating increased bumetanide sensitive Na -K -2 Cl opment with some changes being present at birth and others
(NKCC2) transporter activity in the mTAL.177 Rats were al- manifesting in later life, as reviewed elsewhere.8 In addition,
ready hypertensive at the time of study and, consistent with some of the observed variation likely results from differences
these ndings, administration of furosemide reduced blood in timing and nature of the programming insult. The obser-
pressure in the prenatal low protein diet group compared to vation that programmed hypertension could be modulated
controls, demonstrating that the increased NKCC2 activity by postnatal administration of inhibitors of the renin-angio-
was contributing to the higher blood pressure.177 Interesting- tensin system supports a role for this system in generation
ly, in this study, at 6 weeks of age, expression of the NKCC2 of increased blood pressures, potentially involving altered
was increased in the medulla of the low protein diet group sodium transport, reduced nitric oxide activity, and reactive
but not the prenatal dexamethasone group, despite changes oxygen species generation.183,185 Furthermore, evidence of
in sodium chloride transport being evident in both.177 Simi- lack of suppression of this system in the setting of increased
larly, NKCC2 and thiazide sensitive Na -Cl cotransporter renal sodium transport and plasma volume expansion also
(NCC) protein levels were signi cantly elevated in low birth suggests dysregulation by prenatal programming.8 To our
weight, induced by maternal low protein diet, rat kidneys at knowledge, levels of renin-angiotensin system activity have
4 weeks of age—that is, before the manifestation of hyperten- not been investigated in low birth weight humans.
sion. This occurred even though expression of the proximal
tubule sodium hydrogen exchanger (NHE3) and the epithe- Catch-up Growth and Blood Pressure
lial sodium channel (ENaC) expression were not changed.178 Developmental programming does not only encompass
In another study, prenatal dexamethasone was associ- the intrauterine period but, as discussed previously, early
ated with increased NKCC2 protein expression at 8 weeks, postnatal events may also be critical in renal development.
along with NCC and NHE3, but not ENaC, and their expres- Similarly, early childhood growth, especially after intrauter-
sion was reduced by renal denervation, suggesting modula- ine growth restriction, is emerging as a signi cant risk factor
tion of sodium transporter expression by the renal nerves.179 for subsequent hypertension and cardiovascular disease.186
Other investigators found mRNA expression of the glucocor- In low birth weight male rats with reduced nephron
ticoid receptor, and the glucocorticoid responsive 1- and numbers, induced by maternal gestational protein restriction,
1-subunits of Na /K -ATPase to be increased in offspring postnatal overfeeding resulted in accelerated development of
of rats fed low protein diets during gestation.180 Similarly, hypertension and a signi cant reduction in GFR in adult-
in animals subjected to maternal diabetes, renal Na /K - hood.187 The rapid postnatal weight gain in the low birth
ATPase expression was increased, as well as the and weight overfed rats therefore exacerbated the programmed
subunits of ENaC.168 When these animals were subjected to risk of hypertension, acting as a “second hit” superimposed on
a high salt diet, expression of NHE3 and NCC increased, the low nephron number. Interestingly, appetite, obesity, and
but expression of NKCC2 decreased.168 In yet another mod- energy expenditure are also developmentally programmed,
el of low nephron endowment, there was no difference in compounding the risk of cardiovascular disease in growth
NCC or ENaC expression by immunohistochemistry in kid- restricted individuals.188 In a cohort of adolescents, rapid
neys of Gdnf heterozygous compared to wild type mice.119 weight gain in the rst 2 weeks of life was associated with
Various programming models of low nephron number and reduced ow-mediated dilation of the brachial artery mea-
hypertension, therefore, are associated with some disparities sured at ages 13 to 16, underscoring the long-term impact
in renal sodium transporter expression, but in general it ap- of early nutrition on vascular function.189 Similarly, weight
pears sodium transporters are upregulated and likely contrib- gain within the rst 5 months of life was associated with in-
ute to increased blood pressures. Whether the alteration in creased systolic and diastolic blood pressures at 25 years of
sodium transport is a direct result of reduced nephron num- age, although only systolic blood pressure was inversely as-
bers, single nephron hyper ltration and glomerulotubular sociated with birth weight.190 The combination of low birth
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 663
3

TA B L E

2.5 Difference in Systolic Blood Pressure (mm Hg) at 22 Years per SDs Increase in Birth Weight,
Infant Weight Gain (First Year), and Early Childhood Weight Gain (1–5 Years)a

Regression Coef cient (95% CI)

Weight Growth Variable Without Adjustment for Adult BMI With Adjustment for Adult BMI

Birth weight 1.3 ( 2.3 to 0.3) 1.2 ( 2.2 to 0.3)

Conditional infant weight gain 0.5 ( 0.6 to 1.5) 0.1 ( 1.2 to 0.9)

Conditional early childhood 1.6 (0.6 to 2.7) 0.6 ( 0.5 to 1.7)


weight gain

Adult BMIb ... 2.6 (1.5 to 3.7)

a
With and without adjustment for adult body mass index.
b
Geometric SDs.
CI, con dence interval; BMI, body mass index; SDs, standard deviations.
Reprinted with permission from Law CM, Shiell AW, Newsome CA, et al. Fetal, infant, and childhood growth and adult blood pressure: a longitudinal
study from birth to 22 years of age. Circulation. 2002;105:1088–1092.

weight followed by accelerated childhood growth has also with age, suggesting ongoing tissue stress and accelerated
been consistently found to be associated with higher blood senescence precipitated by catch-up growth. Increased
pressures in childhood and adulthood (Table 2.5).191,192 senescence in the low birth weight kidneys may result from
Several potential mechanisms have been investigated to ongoing hyper ltration injury in kidneys with fewer neph-
explain this ampli cation of cardiovascular risk by rapid rons, exacerbated by a rapid increase in body size. Interest-
weight gain after growth restriction. In obese sheep that ingly in humans, leukocyte telomere length was not different
had been exposed to maternal nutrient restriction during between small for gestational age and appropriate for gesta-
gestation, adipose tissue showed an increase in number tional age newborns, but by 5 years low birth weight children
of necrotic cells, increased secretion of pro-in ammatory had signi cantly shorter telomeres than normal birth weight
factors, and increased evidence of endoplasmic reticulum children, consistent with accelerated senescence.197,198 How-
stress compared to obese controls.188 In addition, the nu- ever, growth trajectories of these children were not reported.
trient restricted obese sheep exhibited abnormal myocardial Markers of endoplasmic reticulum and mitochondrial stress
structure and function.188 One possible connection between were increased in low birth weight rat kidneys after rapid
growth restriction and accelerated catch-up growth is the catch-up growth, which points to increased reactive species
development of premature senescence.193 Chronic diseases generation as a possible mediator of increased senescence.193
such a hypertension, chronic kidney disease, and cardio- Consistent with these animal ndings, in humans there is
vascular disease have all been associated with increased evidence of increased oxidative stress in children born small
expression of senescence markers. In animal models, low for gestational age compared to controls, which was relative-
birth weight followed by accelerated postnatal growth was ly higher in those who experienced catch-up growth.199,200
associated with more rapid telomere shortening and acceler- The link between nephron mass, catch-up growth, prema-
ated senescence in kidneys and aortas, as well as premature ture senescence, and the development of hypertension and
death.194–196 These markers were not correlated directly with renal disease in humans after developmental programming
blood pressures and nephron numbers in the same animals. has not been studied.
However, investigators utilized a standard programming
model of maternal dietary protein restriction, therefore low Renal Function and Nephron Mass
birth weight animals were expected to have low nephron
numbers and high blood pressures. Glomerular Filtration Rate
In another study, there was no difference in expression A reduction in nephron number, and therefore ltration
of the senescence marker p16 between low birth weight and surface area, in the absence of compensatory hyper ltra-
normal birth weight rats, suggesting that senescence is not tion, would be expected to be associated with a reduced
likely a contributor to the reduced nephron numbers.193 p16 whole kidney GFR. Consistent with this possibility, amikacin
was signi cantly increased by weaning in kidneys and hearts clearance, as a surrogate for GFR, was lower in premature
of low birth weight rats, and continued to rise progressively or growth restricted neonates on day 1 of life (i.e., before
64
64 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

any adaptation would have occurred).201 Similarly, nephron maternal diabetes during gestation) compared to those with
number and GFR were measured in newborn growth restrict- diabetic fathers. Subjects were matched for age, gender, BMI,
ed piglets.202 GFR was reduced in proportion with nephron and birth weight. The offspring of diabetic mothers had a
number, suggesting a lack of glomerular adaptation early af- signi cantly reduced renal reserve capacity, suggesting a re-
ter birth. With increasing age, however, as the single nephron nal programming effect due to maternal diabetes.53 Evalua-
GFR increased with hyper ltration, differences in total GFR tion of renal functional reserve may therefore be a sensitive
have not been consistently seen between normal and low method to detect subtle changes in renal function due to
birth weight subjects. Among 6- to 12-year-olds, estimated a reduced nephron number that may not be evident with
creatinine clearance was lower and serum creatinine higher baseline GFR measurements.
in very low birth weight compared to age-matched normal Proteinuria is a marker of glomerular hyper ltration and
birth weight subjects.203 In contrast, estimated GFRs were renal injury and, as such, has been investigated in several low
not different between three groups of 9- to 12-year-olds born birth weight populations. Among children aged 8 to 11 years
with low gestational age or either small for gestational age or of age, low birth weight was associated with signi cantly
appropriate for gestational age at term.106 Interestingly, esti- higher blood pressures and 24-hour urine albumin excre-
mated GFR calculated using cystatin C showed a signi cant tion compared to normal birth weight controls.209 Multiple
linear trend with birth weight compared to creatinine-based other studies have also shown a largely consistent relation-
GFR in a cohort of children divided into quartiles by birth ship between low birth weight and proteinuria, although in
weight. This suggested the potential pitfalls with creatinine some studies albuminuria was associated with thinness at
measurements and/or the need to adapt GFR calculation for- birth, an indicator of intrauterine stress, rather than birth
mulae in young low birth weight subjects.204 weight.94,131,207,210,211 Among Australian Aborigines, albu-
Utilizing iothalamate clearance, GFR was measured in a minuria was strongly associated with low birth weight and
cohort of children at a mean age of 7.6 years who were born the relationship was ampli ed with increasing age.212 Macro-
with a birth weight under 1,000 g or before 30 weeks of albuminuria in this cohort was also associated with a high
gestation, and strati ed according to whether they had expe- risk of renal failure and death.213 In a Finnish population
rienced either intrauterine or extrauterine growth restriction with type 1 diabetes, however, after a mean duration of dia-
or normal perinatal nutrition.55 GFRs were signi cantly low- betes of 19 years, no association was found between pro-
er in both pre- and postnatally growth restricted children, teinuria and birth weight strati ed as low, 10th percentile;
although still within the normal range for their age. This high, 90th percentile; and intermediate, between 10th and
observation further highlights the potential negative impact 90th percentiles.214
of poor extrauterine nutrition on early renal development in In a similar study, among Danish women with type 1
premature infants. In young adults born very premature, the diabetes, with a median duration of 27 years, 75% of those
creatinine-based Cockroft-Gault GFR was found to correlate with birth weight under the 10th percentile had nephropa-
positively with birth weight.131 GFRs measured by 24-hour thy, de ned as persistent urine albumin excretion 300 g
urine creatinine clearance within adult twin pairs were lower per day, compared with 35% of those with birth weights
in the lower birth weight twin, again suggesting an indepen- above the 90th percentile.215 The effect was not present in
dent effect of the intrauterine environment on programming men, although in another study, short stature in men was
of renal function.205 Overall, from several studies, GFRs were associated with a higher risk of macroalbuminuria.216 A U-
estimated to increase by 3.8 to 7.2 mL per minute in males shaped association between birth weight and proteinuria
and 2.6 to 5.7 mL per minute in females per 1-kg increase was found among Pima Indians with type 2 diabetes in the
in birth weight.206,207 United States, suggesting that high birth weight and low
An interesting small study evaluated total GFR, effective birth weight are both risk factors for renal disease in this
renal plasma ow, and ltration fraction before and after low population.94 Interestingly 64% of subjects with high birth
dose dopamine or an oral amino acid load to test renal func- weight were offspring of diabetic mothers versus none of
tional reserve in 20-year-olds born (1) premature with ap- those with low birth weight. The association of high birth
propriate weight for gestational age, (2) premature and small weight with proteinuria was lost after adjustment for mater-
for gestational age, or (3) term and appropriate for gestational nal diabetes, suggesting potentially different programming
age. Intriguingly, although the changes did not reach statis- mechanisms in low birth weight and high birth weight with
tical signi cance, the stimulated increase in GFR was less respect to proteinuria.94
in small for gestational age compared with appropriate for Differences between these studies in diabetic subjects
gestational age and control subjects. Effective renal plasma may re ect altered genetic susceptibility to renal disease in
ow was lower in both small for gestational age and appro- the Pima population, differences between type 1 and type 2
priate for gestational age preterm subjects, suggesting at least diabetes, as well as different durations of diabetes. As men-
a small decrease in renal functional reserve capacity.208 tioned previously, podocyte abnormalities are present in
GFR and effective renal plasma ow were also studied kidneys with developmentally programmed low nephron
before and after an intravenous amino acid infusion in young numbers, which may contribute to the development of pro-
adults who had diabetic mothers (i.e., had been exposed to teinuria in low birth weight subjects.118
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 665
5

Chronic Kidney Disease A direct link between nephron number and renal disease
Although GFRs have been found to be statistically lower in individual human subjects has not been made, however.
in low birth weight populations, this has not always been Nephron number is unlikely to be the sole cause of renal
outside of the normal range, calling into the question true dysfunction in most patients, and other susceptibilities likely
clinical relevance.55,131 A recent meta-analysis examined compound the risk. A low nephron number, however, may
31 studies that reported risk of CKD—including various lower the threshold to reach a critical loss of renal function in
end points, including proteinuria, diabetic nephropathy, response to superimposed renal injury or stress. In support
and reduced GFR—relative to birth weight.207 Overall they of this possibility, low birth weight has been associated with
found a 70% increased risk of CKD in low birth weight in- poorer renal outcomes in patients with nephrotic syndrome,
dividuals, regardless of end point studied.207 Again, the ef- membranous nephropathy, IgA nephropathy, minimal change
fect was stronger in males. Among 12,364 participants in the disease, and diabetic nephropathy.95,219–222
Kidney Early Evaluation Program, among men a U-shaped Interestingly in a model of diabetes superimposed on
curve was found for risk of CKD, de ned as an estimated low nephron number, the increase in glomerular volume in
GFR 60 mL per minute or albumin excretion 30 g per response to hyperglycemia was more exaggerated and mal-
g, and birth weight, with increased risk with birth weights adaptive in the low compared to normal birth weight rats.223
2,500 g and 4,500 g.73 No association was found among Similarly, mesangioproliferative glomerulonephritis was as-
female participants, however. sociated with signi cantly increased glomerulosclerosis in
The clinical relevance of the risk of CKD with low or low birth weight animals with reduced nephron numbers.224
high birth weights is borne out in studies examining risk of Potential molecular mechanisms whereby kidneys with few-
ESRD. Retrospective analysis in over 2 million Norwegians er nephrons adapt differently than normal kidneys include
found the relative risk of ESRD to be 1.7 in males and females an imbalance between apoptosis and cell proliferation, ac-
born below the 10th percentile in weight.217 Interestingly, celerated senescence, reactive oxygen species generation,
when looking at absolute birth weights and risk, the relative and mitochondrial dysfunction.162,193
risk for ESRD was 2.0 with birth weights 2.5 kg, but was
only increased in females with birth weights 4.5 kg.217 In a RELEVANCE OF NEPHRON MASS IN
predominantly black, southern U.S. population, a U-shaped TRANSPLANTATION
curve was again described for risk of ESRD with low and high
birth weights, this time in both men and women.218 Impact of Kidney Donation
Growing numbers of epidemiologic studies therefore In experimental models of low birth weight, as described
support the relationship between high birth weight or low previously, nephron numbers are often reduced by 25% to
birth weight and risk of subsequent renal disease (Table 2.6). 30%, and animals develop spontaneous hypertension and

TA B L E

2.6 Clinical Findings Associated with Birth Weight, Nephron Number, and
Kidney Size in Humans

Low Birth Weight/ Low Nephron Number Reduced Renal Size High Birth Weight/
Prematurity Maternal Diabetes

↑ Blood pressure127 ↑ Blood pressure70 ↑ Blood pressure105 Proteinuria95


Salt sensitivity105,176 ↑ Glomerular volume13 Salt sensitivity105 ↓ Renal functional reserve53
Proteinuria131,210 ? Predisposition to renal ↓ GFR105 End-stage kidney
↓ GFR55,131,201 failure in neonates54 ↓ Renal allograft survival disease217,218
↓ Renal functional if small kidney into
reserve208 large recipient237
Accelerated progression
of primary renal
disease219–222
Chronic kidney disease73,207
End-stage kidney
disease217,218
Death 210

↑, increase; ↓, decrease; ?, unknown.


66
66 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

renal dysfunction.12,37 In humans, donation of a kidney im- forward.229 Indeed, in rat models it has been elegantly shown
plies loss of 50% of nephron mass, and therefore may carry that transplanted nephron mass (i.e., kidneys with vary-
some long-term risk. However, many studies have shown ing nephron numbers) has a signi cant impact on allograft
that kidney donation is safe and former kidney donors have outcome, independent of immunologic barriers.230 As neph-
similar or even better life expectancy and lower risk for ron numbers cannot be determined in vivo, various surro-
ESRD than the general population.153 This paradox might be gates have been examined in humans to assess the impact
due to the very thorough screening of potential donors and of transplanted nephron mass, measurable to some degree
the selection of only the very healthiest subjects. In addition, ex vivo, relative to recipient demand, on allograft outcomes
the studies on long-term outcomes have predominantly been (Table 2.7). Such surrogates include ratios of recipient to do-
done in Caucasians, are limited by signi cant loss to follow- nor body surface area (BSA) or body weight, of kidney vol-
up, and do not use similarly selected, healthy control groups ume to recipient BSA, and of kidney weight to recipient body
for their comparative analysis. weight.231–235 As mentioned previously, kidney mass and kid-
An early retrospective study of 52 kidney donors fol- ney volume do re ect nephron number to some degree, but
lowed after at least 10 years did nd a higher risk of hy- these data should be interpreted with caution, realizing that
pertension and mild proteinuria compared to age-matched BSA is not always proportional to kidney weight, and that
controls and other potential donors, although creatinine two kidneys of the same size may differ in nephron number.
clearance did not deteriorate as a function of time.225 Despite these caveats and the variability of methods
Interestingly the risks of hypertension and proteinuria employed, the evidence shows fairly consistently that small
were greater in men. In contrast, subsequent studies in pre- kidneys, or kidneys from small donors, transplanted into larg-
dominantly Caucasian donors did not nd a signi cantly er recipients, tend to have poorer outcomes.231–235 Duration
increased risk of hypertension and proteinuria, suggesting of follow-up, however, is also a crucial variable, as seen in the
that uninephrectomy is safe.153 Concerns have been raised, donor literature, where differences in hypertension and pro-
however, about possible harm of living kidney donation in teinuria emerge only after many years. Interestingly, an early
other ethnic groups. After a median of 16 years postdona- report in a cohort of renal allograft recipients, with a mean
tion, the incidence of new onset hypertension, CKD, and of 32 months of follow-up, failed to nd any impact of graft
ESRD was signi cantly higher among Australian Aboriginal weight on short-term graft survival.236 With longer follow-up,
kidney donors compared to Caucasians.226 Similarly, among however, in the same cohort, subjects with a low donor kidney
Canadian donors, hypertension was present in 42% of Ab- weight to recipient body weight ratio (DKW/RBW) showed a
originals compared with 19% of Caucasians by a mean of 14 greater adaptive early increase in GFR, which remained stable
years of follow-up, and in 100% of Aboriginal donors by 20 for 7 years, followed by a more rapid loss of GFR as com-
years postdonation.227 Estimated GFR was not different be- pared to the high DKW/RBW group, thus demonstrating the
tween Caucasians and Aboriginals, but proteinuria was more importance of long term follow-up.237 These data suggest that
common among Aboriginal donors. In U.S. cohorts, black smaller kidneys transplanted into larger recipients underwent
kidney donors were found to have signi cantly more hyper- early hyper ltration that could not be sustained inde nitely,
tension and CKD compared to white donors.159,228 resulting in ongoing nephron loss. Over time, the low DKW/
In all of these cohorts donors are presumed to have been RBW group required more antihypertensives, had more pro-
screened and found healthy prior to donation, therefore, teinuria, and on kidney biopsy showed a greater degree of
uninephrectomy in populations with high susceptibility to glomerulosclerosis. Overall, the risk of transplant failure was
hypertension and kidney disease may carry more risk than 55% higher in low DKW/RBW compared to the high DKW/
has thus far been appreciated. Aboriginal Australian and U.S. RBW group at 2 years.237 The authors conclude that mismatch
black populations have lower birth weights than their Cau- between allograft and recipient weight is an independent pre-
casian counterparts, and Canadian Aboriginals have higher dictor of long-term graft survival.
birth weights (World Health Organization). This suggests that These results are consistent with another analysis, which
programming of nephron number may be a factor contribut- was restricted to recipients of living donor kidneys who had
ing to increased hypertension and renal risk postnephrectomy. not experienced any complications within the rst year of
Birth weight and early development history should therefore transplantation. Progressively higher levels of urine protein
be incorporated into a potential donor’s evaluation, and con- excretion and lower GFRs occurred as DKW/RBW fell.238
sideration given to measurement of renal functional reserve, Similarly, a large retrospective analysis of 32,083 recipients
although at present it is likely premature to suggest that deci- of a rst cadaveric kidney transplant, utilizing the ratio of
sions on donor eligibility be based on this information. donor to recipient BSA, found that large recipients of small
kidneys had a 43% increased risk of late allograft failure
Impact on Allograft Function compared to the reference group which was medium sized
The importance of nephron mass as a nonimmunologic de- recipients receiving kidneys from medium sized donors.234
terminant of long-term transplant outcomes has been de- Another study examined the outcomes of kidneys from
bated since the nephron number hypothesis was rst put donors over age 60, presumed to have fewer nephrons by
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 667
7

TA B L E

2.7 Impact of Donor and Recipient Mismatch on Renal Allograft Outcomes

Measurement Allograft Outcome Donor Reference

Donor kidney weight : ↑ Risk of late allograft loss, proteinuria, Cadaveric 237
recipient body weight hypertension, and glomerulosclerosis at
6.2 years with lower ratios
Donor kidney weight : ↓ Creatinine clearance and ↑ proteinuria at Living 238
recipient body weight 3 years with lower ratios
Donor–recipient body ↓ Graft survival at 5 years in low ratio group Living 232
weight ratio
Donor : recipient BSA ↑ Late allograft loss in large recipients who Cadaveric 234a
received kidneys from small donors
Transplant cross-sectional area ↓ Creatinine, trend toward improved Cadaveric 241
(ultrasound): recipient outcome with larger ratios at 12 months
body weight (Tx/W)
Kidney weight (g) ↑ Creatinine clearance with higher kidney Living 231
weight at 12 months

a
More studies reviewed in this reference.
↑, increase; ↓, decrease; BSA, body surface area.

virtue of increased age, relative to recipient BMI, and found clinician’s decision about medication choices, peritransplant
that 5-year allograft survival was signi cantly lower in interventions, and ultimately potentially organ allocation.
recipients with higher BMI or BSA, again suggesting an impact
for mismatch between fewer transplanted functioning neph-
rons and higher donor demand on long-term outcomes.239
THE IMPACT OF PROGRAMMING ON
Not all studies have found consistent results, however, with RELATED ORGAN SYSTEMS
some failing to nd an impact of differing donor to recipi- The interaction of gestational diabetes exposure, birth weight,
ent BSA ratios on outcomes of paired cadaver kidneys.233 The and proteinuria, for example, demonstrates that developmen-
high and low ratios did overlap in this study, however. tal programming may impact multiple organ systems simul-
A more recent study of paired kidneys did nd a donor– taneously.53,95,215 In a Swedish cohort of 18,230 twins, low
recipient size mismatch to be a risk factor for delayed graft birth weight was found to increase the risk of adult type 2 dia-
function.240 Other investigators have examined the ratio of betes with an adjusted odds ratio of 1.44 per 500 g decrease
transplanted kidney cross-sectional area, as measured by in birth weight.242 Similarly, in a Chinese cohort, low birth
ultrasound, relative to recipient weight as a predictor of out- weight was found to be inversely associated with risk of type
comes. They found lower serum creatinines and a trend to- 2 diabetes, and both high and low birth weights were associ-
ward improved graft survival at 5 years in those with higher ated with increased risk of hypertension and abdominal obe-
ratios.241 Overall, therefore, transplanted nephron mass does sity.243 Low birth weight combined with abdominal obesity
appear to have a long-term impact on allograft function. was the strongest predictor of diabetes. Obesity and diabetes
Clinically, however, nephron mass at transplantation is likely are both risk factors for CKD and, therefore, their interaction
impacted by many other factors in addition to nephron en- with low birth weight likely augments this risk. Interestingly,
dowment (e.g., loss through peritransplant injury, immune- low birth weight rats, induced by maternal low protein diet,
mediated injury, donor age, and other donor factors). Kid- suffered more severe cardiac dysfunction after myocardial
neys transplanted with fewer nephrons are likely to have less ischemia and reperfusion compared to normal birth weight
functional reserve and therefore are at risk of declining func- rats.244 In another study, graded surgical reduction in neph-
tion over time. Awareness of this association may impact a ron number in normal rats was associated with progressively
68
68 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

higher systolic blood pressures, left ventricular hypertrophy, does appear to be a strong modi er of risk in various ethnic
and left ventricular systolic and diastolic dysfunction com- groups, as well as under certain clinical conditions (e.g., dia-
pared to sham operated controls.245 In sheep, fetal unine- betes mellitus). As such, surrogate markers for low nephron
phrectomy resulted in hypertension and more severe renal number and adverse perinatal conditions should be screened
dysfunction and albuminuria with age.156 In addition, cardiac for in the clinical setting and appreciated as risk factors that
functional reserve, measured in response to dobutamine in- may not be modi able in the adult. However, they may high-
fusion, was signi cantly reduced by 6 months of age in the light the need to minimize further insults and optimize other
neonatally nephrectomized sheep and left ventricular mass risk factors for hypertension and renal disease. Low birth
was signi cantly increased. In the clinical arena, patients with weight is currently the most useful clinical surrogate for low
CKD often have coexisting diabetes and/or cardiac dysfunc- nephron mass and the developmentally determined risk of
tion, and CKD itself is a known cardiac risk factor. Whether hypertension and renal disease.
some of this association is related to the consequences of renal Much more work is needed to determine the impact of
dysfunction per se—that is, hypertension, volume expansion, high birth weight. The exciting experimental ndings that
proteinuria—or may re ect parallel programming of multiple low nephron numbers can be rescued under some circum-
organ systems in the same individual has yet to be elucidated. stances points to the importance of improving maternal
health before and during gestation, optimizing of neonatal
nutrition, and avoiding nephrotoxins in the early perinatal
CONCLUSION period, as well as raising hope for potential translation of
The concept that nephron mass, at least in part, is deter- therapeutic interventions to the human in the future. From
mined during the perinatal period and has a long-term im- a public health point of view, close attention should be paid
pact on an individual’s subsequent risk of hypertension and to improving perinatal care and early childhood nutrition as
renal disease is now accepted (Fig. 2.10). Nephron number potential tools to stem the growing tides of renal and cardio-
alone is not often enough to result in overt disease, but vascular disease in future generations.

Post-natal Low protein diet, Vit A de ciency, iron


growth de ciency, uterine ischemia, drugs,
restric on alcohol , dexamethasone etc
prematurity Intrauterine
growth
restric on Gene
polymorphisms
Rapid post-natal
catch-up growth Maternal
Low birth weight hyperglycemia

Surrogate markers:
short stature, female, ethnicity,
Low nephron number High birth weight
renal mass, renal volume

↑ Body: kidney size →


↑ Physiologic demand
Reduced ltra on surface area
Altered renal sodium
handling/ salt sensi vity
Accelerated
senescence

↑ glomerular volume
Single nephron
Hypertension
hyper ltra on

Proteinuria
Glomerulosclerosis

Vascular
disease

FIGURE 2.10 Diagram of proposed mechanisms impacting developmental programming of renal disease and hypertension.
(Adapted from Schreuder M, Delemarre-van de Waal H, van Wijk A. Consequences of intrauterine growth restriction for the kidney.
Kidney Blood Press Res. 2006;29:108–125.)
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 669
9

REFERENCES 24. Langley-Evans


32. Wlodek S, Langley-Evans
ME, Westcott K, Siebel AL,A,etMarchand
al. Growth M. Nutritional
restriction beforeprogram-
or after
ming o bloodnephron
birth reduces pressurenumberand renal andmorphology.
increases blood Archpressure
Physiol Biochem.
in male rats2003;111:
. Kidney
Guyton AC,
1. Guyton AC, Coleman
Coleman TG, TG, Y Young
oung DB, DB, et et al.
al. Salt
Salt balance
balance and and long-term
long-term 8–16.
Int. 2008;74:187–195.
blood pressure control. Annu Rev Med. 1980;31:15–27. http://www.ncbi.nlm.nih.gov/pubmed/12715270
33. Moritz KM, Mazzuca MQ, Siebel AL, et al. Uteroplacental insuf ciency
http://www.ncbi.nlm.nih.gov/pubmed/6994602
2. Brenner BM, Garcia DL, Anderson S. Glomeruli and blood pressure. Less 25. Lisle
causes SJ, Lewis
a nephron de RM,cit, Petry
modest CJ,renal
et al.insuf
E ectciency
o maternalbut noiron restriction with
hypertension dur-
2. Brenner
of one, more the BM, Garcia
other? AmDL, Anderson1988;1:335–347.
J Hypertens. S. Glomeruli and blood pressure. Less ing pregnancy
ageing in female onrats
renal morphology
. J Physiol. in the adult rat o spring. Br JNutr. 2003;90:
2009;587:2635–2646.
o 3.
one,McMillen
more theIC, other? Am J Hypertens.
Robinson 1988;1:335–347.
JS. Developmental origins of the metabolic syn- 33–39.
34. Kajantie E, Dunkel L, Turpeinen U, et al. Placental 11 beta-hydroxysteroid
http://www.ncbi.nlm.nih.gov/pubmed/3063284
drome: prediction, plasticity, and programming. Physiol Rev. 2005;85:571–633. 26. Hoppe CC, and
dehydrogenase-2 Evans RG,cortisol/cortisone
fetal Bertram JF, et al.shuttle E ects in o small
dietarypreterm
protein infants
restric-.
3. Hsu
4. McMillen
CY, Lin IC,F,Robinson
Vittinghoff JS.E,Developmental origins o the
et al. Racial differences metabolic
in the syn-
progression tion
J Clinon nephronMetab.
Endocrinol number in the mouse. Am J Physiol Regul Integr Comp Physiol.
2003;88:493–500.
drome:
from prediction,
chronic plasticity,
renal insuf andtoprogramming.
ciency end-stage renal Physiol
diseaseRev.in2005;85:571–633.
the United States. J 2007;292:R1768–R1774.
35. Habib S, Gattineni J, Twombley K, et al. Evidence that prenatal program-
http://www.ncbi.nlm.nih.gov/pubmed/15788706
Am Soc Nephrol. 2003;14:2902–2907. 27. of
ming Bryce J, CoitinhobyD,dietary
hypertension Darnton-Hill
protein I,deprivation
et al. Maternal and child
is mediated by undernutri-
fetal gluco-
4. Fan
5. Hsu Z,CY , Lin F,S,Vittingho
Lipsitz Egan B, etE,al.etThe al. Racial
impactdiof erences
birth weight in theonprogression
the racial tion: e ective
corticoid actionAm
exposure. at national
J Hypertens. level.2011;24(1):96–101.
Lancet. 2008;371:510–526.
rom chronic
disparity renal insu
of end-stage renalciency
diseaseto .end-stage
Ann Epidemiol.renal 2000;10:459.
disease in the United States.J http://www.ncbi.nlm.nih.gov/pubmed/18206224
36. Wintour EM, Moritz KM, Johnson K, et al. Reduced nephron number
Am6.Soc Nephrol.
Fan 2003;14:2902–2907.
ZJ, Lackland DT, Kenderes B, et al. Impact of birth weight on familial in28. Merlet-Benichou
adult sheep, hypertensive C, VilarasJ,aLelievre-Pegorier
result of prenatal M,glucocorticoid
et al. Role o retinoids
treatmentin.
5. Fan Z,ofLipsitz
aggregation end-stageS, Egan
renal B, et al.. Am
disease TheJimpact
Nephrol.o 2003;23:117–120.
birth weight on the racial renal development:
J Physiol. pathophysiological implication. Curr Opin Nephrol Hypertens.
2003;549:929–935.
disparity o end-stage
7. Lackland DT, EganrenalBM,
disease.
SyddallAnn Epidemiol.
HE, et al. 2000;10:459.
Associations between birth 1999;8:39–43.
37. Celsi G, Kistner A, Aizman R, et al. Prenatal dexamethasone causes oligo-
http://www.ncbi.nlm.nih.gov/pubmed/11018370
weight and antihypertensive medication in black and white Medicaid recipients. http://www.ncbi.nlm.nih.gov/pubmed/9914859
nephronia, sodium retention, and higher blood pressure in the offspring. Pediatr
6. Fan ZJ, 2002;39:179–183.
Hypertension. Lackland DT, Kenderes B, et al. Impact o birth weight on amilial 29.1998;44:317–322.
Res. Goodyer P, Kurpad A, Rekha S, et al. E ects o maternal vitamin A status
aggregation
8. Baum oM.end-stage
Role of the renal disease.
kidney in theAmprenatal
J Nephrol. and2003;23:117–120.
early postnatal program- on38.
kidney development:
Bramlage a pilot study.
CP, Schlumbohm PediatrCR,
C, Pryce Nephrol.
et al. 2007;22:209–214.
Prenatal dexamethasone
http://www.ncbi.nlm.nih.gov/pubmed/12481151
ming of hypertension . Am J Physiol Renal Physiol. 2010;298:F235–247. http://www.ncbi.nlm.nih.gov/pubmed/17093988
exposure does not alter blood pressure and nephron number in the young adult
7. Hoy
9. Lackland DT, EganMD,
WE, Hughson BM,Bertram
SyddallJF,HE, et al.etNephron
al. Associations
number, between birth
hypertension, 30. Sutherland
marmoset monkey.MR, Gubhaju L,2009;54:1115–1122.
Hypertension. Yoder BA, et al. The e ects o postnatal reti-
weightdisease,
renal and antihypertensive
and renal failure medication in black 2005;16:2557–2564.
. J Am Soc Nephrol. and white Medicaid recipients. noic
39. acid administration
Schreuder MF, Bueters on nephron
RR, Huigen endowment
MC, etinal. theEffect
preterm baboononkidney.
of drugs renal
Hypertension.
10. Keller G,2002;39:179–183.
Zimmer G, Mall G, et al. Nephron number in patients with pri- Pediatr Res. 2009;65:397–402.
development. Clin J Am Soc Nephrol. 2011;6:212–217.
http://www.ncbi.nlm.nih.gov/pubmed/11799099
mary hypertension. N Engl J Med. 2003;348:101–108. http://www.ncbi.nlm.nih.gov/pubmed/19092718
40. Kent AL, Douglas-Denton R, Shadbolt B, et al. Indomethacin, ibuprofen
8. Schreuder
11. Baum M. Role MF, oLangemeijer
the kidneyME, in the prenatal and
Bokenkamp A, early
et al. postnatal program-
Hypertension and 31.gentamicin
and Bhat PV, Manolescu
administered DC.during
Role olate vitamin
stagesAofinglomerulogenesis
determining nephron do notmass re-
ming o hypertension.
microalbuminuria Am J Physiol
in children with Renal Physiol.
congenital 2010;298:F235–247.
solitary kidneys. J Paediatr Child and
ducepossible
glomerularrelationship
number to at hypertension.
14 days of ageJ Nutr. in the2008;138:1407–1410.
neonatal rat. Pediatr Nephrol.
9. Hoy
Health. WE, Hughson MD, Bertram JF, et al. Nephron number, hypertension,
2008;44:363–368. http://www.ncbi.nlm.nih.gov/pubmed/18641182
2009;24:1143–1149.
renal
12. disease,
Vehaskari and renal
VM, ailure.
Aviles DH,J Manning
Am Soc Nephrol. 2005;16:2557–2564.
J. Prenatal programming of adult hy- 32. Wlodek ME,
41. Komhoff M, Westcott
Wang JL,K,Cheng Siebel AL,HF, etetal.al.Growth restriction be ore or a in-
Cyclooxygenase-2.selective ter
http://www.ncbi.nlm.nih.gov/pubmed/16049069
pertension in the rat. Kidney Int. 2001;59:238–245. birth reduces
hibitors impairnephron number andand
glomerulogenesis increases
renal blood
corticalpressure in male. rats.
development KidneyKidney
Int.
10. Puelles
13. Keller G,VG,Zimmer
Hoy WE, G, Hughson
Mall G, etMD, al. Nephron numbernumber
et al. Glomerular in patients
and with pri-
size vari- Int. 2008;74:187–195.
2000;57:414–422.
mary hypertension.
ability and risk for kidneyN Engldisease.
J Med. 2003;348:101–108.
Curr Opin Nephrol Hypertens. 2011;20:7–15. http://www.ncbi.nlm.nih.gov/pubmed/18432184
42. Nathanson S, Moreau E, Merlet-Benichou C, et al. In utero and in vitro
http://www.ncbi.nlm.nih.gov/pubmed/12519920
14. Fonseca Ferraz ML, Dos Santos AM, Cavellani CL, et al. Histochemical and 33. Moritz
exposure KM, Mazzuca
to beta-lactams MQ,kidney
impair Siebeldevelopment
AL, et al. Uteroplacental
in the rat. J Aminsu ciency
Soc Nephrol.
11. Schreuder MF, Langemeijer
immunohistochemical study of theME, Bokenkamp
glomerular A, et al. in
development Hypertension
human fetuses and. causes a nephron de cit, modest renal insu ciency but no hypertension with
2000;11:874–884.
microalbuminuria
Pediatr in children with congenital solitary kidneys. J Paediatr Child
Nephrol. 2008;23:257–262. ageing in emale rats. J Physiol.
43. Tendron-Franzin A, Gouyon 2009;587:2635–2646.
JB, Guignard JP, et al. Long-term effects of in
Health. 2008;44:363–368.
15. Bertram JF. Counting in the kidney. Kidney Int. 2001;59:792–796. http://www.ncbi.nlm.nih.gov/pubmed/19359373
utero exposure to cyclosporin A on renal function in the rabbit. J Am Soc Nephrol.
http://www.ncbi.nlm.nih.gov/pubmed/18476930
16. Nyengaard JR. Stereologic methods and their application in kidney re- 34. Kajantie E, Dunkel L, Turpeinen U, et al. Placental 11 beta-hydroxysteroid
2004;15:2687–2693.
12. .Vehaskari
search VM, Aviles
J Am Soc Nephrol. DH, Manning J. Prenatal programming o adult hy-
1999;10:1100–1123. dehydrogenase-2
44. Gray SP, Denton and KM, etal Cullen-McEwen
cortisol/cortisoneL,shuttle in smallexposure
et al. Prenatal pretermtoinalcohol
ants. J
pertension
17. Basgen in the
JM,rat. KidneyMW,
Steffes Int. 2001;59:238–245.
Stillman AE, et al. Estimating glomerular Clin Endocrinol
reduces nephron Metab.
number 2003;88:493–500.
and raises blood pressure in progeny. J Am Soc Nephrol.
http://www.ncbi.nlm.nih.gov/pubmed/11135076
number in situ using magnetic resonance imaging and biopsy. Kidney Int. 35. Habib S, Gattineni J, Twombley K, et al. Evidence that prenatal program-
2010;21:1891–1902.
13. Puelles VG, Hoy WE, Hughson MD, et al. Glomerular number and size
1994;45:1668–1672. ming
45. oGrayhypertension
SP, Kenna K, byBertram
dietary protein
JF, et al. deprivation
Repeated ethanol is mediated
exposureby during
etal gluco-
late
vari
18.ability and risk
Nyengaard or kidneyTF
JR, Bendtsen disease.
. Glomerular Curr number
Opin Nephrol
and size Hypertens. 2011;20:
in relation to age, corticoid
gestation exposure. Am J Hypertens.
decreases nephron endowment 2011;24(1):96–101.
in fetal sheep. Am J Physiol Regul Integr
7–15. weight, and body surface in normal man. Anat Rec. 1992;232:194–201.
kidney 36. Physiol.
Comp Wintour2008;295:R568–574.
EM, Moritz KM, Johnson K, et al. Reduced nephron number
14. Hughson
19. Fonseca Ferraz ML, Dos
M, Farris AB,Santos AM, CavellaniR,CL,
Douglas-Denton et etal.al. Glomerular
Histochemical and
num- in46.
adult sheep,
Valero De hypertensive
Bernabe J, Soriano as a result o prenatalR,glucocorticoid
T, Albaladejo et al. Risk factorstreatment.
for lowJ
immunohistochemical
ber and size in autopsystudy o theThe
kidneys: glomerular
relationship development
to birth weight in human etuses.
. Kidney Int. Physiol. 2003;549:929–935.
birth weight: a review. Eur J Obstet Gynecol Reprod Biol. 2004;116:3–15.
Pediatr Nephrol. 2008;23:257–262.
2003;63:2113–2122. http://www.ncbi.nlm.nih.gov/pubmed/12730337
47. Vikse BE, Irgens LM, Leivestad T, et al. Preeclampsia and the risk of end-
http://www.ncbi.nlm.nih.gov/pubmed/17990004
20. Hoy WE, Douglas-Denton RN, Hughson MD, et al. A stereological study 37. renal
stage Celsidisease.
G, Kistner A, Aizman
N Engl R, et al. Prenatal dexamethasone causes oligo-
J Med. 2008;359:800–809.
of15. Bertramnumber
glomerular JF. Counting in the kidney.
and volume: preliminaryKidney ndings
Int. 2001;59:792–796.
in a multiracial study of nephronia,
48. Manalich sodium retention,
R, Reyes and higher
L, Herrera M, et al.blood pressure between
Relationship in the o weight
spring. atPediatr
birth
http://www.ncbi.nlm.nih.gov/pubmed/11168963
kidneys at autopsy. Kidney Int. 2003:S31–S37. Res.
and 1998;44:317–322.
the number and size of renal glomeruli in humans: a histomorphometric
16. Moritz
21. NyengaardKM, JR. Stereologic
Wintour EM, Black methodsMJ, etand their application
al. Factors in uencinginmammalian
kidney re- http://www.ncbi.nlm.nih.gov/pubmed/9727707
study. Kidney Int. 2000;58:770–773.
search. development:
kidney J Am Soc Nephrol. 1999;10:1100–1123.
implications for health in adult life. Adv Anat Embryol Cell 38. Bramlage
49. Amri CP, Schlumbohm
K, Freund N, Vilar J, et al. C, Adverse
Pryce CR, et al.of Prenatal
effects hyperglycemiadexameth asone
on kidney
http://www.ncbi.nlm.nih.gov/pubmed/10232698
Biol. 2008;196:1–78. exposure
development doesinnot alter
rats: blood
in vivo andpressure
in vitroand nephron
studies. number
Diabetes. in the young adult
1999;48:2240–2245.
17. Luyckx
22. Basgen VA,JM, Brenner
Ste es MW, BM. Stillman
NephronAE, et al. Estimating
endowment. In: Brenner glomerular
BM, ed.num-The marmoset
50. Tran monkey.
S, Chen Hypertension.
YW, Chenier I,2009;54:1115–1122.
et al. Maternal diabetes modulates renal mor-
ber in situ
Kidney, 9th using magnetic resonance
ed. Philadelphia: imaging and biopsy. Kidney Int. 1994;45:
Elsevier; 2011. http://www.ncbi.nlm.nih.gov/pubmed/19770406
phogenesis in offspring. J Am Soc Nephrol. 2008;19:943–952.
1668–1672.
23. Lucas SR, Costa Silva VL, Miraglia SM, et al. Functional and morphometric 39. SchreuderA,MF
51. Magaton , Bueters
Gil RR, Huigen
FZ, Casarini DE, etMC, et al. E ect
al. Maternal o drugs
diabetes on renal de-
mellitus—early
http://www.ncbi.nlm.nih.gov/pubmed/7933814
evaluation of offspring kidney after intrauterine undernutrition . Pediatr Nephrol. velopment.
consequences Clin
forJ Am
the Soc Nephrol.
offspring 2011;6:212–217.
. Pediatr Nephrol. 2007;22:37–43.
18. Nyengaard JR, Bendtsen TF. Glomerular number and size in relation to age,
1997;11:719–723. http://www.ncbi.nlm.nih.gov/pubmed/21071516
52. Rocco L, Gil FZ, da Fonseca Pletiskaitz TM, et al. Effect of sodium over-
kidney weight, and body
24. Langley-Evans sur ace in normal
S, Langley-Evans man. Anat
A, Marchand M. Rec. 1992;232:194–201.
Nutritional programming 40. on
load Kent AL, function
renal Douglas-Denton
of offspring R, Shadbolt B, et al.mothers
from diabetic Indomethacin,
. Pediatribupro
Nephrol. en
http://www.ncbi.nlm.nih.gov/pubmed/1546799
of blood pressure and renal morphology. Arch Physiol Biochem. 2003;111:8–16. and gentamicin administered during late stages o glomerulogenesis do not re-
2008;23:2053–2060.
19. Lisle
25. Hughson
SJ, Lewis M, RM,
Farris AB,CJ,Douglas-Denton
Petry et al. Effect of maternalR, et al. ironGlomerular num-
restriction during duce
53. glomerular
Abi Khalil numberC, Travert at 14F, daysFetitao ageS, etin the neonatal
al. Fetal rat. Pediatr
exposure Nephrol.
to maternal
ber and size
pregnancy in autopsy
on renal kidneys:
morphology The
in the relationship
adult rat offspringto. birth
Br J Nutr.weight. Kidney Int.
2003;90:33–39. 2009;24:1143–1149.
type 1 diabetes is associated with renal dysfunction at adult age. Diabetes.
2003;63:2113–2122.
26. Hoppe CC, Evans RG, Bertram JF, et al. Effects of dietary protein restric- 41. Komho M, Wang JL, Cheng HF, et al. Cyclooxygenase-2.selective in-
2010;59(10):2631–2636.
20. onHoy
tion WE, Douglas-Denton
nephron number in the mouse. RN, Hughson MD, etRegul
Am J Physiol al. AIntegr
stereological study
Comp Physiol. hibitors impair glomerulogenesis
54. Rodriguez MM, Gomez AH, Abitbol and renal CL, cortical development. Kidney
et al. Histomorphometric analysisInt.
o glomerular number and volume: preliminary ndings in a multiracial study o
2007;292:R1768–R1774. 2000;57:414–422.
of postnatal glomerulogenesis in extremely preterm infants. Pediatr Dev Pathol.
kidneys
27. Bryceat autopsy.
J, CoitinhoKidneyD, Int. 2003:S31–S37.
Darnton-Hill I, et al. Maternal and child undernutri- http://www.ncbi.nlm.nih.gov/pubmed/10652018
2004;7:17–25.
21. effective
tion: Moritz KM,action Wintour EM, level
at national Black MJ, et 2008;371:510–526.
. Lancet. al. Factors inf uencing mammalian 42. NathansonJ, S,Harambat
55. Bacchetta Moreau E, J, Merlet-Benichou
Dubourg L, et al.C,Both et al.extrauterine
In utero and andinintra-
vitro
kidney development: implications
28. Merlet-Benichou or health in adult
C, Vilar J, Lelievre-Pegorier M,liete.al.
Adv RoleAnat
of Embryol
retinoidsCell
in exposure to beta-lactams
uterine growth restrictionimpairimpairkidney
renal development
function in childrenin the rat. J Am
born Socpreterm
very Nephrol..
Biol. 2008;196:1–78.
renal development: pathophysiological implication. Curr Opin Nephrol Hypertens. 2000;11:874–884.
Kidney Int. 2009;76:445–452.
22. Luyckx VA, Brenner BM. Nephron endowment. In: Brenner BM, ed. The
1999;8:39–43. http://www.ncbi.nlm.nih.gov/pubmed/10770965
56. Csaicsich D, Russo-Schlaff N, Messerschmidt A, et al. Renal failure, comor-
Kidney, 9th ed. Philadelphia:
29. Goodyer P, Kurpad A, Rekha Elsevier; S, 2011.
et al. Effects of maternal vitamin A status 43. Tendron-Franzin
bidity and mortality in preterm A, Gouyon JB, Guignard
infants. JP, et al. Long-term
Wien Klin Wochenschr. e ects o in
2008;120:153–157.
on23. Lucas
kidney SR, Costa Silva
development: VL, Miraglia
a pilot SM, etNephrol.
study. Pediatr al. Functional and morphometric
2007;22:209–214. utero
57. exposure
Askenazi to DJ,cyclosporin
Grif n R, McGwinA on renalG, unction in thekidney
et al. Acute rabbit.injury
J Am Soc Nephrol.
is indepen-
evaluation o o spring
30. Sutherland kidney a L,
MR, Gubhaju terYintrauterine
oder BA, et al. undernutrition.
The effects ofPediatr Nephrol.
postnatal reti- 2004;15:2687–2693.
dently associated with mortality in very low birthweight infants: a matched case-
1997;11:719–723.
noic acid administration on nephron endowment in the preterm baboon kidney. 44. Gray
control SP, Denton
analysis . PediatrKM, Cullen-McEwen
Nephrol. 2009;24:991–997. L, et al. Prenatal exposure to alcohol
http://www.ncbi.nlm.nih.gov/pubmed/9438651
Pediatr Res. 2009;65:397–402. reduces nephronDJ,
58. Askenazi number and raises
Ambalavanan N,blood pressure
Goldstein in progeny.
SL. Acute kidney J Am Soc Nephrol.
injury in criti-
31. Bhat PV, Manolescu DC. Role of vitamin A in determining nephron mass 2010;21:1891–1902.
cally ill newborns: what do we know? What do we need to learn? Pediatr Nephrol.
and possible relationship to hypertension . J Nutr. 2008;138:1407–1410. 2009;24:265–274.
70
70 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

45. Kemper
59. Gray SP,MJ, Kenna K, Bertram DE.
Muller-Wiefel JF, etRenal
al. Repeated
functionethanol exposure
in congenital during late
anomalies of 87. Hinchli eR,SA,
68. Mansano Desai Lynch
M, GargMR,A,Sargent PH, et al.
et al. Enhanced The e ect oin intrauterine
nephrogenesis offspring of
gestation
the kidneydecreases
and urinary nephrontract. endowment
Curr Opin Urol. in etal sheep. Am J Physiol Regul Integr
2001;11:571–575. growth retardation
water-restricted on theAm
rat dams. development o renal
J Obstet Gynecol. nephrons. Br J Obstet Gynaecol.
2007;196:480e1–6.
Comp Physiol. 2008;295:R568–574.
60. Dziarmaga A, Clark P, Stayner C, et al. Ureteric bud apoptosis and renal 1992;99:296–301.
88. Okada T, Yamagishi T, Morikawa Y. Morphometry of the kidney in rat pups
46. ValeroinDe
hypoplasia Bernabe PAX2.Bax
transgenic J, Sorianofetal T, Albaladejo
mice mimics R, ettheal.renal-coloboma
Risk actors orsyn- low http://www.ncbi.nlm.nih.gov/pubmed/1581274
from uninephrectomized mothers. Anat Rec. 1994;240:120–124.
birth weight:
drome . J Am Soc a review.
Nephrol. Eur2003;14:2767–2774.
J Obstet Gynecol Reprod Biol. 2004;116:3–15. 69. Averbukh
89. Hinchli e Z, SA, Sargent
Bogin E, CohnPH, Howard
M, et al. CV The, etrenotrophic
al. Humanfactor,intrauterine renal
a persistent
http://www.ncbi.nlm.nih.gov/pubmed/15294360
61. Cain JE, Di Giovanni V, Smeeton J, et al. Genetics of renal hypoplasia: growth expressed
stimulus that crosses in absolute
the placenta number o glomeruli
in mice . J Physiol.assessed by the disector meth-
1988;404:31–38.
47. Vikse
insights intoBE, theIrgens LM, Leivestad
mechanisms T, et al.nephron
controlling Preeclampsia and the
endowment risk o end-
. Pediatr Res. od andFulladosa
90. CavalieriX, principle.
Moreso Lab Invest. 1991;64:777–784.
F, Narvaez JA, et al. Estimation of total glomerular
stage renal disease. N Engl J Med. 2008;359:800–809.
2010;68:91–98. http://www.ncbi.nlm.nih.gov/pubmed/2046329
number in stable renal transplants. J Am Soc Nephrol. 2003;14:2662–2668.
http://www.ncbi.nlm.nih.gov/pubmed/18716297
62. Sanyanusin P, McNoe LA, Sullivan MJ, et al. Mutation of PAX2 in two 70. Y
91. Hoy
iu V,WE,BukaBertram JF, Denton
S, Zurakowski D, RD,
et al.etRelationship
al. Nephronbetween number,birthweight
glomerular and vol-
48. Manalich
siblings R, Reyes L, Herrera
with renal-coloboma syndrome.M, et Humal. Relationship
Mol Genet. between weight at birth
1995;4:2183–2184. ume, renal
blood pressure disease and hypertension.
in childhood. Am J Kidney Curr
Dis.Opin Nephrol Hypertens. 2008;17:
1999;33:253–260.
and
63. the number
Quinlan and sizeM,o Hudson
J, Lemire renal glomeruli
T, et al. inA humans:
common avariant histomorphometric
of the PAX2 258–
92. 265.
Blumenshine P, Egerter S, Barclay CJ, et al. Socioeconomic disparities in
study. isKidney
gene Int. 2000;58:770–773.
associated with reduced newborn kidney size. J Am Soc Nephrol. http://www.ncbi.nlm.nih.gov/pubmed/18408476
adverse birth outcomes: a systematic review. Am J Prev Med. 2010;39:263–272.
http://www.ncbi.nlm.nih.gov/pubmed/10916101
2007;18:1915–1921. 71. Surkan
93. Gilbert PJ, JS,Hsieh
Nijland CC,MJ. Sex di AL,
Johansson erences
et al.inReasons
the developmental
for increasingorigins
trends in o
49. Zhang
64. Amri K, Z, Freund
QuinlanN,J, Vilar
Hoy J, W,etetal.al.Adverse
A common e ectsRETo hyperglycemia on kidney
variant is associated with hypertension
large and cardiorenal
for gestational disease.Gynecol.
age births. Obstet Am J Physiol Regul Integr Comp Physiol.
2004;104:720–726.
development
reduced newborn in rats: in vivo
kidney sizeandand in vitro studies.
function . J Am Soc Diabetes.
Nephrol.1999;48:2240–2245.
2008;19:2027–2034. 2008;295:R1941–1952.
94. Nelson RG, Morgenstern H, Bennett PH. Birth weight and renal disease in
http://www.ncbi.nlm.nih.gov/pubmed/10535460
65. Zhang Z, Quinlan J, Grote D, et al. Common variants of the glial cell- 72. Indians
Pima Moritz KM, with Cu typee2JS, Wilsonmellitus.
diabetes LB, et al.Am Review: Sex speci
J Epidemiol. c programming:
1998;148:650–656.
50. Tran
derived S, Chen YW,
neurotrophic Chenier
factor geneI, doet al.notMaternal
in uence diabetes
kidneymodulates
size of the renal mor-
healthy a 95.
critical
Nelsonrole RG,or the renal renin-angiotensin
Morgenstern H, Bennett PH. system. Placenta. 2010;31
Intrauterine diabetes Suppl:
expo-
phogenesis
newborn in o spring.
. Pediatr Nephrol.J Am Soc Nephrol. 2008;19:943–952.
2009;24:1151–1157. S40–and
sure S46.the risk of renal disease in diabetic Pima Indians. Diabetes. 1998;47:
http://www.ncbi.nlm.nih.gov/pubmed/18305124
66. Hinchliffe SA, Howard CV, Lynch MR, et al. Renal developmental arrest in 73. Li S, Chen SC, Shlipak M, et al. Low birth weight is associated with chron-
1489–1493.
51. Magaton
sudden infant death A, Gil FZ, Casarini
syndrome. PediatrDE,Pathol.
et al. Maternal diabetes mellitus—early
1993;13:333–343. ic96.
kidney disease
Gilbert only AL,
JS, Lang in men.
GrantKidney
AR, etInt.al. 2008;73:637–642.
Maternal nutrient restriction in sheep:
consequences
67. Gubhaju or L, the o spring.
Sutherland MR, Pediatr
YoderNephrol.
BA, et 2007;22:37–43.
al. Is nephrogenesis affected by http://www.ncbi.nlm.nih.gov/pubmed/18094674
hypertension and decreased nephron number in offspring at 9 months of age.
http://www.ncbi.nlm.nih.gov/pubmed/16967284
preterm birth? Studies in a non-human primate model. Am J Physiol Renal Physiol. 74. Hughson
J Physiol. MD, Douglas-Denton R, Bertram JF, et al. Hypertension, glomer-
2005;565:137–147.
52. Rocco L, Gil FZ, da Fonseca Pletiskaitz TM, et al. E ect o sodium over-
2009;297:F1668–F1677. ular
97. number,
Jones SE,and birth weight
Nyengaard in A ricanA,Americans
JR, Flyvbjerg et al. Birthand white
weight hassubjects in the
no in uence
load
68. on renal unction
Hinchliffe SA, Lynch o MR, o spring
Sargent romPH,diabetic
et al. Themothers.
effect Pediatr Nephrol.
of intrauterine southeastern
on glomerularUnited number States. Kidney Int.
and volume 2006;69:671–678.
. Pediatr Nephrol. 2001;16:340–345.
2008;23:2053–2060.
growth retardation on the development of renal nephrons. Br J Obstet Gynaecol. http://www.ncbi.nlm.nih.gov/pubmed/16395270
98. Hoy WE, Hughson MD, Singh GR, et al. Reduced nephron number and
http://www.ncbi.nlm.nih.gov/pubmed/18574600
1992;99:296–301. 75. McNamara in
glomerulomegaly BJ,Australian
Diou B, Douglas-Denton
Aborigines: a group RN, at et al. A comparison
high risk for renalodisease
neph-
53. Hinchliffe
69. Abi KhalilSA, C, Sargent
Travert PH, F, Fetita
Howard S, CVet, al. Fetal
et al. Humanexposure to maternal
intrauterine renal ron number,
and hypertension glomerular
. Kidneyvolume and kidney weight in Senegalese A ricans and
Int. 2006;70:104–110.
type 1 expressed
growth diabetes isinassociated with renal
absolute number of dys unctionassessed
glomeruli at adultbyage. the Diabetes.
disector A99.
ricanDeutinger
Americans. Nephrol
J, Bartl W, Dial Transplant.
Pfersmann C, et2010;25:1514–1520.
al. Fetal kidney volume and urine
2010;59(10):2631–2636.
method and Cavalieri principle. Lab Invest. 1991;64:777–784. 76. Collins
production in JW,
cases Rankin
of fetalKM,growthDavidretardation.
RJ. Low birth weight
J Perinat across
Med. generations: the
1987;15:307–315.
54. Hoy
70. Rodriguez MM, Gomez
WE, Bertram AH, Abitbol
JF, Denton RD, et al. CL,Nephron
et al. Histomorphometric
number, glomerular analysis
vol- e ect Kurjak
100. o economic environment.
A, Kirkinen P, Latin VMatern Child Health
, et al. Ultrasonic J. 2011;15(4):438–445.
assessment of fetal kidney func-
o postnatal
ume, renal diseaseglomerulogenesis
and hypertension. in extremely
Curr Opin preterm
Nephrol inHypertens.
ants. Pediatr Dev Pathol.
2008;17:258– http://www.ncbi.nlm.nih.gov/pubmed/20390329
tion in normal and complicated pregnancies. Am J Obstet Gynecol. 1981;141:266–270.
2004;7:17–25.
265. 77. Silver
101. Horta LE,BL, Decamps
Gigante DP , Osmond
PJ, Korst LM, C, etet al.
al. Intrauterine
Intergenerational growth e ect o weight
restriction is
http://www.ncbi.nlm.nih.gov/pubmed/17827558
71. Gilbert JS, Nijland MJ. Sex differences in the developmental origins of gain in childhood
accompanied on o spring
by decreased renalbirthweight.
volume in the Inthuman
J Epidemiol.fetus.2009;38:724–732.
Am J Obstet Gynecol.
55. Bacchetta
hypertension andJ, cardiorenal
Harambat J,disease. Dubourg AmL,J et al. Both
Physiol Regulextrauterine
Integr Comp andPhysiol.
intra- http://www.ncbi.nlm.nih.gov/pubmed/19376883
2003;188:1320–1325.
uterine growth restriction impair renal unction in children born very preterm.
2008;295:R1941–1952. 78. Konje
102. Vikse JC,BE, Bell
Irgens
SC,LM,MortonBostad L, al.
JJ, et et al.
HumanAdverse fetalperinatal outcome and dur-
kidney morphometry later
Kidney Int. 2009;76:445–452.
72. Moritz KM, Cuffe JS, Wilson LB, et al. Review: Sex speci c program- kidney
ing biopsyand
gestation in thethemother. J Am Soc
relationship Nephrol.
between 2006;17:837–845.
weight, kidney morphometry and
56. Csaicsich
ming: a critical D, roleRusso-Schla
for the renalN,renin-angiotensin
Messerschmidt A,system. et al. Renal ailure,2010;31
Placenta. comor- http://www.ncbi.nlm.nih.gov/pubmed/16421228
plasma active renin concentration at birth. Clin Sci (Lond). 1996;91:169–175.
bidity and mortality
Suppl:S40– S46. in preterm in ants. Wien Klin Wochenschr. 2008;120:153–157. 79. Schmidt
103. Munkhaugen J, Lydersen
IM, Damgaard IN,S,Boisen
Romundstad
KA, et al. PR, et al. Kidney
Increased kidneyunction
growthandin for-u-
http://www.ncbi.nlm.nih.gov/pubmed/18365155
73. Li S, Chen SC, Shlipak M, et al. Low birth weight is associated with chronic ture risk versus
mula-fed or adverse pregnancy
breast-fed healthy outcomes: a population-based
infants. Pediatr study rom HUNT
Nephrol. 2004;19:1137–1144.
57. Askenazi
kidney disease only DJ, Gri
in men n .R,Kidney
McGwin G, et al. Acute kidney injury is indepen-
Int. 2008;73:637–642. II, Norway.
104. Spencer Nephrol
J, Wang DialZ,Transplant.
Hoy W. Low 2009;24(12):3744–3750.
birth weight and reduced renal volume in
dently associated
74. Hughson MD,with mortality in very
Douglas-Denton R, low birthweight
Bertram JF, et al.inHypertension,
ants: a matched case-
glomer- 80. Harrison
Aboriginal children.M, Langley-Evans
Am J Kidney Dis.SC. Intergenerational programming o im-
2001;37:915–920.
control
ular analysis.
number, andPediatr Nephrol.in2009;24:991–997.
birth weight African Americans and white subjects in the pairedSimonetti
105. nephrogenesis GD, Raio andL,hypertension
Surbek D, et in al. rats ollowing maternal
Salt sensitivity of childrenprotein
with low re-
http://www.ncbi.nlm.nih.gov/pubmed/19238451
southeastern United States. Kidney Int. 2006;69:671–678. striction
birth during
weight. pregnancy. 2008;52:625–630.
Hypertension. Br J Nutr. 2009;101:1020–1030.
58. McNamara
75. Askenazi DJ, BJ,Ambalavanan N, GoldsteinRN,
Diouf B, Douglas-Denton SL. etAcute
al. Akidney injuryofinneph-
comparison criti- 81. Rakow
106. WlodekA,ME, Mibus A,
Johansson Tan A, et L,
S, Legnevall al. etNormal
al. Renal lactational
volume environment
and function re- in
callynumber,
ron ill newborns: what do
glomerular we know?
volume What doweight
and kidney we need to learn? Pediatr
in Senegalese Nephrol.
Africans and stores nephron
school-age endowment
children born pretermand prevents
or small hypertension
for gestational a terage
placental
. Pediatrrestric tion
Nephrol.
2009;24:265–274.
African Americans. Nephrol Dial Transplant. 2010;25:1514–1520. in the rat. J Am Soc Nephrol. 2007;18:1688–1696.
2008;23:1309–1315.
59. Collins
76. KemperJW, MJ,Rankin
Muller-Wie
KM, David el DE.RJ.Renal
Low birthunction in congenital
weight anomaliesthe
across generations: o 82. Keijzer-Veen
107. Makrakis J, MG, ZimanyiDulger MA, Black MJ.
A, Dekker FW,Retinoic
et al. Very acid enhances
preterm birth nephron
is a risk
the kidney
effect and urinary
of economic tract. CurrMatern
environment. Opin Urol.
Child2001;11:571–575.
Health J. 2011;15(4):438–445. endowment
factor in ratssystolic
for increased exposed bloodto maternal
pressure atprotein
a young restriction.
adult age. Pediatr Nephrol.
Pediatr Nephrol.
http://www.ncbi.nlm.nih.gov/pubmed/11734692
77. Horta BL, Gigante DP, Osmond C, et al. Intergenerational effect of weight 2007;22:1861–1867.
2010;25:509–516.
60.inDziarmaga
gain childhood A, on Clark P, Stayner
offspring birthweightC, et. al.
Int Ureteric
J Epidemiol.bud2009;38:724–732.
apoptosis and renal http://www.ncbi.nlm.nih.gov/pubmed/17849154
108. Kent AL, Jyoti R, Robertson C, et al. Are renal volumes measured by mag-
hypoplasia
78. Vikse in BE,transgenic
Irgens LM,PAX2.BaxBostad L, etal et al.mice mimics
Adverse the renal-coloboma
perinatal outcome and later syn- 83. resonance
netic El Kares R, Manolescu
imaging DC, Lakhal-Chaiebultrasound
and three-dimensional L, et al. Ainhuman
the term ALDH1A2
neonate
drome. biopsy
kidney J Am Soc in Nephrol.
the mother 2003;14:2767–2774.
. J Am Soc Nephrol. 2006;17:837–845. gene variant isPediatr
comparable? associated
Nephrol.with2010;25:913–918.
increased newborn kidney size and serum retinoic
http://www.ncbi.nlm.nih.gov/pubmed/14569086
79. Munkhaugen J, Lydersen S, Romundstad PR, et al. Kidney function and acid. Kidney
109. BrennerInt. BM,2010;78:96–102.
Mackenzie HS. Nephron mass as a risk factor for progression
61. Cain
future risk for JE, Di Giovanni
adverse V, Smeeton
pregnancy outcomes:J, et al. Genetics o renal study
a population-based hypoplasia:
from http://www.ncbi.nlm.nih.gov/pubmed/20375987
of renal disease. Kidney Int. Suppl 1997;63:S124–127.
insightsII,into
HUNT the mechanisms
Norway. controlling2009;24(12):3744–3750.
Nephrol Dial Transplant. nephron endowment. Pediatr Res. 84. Tan
110. Li J,JC,
Khodus
WorkenehG, Kruusmagi
B, BusqueM,S,etetal. al.Ouabain
Glomerular protects against
function, adverseand
structure, de-
2010;68:91–98.
80. Harrison M, Langley-Evans SC. Intergenerational programming of im- velopmental
number programming
in renal o theolder
allografts from kidney. Nat Commun.
deceased donors.2010;1:42.
J Am Soc Nephrol. 2009;
http://www.ncbi.nlm.nih.gov/pubmed/20421843
paired nephrogenesis and hypertension in rats following maternal protein re- http://www.ncbi.nlm.nih.gov/pubmed/20975704
20:181–188.
62. Sanyanusin
striction during pregnancyP, McNoe . BrLA, Sullivan
J Nutr. MJ, et al. Mutation o PAX2 in two
2009;101:1020–1030. 85. McNamara
111. Vasarhelyi B, BJ,Dobos
DioufM, B, Reusz
Hughson GS, MD,
et al. etNormal kidney unction
al. Associations between andage,el-
siblings
81. Wlodek with renal-coloboma
ME, Mibus A,syndrome. Tan A, etHum al. Mol Genet.lactational
Normal 1995;4:2183–2184.
environment evatedsize
body natriuresis
and nephron in youngnumber men with bornindividual
with low glomerular
birth weight. PediatrinNephrol.
volumes urban
http://www.ncbi.nlm.nih.gov/pubmed/8589702
restores nephron endowment and prevents hypertension after placental restric- 2000;15:96–100.
West African males. Nephrol Dial Transplant. 2009;24:1500–1506.
63.inQuinlan
tion the rat. JJ,Am Lemire M, Hudson
Soc Nephrol. T, et al. A common variant o the PAX2
2007;18:1688–1696. http://www.ncbi.nlm.nih.gov/pubmed/11095022
112. Hoy WE, Hughson MD, Zimanyi M, et al. Distribution of volumes of indi-
gene is associated
82. Makrakis with reduced
J, Zimanyi MA, Black newborn kidney size.
MJ. Retinoic J Am Soc nephron
acid enhances Nephrol. 86. Li
vidual J, Khodus
glomeruli GR, Kruusmagi
in kidneys at autopsy: M, et al. Ouabain
association withprotects againstnumber,
age, nephron adverse
2007;18:1915–1921.
endowment in rats exposed to maternal protein restriction . Pediatr Nephrol. developmental
birth weight andprogramming
body mass index. o theClin kidney. Nat Commun.
Nephrol. 2010;1:1–7.
2010;74:105–112.
64. Zhang Z, Quinlan J, Hoy W, et al. A common RET variant is associated
2007;22:1861–1867. http://www.ncbi.nlm.nih.gov/pubmed/20975704
113. Zimanyi MA, Hoy WE, Douglas-Denton RN, et al. Nephron number and
with
83. reduced
El Kares newbornR, Manolescu kidney DC,size and unction.L,JetAm
Lakhal-Chaieb al.Soc Nephrol.ALDH1A2
A human 2008;19: 87. Mansano
individual R, Desai
glomerular M, GarginA,male
volumes et al.Caucasian
Enhancedand nephrogenesis in o spring
African American sub-
2027–2034.
gene variant is associated with increased newborn kidney size and serum retinoic o water-restricted
jects. rat dams. Am
Nephrol Dial Transplant. J Obstet Gynecol. 2007;196:480e1–6.
2009;24:2428–2433.
65. Kidney
acid. ZhangInt. Z, 2010;78:96–102.
Quinlan J, Grote D, et al. Common variants o the glial cell- http://www.ncbi.nlm.nih.gov/pubmed/17466712
114. Ritz E, Koleganova N. African Americans compared to Senegalese—same
derived
84. Li neurotrophic
J, Khodus G, actor Kruusmagigene do M, notet al.infOuabain
uence kidney protectssizeagainst
o the adverse
healthy 88. Okada
number T, Yamagishi
of glomeruli, T, Morikawa
but greater glomerular Y. Morphometry
size. What doesothis thetell
kidney in rat
us? Nephrol
newborn. Pediatr
developmental Nephrol. 2009;24:1151–1157.
programming of the kidney. Nat Commun. 2010;1:42. pupsTransplant.
Dial rom uninephrectomized
2010;25:1368–1370. mothers. Anat Rec. 1994;240:120–124.
66. Vasarhelyi
85. Hinchli e SA, HowardM,CV
B, Dobos , LynchGS,
Reusz MR,etetal.al.Normal
Renal developmental
kidney function arrestandin http://www.ncbi.nlm.nih.gov/pubmed/7810908
115. Abdi R, Slakey D, Kittur D, et al. Baseline glomerular size as a predictor of
sudden innatriuresis
elevated ant deathinsyndrome.
young men Pediatr
bornPathol.
with low 1993;13:333–343.
birth weight. Pediatr Nephrol. 89. Averbukh
function in human Z, renal
Bogintransplantation.
E, Cohn M, et al. The renotrophic
Transplantation. actor, a persistent
1998;66:329–333.
http://www.ncbi.nlm.nih.gov/pubmed/8516228
2000;15:96–100. stimulus
116. that crosses
Hughson MD, Gobethe placenta
GC, Hoy in WE,
mice.etJ al.
Physiol. 1988;404:31–38.
Associations of glomerular num-
67. Li
86. Gubhaju
J, Khodus L, Sutherland
GR, Kruusmagi MR, YM, oderetBA, et al. Is nephrogenesis
al. Ouabain protects against a ected
adverse by http://www.ncbi.nlm.nih.gov/pubmed/3253434
ber and birth weight with clinicopathological features of African Americans and
preterm birth? Studies
developmental programming in a non-human
of the kidney. primateNat model.
Commun. Am2010;1:1–7.
J Physiol Renal Physiol. 90. Fulladosa
whites. Am J Kidney X, Moreso F, Narvaez JA, et al. Estimation o total glomerular
Dis. 2008;52:18–28.
2009;297:F1668–F1677. number in stable renal transplants. J Am Soc Nephrol. 2003;14:2662–2668.
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 771
1

91. Hodgin
117. Yiu V, Buka S, Zurakowski
JB, Rasoulpour M,D,Markowitz
et al. Relationship
GS, et al.between
Very low birthweight and
birth weight 114. Ritz E,NS,
145. Levitt Koleganova
Steyn K, N. DeAWet ricanT,Americans compared
et al. An inverse to Senegalese—same
relation between blood
blood
is a riskpressure
factor forin childhood.
secondary Am focalJ Kidney
segmentalDis. 1999;33:253–260.
glomerulosclerosis. Clin J Am Soc number
pressureoand glomeruli,
birth weight but greater
among glomerular size. What
5 year old children from does this tell
Soweto, us? Nephrol
South Africa.
http://www.ncbi.nlm.nih.gov/pubmed/10023635
Nephrol. 2009;4:71–76. Dial Transplant.
J Epidemiol 2010;25:1368–1370.
Community Health. 1999;53:264–268.
92. Villar-Martini
118. Blumenshine PVC, , Egerter
CarvalhoS, Barclay
JJ, NevesCJ, et MFal.
, etSocioeconomic
al. Hypertension disparities
and kid-in http://www.ncbi.nlm.nih.gov/pubmed/20228072
146. Gilbert T, Lelievre-Pegorier M, Merlet-Benichou C. Long-term effects of
adverse
ney birth outcomes:
alterations a systematic
in rat offspring from low review. Am Jpregnancies.
protein Prev Med. 2010;39:263–272.
J Hypertens Suppl. 115. Abdi R, Slakey D,
mild oligonephronia Kittur D,
induced in et al. Baseline
utero glomerular
by gentamicin in size as a. Pediatr
the rat predictor o
Res.
http://www.ncbi.nlm.nih.gov/pubmed/20709259
2009;27:S47–51. unction in human renal transplantation. Transplantation. 1998;66:329–333.
1991;30:450–456.
93. Benz
119. SurkanK,PJ,Campean
Hsieh CC,V,Johansson
Cordasic AL, N, etetal.al.Reasons or increasingalterations
Early glomerular trends in http://www.ncbi.nlm.nih.gov/pubmed/9721801
147. Langley-Evans SC. Intrauterine programming of hypertension in the rat:
large
in or gestational
genetically age births.
determined lowObstet
nephron Gynecol.
number2004;104:720–726.
. Am J Physiol Renal Physiol. 116.
nutrientHughson MD, Gobe
interactions. CompGC, Hoy Physiol
Biochem WE, etAal.Physiol.
Associations o glomerular num-
1996;114:327–333.
http://www.ncbi.nlm.nih.gov/pubmed/15458892
2011;300(2):F521–530. ber
148.and birth
Ortiz LA,weight
Quanwith clinicopathological
A, Weinberg A, et al. Effect eatures o A rican
of prenatal Americans and
dexamethasone on
94. Jones
120. NelsonSE, RG, Morgenstern
White KE, FlyvbjergH, Bennett
A, et al.PH.The Birth weight
effect and renal disease
of intrauterine environ-in whites.
rat renalAm J Kidney Dis.
development 2008;52:18–28.
. Kidney Int. 2001;59:1663–1669.
Pima Indians
ment and lowwith type 2 diabetes
glomerular numbermellitus. Am J Epidemiol.
on the histological 1998;148:650–656.
changes in diabetic glo- http://www.ncbi.nlm.nih.gov/pubmed/18514988
149. Woods LL, Ingel nger JR, Nyengaard JR, et al. Maternal protein restriction
http://www.ncbi.nlm.nih.gov/pubmed/9778171
merulosclerosis. Diabetologia. 2006;49:191–199. 117. Hodgin
suppresses theJB, Rasoulpour
newborn M, Markowitz
renin-angiotensin GS, etand
system al. Very
programslow birth
adultweight is a
hyperten-
95. Barker
121. Nelson DJ. RG, Morgenstern
The fetal origins H, Bennett PH. Intrauterine
of adult hypertension. diabetes exposure
J Hypertens and
Suppl. risk
sion inactor
rats.orPediatr
secondary ocal segmental glomerulosclerosis. Clin J Am Soc Neph-
Res. 2001;49:460–467.
the risk o renal disease in diabetic Pima Indians. Diabetes. 1998;47: 1489–1493.
1992;10:S39–44. rol.
150.2009;4:71–76.
Alexander BT. Placental insuf ciency leads to development of hyperten-
96. Curhan
122. Gilbert JS,GC,Lang AL,WC,
Willett GrantRimm AR, etEB,al. et
Maternal
al. Birthnutrient
weight and restriction in sheep:
adult hyperten- http://www.ncbi.nlm.nih.gov/pubmed/21137017
sion in growth-restricted offspring. Hypertension. 2003;41:457–462.
hypertension
sion, diabetes and decreased
mellitus, nephron
and obesity number
in US men. in o spring 1996;94:3246–3250.
Circulation. at 9 months o age. J 118. Villar-Martini VC,
151. Langley-Evans SC, Carvalho
Jackson AA. JJ, Neves
Rats with MF, et al. Hypertension
hypertension induced andbykid-in
Physiol.
123. 2005;565:137–147.
Doyle LW, Faber B, Callanan C, et al. Blood pressure in late adolescence ney
uteroalterations
exposure in to rat o spring
maternal rom low diets
low-protein protein failpregnancies.
to increase blood J Hypertens
pressureSuppl.
in
http://www.ncbi.nlm.nih.gov/pubmed/15790663
and very low birth weight. Pediatrics. 2003;111:252–257. 2009;27:S47–51.
response to a high salt intake. Ann Nutr Metab. 1996;40:1–9.
97. Langley
124. Jones SE,SC, Nyengaard
Jackson JR, AA. Flyvbjerg
Increased A, etsystolic
al. Birthblood
weightpressure
has no infin uence
adult 119. Benz K, FCampean
152. Boubred , Buffat C, V , CordasicJM,
Feuerstein N,etetal.al. Earlyofglomerular
Effects early postnatal alterations
hyper-
on glomerular
rats induced by number and volume.
fetal exposure Pediatr Nephrol.
to maternal low protein2001;16:340–345.
diets. Clin Sci (Lond). in genetically
nutrition on nephrondetermined
number lowandnephron
long-term number. Am J Physiol
renal function Renal Physiol.
and structure in rats.
http://www.ncbi.nlm.nih.gov/pubmed/11354778
1994;86:217–222; discussion 121. 2011;300(2):F521–530.
Am J Physiol Renal Physiol. 2007;293:F1944–1949.
98. Longo-Mbenza
125. Hoy WE, Hughson MD, Singh
B, Ngiyulu GR, et M,
R, Bayekula al. et
Reduced
al. Lownephron
birth weight number and
and risk 120. Jones SE,HN,
153. Ibrahim White KE, R,
Foley Flyvbjerg
Tan L, A, et et
al. al. The e ectconsequences
Long-term o intrauterineofenviron-
kidney
glomerulomegaly
of hypertension ininAfrican Australian
school Aborigines:
children. Ja Cardiovasc
group at high Risk.risk or renal disease
1999;6:311–314. ment and N
donation. lowEngl glomerular number on the histological changes in diabetic glo-
J Med. 2009;360:459–469.
and hypertension.
126. Manning J, VehaskariKidney Int. VM.2006;70:104–110.
Low birth weight-associated adult hypertension merulosclerosis.
154. Kett MM, Bertram Diabetologia. 2006;49:191–199.
JF. Nephron endowment and blood pressure: what do
http://www.ncbi.nlm.nih.gov/pubmed/16723986
in the rat. Pediatr Nephrol. 2001;16:417–422. http://www.ncbi.nlm.nih.gov/pubmed/16365725
we really know? Curr Hypertens Rep. 2004;6:133–139.
99. Huxley
127. Deutinger RR,J,Shiell
Bartl W,AW,P Law
ersmannCM. C, Theet role
al. Fetal kidney
of size volume
at birth and and urine
postnatal 121. Barker KM,
155. Moritz DJ. Wintour
The etalEM, origins
DodicoM.adult hypertension. J leads
Fetal uninephrectomy Hypertens Suppl.
to postnatal
production
catch-up in cases
growth in odetermining
etal growth retardation.
systolic blood Jpressure:
Perinat Med. 1987;15:307–315.
a systematic review of 1992;10:S39–44.
hypertension and compromised renal function. Hypertension. 2002;39:1071–1076.
http://www.ncbi.nlm.nih.gov/pubmed/3323459
the literature. J Hypertens. 2000;18:815–831. 122. CurhanRR,GC,
156. Singh Willett
Denton KM,WC, RimmJF, EB,
Bertram et al.et Development
al. Birth weight and adult
of cardiovascu-
100. Chen
128. KurjakW,A,Srinivasan
Kirkinen PSR, , Latin V, et al.
Berenson GS.Ultrasonic
Ampli cation assessment
of theoassociation
etal kid- hyperten
lar disease sion,due diabetes
to renal insufmellitus,
ciencyand obesity
in male sheepin following
US men. fetalCirculation.
unilateral
ney uncbirthweight
between tion in normal and complicated
and blood pressure with pregnancies. AmJ Obstet
age: the Bogalusa HeartGynecol.
Study. 1996;94:3246–3250.
nephrectomy. J Hypertens. 2009;27:386–396.
J1981;141:266–270.
Hypertens. 2010;28:2046–2052. http://www.ncbi.nlm.nih.gov/pubmed/8989136
157. Woods LL, Weeks DA, Rasch R. Hypertension after neonatal uninephrec-
101. V
129. Silver
ancheri LE, Decamps
F, Alletto PJ, Korst
M, Burgio A, etLM, et al. Intrauterine
al. [Inverse relationshipgrowth
betweenrestriction
fetal growthis 123.
tomy in Doyle LW, Faberglomerular
rats precedes B, Callanan C, et al.
damage. Blood pressure
Hypertension. in late adolescence
2001;38:337–342.
accompanied
and by decreased
arterial pressure renal
in children andvolume
adults].in Gthe human
Ital Cardiol.etus. Am J Obstet Gynecol.
1995;25:833–841. and
158.very low birthJR.
Nyengaard weight.
Number Pediatrics. 2003;111:252–257.
and dimensions of rat glomerular capillaries in
2003;188:1320–1325.
130. Yang S, Bergvall N, Cnattingius S, et al. Gestational age differences in http://www.ncbi.nlm.nih.gov/pubmed/12563047
normal development and after nephrectomy. Kidney Int. 1993;43:1049–1057.
102. Konje
health JC, Bell SC,among
and development Morton youngJJ, et al. Human
Swedish men born etal atkidney
term. Intmorphometry
J Epidemiol. 124. Langley EM,
159. Gibney SC, Parikh
JacksonCR, AA.Garg Increased
AX. Age, systolic
gender, blood
race,pressure in adult
and associations
during gestation and the relationship between weight, kidney morphometry and
2010;39:1240–1249. rats
with induced by etal
kidney failure exposure
following to maternal
living low protein
kidney donation diets. Clin
. Transplant Proc.Sci (Lond).
2008;40:
plasmaKeijzer-V
131. active renin
een MG, concentration
Schrevel M,atFinkenbirth. Clin
MJ, et Scial.
(Lond). 1996;91:169–175.
Microalbuminuria and low- 1994;86:217–222;
1337–1340. discussion 121.
103.
er Schmidt ltration
glomerular IM, Damgaard IN, Boisen
rate at young adult KA,age inet subjects
al. Increased
born verykidney growth
premature http://www.ncbi.nlm.nih.gov/pubmed/8143432
160. Abdi R, Slakey D, Kittur D, et al. Heterogeneity of glomerular size in nor-
in or
and mula-
after ed versus
intrauterine breast-
growth ed healthy
retardation . J Am inSoc
ants. Pediatr
Nephrol. Nephrol. 2004;19:
2005;16:2762–2768. 125. Longo-Mbenza
mal donor kidneys: Impact B, Ngiyulu
of race.R, Am
Bayekula
J Kidney M,Dis.
et al.1998;32:43–46.
Low birth weight and risk
1137–1144.
132. Schmidt IM, Chellakooty M, Boisen KA, et al. Impaired kidney growth in o161.
hypertension
Baum M. inOverview A rican school children.
of chronic kidneyJ Cardiovasc
disease Risk. 1999;6:311–314.
in children. Curr Opin
http://www.ncbi.nlm.nih.gov/pubmed/15309602
low-birth-weight children: distinct effects of maturity and weight for gestational 126.
Pediatr.Manning J, Vehaskari VM. Low birth weight-associated adult hypertension
2010;22:158–160.
104.Kidney
age. SpencerInt.J,2005;68:731–740.
Wang Z, Hoy W. Low birth weight and reduced renal volume in in the Luyckx
162. rat. Pediatr VA,Nephrol.
Brenner 2001;16:417–422.
BM. The clinical importance of nephron mass. J Am
Aboriginal
133. children.A,
Siewert-Delle AmLjungman
J Kidney Dis.S. The2001;37:915–920.
impact of birth weight and gestational http://www.ncbi.nlm.nih.gov/pubmed/11405116
Soc Nephrol. 2010;21:898–910.
http://www.ncbi.nlm.nih.gov/pubmed/11325672
age on blood pressure in adult life: a population-based study of 49–year-old 127. Huxley
163. Ligi RR, Shiell AW,I,Law
I, Grandvuillemin AndresCM.V,Theet al.role
Lowo birth
size weight
at birthinfants
and postnatal
and the
105. Am
men. Simonetti GD, Raio
J Hypertens. L, Surbek D, et al. Salt sensitivity o children with low
1998;11:946–953. catch-up
developmentalgrowthprogramming
in determining systolic blooda pressure:
of hypertension: a systematic
focus on vascular review
factors. Semino
birth weight.
134. Wen X,Hypertension.
Triche EW, Hogan 2008;52:625–630.
JW, et al. Association between placental mor- the literature.
Perinatol. J Hypertens. 2000;18:815–831.
2010;34:188–192.
http://www.ncbi.nlm.nih.gov/pubmed/18695145
phology and childhood systolic blood pressure. Hypertension. 2011;57:48–55. http://www.ncbi.nlm.nih.gov/pubmed/10930178
164. Nuyt AM. Mechanisms underlying developmental programming of elevat-
106. Bergvall
135. Rakow A, N,Johansson
Iliadou A, S, LegnevallS, L,
Johansson et et
al.al. Renal and
Genetic volumeshared andenvironmen-
unction in 128.
ed bloodChen W Srinivasan
pressure and vascularSR, Berenson
dysfunction: GS.evidence
Ampli cation from humano the studies
association
and
school-age
tal factors do children born preterm
not confound or small between
the association or gestational age. Pediatr
birth weight Nephrol.
and hyperten- between
experimental birthweight and blood
animal models. Clinpressure
Sci (Lond).with age: the Bogalusa Heart Study. J
2008;114:1–17.
2008;23:1309–1315.
sion: a study among Swedish twins. Circulation. 2007;115:2931–2938. Hypertens.
165. Franco 2010;28:2046–2052.
MC, Christofalo DM, Sawaya AL, et al. Effects of low birth weight
http://www.ncbi.nlm.nih.gov/pubmed/18491148
136. Levine RS, Hennekens CH, Jesse MJ. Blood pressure in prospective popu- http://www.ncbi.nlm.nih.gov/pubmed/20616754
in 8- to 13-year-old children: implications in endothelial function and uric acid
107. based
lation Keijzer-V een MG,
cohort Dulger and
of newborn A, Dekker FW, et. Br
infant twins al.Med
VeryJ.preterm birth is a risk
1994;308:298–302. 129.
levels. Vancheri
Hypertension. F, Alletto M, Burgio A, et al. [Inverse relationship between etal
2006;48:45–50.
actor Chen
137. or increased systolic SR,
W, Srinivasan blood pressureDM,
Hallman at a et
young adultrelationship
al. The age. Pediatr between
Nephrol. growth and arterial
166. Vehaskari VM.pressure
Programming in children and adults].theG nervous
of hypertension: Ital Cardiol.
kidney1995;25:
. Am J
2010;25:509–516.
birthweight and longitudinal changes of blood pressure is modulated by 833–841.
Physiol Renal Physiol. 2008;295:F27–28.
http://www.ncbi.nlm.nih.gov/pubmed/20012998
beta-adrenergic receptor genes: the Bogalusa Heart Study. J Biomed Biotechnol. http://www.ncbi.nlm.nih.gov/pubmed/7557032
167. Dodic M, Moritz K, Koukoulas I, et al. Programmed hypertension: kidney,
108. Kent AL, Jyoti R, Robertson C, et al. Are renal volumes measured by mag-
2010;2010:543514. 130.
brain orYangboth? S, Trends
Bergvall N, Cnattingius
Endocrinol S, et al. Gestational age di erences in
Metab. 2002;13:403–408.
netic resonance
138. Falkner B, imaging
Hulman and three-dimensional
S, Kushner H. Effect ofultrasound
birth weightinon theblood
termpressure
neonate health and development
168. Nehiri T, Duong Van among
Huyen young Swedish
JP, Viltard M,menet al.born at term.
Exposure toInt J Epidemiol.
maternal dia-
comparable?
and body sizePediatr
in early Nephrol. 2010;25:913–918.
adolescence. Hypertension. 2004;43:203–207. 2010;39:1240–1249.
betes induces salt-sensitive hypertension and impairs renal function in adult rat
109. Huxley
139. BrennerR, BM, NeilMackenzie
A, Collins HS.R.Nephron
Unravelling massthe as afetal
risk origins
actor or progression
hypothesis: is http://www.ncbi.nlm.nih.gov/pubmed/20483833
offspring. Diabetes. 2008;57:2167–2175.
o renal
there disease.
really Kidney association
an inverse Int. Suppl 1997;63:S124–127.
between birthweight and subsequent blood 131. Keijzer-Veen
169. Sanders MW, MG, FazziSchrevel
GE, Janssen M, Finken
GM, et MJ, et al.sodium
al. High Microalbuminuria
intake increasesand
110. TanLancet.
pressure? JC, Workeneh B, Busque S, et al. Glomerular unction, structure, and
2002;360:659–665. low er glomerular
blood pressure andltration rate atfunction
alters renal young adult age in subjects
in intrauterine born very rats.
growth-retarded pre-
number
140. in renal
Muhle allogra
A, Muhle C, ts rom older
Amann K, et al.deceased
No juvenile donors. J Am
arterial Soc Nephrol.
hypertension in 2009;
sheep mature and a2005;46:71–75.
Hypertension. ter intrauterine growth retardation. J Am Soc Nephrol. 2005;16:
20:181–188.
multiples despite reduced nephron numbers. Pediatr Nephrol. 2010;25:1653–1661. 2762–2768.
170. Zimanyi MA, Bertram JF, Black MJ. Does a nephron de cit in rats predispose
http://www.ncbi.nlm.nih.gov/pubmed/18815243
141. Rostand SG, Cliver SP, Goldenberg RL. Racial disparities in the association http://www.ncbi.nlm.nih.gov/pubmed/15987756
to salt-sensitive hypertension? Kidney Blood Press Res. 2004;27:239–247.
111.
of foetalMcNamara BJ, Diou toB,childhood
growth retardation Hughsonblood MD, pressure.
et al. Associations
Nephrol Dial between age,
Transplant. 132. SchmidtJS.IM,
171. Gilbert Chellakooty
Sex, M, Boisen KA,
salt, and senescence: sortinget al.
outImpaired
mechanisms kidney growth
of the devel-in
body size and nephron number with individual glomerular volumes in urban
2005;20:1592–1597. low-birth-weight
opmental origins children: distinctHypertension.
of hypertension. e ects o maturity and weight or gestational
2008;51:997–999.
West A
142. rican males.
McCormick Nephrol
Covelli M.Dial
TheTransplant.
relationship2009;24:1500–1506.
of low birth weight to blood pres- age.
172.Kidney
Salazar Int.F,2005;68:731–740.
Reverte V, Saez F, et al. Age- and sodium-sensitive hyperten-
http://www.ncbi.nlm.nih.gov/pubmed/19028752
sure, cortisol levels, and reactivity in African American adolescents: a pilot study. http://www.ncbi.nlm.nih.gov/pubmed/16014050
sion and sex-dependent renal changes in rats with a reduced nephron number.
112. Compr
Issues Hoy WE, Hughson
Pediatr Nurs. MD, Zimanyi M, et al. Distribution o volumes o indi-
2006;29:173–187. 133. Siewert-Delle
Hypertension. A, Ljungman S. The impact o birth weight and gestational
2008;51:1184–1189.
vidualForrester
143. glomeruliT.inHistoric
kidneysand at autopsy:
early lifeassociation
origins ofwith age, nephron
hypertension number,.
in Africans age
173.onMagalhaes
blood pressure JC, da in Silveira
adult li e: AB,a population-based
Mota DL, et al. Renal studyfunction
o 49-year-old men.
in juvenile
Jbirth
Nutr.weight and body mass index. Clin Nephrol. 2010;74:105–112.
2004;134:211–216. Am
rats Jsubjected
Hypertens.to1998;11:946–953.
prenatal malnutrition and chronic salt overload. Exp Physiol.
113. Hemachandra
144. Zimanyi MA, AH, Hoy Klebanoff
WE, Douglas-Denton
MA, Furth SL.RN, et al.
Racial Nephroninnumber
disparities and
the associa- 134. Wen X, Triche EW, Hogan JW, et al. Association between placental
2006;91:611–619.
individual
tion between glomerular
birth weight volumes in male
in the term infantCaucasian
and bloodand A rican
pressure at American
age 7 years:sub-
re- mor
174.phology
Stewart and childhood
T, Ascani J, Craver systolic
RD, etblood
al. Rolepressure. Hypertension.
of postnatal 2011;57:
dietary sodium in
jects.from
sults Nephrol Dial Transplant.
the collaborative 2009;24:2428–2433.
perinatal project. J Am Soc Nephrol. 2006;17:2576–2581. 48–55.
prenatally programmed hypertension . Pediatr Nephrol. 2009;24:1727–1733.
http://www.ncbi.nlm.nih.gov/pubmed/19297355 http://www.ncbi.nlm.nih.gov/pubmed/21079045
72
72 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

135. Ruta
175. BergvallLA, N, Iliadou A,H,Johansson
Dickinson Thomas MC, S, et et
al. al.
Genetic
High-saltand shared environmen-
diet reveals the hy- 201. Singh RR, MF
156. Schreuder Denton
, Wilhelm KM, AJ,
Bertram
BokenkampJF, et al.A,Development
et al. Impact of o gestational
cardiovascuage lar
tal actors do
pertensive andnot coneffects
renal ound of thereduced
association between
nephron birth weight
endowment . Am Jand hyperten-
Physiol Renal disease
and birthdue to renal
weight insu f ciency
on amikacin in male
clearance on sheep
day 1 ofollowing
life. Clin etal
J Amunilateral
Soc Nephrol.ne-
sion: a study
Physiol. among Swedish twins. Circulation. 2007;115:2931–2938.
2010;298:F1384–1392. phrectomy. J Hypertens. 2009;27:386–396.
2009;4:1774–1778.
http://www.ncbi.nlm.nih.gov/pubmed/17515462
176. de Boer MP, Ijzerman RG, de Jongh RT, et al. Birth weight relates to salt http://www.ncbi.nlm.nih.gov/pubmed/19155792
202. Bauer R, Walter B, Bauer K, et al. Intrauterine growth restriction reduces
136. Levine
sensitivity RS, Hennekens
of blood pressure inCH, Jesse adults.
healthy MJ. Blood pressure in2008;51:928–932.
Hypertension. prospective popu- 157. Woods
nephron number LL, Weeks
and renal DA,excretory
Rasch R.function
Hypertension a ter neonatal
in newborn piglets. uninephrec-
Acta Physiol
lation Dagan
177. based cohort
A, Habib o newborn
S, Gattineni and J,in etantal.twins.
PrenatalBr Med J. 1994;308:298–302.
programming of rat thick tomy in2002;176:83–90.
Scand. rats precedes glomerular damage. Hypertension. 2001;38:337–342.
137. Chenlimb
ascending W, chloride
Srinivasan SR, Hallman
transport DM, et diet
by low-protein al. The relationship between
and dexamethasone. Am J http://www.ncbi.nlm.nih.gov/pubmed/11566901
203. Rodriguez-Soriano J, Aguirre M, Oliveros R, et al. Long-term renal follow-
birthweight
Physiol and longitudinal
Regul Integr Comp Physiol.changes o blood pressure is modulated by
2009;297:R93–99. 158.
up Nyengaardlow
of extremely JR.birth
Numberweight and dimensions
infants . Pediatr oNephrol.
rat glomerular capillaries in
2005;20:579–584.
beta-adrenergic
178. Manning J,receptor
Beutler genes:
K, Knepperthe Bogalusa
MA, et Heart Study. J Biomed
al. Upregulation Biotechnol.
of renal BSC1 normal
204. development
Franco MC, Nishida and a ter SK,nephrectomy.
Sesso R. GFR Kidney Int. 1993;43:1049–1057.
estimated from cystatin C ver-
2010;2010:543514.
and TSC in prenatally programmed hypertension . Am J Physiol Renal Physiol. 159.creatinine
sus Gibney EM, Parikh CR,
in children bornGargsmallAX. forAge, gender, age.
gestational race,AmandJ associations
Kidney Dis.
http://www.ncbi.nlm.nih.gov/pubmed/20467565
2002;283:F202–206. with kidney ailure ollowing living kidney donation. Transplant Proc. 2008;40:
2008;51:925–932.
138. Dagan
179. FalknerA,B,Kwon Hulman HM,S,Dwarakanath
Kushner H. EV,ect o birth
et al. Effectweight
of renal ondenervation
blood pressure on 1337–1340.
205. Gielen M, Pinto-Sietsma SJ, Zeegers MP, et al. Birth weight and creatinine
and bodyprogramming
prenatal size in early adolescence.
of hypertension Hypertension. 2004;43:203–207.
and renal tubular transporter abundance. 160. Abdi
clearance in R, Slakey
young D, Kittur
adult twins:D, in etuence
al. Heterogeneity o glomerular
of genetic, prenatal, size in nor-
and maternal fac-
http://www.ncbi.nlm.nih.gov/pubmed/14676220
Am J Physiol Renal Physiol. 2008;295:F29–34. mal donor
tors. J Am Soc kidneys:
Nephrol. Impact o race. Am J Kidney Dis. 1998;32:43–46.
2005;16:2471–2476.
139. Bertram
180. Huxley C, R, Trowern
Neil A, Collins
AR, Copin R. N,
Unravelling the etal diet
et al. The maternal origins
duringhypothesis:
pregnancy is http://www.ncbi.nlm.nih.gov/pubmed/10407369
206. Hallan S, Euser AM, Irgens LM, et al. Effect of intrauterine growth
there really
programs an inverse
altered association
expression of the between
glucocorticoidbirthweight
receptor andand subsequent
type 2 11beta-blood 161. BaumonM.kidney
restriction Overview o chronic
function at young kidney disease
adult in children.
age: the Curr Opin
Nord Trondelag Pedi-
Health
pressure? Lancet.dehydrogenase:
hydroxysteroid 2002;360:659–665. potential molecular mechanisms underlying the atr. 2010;22:158–160.
(HUNT 2) Study. Am J Kidney Dis. 2008;51:10–20.
http://www.ncbi.nlm.nih.gov/pubmed/12241871
programming of hypertension in utero. Endocrinology. 2001;142:2841–2853. http://www.ncbi.nlm.nih.gov/pubmed/20299869
207. White SL, Perkovic V, Cass A, et al. Is low birth weight an antecedent of
140. Alwasel
181. Muhle A, SH,Muhle
KaleemC,I, AmannSahajpalK,V, et et al. Maternal
No juvenile arterial
protein hyperten-
restriction re- 162. inLuyckx
CKD later life?VA,ABrenner
systematic BM.review
The clinical importancestudies.
of observational o nephron mass. JDis.
Am J Kidney Am
sion inangiotensin
duces sheep multiples
II AT(1)despite
and AT(2) reduced
receptor nephron
expression numbers.
in the Pediatr
fetal rat Nephrol.
kidney. Soc Nephrol. 2010;21:898–910.
2009;54:248–261.
2010;25:1653–1661.
Kidney Blood Press Res. 2010;33:251–259. http://www.ncbi.nlm.nih.gov/pubmed/20150537
208. Keijzer-Veen MG, Kleinveld HA, Lequin MH, et al. Renal function and size
http://www.ncbi.nlm.nih.gov/pubmed/20386927
182. Grigore D, Ojeda NB, Robertson EB, et al. Placental insuf ciency results in 163.
at young Ligiadult
I, Grandvuillemin
age after intrauterineI, Andres V, et restriction
growth al. Low birth andweight in ants and
very premature the
birth.
141. Rostand
temporal SG, Cliver
alterations in theSP , Goldenberg
renin angiotensin RL.system
Racial in disparities in the association
male hypertensive growth developmental
Am J Kidney Dis.programming
2007;50:542–551. o hypertension: a ocus on vascular actors. Semin
o oetal growth
restricted retardation
offspring. to childhood
Am J Physiol Regul Integr bloodComp pressure.
Physiol.Nephrol Dial Transplant.
2007;293:R804–811. Perinatol.
209. 2010;34:188–192.
Salgado CM, Jardim PC, Teles FB, et al. In uence of low birth weight
2005;20:1592–1597.
183. Manning J, Vehaskari VM. Postnatal modulation of prenatally programmed http://www.ncbi.nlm.nih.gov/pubmed/20494734
on microalbuminuria and blood pressure of school children. Clin Nephrol.
http://www.ncbi.nlm.nih.gov/pubmed/15840672
hypertension by dietary Na and ACE inhibition. Am J Physiol Regul Integr Comp 164. Nuyt AM. Mechanisms underlying developmental programming o elevat-
2009;71:367–374.
142. McCormick
Physiol. 2005;288:R80–84.Covelli M. The relationship o low birth weight to blood pres- ed blood
210. Hoypressure
WE, Wang andZ,vascular
VanBuynder dys unction:
P, et al. Theevidence
naturalrom human
history studies
of renal and
disease
sure, cortisol
184. Cornocklevels, and reactivitySC,
R, Langley-Evans in A rican American
Mobasheri A, et al.adolescents:
The impactaof pilot study.
maternal experimental
in Australian animalAborigines.models. PartClin Sci (Lond). 2008;114:1–17.
2. Albuminuria predicts natural death and renal
Issues Compr
protein Pediatr
restriction Nurs.rat
during 2006;29:173–187.
pregnancy upon renal expression of angiotensin re- 165. .Franco
failure KidneyMC, Christo alo DM, Sawaya AL, et al. E ects o low birth weight
Int. 2001;60:249–256.
http://www.ncbi.nlm.nih.gov/pubmed/16923680
ceptors and vasopressin-related aquaporins. Reprod Biol Endocrinol. 2010;8:105. in 8- to
211. 13-year-old
Yudkin JS, Martynchildren:
CN, implications
Phillips DI, etinal. endothelial
Associations unction and uric acid
of micro-albumin-
143. Gwathmey
185. Forrester T.TM, Historic
ShaltoutandHA, earlyRoseli e JC,
origins
et al.oGlucocorticoid-induced
hypertension in A ricans. fetalJ levels.
uria Hypertension.
with intra-uterine 2006;48:45–50.
growth retardation. Nephron. 2001;89:309–314.
Nutr. 2004;134:211–216.
programming alters the functional complement of angiotensin receptor subtypes http://www.ncbi.nlm.nih.gov/pubmed/17913201
212. Hoy WE, Mathews JD, McCredie DA, et al. The multidimensional nature
http://www.ncbi.nlm.nih.gov/pubmed/14704321
within the kidney. Hypertension. 2011;57:620–626. 166.
of renalVehaskari
disease: rates VM. and Programming
associationso ofhypertension:
albuminuria inthe an nervous
Australiankidney. Am J
Aboriginal
144. Barker
186. Hemachandra
DJ, Osmond AH, C, Klebano
Forsen TJ, MA, Furth
et al. SL. Racial
Trajectories disparities
of growth amonginchil- the Physiol RenalKidney
community. Physiol.Int.2008;295:F27–28.
1998;54:1296–1304.
associa
dren tionhave
who between
coronary birth weight
events in theNterm
as adults. Engl Jin ant 2005;353:1802–1809.
Med. and blood pressure at 167. Hoy
213. Dodic WE, M, Wang
MoritzZ,K,VanBuynder
Koukoulas P I,, et al. The
Programmed hypertension:
natural history kidney,
of renal disease
age 7 Boubred
187. years: results
F, DanielromL,the collaborative
Buffat perinatal
C, et al. Early project.
postnatal J Am Soc Nephrol.
overfeeding induces brain
in or both?Aborigines.
Australian Trends Endocrinol Part 1.Metab.
Changes 2002;13:403–408.
in albuminuria and glomerular ltra-
2006;17:2576–2581.
early chronic renal dysfunction in adult male rats. Am J Physiol Renal Physiol. http://www.ncbi.nlm.nih.gov/pubmed/12367823
tion rate over time. Kidney Int. 2001;60:243–248.
http://www.ncbi.nlm.nih.gov/pubmed/16870709
2009;297:F943–951. 168. Fagerudd
214. Nehiri T, Duong J, ForsblomVan Huyen JP, Viltard M, et al. K,
C, Pettersson-Fernholm Exposure to maternal
et al. Low birth weightdia-
145. Fainberg
188. Levitt NS, HP,Steyn
BudgeK,H,De Wet T, ME.
Symonds et al.The
Ancon inverse
ictingrelation
effects of between
maternal blood
nu- betes not
does induces salt-sensitive
increase the risk of hypertension
nephropathy andin impairs
Finnishrenaltype 1unction
diabeticin patients.
adult rat
pressure
trient and birth
restriction andweight among
early-life 5 year
obesity old children
on renal health. Proc romNutr
Soweto, South A rica.
Soc. 2011:1–8. o spring.
Nephrol DialDiabetes.
Transplant.2008;57:2167–2175.
2006;21:2159–2165.
J Epidemiol
189. Singhal Community
A, Cole TJ, Health. 1999;53:264–268.
Fewtrell M, et al. Is slower early growth bene cial for http://www.ncbi.nlm.nih.gov/pubmed/18443204
215. Rossing P, Tarnow L, Nielsen FS, et al. Low birth weight. A risk factor for
http://www.ncbi.nlm.nih.gov/pubmed/10396531
long-term cardiovascular health? Circulation. 2004;109:1108–1113. 169. SandersofMW,
development Fazzinephropathy?
diabetic GE, Janssen Diabetes.
GM, et al.1995;44:1405–1407.
High sodium intake increases
146. Ben-Shlomo
190. Gilbert T, Lelievre-Pegorier
Y, McCarthy A, Hughes M, Merlet-Benichou
R, et al. Immediate C. Long-term
postnatal growth e ects blood Rossing
216. pressureP,and altersL,renal
Tarnow Nielsenunction
FS, etinal.intrauterine
Short stature growth-retarded rats. Hy-
and diabetic nephropa-
o associated
is mild oligonephronia
with bloodinduced pressure in in
utero
youngby gentamicin
adulthood: inthetheBarry rat. Pediatr
CaerphillyRes. pertension.
thy. Br Med2005;46:71–75.
J. 1995;310:296–297.
1991;30:450–456.
Growth Study. Hypertension. 2008;52:638–644. http://www.ncbi.nlm.nih.gov/pubmed/15956110
217. Vikse BE, Irgens LM, Leivestad T, et al. Low birth weight increases risk for
http://www.ncbi.nlm.nih.gov/pubmed/1754301
191. Hemachandra AH, Howards PP, Furth SL, et al. Birth weight, postnatal 170. Zimanyi
end-stage renal MA,diseaseBertram
. J Am JF , Black
Soc Nephrol.MJ. 2008;19:151–157.
Does a nephron def cit in rats predis-
147. Langley-Evans
growth, and risk for high SC. blood
Intrauterine
pressure programming
at 7 years ofoage: hypertension
results frominthe theCol-
rat: pose to
218. salt-sensitive
Lackland hypertension?
DT, Bendall HE, OsmondKidney Blood
C, et Press Res.birth
al. Low 2004;27:239–247.
weights contrib-
nutrient interactions.
laborative Comp Pediatrics.
Perinatal Project. Biochem Physiol A Physiol. 1996;114:327–333.
2007;119:e1264–1270. http://www.ncbi.nlm.nih.gov/pubmed/15273426
ute to high rates of early-onset chronic renal failure in the Southeastern United
http://www.ncbi.nlm.nih.gov/pubmed/8759281
192. Law CM, Shiell AW, Newsome CA, et al. Fetal, infant, and childhood 171. Gilbert
States. Arch InternJS. Sex,
Med.salt, and senescence: sorting out mechanisms o the devel-
2000;160:1472–1476.
148. Ortiz
growth LA, Quan
and adult bloodA,pressure:
Weinberga A, et al. E ectstudy
longitudinal o prenatal
from birthdexamethasone
to 22 years on of opmental
219. Duncan originsRC,o Basshypertension.
PS, GarrettHypertension.
PJ, et al. Weight 2008;51:997–999.
at birth and other factors
rat renal
age. development.
Circulation. Kidney Int. 2001;59:1663–1669.
2002;105:1088–1092. http://www.ncbi.nlm.nih.gov/pubmed/18259036
in uencing progression of idiopathic membranous nephropathy. Nephrol Dial
http://www.ncbi.nlm.nih.gov/pubmed/11318936
193. Luyckx VA, Compston CA, Simmen T, et al. Accelerated senescence in kid- 172. Salazar
Transplant. F, Reverte V, Saez F, et al. Age- and sodium-sensitive hyperten sion
1994;9(7):875.
149. ofWoods
neys LL, Ingelf nger
low-birth-weight ratsJR, Nyengaard
after catch-upJR, et al..Maternal
growth Am J Physiol proteinRenalrestriction
Physiol. and sex-dependent
220. Na YW, Yang HJ, renalChoichanges
JH, etinal.rats with
Effect ofaintrauterine
reduced nephrongrowthnumber.
retardationHyper-
on
suppresses the newborn renin-angiotensin system and programs adult hyperten-
2009;297:F1697–1705. tension.
the 2008;51:1184–1189.
progression of nephrotic syndrome. Am J Nephrol. 2002;22:463–467.
sion inOzanne
194. rats. Pediatr Res. 2001;49:460–467.
SE, Hales CN. Lifespan: catch-up growth and obesity in male http://www.ncbi.nlm.nih.gov/pubmed/18259039
221. Teeninga N, Schreuder MF, Bokenkamp A, et al. In uence of low birth
http://www.ncbi.nlm.nih.gov/pubmed/11264427
mice. Nature. 2004;427:411–412. 173. Magalhaes
weight on minimal JC,change
da Silveira AB, Mota
nephrotic DL, etinal.children,
syndrome Renal unction
including in juvenile
a meta-
150. Tarry-Adkins
195. Alexander BT.JL,Placental insu f ciency
Martin-Gronert MS, Chenleads JH,to development
et al. Maternal o diet
hyperten-
in u- rats subjected
analysis. Nephrol to Dial
prenatal malnutrition
Transplant. and chronic salt overload. Exp Physiol.
2008;23:1615–1620.
sion inDNA
ences growth-restricted
damage, aortico telomere
spring. Hypertension.
length, oxidative 2003;41:457–462.
stress, and antioxidant de- 2006;91:611–619.
222. Zidar N, Cavic MA, Kenda RB, et al. Effect of intrauterine growth retarda-
http://www.ncbi.nlm.nih.gov/pubmed/12623943
fense capacity in rats. FASEB J. 2008;22:2037–2044. http://www.ncbi.nlm.nih.gov/pubmed/16513822
tion on the clinical course and prognosis of IgA glomerulonephritis in children.
151. Tarry-Adkins
196. Langley-EvansJL,SC, Jackson
Ozanne SE,AA. NordenRats A, with hypertension
et al. Lower antioxidantinducedcapac- by in 174. Stewart
Nephron. T, Ascani J, Craver RD, et al. Role o postnatal dietary sodium in
1998;79:28–32.
utero
ity andexposure
elevatedtop53 maternal
and p21 low-protein
may be adiets ail to increase
link between genderblood pressure
disparity in re-in prenatally
223. Jonesprogrammed
SE, Bilous RW, hypertension.
Flyvbjerg A, Pediatr
et al.Nephrol. 2009;24:1727–1733.
Intra-uterine environment in u-
response
nal telomere to ashortening,
high salt intake. Ann Nutrand
albuminuria, Metab. 1996;40:1–9.
longevity . Am J Physiol Renal Physiol. 175. glomerular
ences Ruta LA, Dickinsonnumber and H, Thomas
the acuteMC, renaletadaptation
al. High-salt diet reveals the
to experimental hy-
diabe-
152. Boubred F, Bu at C, Feuerstein JM, et al. E ects o early postnatal hyper-
2006;290:F509–516. pertensive
tes. and renal
Diabetologia. e ects o reduced nephron endowment. Am J Physiol Renal
2001;44:721–728.
nutrition
197. Akkadon nephron
A, Hastings number R, and
Konje long-term
JC, et renal unction length
al. Telomere and structure in rats.
in small-for- Physiol.
224. 2010;298:F1384–1392.
Plank C, Nusken KD, Menendez-Castro C, et al. Intrauterine growth re-
Am J Physiol Renal
gestational-age Physiol.
babies. BJOG.2007;293:F1944–1949.
2006;113:318–323. 176. de following
striction Boer MP,ligation Ijzerman RG,uterine
of the de Jongh RT, leads
arteries et al.toBirth
moreweight
severe relates
glomeru- to
153. Raqib
198. Ibrahim HN, Foley
R, Alam R, TanP, L,
DS, Sarker et Low
et al. al. Long-term
birth weight consequences
is associatedo with kidney
al- salt sensitivity
losclerosis o blood pressure inglomerulonephritis
after mesangioproliferative healthy adults. Hypertension.
in the offspring2008;51:
. Am J
donation.
tered immune N Engl J Med. in
function 2009;360:459–469.
rural Bangladeshi children: a birth cohort study. Am J 928–932.2010;32:287–295.
Nephrol.
http://www.ncbi.nlm.nih.gov/pubmed/19179315
Clin Nutr. 2007;85:845–852. http://www.ncbi.nlm.nih.gov/pubmed/18287343
225. Hakim RM, Goldszer RC, Brenner BM. Hypertension and protein-
154. Mohn
199. Kett MM, Bertram JF. VNephron
A, Chiavaroli , Cerrutoendowment and bloodoxidative
M, et al. Increased pressure:stresswhat do in 177. long-term
uria: Dagan A, sequelaeHabib S, of Gattineni J, et al. Prenatal
uninephrectomy in humans programming
. Kidney Int.o 1984;25:
rat thick
we really know?
prepubertal Curr born
children Hypertens
smallRep. for2004;6:133–139.
gestational age. J Clin Endocrinol Metab. ascending limb chloride transport by low-protein diet and dexamethasone. Am J
930–936.
http://www.ncbi.nlm.nih.gov/pubmed/15010018
2007;92:1372–1378. PhysiolRogers
226. Regul Integr
NM, LawtonComp Physiol.
PD, Jose 2009;297:R93–99.
MD. Indigenous Australians and living kid-
155. Franco
200. Moritz KM,MC, Wintour
Kawamoto EM, EM,Dodic M. Fetal
Gorjao R, et uninephrectomy
al. Biomarkers ofleads to postnatal
oxidative stress 178.donation.
ney ManningN J,Engl Beutler
J Med.K,2009;361:1513–1516.
Knepper MA, et al. Upregulation o renal BSC1
hypertension
and antioxidant andstatus
compromised
in childrenrenalbornunction.
smallHypertension.
for gestational 2002;39:1071–1076.
age: evidence of and TSC
227. in prenatally
Storsley LJ, Youngprogrammed
A, Rush DN, et hypertension.
al. Long-termAm J Physiol
medical Renal Physiol.
outcomes among
http://www.ncbi.nlm.nih.gov/pubmed/12052844
lipid peroxidation . Pediatr Res. 2007;62:204–208. 2002;283:F202–206.
Aboriginal living kidney donors. Transplantation. 2010;90:401–406.
CHAPTER 2 CLINICAL IMPORTANCE OF NEPHRON MASS 773
3

179. Lentine
228. Dagan A, KL, Kwon HM, Dwarakanath
Schnitzler MA, Xiao H, V et, et
al.al. E ectvariation
Racial o renal in denervation
medical out-on 200. Franco MC,
239. Nakatani KawamotoK,EM,
T, Sugimura Gorjao R, H,
Kawashima et al.
et Biomarkers
al. The in uenceo oxidative stress
of recipient
prenatal
comes programming
among living kidneyo hypertension
donors. N England Jrenal
Med.tubular transporter abundance.
2010;363:724–732. and
bodyantioxidant
mass on the status in children
outcome born kidney
of cadaver small or gestationalClin
transplants. age:Exp
evidence o
Nephrol.
Am J Physiol
229. Brenner Renal
BM,Physiol.
Milford2008;295:F29–34.
EL. Nephron underdosing: A programmed cause of lipid peroxidation. Pediatr Res. 2007;62:204–208.
2002;6:158–162.
180. Bertram
chronic C, Trowern
renal allograft failure.AR,
Am JCopin
KidneyN, Dis.et1993;21:66–72.
al. The maternal diet during http://www.ncbi.nlm.nih.gov/pubmed/17597662
240. Doshi MD, Garg N, Reese PP, et al. Recipient risk factors associated with de-
pregnancy
230. Szaboprograms
AJ, Mulleraltered
V, Chenexpression o the glucocorticoid
GF, et al. Nephron number determines receptor and
suscepti- 201. Schreuder
layed graft MF,aWilhelm
function: AJ, Bokenkamp
paired kidney A, et al. Impact
analysis. Transplantation. o gestational age
2011;91:666–671.
type to
bility 2 renal
11beta-hydroxysteroid
mass reduction-induced dehydrogenase:
CKD in Lewis potential
and Fisher molecular
344 rats:mecha-
impli- and
241.birth weightML,
Nicholson on amikacin
Windmillclearance on dayT,1eto al.li In
DC, Horsburgh e. Clin
uenceJ Am Soc Nephrol.
of allograft size
nisms underlying
cations for development the programming
of experimentally o hypertension
induced chronic in utero. Endocrinology.
allograft nephropa- 2009;4:1774–1778.
to recipient body-weight ratio on the long-term outcome of renal transplanta-
2001;142:2841–2853.
thy. Nephrol Dial Transplant. 2008;23:2492–2495. http://www.ncbi.nlm.nih.gov/pubmed/19713296
tion. Br J Surg. 2000;87:314–319.
181. Douverny
231. Alwasel SH, JB, Kaleem I, Sahajpal
Baptista-Silva V, et al.
JC, Pestana JO,Maternal protein restriction
et al. Importance of renal massre- 202. Bauer R, Walter
242. Johansson B, Bauer
S, Iliadou K, et al. N,
A, Bergvall Intrauterine
et al. Thegrowth restriction
association reduces
between low
duces
on angiotensin
graft II AT(1) and
function outcome afterAT(2) receptor
12 months of expression
living donorin kidney
the etaltransplanta-
rat kidney. nephron
birth weightnumber and renal
and type excretory
2 diabetes: unction in
contribution of newborn piglets.Epidemiology.
genetic factors. Acta Physiol
Kidney
tion. Blood Press
Nephrol Res. 2010;33:251–259.
Dial Transplant. 2007;22:3646–3651. Scand. 2002;176:83–90.
2008;19:659–665.
http://www.ncbi.nlm.nih.gov/pubmed/20606474
232. el-Agroudy AE, Hassan NA, Bakr MA, et al. Effect of donor/recipient body http://www.ncbi.nlm.nih.gov/pubmed/12354166
243. Tian JY, Cheng Q, Song XM, et al. Birth weight and risk of type 2 diabetes,
182. Grigore
weight mismatch D,onOjeda
patientNB, andRobertson EB, in
graft outcome et living-donor
al. Placentalkidneyinsu transplan-
ciency re- 203.
abdominalRodriguez-Soriano J, Aguirre M, among
obesity and hypertension OliverosChinese
R, et al.adults.
Long-termEur renal ollow-
J Endocrinol.
sults inAm
tation. temporal
J Nephrol. alterations in the renin angiotensin system in male hyper-
2003;23:294–299. up o extremely low birth weight in ants. Pediatr Nephrol. 2005;20:579–584.
2006;155:601–607.
tensiveGaston
233. growth RS,restricted
Hudson SL, o spring. Am et
Julian BA, J Physiol
al. Impact Regul of Integr Comp Physiol.
donor/recipient size http://www.ncbi.nlm.nih.gov/pubmed/15782301
244. Elmes MJ, McMullen S, Gardner DS, et al. Prenatal diet determines suscep-
2007;293:R804–811.
matching on outcomes in renal transplantation. Transplantation. 1996;61: 204.
tibilityFranco MC,ischaemia-reperfusion
to cardiac Nishida SK, Sesso R. GFRfollowing
injury estimatedtreatment
rom cystatin C ver-
with diethyl-
183. Manning J, Vehaskari VM. Postnatal modulation o prenatally programmed
383–388. sus
maleiccreatinine
acid andinN-acetylcysteine.
children born Life small
Sci. or gestational age. Am J Kidney Dis.
2008;82:149–155.
hypertension
234. Kasiske byBL,dietary
SnyderNa and ACE inhibition.
JJ, Gilbertson D. Inadequate Am Jdonor
Physiolsize
Regul Integr Comp
in cadaver kid- 2008;51:925–932.
245. Rothermund L, Lorenz M, Schnieber A, et al. Impact of nephron number
Physiol.
ney 2005;288:R80–84.
transplantation . J Am Soc Nephrol. 2002;13:2152–2159. http://www.ncbi.nlm.nih.gov/pubmed/18455848
dosing on cardiorenal damage and effects of ACE inhibition. Am J Hypertens.
184. Kim
235. CornockYS, KimR, Langley-Evans
MS, Han DS,SC, et Mobasheri
al. Evidence A, that
et al.the
Theratio
impact o maternal
of donor kid- 205. Gielen M, Pinto-Sietsma SJ, Zeegers MP, et al. Birth weight and creatinine
2011;24(4):474–481.
protein
ney restriction
weight duringbody
to recipient rat pregnancy upon renal
weight, donor age, andexpression
episodes o of
angiotensin re-
acute rejec- clearance in young adult
246. Langley-Evans SC. twins: inf uence
Nutritional o genetic, of
programming prenatal,
disease:and maternal the
unravelling ac-
ceptors
tion and vasopressin-related
correlate independently with aquaporins.
live-donor ReprodgraftBiolfunction.
Endocrinol. 2010;8:105.
Transplantation. tors. J Am SocJ Nephrol.
mechanism. 2005;16:2471–2476.
Anat. 2009;215:36–51.
http://www.ncbi.nlm.nih.gov/pubmed/20807409
2002;72:280–283. http://www.ncbi.nlm.nih.gov/pubmed/15944342
247. Amri K, Freund N, Van Huyen JP, et al. Altered nephrogenesis due to
185. Giral
236. Gwathmey TM, Shaltout
M, Nguyen JM, Karam HA,G,Rose JC,Impact
et al. et al. Glucocorticoid-induced
of graft mass on the clinical etal 206.
maternal Hallan S, Euser
diabetes AM, Irgens
is associated LM,increased
with et al. E ect o intrauterine
expression growth restric-
of IGF-II/mannose-6-
programming
outcome alterstransplants
of kidney the unctional . J Amcomplement
Soc Nephrol.o 2005;16:261–268.
angiotensin receptor subtypes tion on kidney
phosphate unction
receptor in theatfetal
young adultDiabetes.
kidney. age: the 2001;50:1069–1075.
Nord Trondelag Health (HUNT
withinGiral
237. the kidney. Hypertension.
M, Foucher Y, Karam 2011;57:620–626.
G, et al. Kidney and recipient weight in- 2)
248.Study. Am JT,Kidney
Gilbert CibertDis. 2008;51:10–20.
C, Moreau E, et al. Early defect in branching morphogen-
http://www.ncbi.nlm.nih.gov/pubmed/21220702
compatibility reduces long-term graft survival. J Am Soc Nephrol. 2010;21: http://www.ncbi.nlm.nih.gov/pubmed/18155528
esis of the ureteric bud in induced nephron de cit. Kidney Int. 1996;50:783–795.
186. Barker DJ, Osmond C, Forsen TJ, et al. Trajectories o growth among chil-
1022–1029. 207. White SL, Perkovic
249. Schreuder V, Cass A, et
M, Delemarre-van deal.Waal
Is lowH,birth
van weight
Wijk A. an antecedent
Consequenc- o
dren who
238. Kim have
YS, Mooncoronary events
JI, Kim DK,asetadults. N Engl
al. Ratio J Med.
of donor 2005;353:1802–1809.
kidney weight to recipient CKD
es of inintrauterine
later li e? Agrowth
systematic review oforobservational
restriction the kidney. studies.
Kidney Am J Kidney
Blood Dis.
Press Res.
http://www.ncbi.nlm.nih.gov/pubmed/16251536
bodyweight as an index of graft function . Lancet. 2001;357:1180–1181. 2009;54:248–261.
2006;29:108–125.
187. Boubred F, Daniel L, Bu at C, et al. Early postnatal over eeding induces http://www.ncbi.nlm.nih.gov/pubmed/19339091
early chronic renal dys unction in adult male rats. Am J Physiol Renal Physiol. 208. Keijzer-Veen MG, Kleinveld HA, Lequin MH, et al. Renal unction and size
2009;297:F943–951. at young adult age a ter intrauterine growth restriction and very premature birth.
188. Fainberg HP, Budge H, Symonds ME. The conf icting e ects o maternal Am J Kidney Dis. 2007;50:542–551.
nu trient restriction and early-li e obesity on renal health. Proc Nutr Soc. 2011: http://www.ncbi.nlm.nih.gov/pubmed/17900453
1–8. 209. Salgado CM, Jardim PC, Teles FB, et al. Inf uence o low birth weight
http://www.ncbi.nlm.nih.gov/pubmed/21232171 on microalbuminuria and blood pressure o school children. Clin Nephrol.
189. Singhal A, Cole TJ, Fewtrell M, et al. Is slower early growth bene cial or 2009;71:367–374.
long-term cardiovascular health? Circulation. 2004;109:1108–1113. http://www.ncbi.nlm.nih.gov/pubmed/19356368
http://www.ncbi.nlm.nih.gov/pubmed/14993136 210. Hoy WE, Wang Z, VanBuynder P, et al. The natural history o renal disease
190. Ben-Shlomo Y, McCarthy A, Hughes R, et al. Immediate postnatal growth in Australian Aborigines. Part 2. Albuminuria predicts natural death and renal
is associated with blood pressure in young adulthood: the Barry Caerphilly ailure. Kidney Int. 2001;60:249–256.
Growth Study. Hypertension. 2008;52:638–644. http://www.ncbi.nlm.nih.gov/pubmed/11422758
http://www.ncbi.nlm.nih.gov/pubmed/18768401 211. Yudkin JS, Martyn CN, Phillips DI, et al. Associations o micro-albumin-
191. Hemachandra AH, Howards PP, Furth SL, et al. Birth weight, postnatal uria with intra-uterine growth retardation. Nephron. 2001;89:309–314.
growth, and risk or high blood pressure at 7 years o age: results rom the Col- http://www.ncbi.nlm.nih.gov/pubmed/11598395
laborative Perinatal Project. Pediatrics. 2007;119:e1264–1270. 212. Hoy WE, Mathews JD, McCredie DA, et al. The multidimensional nature
192. Law CM, Shiell AW, Newsome CA, et al. Fetal, in ant, and childhood o renal disease: rates and associations o albuminuria in an Australian Aboriginal
growth and adult blood pressure: a longitudinal study rom birth to 22 years o community. Kidney Int. 1998;54:1296–1304.
age. Circulation. 2002;105:1088–1092. http://www.ncbi.nlm.nih.gov/pubmed/9767547
193. Luyckx VA, Compston CA, Simmen T, et al. Accelerated senescence in kid- 213. Hoy WE, Wang Z, VanBuynder P, et al. The natural history o renal disease
neys o low-birth-weight rats a ter catch-up growth. Am J Physiol Renal Physiol. in Australian Aborigines. Part 1. Changes in albuminuria and glomerular ltra-
2009;297:F1697–1705. tion rate over time. Kidney Int. 2001;60:243–248.
194. Ozanne SE, Hales CN. Li espan: catch-up growth and obesity in male http://www.ncbi.nlm.nih.gov/pubmed/11422757
mice. Nature. 2004;427:411–412. 214. Fagerudd J, Forsblom C, Pettersson-Fernholm K, et al. Low birth weight
http://www.ncbi.nlm.nih.gov/pubmed/14749819 does not increase the risk o nephropathy in Finnish type 1 diabetic patients.
195. Tarry-Adkins JL, Martin-Gronert MS, Chen JH, et al. Maternal diet inf u- Nephrol Dial Transplant. 2006;21:2159–2165.
ences DNA damage, aortic telomere length, oxidative stress, and antioxidant de- 215. Rossing P, Tarnow L, Nielsen FS, et al. Low birth weight. A risk actor or
ense capacity in rats. FASEBJ. 2008;22:2037–2044. development o diabetic nephropathy? Diabetes. 1995;44:1405–1407.
http://www.ncbi.nlm.nih.gov/pubmed/14749819 http://www.ncbi.nlm.nih.gov/pubmed/7589846
196. Tarry-Adkins JL, Ozanne SE, Norden A, et al. Lower antioxidant capac- 216. Rossing P, Tarnow L, Nielsen FS, et al. Short stature and diabetic ne-
ity and elevated p53 and p21 may be a link between gender disparity in re- phropa- thy. Br Med J. 1995;310:296–297.
nal telomere shortening, albuminuria, and longevity. Am J Physiol Renal Physiol. http://www.ncbi.nlm.nih.gov/pubmed/7866171
2006;290:F509–516. 217. Vikse BE, Irgens LM, Leivestad T, et al. Low birth weight increases risk or
197. Akkad A, Hastings R, Konje JC, et al. Telomere length in small- or- gesta- end-stage renal disease. J Am Soc Nephrol. 2008;19:151–157.
tional-age babies. BJOG. 2006;113:318–323. http://www.ncbi.nlm.nih.gov/pubmed/18057216
http://www.ncbi.nlm.nih.gov/pubmed/16487204 218. Lackland DT, Bendall HE, Osmond C, et al. Low birth weights contrib-
198. Raqib R, Alam DS, Sarker P, et al. Low birth weight is associated with al- ute to high rates o early-onset chronic renal ailure in the Southeastern United
tered immune unction in rural Bangladeshi children: a birth cohort study. Am J States. Arch Intern Med. 2000;160:1472–1476.
Clin Nutr. 2007;85:845–852. 219. Duncan RC, Bass PS, Garrett PJ, et al. Weight at birth and other actors
http://www.ncbi.nlm.nih.gov/pubmed/17344508 inf uencing progression o idiopathic membranous nephropathy. Nephrol Dial
199. Mohn A, Chiavaroli V, Cerruto M, et al. Increased oxidative stress in Transplant. 1994;9(7):875.
prepubertal children born small or gestational age. J Clin Endocrinol Metab. 220. Na YW, Yang HJ, Choi JH, et al. E ect o intrauterine growth retardation on
2007;92:1372–1378. the progression o nephrotic syndrome. Am J Nephrol. 2002;22:463–467.
http://www.ncbi.nlm.nih.gov/pubmed/17264184 http://www.ncbi.nlm.nih.gov/pubmed/12381944
(contd.)
73
74
72 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

221. Ruta
175. Teeninga N, Schreuder
LA, Dickinson MF, Bokenkamp
H, Thomas MC, et al.A,High-salt
et al. Infdiet
uence o low
reveals thebirth
hy- tion correlate
201. Schreuderindependently
MF, Wilhelm AJ, with live-donorA,gra
Bokenkamp t Impact
et al. unction.of Transplantation.
gestational age
weight on and
pertensive minimal
renalchange nephrotic
effects of reduced syndrome in children,. Am
nephron endowment including
J Physiola Renal
meta- 2002;72:280–283.
and birth weight on amikacin clearance on day 1 of life. Clin J Am Soc Nephrol.
analysis.2010;298:F1384–1392.
Physiol. Nephrol Dial Transplant. 2008;23:1615–1620. http://www.ncbi.nlm.nih.gov/pubmed/12151743
2009;4:1774–1778.
http://www.ncbi.nlm.nih.gov/pubmed/18065792
176. de Boer MP, Ijzerman RG, de Jongh RT, et al. Birth weight relates to salt 236. Bauer
202. Giral M, NguyenB,JM,
R, Walter Karam
Bauer K, etG,al.et Intrauterine
al. Impact ogrowth gra t mass on thereduces
restriction clinical
222. ZidarofN,
sensitivity Cavic
blood MA, Kenda
pressure RB, etadults.
in healthy al. E ect o intrauterine
Hypertension. growth retarda-
2008;51:928–932. outcome number
nephron o kidneyand transplants. J Am Soc
renal excretory Nephrol.in2005;16:261–268.
function newborn piglets. Acta Physiol
tion on
177. the clinical
Dagan A, Habib course and prognosis
S, Gattineni o IgA
J, et al. glomerulonephritis
Prenatal programming of in rat
children.
thick http://www.ncbi.nlm.nih.gov/pubmed/15563571
Scand. 2002;176:83–90.
Nephron. 1998;79:28–32.
ascending limb chloride transport by low-protein diet and dexamethasone. Am J 237. Rodriguez-Soriano
203. Giral M, Foucher J,Y,Aguirre Karam M, G,Oliveros
et al. Kidney
R, et al.and recipientrenal
Long-term weight in-
follow-
http://www.ncbi.nlm.nih.gov/pubmed/9609458
Physiol Regul Integr Comp Physiol. 2009;297:R93–99. compatibility
up of extremelyreduces
low birth long-term gra t .survival.
weight infants J Am Soc
Pediatr Nephrol. Nephrol. 2010;21:
2005;20:579–584.
223. Manning
178. Jones SE,J,Bilous RW,K,Flyvbjerg
Beutler Knepper A, et al.
MA, et Intra-uterine
al. Upregulation environment inf u-
of renal BSC1 1022–1029.
204. Franco MC, Nishida SK, Sesso R. GFR estimated from cystatin C ver-
encesTSC
and glomerular number
in prenatally and the acute
programmed renal adaptation
hypertension . Am JtoPhysiol
experimental diabe-
Renal Physiol. 238.creatinine
sus Kim YS, Moon JI, Kimborn
in children DK, etsmall
al. Ratio
for ogestational
donor kidney age. weight to recipient
Am J Kidney Dis.
tes. Diabetologia. 2001;44:721–728.
2002;283:F202–206. bodyweight as an index o gra t unction. Lancet. 2001;357:1180–1181.
2008;51:925–932.
224. Dagan
179. Plank C, NuskenHM,
A, Kwon KD,Dwarakanath
Menendez-Castro V, et al. C,Effect
et al. of
Intrauterine growth on
renal denervation re- 239. Gielen
205. Nakatani M, T, Sugimura K,SJ,Kawashima
Pinto-Sietsma Zeegers MP,H, et et
al. al.
BirthThe inf uence
weight o recipi-
and creatinine
striction programming
prenatal ollowing ligation o the uterine
of hypertension andarteries leads totransporter
renal tubular more severe glomeru-.
abundance ent body mass
clearance in youngon theadultoutcome
twins: ino cadaver
uence ofkidneygenetic, transplants.
prenatal, andClinmaternal
Exp Nephrol.
fac-
losclerosis
Am J Physiola Renal
ter mesangioproli erative glomerulonephritis in the o spring. Am J
Physiol. 2008;295:F29–34. 2002;6:158–162.
tors. J Am Soc Nephrol. 2005;16:2471–2476.
Nephrol.
180. 2010;32:287–295.
Bertram C, Trowern AR, Copin N, et al. The maternal diet during pregnancy 240. Hallan
206. Doshi MD, Garg N,
S, Euser AM,Reese PP, et
Irgens LM, al. et
Recipient
al. Effectriskofactors associated
intrauterine with
growth
http://www.ncbi.nlm.nih.gov/pubmed/20714134
programs altered expression of the glucocorticoid receptor and type 2 11beta- delayed gra
restriction ont kidney
unction: a paired
function kidney
at young analysis.
adult age: theTransplantation.
Nord Trondelag 2011;91:
Health
225. Hakim RM,
hydroxysteroid Goldszer RC,
dehydrogenase: Brenner
potential BM. Hypertension
molecular and protein-
mechanisms underlying the 666–671.
(HUNT 2) Study. Am J Kidney Dis. 2008;51:10–20.
uria: long-term
programming of sequelae
hypertension o uninephrectomy in humans.
in utero. Endocrinology. Kidney Int. 1984;25:
2001;142:2841–2853. 241. White
207. Nicholson ML, Windmill
SL, Perkovic DC,
V, Cass A,Horsburgh
et al. Is low T, birth
et al. Inf uence
weight anoantecedent
allogra t size
of
930–936.
181. Alwasel SH, Kaleem I, Sahajpal V, et al. Maternal protein restriction re- to recipient
CKD in laterbody-weight
life? A systematicratio onreview
the long-term outcomestudies.
of observational o renalAmtransplanta
J Kidneytion.
Dis.
http://www.ncbi.nlm.nih.gov/pubmed/6381857
duces angiotensin II AT(1) and AT(2) receptor expression in the fetal rat kidney. Br J Surg. 2000;87:314–319.
2009;54:248–261.
226. Rogers
Kidney NM,Res.
Blood Press Lawton PD, Jose MD. Indigenous Australians and living kid-
2010;33:251–259. http://www.ncbi.nlm.nih.gov/pubmed/10718800
208. Keijzer-Veen MG, Kleinveld HA, Lequin MH, et al. Renal function and size
ney donation.
182. Grigore D, N Engl
Ojeda J Med. 2009;361:1513–1516.
NB, Robertson EB, et al. Placental insuf ciency results in 242.
at youngJohansson
adult ageS,after Iliadou A, Bergvall
intrauterine growth N, restriction
et al. Theand association betweenbirth.
very premature low
http://www.ncbi.nlm.nih.gov/pubmed/19812415
temporal alterations in the renin angiotensin system in male hypertensive growth birthJ Kidney
Am weightDis. and2007;50:542–551.
type 2 diabetes: contribution o genetic actors. Epidemiology.
227. Storsley
restricted LJ, Young
offspring. Am J A, RushRegul
Physiol DN, Integr
et al. Long-term
Comp Physiol. medical outcomes among
2007;293:R804–811. 2008;19:659–665.
209. Salgado CM, Jardim PC, Teles FB, et al. In uence of low birth weight
Aboriginal
183. Manningliving kidney donors.
J, Vehaskari Transplantation.
VM. Postnatal modulation 2010;90:401–406.
of prenatally programmed http://www.ncbi.nlm.nih.gov/pubmed/18714437
on microalbuminuria and blood pressure of school children. Clin Nephrol.
http://www.ncbi.nlm.nih.gov/pubmed/20562735
hypertension by dietary Na and ACE inhibition. Am J Physiol Regul Integr Comp 243. Tian JY, Cheng Q, Song XM, et al. Birth weight and risk o type 2 diabe-
2009;71:367–374.
228. Lentine
Physiol. KL, Schnitzler MA, Xiao H, et al. Racial variation in medical out-
2005;288:R80–84. tes, abdominal
210. Hoy WE, obesity
Wang Z,and hypertension
VanBuynder P, etamong
al. TheChinese
natural adults.
historyEur J Endocrinol.
of renal disease
comesCornock
184. among living kidney donors.
R, Langley-Evans SC,NMobasheri
Engl J Med. A,2010;363:724–732.
et al. The impact of maternal 2006;155:601–607.
in Australian Aborigines. Part 2. Albuminuria predicts natural death and renal
http://www.ncbi.nlm.nih.gov/pubmed/20818874
protein restriction during rat pregnancy upon renal expression of angiotensin re- http://www.ncbi.nlm.nih.gov/pubmed/16990660
failure . Kidney Int. 2001;60:249–256.
229. Brenner
ceptors BM, Mil ord EL. aquaporins.
and vasopressin-related Nephron underdosing: A programmed
Reprod Biol Endocrinol. cause o
2010;8:105. 244. Y
211. Elmes
udkinMJ, JS, McMullen
Martyn CN, S, Gardner DS, et
Phillips DI, et al. Prenatal diet determines
Associations suscep-
of micro-albumin-
chronic
185. renal allogra
Gwathmey TM,t Shaltout
ailure. AmHA,J Kidney
Rose JC, Dis.et1993;21:66–72.
al. Glucocorticoid-induced fetal tibility
uria withto intra-uterine
cardiac ischaemia-reper usion injury
growth retardation. ollowing
Nephron. treatment with diethyl-
2001;89:309–314.
http://www.ncbi.nlm.nih.gov/pubmed/8494022
programming alters the functional complement of angiotensin receptor subtypes maleicHoy
212. acidWE,
and Mathews
N-acetylcysteine. Life Sci.DA,
JD, McCredie 2008;82:149–155.
et al. The multidimensional nature
230. Szabo
within AJ, Muller
the kidney. V, Chen GF
Hypertension. , et al. Nephron number determines suscepti-
2011;57:620–626. http://www.ncbi.nlm.nih.gov/pubmed/18062993
of renal disease: rates and associations of albuminuria in an Australian Aboriginal
bility to
186. renalDJ,
Barker mass reduction-induced
Osmond C, Forsen TJ,CKD et al.inTrajectories
Lewis and of Fisher
growth344among
rats: impli-
chil- 245. Rothermund
community. Kidney L, Int.Lorenz M, Schnieber A, et al. Impact o nephron number
1998;54:1296–1304.
cations
dren whoorhavedevelopment o experimentally
coronary events as adults. Ninduced
Engl J Med. chronic allogra t nephropa-
2005;353:1802–1809. dosingHoy
213. on WE,
cardiorenal
Wang Z,damage VanBuynderand eP, ects
et al.oThe ACE inhibition.
natural historyAm J Hypertens.
of renal disease
thy. Nephrol
187. Boubred Dial
F, Transplant. 2008;23:2492–2495.
Daniel L, Buffat C, et al. Early postnatal overfeeding induces 2011;24(4):474–481.
in Australian Aborigines. Part 1. Changes in albuminuria and glomerular ltra-
231. chronic
early Douverny JB, dysfunction
renal Baptista-SilvainJC, Pestana
adult maleJO, et. al.
rats AmImportance o renal
J Physiol Renal mass
Physiol. http://www.ncbi.nlm.nih.gov/pubmed/20864942
tion rate over time. Kidney Int. 2001;60:243–248.
on gra t unction outcome a ter 12 months o living donor kidney transplanta-
2009;297:F943–951. 246. Fagerudd
214. Langley-Evans SC. Nutritional
J, Forsblom programming oK, disease:
C, Pettersson-Fernholm et al. Lowunravelling
birth weight the
tion. Nephrol
188. FainbergDialHP,Transplant. 2007;22:3646–3651.
Budge H, Symonds ME. The con icting effects of maternal nu- mechanism.
does J Anat.the
not increase 2009;215:36–51.
risk of nephropathy in Finnish type 1 diabetic patients.
http://www.ncbi.nlm.nih.gov/pubmed/17704114
trient restriction and early-life obesity on renal health. Proc Nutr Soc. 2011:1–8. http://www.ncbi.nlm.nih.gov/pubmed/19175805
Nephrol Dial Transplant. 2006;21:2159–2165.
232. Singhal
189. el-Agroudy AE, TJ,
A, Cole Hassan NA, M,
Fewtrell Bakret MA,
al. Isetslower
al. E ect o growth
early donor/recipient
bene cialbodyfor 247. Rossing
215. Amri K,P,Freund
TarnowN, Van Huyen
L, Nielsen FS, etJP,al.etLowal. Altered nephrogenesis
birth weight. A risk factorduefor
to
weight mismatch
long-term on patient
cardiovascular and gra
health? t outcome
Circulation. in living-donor kidney transplan-
2004;109:1108–1113. maternal diabetes
development is associated
of diabetic with increased
nephropathy? expression o IGF-II/mannose-
Diabetes. 1995;44:1405–1407.
tation.Ben-Shlomo
190. Am J Nephrol. Y, 2003;23:294–299.
McCarthy A, Hughes R, et al. Immediate postnatal growth 6-phosphate
216. Rossingreceptor
P, Tarnow in L,the etal kidney.
Nielsen FS, et al. Diabetes. 2001;50:1069–1075.
Short stature and diabetic nephropa-
http://www.ncbi.nlm.nih.gov/pubmed/12902614
is associated with blood pressure in young adulthood: the Barry Caerphilly 248.BrGilbert
thy. T, Cibert C, Moreau E, et al. Early de ect in branching morphogen-
Med J. 1995;310:296–297.
233. Gaston
Growth Study. RS, Hudson SL,
Hypertension. Julian BA, et al. Impact o donor/recipient size
2008;52:638–644. esis o Vikse
217. the ureteric bud LM,
BE, Irgens in induced nephron
Leivestad T, et al.deLow cit. birth
Kidney Int. 1996;50:783–795.
weight increases risk for
matching
191. on outcomes
Hemachandra in renal PP
AH, Howards transplantation.
, Furth SL, et al.Transplantation. 1996;61:
Birth weight, postnatal http://www.ncbi.nlm.nih.gov/pubmed/8872952
end-stage renal disease. J Am Soc Nephrol. 2008;19:151–157.
383–388.
growth, and risk for high blood pressure at 7 years of age: results from the Col- 249. Lackland
218. SchreuderDT, M, Bendall
Delemarre-van
HE, Osmondde Waal C, H, vanLow
et al. Wijk A. Consequenc
birth weights contrib-es o
234. Kasiske
laborative BL, Snyder
Perinatal JJ,Pediatrics.
Project. Gilbertson2007;119:e1264–1270.
D. Inadequate donor size in cadaver kid- intrauterine
ute to high rates growth restriction
of early-onset or renal
chronic the kidney.
failure inKidney Blood Press
the Southeastern Res.
United
ney transplantation.
192. Law CM, Shiell J Am
AW,SocNewsome
Nephrol. 2002;13:2152–2159.
CA, et al. Fetal, infant, and childhood 2006;29:108–125
States. Arch Intern Med. 2000;160:1472–1476.
http://www.ncbi.nlm.nih.gov/pubmed/12138149
growth and adult blood pressure: a longitudinal study from birth to 22 years of http://www.ncbi.nlm.nih.gov/pubmed/16837795.
219. Duncan RC, Bass PS, Garrett PJ, et al. Weight at birth and other factors
235.Circulation.
age. Kim YS, Kim MS, Han DS, et al. Evidence that the ratio o donor kid-
2002;105:1088–1092. in uencing progression of idiopathic membranous nephropathy. Nephrol Dial
ney weight
193. LuyckxtoVA, recipient
Compston body CA,weight,
Simmen donor
T, et age, and episodes
al. Accelerated o acuteinrejec-
senescence kid- Transplant. 1994;9(7):875.
neys of low-birth-weight rats after catch-up growth . Am J Physiol Renal Physiol. 220. Na YW, Yang HJ, Choi JH, et al. Effect of intrauterine growth retardation on
2009;297:F1697–1705. the progression of nephrotic syndrome. Am J Nephrol. 2002;22:463–467.
194. Ozanne SE, Hales CN. Lifespan: catch-up growth and obesity in male 221. Teeninga N, Schreuder MF, Bokenkamp A, et al. In uence of low birth
mice. Nature. 2004;427:411–412. weight on minimal change nephrotic syndrome in children, including a meta-
195. Tarry-Adkins JL, Martin-Gronert MS, Chen JH, et al. Maternal diet in u- analysis. Nephrol Dial Transplant. 2008;23:1615–1620.
ences DNA damage, aortic telomere length, oxidative stress, and antioxidant de- 222. Zidar N, Cavic MA, Kenda RB, et al. Effect of intrauterine growth retarda-
fense capacity in rats. FASEB J. 2008;22:2037–2044. tion on the clinical course and prognosis of IgA glomerulonephritis in children.
196. Tarry-Adkins JL, Ozanne SE, Norden A, et al. Lower antioxidant capac- Nephron. 1998;79:28–32.
ity and elevated p53 and p21 may be a link between gender disparity in re- 223. Jones SE, Bilous RW, Flyvbjerg A, et al. Intra-uterine environment in u-
nal telomere shortening, albuminuria, and longevity. Am J Physiol Renal Physiol. ences glomerular number and the acute renal adaptation to experimental diabe-
2006;290:F509–516. tes. Diabetologia. 2001;44:721–728.
197. Akkad A, Hastings R, Konje JC, et al. Telomere length in small-for- 224. Plank C, Nusken KD, Menendez-Castro C, et al. Intrauterine growth re-
gestational-age babies. BJOG. 2006;113:318–323. striction following ligation of the uterine arteries leads to more severe glomeru-
198. Raqib R, Alam DS, Sarker P, et al. Low birth weight is associated with al- losclerosis after mesangioproliferative glomerulonephritis in the offspring. Am J
tered immune function in rural Bangladeshi children: a birth cohort study. Am J Nephrol. 2010;32:287–295.
Clin Nutr. 2007;85:845–852. 225. Hakim RM, Goldszer RC, Brenner BM. Hypertension and protein-
199. Mohn A, Chiavaroli V, Cerruto M, et al. Increased oxidative stress in uria: long-term sequelae of uninephrectomy in humans. Kidney Int. 1984;25:
prepubertal children born small for gestational age. J Clin Endocrinol Metab. 930–936.
2007;92:1372–1378. 226. Rogers NM, Lawton PD, Jose MD. Indigenous Australians and living kid-
200. Franco MC, Kawamoto EM, Gorjao R, et al. Biomarkers of oxidative stress ney donation. N Engl J Med. 2009;361:1513–1516.
and antioxidant status in children born small for gestational age: evidence of 227. Storsley LJ, Young A, Rush DN, et al. Long-term medical outcomes among
lipid peroxidation . Pediatr Res. 2007;62:204–208. Aboriginal living kidney donors. Transplantation. 2010;90:401–406.
C H A P T E R

3 Renal Circulation and Glomerular


Hemodynamics
William J. Arendshorst • L. Gabriel Navar

T
he capability of the kidneys to achieve their sophis- shown in Table 3.1, blood ow per gram of kidney weight is
ticated homeostatic function is optimized by an in- about 4 mL per minute, which is 5 to 50 times greater than
tricate microvascular system that adjusts vascular the ow through other organs and circulatory beds. Based
resistance to maintain an appropriate control of the intra- on a total of 1 million glomeruli in each kidney or a glomer-
capillary and interstitial forces that govern renal blood ow ular density of 7,000 glomeruli per gram, the average blood
in the cortex and medulla as well as the glomerular ltra- ow and glomerular ltration rate (GFR) per glomerulus is
tion rate. A combination of intrinsic and extrinsic regula- 570 nL per minute and 62 nL per minute, respectively. This
tory mechanisms are responsible for controlling the one- fth large ow, coupled with the maintenance of a high hydro-
of the cardiac output that circulates through the kidneys. static pressure in glomerular capillaries, allows the ltration
Essentially all the renal blood ow (RBF) traverses through of about 20% of the plasma, which amounts to an average
the glomerular capillaries, where about 20% of the plasma is GFR of 120 mL per minute, or 170 L per day.1,2
ltered. The complex glomerular ltration apparatus is truly The extraordinarily high RBF is in marked excess of
unique in having both a very high hydraulic conductivity that simply required to provide the renal parenchyma with
and a remarkably low permeability to plasma proteins. One adequate supplies of oxygen and nutrients. For this reason,
major function of the renal vasculature is to regulate the in- it is generally recognized that RBF is regulated primarily to
traglomerular forces so that an adequate, yet not excessive, maintain the glomerular and peritubular intrarenal hemody-
volume is ltered into the urinary tubules. In this chapter, namic environments at levels compatible with the optimum
we discuss the characteristics of the ltering and the reab- delivery of ltrate to the nephrons and appropriate reabsorp-
sorbing microcirculatory structures in the normal kidney. tion of uid back into the systemic vasculature.
Particular emphasis is placed on the dynamic interactions
among the intrarenal paracrine and extrarenal homeostatic The Relationship of Renal Blood Flow to
mechanisms that participate in regulating these processes. Oxygen Consumption
To allow a better appreciation of basic mechanisms, some Although oxygen (O2) use is not a major determinant of RBF,
structural relationships and fundamental concepts related to O2 consumption by the kidneys is still quite high because
vascular smooth muscle, endothelial cells, and other com- of the very high metabolic activity of the tubules. Over 99%
ponents of the renal microvascular network are discussed. of the ltered uid, electrolytes, and essential organic nutri-
A detailed discussion of the anatomic features of the kidney ents are normally reabsorbed by the tubules and returned
is provided in Chapter 1. to the circulation via the peritubular capillaries. The tubular
reabsorptive processes depend on the integrity of the epi-
THE MAGNITUDE OF RENAL thelial transport systems, in particular the energy requiring
Na-K-ATPase. Such tubular enzyme systems account for the
BLOOD FLOW AND GLOMERULAR majority of the O2 consumption by the kidneys.
FILTRATION RATE RBF is about 400 mL/min/100 g of tissue, and the arte-
The multiple intrarenal parallel arteriolar pathways provide riovenous O2 difference is relatively low, only 1 to 2 mL per
the kidneys with a very low vascular resistance. They nor- deciliter of blood. Thus, O2 consumption by the kidney is
mally receive about 20% of the cardiac output. This amounts about 8 mL of O2 per minute or 400 m of O2/min/100 g,
to a blood ow of 1,000 to 1,200 mL per minute in a 70- to which amounts to 6% to 8% of the whole body O2 con-
75-kg person. RBF is even more impressive when considered sumption. This level of O2 use is relatively constant and is
per unit of kidney weight, because the kidneys account for not reduced by moderate hypoxemia. Under physiologic
only 0.5% of the total body weight, or about 300 g. Thus, as conditions, there is a consistent relationship between RBF
74
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 775
5

TA B L E

3.1 Renal Hemodynamic Function in Humans

Per Kilogram Per Gram


Total Body Weight Kidney Weight

Renal blood ow 1,200 mL/min 17 mL/min 4 mL/min


Renal plasma ow 670 mL/min 9.6 mL/min 2.2 mL/min
Glomerular ltration rate 130 mL/min 1.9 mL/min 0.45 mL/min
Number of glomeruli 2 million 28,500 7,000
Oxygen consumption 1,200 MO2/min 17 MO2/min 4 MO2/min

and renal O2 consumption. However, this relationship is a O2 (pO2). A cortical–medullary gradient of oxygenation
consequence of the associated changes in GFR and ltered exists in the kidney. In the super cial cortex, pO2 in effer-
sodium load, re ecting a direct causal relationship be- ent arterioles is 40 to 45 mm Hg, with values of 40 mm
tween tubular sodium reabsorption and O2 consumption, Hg recorded in other cortical structures (proximal tubule,
as shown in Figure 3.1. The rate of actively transported so- distal tubule, super cial cortical tissue), and 30 mm Hg in
dium appears to be the primary determinant of the rate of O2 the deep cortex. The fact that pO2 in the renal vein exceeds
consumption. About 20% of the consumption, or approxi- that of any site in the cortex indicates precapillary shunt-
mately 100 m of O2/min/100 g of kidney, is used for basal ing of O2 from artery to vein. Renal medullary pO2 is about
metabolic purposes and continues even in the absence of 20 mm Hg, with cells functioning in a climate of constant
ltration. Above this basal rate, there is a linear relationship. relative hypoxia. Recent mathematical analysis indicates that
Approximately 27 to 35 mEq of sodium is reabsorbed per arteriovenous O2 shunting in the cortex is substantial. The
millimole of O2 consumed, depending on the contribution shunting contributes to the stabilization of tissue pO2 levels.
of passive transport via paracellular pathways, which may be Cortical ischemia may exacerbate medullary hypoxia even
about 40%. In contrast to other organs, the kidneys do not when medullary perfusion is maintained.5–8
have a hyperemic response to hypoxia, making them more In the renal medullary microcirculation, the net amount
susceptible to hypoxemia.3,4 of O2 reabsorbed from vasa recta into the interstitium is sig-
The balance of O2 consumption for sodium reabsorp- ni cantly lower than estimated medullary O2 requirements
tion and O2 delivery is re ected by the tissue pressure of based on active sodium reabsorption. Low inner medullary
pO2 results from the countercurrent arrangement of vasa
recta and high vascular permeability to O2, as well as high
metabolic needs. Diffusional shunting of O2 between des-
cending and ascending vasa recta explains why a 20-mm Hg
decrease in initial pO2 at the corticomedullary border only
leads to a small drop in pO2 at the papillary tip ( 2 mm Hg
with baseline parameter values). In contrast, small decreas-
es in medullary blood ow, hematocrit, and O2 consump-
tion by tubules markedly reduce interstitial pO2. Without
erythrocytes, papillary tip pO2 cannot be maintained above
10 mm Hg, even when O2 consumption is zero. An increase
in medullary blood ow during water diuresis improves
medullary O2 delivery.
The renal medulla normally functions in an hypoxic en-
vironment. Tissue hypoxia impacts on O2-regulated genes
and leads to the renal production of adrenomedullin and
FIGURE 3.1 The relationship between tubular sodium reab- erythropoietin. Hypoxia-inducible factor-1alpha (HIF-1 )
sorption and oxygen consumption by the kidney. The primary is a transcription factor that regulates the O2-dependent
determinant of oxygen consumption above basal levels is the expression of many genes. This transcription factor may
rate of active sodium transport. contribute to gene expression in renal medullary cells that
76
76 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

function normally under hypoxic conditions. In this regard,


the loop diuretic furosemide markedly increases renal med-
ullary pO2 levels (20 to 50 mm Hg) in association with the
inhibition of reabsorption along the ascending limb of Henle
loop and a reduction in HIF-1 .9,10
The ef ciency of coupling between tubular transport and
O2 consumption is modi ed by paracrine/autocrine factors.
Nitric oxide (NO) normally suppresses O2 consumption by
epithelial mitochondria. The inhibition of NO synthesis in-
creases O2 extraction and O2 consumption and reduces the
ef ciency of sodium transport. The regulation of renal O2
consumption by NO may become impaired during oxidative
stress when superoxide production is excessive. Oxidative
stress and decreased availability/activity of NO can lead to
reduced intrarenal pO2 due to enhanced O2 usage relative
to tubular sodium transport. A low sodium intake leads to
increased renal medullary oxygenation.11
Endothelial-dependent NO plays a role in the regulation
of renal O2 consumption in normal kidneys. Baseline corti-
cal O2 consumption is about 600 nmol per minute per gram
of tissue. Stimulation of NO production by bradykinin or
administration of a NO donor reduces O2 consumption 25%
to 30% in the renal cortex and 30% in the medulla. Super-
oxide scavenging of NO attenuates the stimulatory effect of FIGURE 3.2 Renal microcirculation showing branching of affer-
bradykinin or NO donors.3,5,8 ent arterioles from arcuate arteries, glomerular capillary tufts, ef-
ferent arterioles, peritubular capillaries, some initial portions of
the vasa recta, and the venous system. The vessels are lled with
CHARACTERISTICS OF THE dark elastic polymer (Micro ll), and two tubules are lled with
CONTRACTILE PROCESS light polymer, showing the Bowman capsule (single arrow) and
Structural–Functional Aspects the proximal tubules (double arrow) and parts of loop of Henle.
AA, afferent arteriole; ArA, arcuate artery; PC, peritubular capil-
There are important interactions among various cell types in
lary; V, venule; VR, vasa recta. (Courtesy of Dr. Daniel Casellas.)
the microvasculature that determine the caliber of the small
diameter resistance vessels. The complex vasculature of the
kidney (Fig. 3.2) allows the ne regulation of the intrarenal
hemodynamic environment. Smooth muscle cells surround and the larger arterioles leading to the super cial nephrons
all vascular structures from the main renal artery to the in- contribute more to this pressure drop. Overall, the pressure
dividual afferent and efferent arterioles. The preglomerular drop up to the glomerular capillary tuft is much lower than
vasculature also has extensive innervation and responds in other vascular beds. This allows for high hydrostatic pres-
to renal nerve stimulation and many different hormonal, sure in the glomerular capillaries, which is much greater
paracrine, and physical stimuli. Nevertheless, most of the than the plasma colloid osmotic pressure and is thus respon-
ne control of preglomerular resistance occurs in the small sible for the ultra ltration of uid into the Bowman space.1
diameter afferent arterioles. The afferent arterioles vary in The terminal portion of an afferent arteriole contains
length and in the angle at which they branch from the inter- modi ed granular epithelioid cells that form part of the
lobular arteries; those in juxtamedullary portions branch at juxtaglomerular apparatus. The granules contain renin and
a much sharper angle. In addition, smooth muscle cells of other components of the renin–angiotensin system; the ex-
the afferent arterioles are modi ed as the vessels approach tent of granulation varies inversely with sodium intake. There
the glomerulus. The proximal portions of afferent arterioles is a reciprocal relationship between the amount of renin and
possess typical elongated smooth muscle cells, whereas cells actin, and the granular cells may have reduced contractile
closer to the glomerulus are more rounded and many pos- capability. As is shown in Figure 3.4, the juxtaglomerular
sess renin-containing granules.1,2 granular cells are adjacent to the tubular macula densa seg-
As is shown in Figure 3.3, the magnitude of the hydro- ment at the end of the ascending loop of Henle, and they are
static pressure drop along the arterial tree is relatively small associated with the nongranular extraglomerular mesangial
up to the terminal segments of the afferent arterioles. About cells between the afferent and efferent arterioles. The appear-
70% of the preglomerular pressure drop occurs in the termi- ance of the macula densa cells with large nuclei along with
nal portion of the afferent arteriole. In rodents, the arteries their close apposition to the glomerular vessels provide the
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 777
7

FIGURE 3.3 A representative pressure


pro le along the renal microvasculature in
a normal kidney. The segments are depicted
at the bottom of the graph, and the range of
ideal pulse pressures is represented by the
stippled area.

FIGURE 3.4 A drawing of the glomerulus and the juxta-


glomerular complex consisting of the afferent arteriole
(AA) with the granular cells of the juxtaglomerular ap-
paratus (JGC), the extraglomerular mesangial cells (EMC),
the macula densa (MDC) segment of the ascending loop
of Henle, and the efferent arteriole (EA). Also shown are the
thick ascending limb (TAL), the proximal tubule (PT), the
Bowman space (BS), glomerular capillaries (GC), peritubular
capillaries (PC), mesangial cells (MC), and nerve bers (NF).
(Drawing by Dr. Daniel Casellas.)
78
78 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

morphologic basis for the in uence of alterations in ow or because glomerular pressure and blood ow change in op-
composition of the tubular uid to generate signals that are posite directions. The maintenance of appropriate efferent
transmitted to the afferent arteriole or juxtaglomerular cells arteriolar tone serves to keep glomerular capillary pressure
to control vascular tone and renin release.12–14 suf ciently high to provide an adequate hydrostatic pressure
As an afferent arteriole approaches a glomerulus, the for ltration. The efferent arterioles also are responsible for
muscle cells surrounding the vessel intermingle with extra- the marked decrease in pressure at the peritubular capillar-
glomerular and intraglomerular mesangial cells. As it enters ies, which allows the reabsorptive force of the plasma colloid
the glomerulus, the arteriole expands into a manifold lined osmotic pressure to predominate.1
by endothelial cells, which, in turn, gives rise to a series of There are important regional differences in the circula-
glomerular capillary loops. The loops subdivide further into tion within the kidney, which have considerable functional
a branching system of exchange channels. Finally, the chan- signi cance. The relative distribution of glomerular and post-
nels coalesce into a small number of terminal capillaries, glomerular blood ow is depicted in Figure 3.5. Glomeru-
which join to form the efferent arteriole. Greater structural lar blood ow is proportional to the size of the glomeruli.
detail regarding the glomerular capillaries subserving the l- The larger deep juxtamedullary nephrons have higher ows
tration process is provided in Chapter 1. than the super cial or midcortical glomeruli. These juxta-
Efferent arterioles originate deep within the glomeruli medullary glomeruli give rise to muscular efferent arterioles
and vary with regard to length, diameter, and density of that descend toward the medulla and branch into the vasa
smooth muscle cells. In the outer cortex, these vessels are recta bundles, which are intimately associated with the sur-
relatively short, have a smaller diameter, and have a less rounding concentric rings of loops of Henle and collecting
well-developed muscular wall than efferent arterioles in ducts. With regard to the postglomerular blood ow, about
deeper cortical regions. The smooth muscle cells of the 75% to 85% of the total RBF is distributed to the peritubular
super cial efferent arterioles resemble pericytes that often capillaries in the cortex, whereas 15% to 25% goes to the
extend onto the peritubular capillaries. In the midcortex, medullary region. Blood ow throughout the cortex is much
the efferent arterioles are usually longer and have a greater higher than in the medulla and is higher in the outer cortex
degree of smooth muscle development. In the deeper por- than in the inner cortex. Overall cortical blood ow averages
tion of the cortex, the efferent arterioles are more variable 4 to 6 mL/min/g of tissue. Medullary blood ow ranges from
in length. Some efferent arterioles of juxtamedullary neph- 2.0 to 3.5 mL/min/g in the outer medulla to much lower
rons give rise to typical cortical peritubular capillary sys- values of approximately 0.2 to 1.0 mL/min/g of tissue in the
tems, whereas others are much longer and descend toward inner medulla and papilla. Regional mean transit times of
the medulla. These two distinct capillary networks subserve intravascular indicators are 1 to 3 seconds for the cortex,
the reabsorptive functions of the cortex and medulla, re-
spectively, and may be subject to independent regulation.
At the corticomedullary border, the efferent arterioles break
up into vascular bundles that branch into numerous de-
scending vasa recta. Vasa recta branch to form a capillary
plexus at each level within the medulla. There are three
distinct capillary plexuses, with the densest found in the
inner stripe of the outer medulla. The ascending vasa recta
are morphologically distinct from the descending vasa recta
and ascend within vascular bundles to drain into the ar-
cuate veins. The ascending vasa recta are more numerous
and have a highly fenestrated, thin endothelium, whereas
the descending vasa recta have a continuous thick endothe-
lium. These anatomic differences suggest that the ascending
vasa recta have a much greater permeability to macromol-
ecules than the descending vasa recta.1,2,15,16
Each glomerular resistance segment contributes to
the regulation of glomerular blood ow and pressure in a
unique manner because the glomerular capillary is “nested” FIGURE 3.5 The distribution of glomerular and postglomerular
between the afferent and efferent arterioles. Although a ow and of cortical (C1, C2, C3) and medullary (M) blood ow. The
more quantitative analysis of their respective roles is pre- distribution of glomerular ow is expressed as a percentage of
sented in a following section, it should be appreciated that total blood ow; preglomerular ow is presented on the left,
alterations in preglomerular resistance produce changes in and postglomerular ow on the right. The deep cortical ow is
glomerular blood ow, pressure, and GFR, which are di- subdivided to account for medullary ow distribution. As noted
rectionally similar. In contrast, selective changes in efferent from the arrows, there is a general shift of the postglomerular
arteriolar resistance cause more complex GFR responses blood ow toward deeper areas.
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 779
9

4 to 6 seconds for the outer medulla, and 10 to 30 seconds the formation of cAMP, and attenuates activity of protein ki-
for the inner medulla.15,17–19. nase A (PKA), whereas binding to a 1- or 2-adrenoceptor
activates adenylate cyclase to increase cAMP/PKA signaling.
Norepinephrine also binds to 1-adrenoceptors and acti-
Receptor Activation and Intracellular vates a G q protein, which is coupled to PLC, leading to
Signaling the formation of inositol triphosphate (IP3) and the release
Contractile responses at various sites along the vascular net- of Ca2 from the sarcoplasmic reticulum. 1-Adrenoceptors
work have different functional characteristics, depending also stimulate nicotinamide adenine dinucleotide phosphate
on the expression of receptor populations and/or activation (NADPH) oxidase to increase superoxide oxide production
mechanisms. The actions of circulating hormones and neu- and adenosine diphosphate (ADP) ribosyl cyclase generation
ral stimuli combined with local paracrine factors from en- of cyclic ADP ribose that sensitizes ryanodine receptors to
dothelial and epithelial cells are expressed through different release Ca2 from sarcoplasmic reticular stores.20–24
effector mechanisms to provide a highly integrated regula- GPCRs and/or their immediate signaling partners
tion of the renal microcirculation and the interstitial envi- (G proteins) are concentrated in caveolae, ask-shaped plasma
ronment. Many vasoactive agents interact with membrane membrane invaginations (50 to 100 nm in diameter) that are
receptors on the vascular smooth muscle and the endothelial subcellular microdomains of lipid rafts and caveolae, enriched
and mesangial cells. in glycosphingolipids and cholesterol and the protein caveolin.
Plasma membrane surface receptors can be subdivided Caveolae are complexes with a high concentration of signaling
into three main groups in which the receptor is coupled to a molecules. Caveolin is a scaffolding protein that anchors recep-
guanine nucleotide binding protein (G protein), regulates tors (e.g., angiotensin II subtype 1 [AT1], endothelin [ET-1],
enzyme activity, or serves as part of an ion channel. Exam- epidermal growth factor [EGF]) signaling and traf cking pro-
ples of the latter two groups are the atrial natriuretic pep- teins (G q/11) as well as ion channels (K and transient recep-
tide (ANP) receptor, guanylate cyclase, in vascular smooth tor potential [TRP] channels) and enzymes (endothelial nitric
muscle, and the nicotinic-acetylcholine receptor that di- oxide synthase [eNOS], protein kinase C [PKC], phospholi-
rectly activates a cation channel at the neuromuscular junc- pase C [PLC], NADPH oxidase, ADP ribosyl cyclase) involved
tion. Almost all known vasoactive agents affect vasomotor in controlling vasomotor tone. Endothelial cells and broblasts
tone via receptor coupling to G proteins. G protein-coupled are rich in caveolins 1 and 2; smooth muscle cells express all
receptors (GPCR) share several common structural features three caveolins (Cav-1, -2, and -3). In vascular smooth muscle
with seven transmembrane domains with three extracellular cells, Cav-1 serves as a scaffold or chaperone to target AT1 re-
and three intracellular loops. The extracellular loops act in ceptors to caveolae to activate downstream signaling; Cav-1 is
concert with the transmembrane domains to bind the ago- required for ef cient coupling of G q/ll proteins and PLC to
nist. The intracellular loops function to activate a G protein. promote Ca2 mobilization from the sarcoplasmic reticulum.
G proteins are heterotrimeric proteins consisting of , , Cav-1 links AT1 receptors to NADPH oxidases and promotes
and subunits. The G subunit is unique for each receptor recruitment of Rac1 to activate reactive oxygen species (ROS)
and is responsible for generating a speci c intracellular sig- formation. Cav-1 couples IP3 receptors and TRPC3 channels
nal. The and subunits share a high degree of homology for store-operated Ca2 entry. It also suppresses KATP channel
among G proteins; together they function to modulate the activity and negatively regulates Ang II–induced EGF receptor
ability of the subunit to generate the signal. transactivation. Most caveolae in vascular smooth muscle cells
G proteins undergo a conformational change follow- have nanocontacts with the sarcoplasmic reticulum, providing
ing agonist binding to a membrane receptor, which in turn a direct link to intracellular Ca2 mobilization and excitation–
enables guanosine triphosphate (GTP) to replace guanosine contraction coupling.
diphosphate (GDP) on the subunit. The G -GTP com- In endothelial cells, Cav-1 binds eNOS, limiting the
plex then dissociates from the and subunits and inter- translocation and activation of eNOS. Caveolin-de cient ani-
acts with an effector such as an enzyme or a channel. The mals exhibit unusual endothelial dysfunction in that NO pro-
intrinsic GTPase activity of the G subunit then hydrolyzes duction is unopposed, leading to marked vasodilation. Cav-1
GTP to GDP, and the G -GDP complex reassociates with the knockout animals display attenuated vasoconstriction to phen-
and subunits, which terminates the response. G pro- ylephrine and a lack of myogenic tone, largely due to excessive
teins linked to adenylate cyclase are classi ed as G s or G i, NO production. Mice lacking caveolin show that caveolae and
depending on whether they stimulate or inhibit adenylate caveolins play a prominent role in various pathophysiologic
cyclase and cyclic adenosine monophosphate (cAMP) gen- conditions, especially those related to the cardiovascular sys-
eration. G q11/12 activate membrane-bound phospholipase C tem. These disease phenotypes include atherosclerosis, car-
(PLC) or activate membrane Ca2 channels, or both. An ex- diac hypertrophy, cardiomyopathy, diabetes, and neointimal
ample of multiple effects an agonist can produce depending hyperplasia (smooth muscle cell proliferation).25–28
on receptor coupling to different G proteins (e.g., G q11/12) is GPCR desensitization reduces receptor downstream
provided by norepinephrine. The binding of norepinephrine signaling and effector response. G-protein coupled recep-
to an 2-adrenoceptor inhibits adenylate cyclase, reduces tor kinases (GRKs), a family of serine/threonine protein
80
80 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

kinases, initiate receptor-speci c, homologous desensitiza- in mediating the contractile response in vascular smooth
tion that curtails receptor signaling by phosphorylating the muscle cells is an increase in the cytosolic concentration
C-terminal tail of agonist-bound GPCR to promote docking of free ionized calcium ([Ca2 ]i) above its very low basal
with inhibitory -arrestins. -Arrestins not only uncouple value of 10 7 M. (This is approximately 0.01% of the ionic
receptors from heterotrimeric G-proteins, they also target Ca2 levels of plasma and extracellular uid, 1 mM). As
GPCRs to clathrin-coated vesicles for internalization. En- shown in Figure 3.5, cytosolic Ca2 binds with calmodulin.
docytosed receptors are either resensitized and recycled to The Ca2 -calmodulin complex activates myosin light chain
the plasma membrane or degraded in lysosomes. GPCRs in- (MLC) kinase, leading to the phosphorylation of MLC that
ternalize as a stable complex with -arrestin with signaling interacts with actin and adenosine triphosphate (ATP) to
potential and gene transcription.22,29–31 elicit tension development. Increased [Ca2 ]i also phos-
Although GRKs regulate GPCR activity, regulators of G phorylates CPI-17, a phosphoprotein that inhibits MLC.
protein signaling (RGS) proteins directly control the activity GPCR activation of PKC enhances Ca2 sensitivity of the
of G -protein subunits, functioning as endogenous negative contractile apparatus by potentiating the ef ciency of Ca2
regulators of GPCR signaling by accelerating GTP hydrolysis stimulation of CPI-17. In addition, GPCR couples to the
by G -subunits and thereby attenuating signaling. The most G q/11 signal through the monomeric GTPase RhoA/Rho
well-characterized RGS proteins (e.g., RGS2, PGS4, PGS5 of kinase pathway to decrease the activity of MLC phospha-
the R4 family) determine signaling speci city of G q- and tase, resulting in increased Ca2 sensitivity of myo laments
G i-coupled receptors. RGS2 is a GTPase activating protein and enhanced vasoconstriction for a given level of cytosolic
that binds to both G q/11 and G i subunits of GPCRs to ac- Ca2 concentration. Relaxation occurs as a consequence of
celerate their intrinsic GTPase activity. As a result, they are the removal or sequestration of Ca2 from the cytosol and/
deactivated with a reduced stimulation of PLC -mediated or MLC dephosphorylation. Decreases in Ca2 signaling oc-
Ca2 release and subsequent vasoconstriction. RGS2 and/ cur following dissociation of ligand from GPCR surface re-
or RGS5 reduce Ca2 signaling initiated by AT1 and ET-1 ceptor, receptor inactivation, or internalization.2,35
and 1-adrenoceptors and subsequent contraction. RGS5 Various hormones and drugs activate plasma membrane
mRNA, a regulator of vascular remodeling, is expressed in receptors to induce contraction by eliciting an increase in
medial smooth muscle of afferent arterioles and the main [Ca2 ]i. Cytosolic Ca2 is increased through a combination
renal artery in nonhuman primates. RGS proteins may play of Ca2 entry from the extracellular environment and mobi-
a role in mechanosensation and stretch-induced myogenic lization of Ca2 from internal stores. Ca2 can be released
responses, as well as coordinating actions of vasoconstrictor from intracellular stores via release channels, activated by
hormones and paracrine agents. IP3 or by a ryanodine (Ry)-like ligand. Ca2 is released from
RGS2 activity is acutely stimulated by NO and cGMP a common sarcoplasmic reticular pool, with uptake medi-
signaling to promote vascular relaxation by attenuating ated by a common sarcoplasmic–endoplasmic reticular Ca2
Ca2 signaling in response to vasoconstrictor agents. RGS2 ATPase (SERCA). Many vasoconstrictor agents increase in-
expression in vascular smooth muscle is upregulated by tracellular Ca2 by activating a G protein that stimulates
Ang II and is downregulated in hypertension. RGS2 knock- PLC to break down membrane-bound phosphatidylinosi-
out mice are hypertensive with enhanced vasoconstriction tol 4,5-diphosphate (PIP2) into IP3 and 1,2-diacylglycerol
produced by Ang II and 1-adrenoceptor agonists. The ex- (DAG). The soluble IP3 binds to an IP3 receptor located on
aggerated G q/11 signaling in mutant animals is associated the sarcoplasmic reticulum, leading to the activation of Ca2
with renovascular abnormalities, exaggerated vasoconstric- channels and the release of Ca2 into the cytoplasm. The
tion, hypertension, thickening of the vascular wall of the lipophilic DAG remains within the membrane environment
aorta and renal interlobular arteries, and cardiac hypertro- and activates PKC isoform that phosphorylates various regu-
phy. Kidney cross-transplantation studies demonstrate that latory proteins. DAG may also be generated by the actions of
a speci c loss of RGS2 in the kidney causes hypertension, phospholipase D on phosphatidylcholine that is not accom-
whereas the absence of RGS2 from all extrarenal tissues, panied by concurrent IP3 generation and the associated in-
including the peripheral vasculature, does not affect arte- crease in [Ca2 ]i. Agents that activate PKC, such as phorbol
rial pressure. Isolated perfused kidneys of RGS4 knockout esters, induce slowly developing, sustained contractions or
mice show increased renal vasoconstrictor responses to enhance contractile responsiveness to other stimuli.2,36
ET-1.21,32–34 Activation of cell surface GPCRs also leads to the stim-
ulation of plasma membrane-bound ADP-ribosyl cyclase
Regulation of Microvascular Contractility that converts the substrate nicotinamide adenine dinucleo-
Changes in vascular perfusion are mediated by smooth tide (NAD ) to adenosine 5 -cyclic diphosphate (cADP)-
muscle cell contraction or relaxation that elicits a change ribose, a potent Ca2 mobilizing agent that acts on a RyR
in vessel radius and in vascular resistance. Multiple steps to trigger Ca2 release from the sarcoplasmic reticulum.
and enzymatic cascades are involved in the contractile pro- cADP-ribose is a calmodulin-dependent Ca2 -mobilizing
cess, and many of these mechanisms interact with one an- second messenger system that acts independently of IP3,
other to modulate the contractile response. A pivotal step with RyR-mediated Ca2 release to amplify IP3-mediated
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 881
1

Ca2 mobilization. Ryanodine receptors are extremely sen- observation, genetic deletion of Na /Ca2 exchanger NCX1
sitive to [Ca2 ]i and exhibit Ca2 -induced Ca2 release in smooth muscle cells reduces Ang II–induced renal va-
(CICR), a form of autopotentiation. Concentrations of Ca2 soconstriction in vivo, presumably due to less Ca2 entry
over a wide range (5 to 100 M) enhance the open prob- into smooth muscle cells. Nevertheless, the pharmacologic
ability of RyR, which contrasts with a narrower range (180 blockade of NCX or lowering extracellular Na concentra-
to 220 nM) for the IP3R.20 tion causes renal vasoconstriction and exaggerated Ang II–
Calcium entry occurs through a variety of pathways, in- induced constriction of the isolated perfused kidney. Smooth
cluding voltage-operated channels (VOCs) that are activated muscle Na /Ca2 activity responds to Na /K -ATPase inhi-
upon membrane depolarization and voltage-independent bition by an endogenous ouabainlike glycoside to extrude
receptor-operated channels (ROCs) and store-operated the high cellular Na in exchange for Ca2 entry in hyper-
channels (SOCs). Membrane depolarization downstream of tensive states.43,48–50
GPCR activation results from the activation of Cl channels Several different K channels have been identi ed; the
or the inactivation of K channels. Voltage-independent most prominent are Ca2 -activated and ATP-dependent K
ROCs and SOCs also contribute to agonist-induced Ca2 en- channels, which mediate relaxation by hyperpolarizing the
try in both preglomerular and efferent arterioles. ROCs are cell membrane and reducing Ca2 entry through voltage-
activated downstream of cell surface GPCR, independent of gated Ca2 channels. Importantly, increases in intracellular
Ca2 mobilization, via signaling mechanisms involving DAG ATP inhibit K channels, thus causing depolarization. Chan-
and calmodulin, and other possible intermediates. SOCs are nel activity is reduced by intracellular ATP concentrations
known to contribute to agonist-induced Ca2 entry in renal normally present, suggesting that the activity of this channel
vascular smooth muscle cells; they are also termed capacita- is quite low in normal cells and may serve a primary protec-
tive as they respond to depletion of Ca2 stores.37,38 tive role during the depletion of energy reserves.2,51–54
Among the identi ed families of TRP proteins, the ca- At the whole kidney level, Rho-kinase inhibition dilates
nonical TRP (TRPC) family (TRPC1 through TRPC7) is the renal vasculature under basal conditions and attenuates
thought to be involved in Ca2 entry in vascular smooth renal vasoconstriction produced by intrarenal infusion of Ang
muscle cells, most likely functioning as voltage-independent II, arginine vasopressin (AVP), or norepinephrine, as well as
SOCs and/or ROCs. TRPC1 proteins in vascular smooth increased renal perfusion pressure. Frequency analysis of re-
muscle cells form tetrameric channels in association with nal vascular admittance indicates that Rho-kinase strengthens
TRPC4 or TRPC5. TRPC3, -6, and -7 are activated by DAG the myogenic response. Ca2 sensitivity of the afferent
and thus are attractive possibilities as ROCs. TRPC6 is an arteriole is increased by adenosine and norepinephrine.
essential component of 1-adrenoceptor–activated cation Adenosine increases Ca2 sensitivity by PKC, Rho-kinase,
channels in portal venous smooth muscle cells. Preglomeru- and p38 mitogen activated protein (MAP) kinase signaling
lar arterioles have a predominance of TRPC3 and -6 sub- pathways. This provides a mechanism by which different
units that may comprise SOCs and/or ROCs in these vessels. vasoactive agents can modulate reactivity to other stimuli.
Evidence to support TRPC channel function is attenuation Interestingly, adenosine enhances Ang II–induced afferent
of Ca2 responses of the afferent arteriole to norepineph- arteriolar contraction but does not potentiate the contractile
rine by the blockers of voltage-insensitive Ca2 entry Gd 3 responses to ET-1 or norepinephrine. Reactivity to Ang II is
and SKF 96365. Activation of TRPC6 in the afferent arteriole enhanced by the ability of norepinephrine to increase Ca2
leads to increased [Ca2 ]i. The Ca2 permeable TRPC6 is sensitivity with increased MLC phosphorylation.
a key signaling component in a functional slit diaphragm Rho-kinase participates in pressure-induced interlobu-
formed by podocytes in the glomerular ltration barrier. lar arterial and afferent arteriolar myogenic behavior and
Gain-of-function mutations in TRPC6 are the cause for pro- vasoconstrictor responses evoked by Ang II, adenosine A1,
gressive kidney failure with urinary protein loss and focal endothelin ETB, and purinergic P2X1 receptor activation as
segmental glomerular sclerosis. SOCs are activated by Ca2 well as membrane depolarization. Ca2 sensitization due to
mobilization to speci cally restore Ca2 content in sarco- Rho-kinase also contributes to Ang II–induced constriction
plasmic reticulum. Current interest focuses on stromal in- of efferent arterioles. Rho-kinase inhibition dilates preglo-
teraction molecules (STIM) in the sarcoplasmic reticulum, merular and postglomerular arterioles and attenuates va-
which sense SR Ca2 depletion and then translocate to junc- soconstriction elicited by adenosine A1 or endothelin ETB
tions near the plasma membrane to organize Orai proteins, receptor stimulation. Ang II, AVP, and TxA2 constrict pre-
the Ca2 release-activated Ca2 channels, to form a pore and glomerular arcuate and interlobular arteries in the hydrone-
increase Ca2 entry.37,39–47 phrotic kidney in part via Rho-kinase. The afferent arteriolar
Na /Ca2 exchange (NCX) contributes to the regulation myogenic response in this preparation is markedly attenu-
of [Ca2 ]i, working reversibly in the exit mode to extrude ated by Rho-kinase inhibition.55–59
Ca2 or in the entry mode to facilitate Ca2 entry. Exchanger As mentioned earlier, cAMP also activates Ca2 trans-
activity is normally higher in afferent then in efferent arte- location mechanisms that increase extrusion of Ca2 out of
rioles. Ang II stimulates [Ca2 ]i in afferent arterioles by ac- the cell, return Ca2 to the sarcoplasmic reticulum, or in-
tivating NCX to promote Ca2 entry. Consistent with this hibit IP3-mediated mobilization of Ca2 from sarcoplasmic
82
82 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Age nts tha t incre a s e cytos olic ca lcium: Age nts tha t incre a s e cAMP (or cGMP ):

Ang II, AVP , Epine phrine (α ), TXA2 , Epine phrine (β), P TH, P GI2 , P GE 2 ,
Le ukotrie ne s , Nore pine phrine , ANP , Dopa mine , Bra dykinin, Nitric Oxide ,
Endothe lin, ATP Ade nos ine (A2 )

Cl− Ca 2+ K+
ROC Ca 2+ Ca 2+
ADP ribos yl
cycla s e S OC VOC

Gα Gβγ Gs
P LC
NAD+ Ad Cy

RhoA
IP s Ca 2+ cAMP
DAG + IP 3 IP 3 R
cADP ribos e

Ca 2+
Ca -ATPa s e
P KA

RyR
Active MLCK Ina ctive MLCK
Ca lmodulin Ca 2+ − Ca l (phos phoryla te d)
+
Myos in light P hos phoryla te d Te ns ion
cha in (MLC) MLC P hos pha ta s e MLC de ve lopme nt

P KC
Actin
Rho Kina s e

FIGURE 3.6 Primary intracellular signaling systems mediating smooth muscle cell or mesangial cell contraction, with effects of vari-
ous hormones and vasoactive agents on the two major types of receptor systems. For ease of presentation, only two receptor mecha-
nisms are depicted, but each agent acts on its own receptor system.

reticulum stores (Fig. 3.6). Drugs or agents such as PGE2 cGMP/PKG can block RhoA activation and can reduce MLC
and PGI2 that increase cAMP via G s signaling produce activity by inhibiting CPI-17 phosphorylation and activating
increases in RBF and GFR. Small amounts of such ligands MLC phosphatase. cGMP also may stimulate Ca2 -activated
can buffer the action of vasoconstrictor agents without af- K channels, which leads to hyperpolarization.33,62
fecting baseline vascular tone. In contrast, ligand-receptor The arachidonic acid pathways constitute another in-
complexes coupled to the inhibitory G protein (G i) reduce tracellular signaling system. Increased [Ca2 ]i can activate
cAMP levels and cause greater contraction for a given level phospholipase A2 and can release arachidonic acid from
of [Ca2 ]i. G i proteins are reported to activate PLC and mo- membrane phospholipids, resulting in the production of
bilize Ca2 in the afferent arteriole.60,61 various metabolites that lead to vasodilation or vasocon-
Another family of receptors operates through the G striction. Arachidonic acid metabolites exert effects through
protein-dependent activation of guanylate cyclase, the gen- multiple pathways, including cAMP, cytosolic Ca2 , and in-
eration of cGMP, and the activation of protein kinase C to hibition of K channels. Arachidonic acid itself may increase
mediate vasodilation. In addition, two major guanylate cy- Ca2 entry via a noncapacitative Ca2 entry channel. These
clase activators are not G protein-dependent. NO derived actions will be discussed in detail later in this chapter.
from endothelial cells directly interacts with soluble guanyl- Differences in cellular sites and mechanisms of smooth
ate cyclase. Also, ANP directly activates particulate guanylate muscle activation may be partially responsible for the large
cyclase in vascular smooth muscle cells. The mechanisms me- variety of renal hemodynamic responses produced by dif-
diating cGMP-dependent vasorelaxation are similar to those ferent vasoactive agents. There is a major difference in the
used by cAMP. cGMP-dependent kinases lead to the inhibi- mechanisms leading to Ca2 activation in the vascular
tion of voltage-gated L-type Ca2 channels, activation of a smooth muscle cells of the afferent and efferent arterioles.
Na /Ca2 exchanger, stimulation of Ca2 -ATPase, inhibition Preglomerular vessels have a strong dependence on L-type
of IP3 formation, and phosphorylation of phospholamban, voltage-gated Ca2 channels, whereas their in uence is
resulting in increased Ca2 -ATPase activity in the sarcoplas- not readily apparent in efferent arterioles. Antagonists of
mic reticulum. Ca2 desensitization of the contractile ma- Ca2 in ux through L-type, dihydropyridine-sensitive Ca2
chinery, independent of [Ca2 ]i changes, takes place because channels selectively block agonist-induced constriction of
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 883
3

the preglomerular arterioles, including the afferent arteri- effects of Ang II. Nevertheless, the structural mechanism
ole, without affecting efferent arteriolar contraction. This by which mesangial cell contraction actually alters Kf re-
is the case for Ang II, ET-1, norepinephrine, and potassium mains unclear.2,68–70
chloride–induced depolarization. Agents that block L-type
Ca2 channels such as nifedipine, diltiazem, and verapamil
primarily cause afferent vasodilation and impair autoreg- Endothelial Interactions with Vascular
ulatory responses to changes in renal perfusion pressure, Smooth Muscle
affecting both myogenic and tubuloglomerular feedback The vasculature is lined with a continuous layer of endo-
(TGF responses). In contrast, T-type Ca2 channels are ac- thelial cells, which has many functions including serving
tive at both afferent and efferent arteriolar sites and in u- as a diffusion barrier and preventing vascular thrombosis.
ence vascular responsiveness. T-type Ca2 channel blockers Endothelial cells are dynamic metabolic units having mem-
vasodilate afferent and efferent arterioles and prevent con- brane receptors and membrane-bound enzymes, which
tractile responses to various stimuli at both sites. In affer- allow them to respond to and contribute to changes in
ent arterioles, T-type channels may act cooperatively with the concentration of humoral agents. Membrane-bound
L-type channels to bring about membrane depolarization ectoenzymes form or degrade, circulating vasoactive sub-
and Ca2 entry. A primary action on the preglomerular vas- stances such as Ang II (angiotensin converting enzyme
culature also explains the increases in GFR and glomerular [ACE]), ET-1 (endothelin converting enzyme and metal-
capillary pressure produced by Ca2 entry blockers as well lopeptidase), bradykinin (kininase II), and adenonucleo-
as inhibition of the TGF system. An example of a hormone tides (three ectonucleotidases convert ATP, ADP, and AMP).
that exerts its effects through different mechanisms is Ang Localization of ACE in preglomerular vessels allows the
II. Its effects are mediated by at least two mechanisms. conversion of systemically delivered Ang I, an inactive
Afferent arteriolar responses are highly dependent on Ca2 decapeptide, to the biologically active octapeptide Ang II,
entry via L-type channels, whereas the efferent arteriolar that can then induce vasoconstriction locally or in down-
response is in uenced by Ca2 mobilization and Ca2 entry stream segments.2
via T-type channels and through store-operated channels The vascular endothelium serves an important para-
in the absence of any entry through voltage-gated L-type crine role (Fig. 3.7). Endothelial cells participate in con-
channels. NO may suppress voltage-sensitive Ca2 entry tractile and dilator mechanisms by responding to a variety
in both afferent and efferent arterioles. Ca2 entry in outer of stimuli and increasing or decreasing formation of po-
medullary vasa recta is mediated by a combination of L- tent vasoactive substances that act locally to modulate the
and T-type Ca2 channels as well as store-operated cation tone of adjacent smooth muscle cells. General classes con-
channels.63–67 sist of endothelium-derived relaxing factors (EDRF) and
In addition to the smooth muscle cells of the resis- endothelium-derived contracting factors (EDCF). Speci c
tance vessels, the mesangial cells within the glomerular examples of relaxing factors that cause renal vasodilation are
tufts possess contractile capability, which may contribute NO, PGE2, and PGI2 (prostacyclin), carbon monoxide (CO),
not only to the regulation of blood ow through the glo- and an epoxyeicosatrienoic acid (EET), a hyperpolarizing
merulus but also to the ltering capacity. Mesangial con- factor that is a metabolite of the cytochrome P450 path-
traction is postulated to reduce the glomerular ltration way. Examples of EDCF include Ang II, ET-1, thromboxane
coef cient (Kf) by decreasing the radius of capillaries or (TxA2), and oxygen-free radicals. These paracrine factors act
the surface area available for ltration, or both, but the on smooth muscle cells to modify vasomotor tone, prolifera-
precise mechanism is not clear. Many agents, including tive state, and provide a balance between antioxidant de-
Ang II and vasopressin, reduce Kf. Low Kf values have also fense mechanisms and excess generation of O2-derived free
been observed during sodium depletion when plasma and radicals. Thus, the endothelial cells are intimately involved
local concentrations of endogenous Ang II are elevated. in controlling the renal microcirculation.71,72
These responses re ect speci c receptor-mediated effects One of the most studied interactions between endo-
on mesangial cells, because the response can be reversed thelial cells and smooth muscle cells involves the ability
by selective receptor antagonists in vivo and in cultured of the endothelium to modify the vascular responses to
mesangial cells. However, podocytes are closely associated acetylcholine and other agents. Acetylcholine is a power-
with mesangial cells in vivo and also respond to Ang II, ful vasodilator in vivo and also in isolated smooth muscle
suggesting that part of the actions of Ang II on Kf could preparations that have an intact endothelium because
be due to responses by podocytes. Some mesangial cell it stimulates endothelial cells to produce vasodilatory
receptors exert stabilizing effects on mesangial cell con- paracrine substances. However, when applied to vascu-
traction and counteract the in uence of excessive levels lar preparations whose endothelium has been removed,
of vasoconstrictor agents or serve metabolic functions. - acetylcholine induces vasoconstriction by acting directly
Adrenergic agonists and vasodilator prostaglandins (PGE2 on muscarinic receptors on smooth muscle cells. Many
and PGI2) increase cAMP in isolated glomeruli and mesan- other substances have now been shown to stimulate
gial cells and they directly oppose the apparent contractile the release of endothelium-derived vasoactive factors.
84
84 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 3.7 The interaction of endothelial cells with smooth muscle or mesangial cells. Agents that are known to in uence endothelial-
derived relaxing factors or nitric oxide (NO) and endothelial-derived constricting factors (EDCF) production by endothelial cells are
shown. Endothelial cells also produce several vasoconstrictor and vasodilator agents, as shown in the gure and described in the text.

One of the major relaxing factors is NO derived from pathway for renin entry into the circulation from juxtaglo-
L-arginine. These paracrine systems will be discussed in merular granular cells.2,76
later sections. 2,73,74
Endothelial cells have a remarkable capability to
transport substances across their layers through a variety TRANSCAPILLARY EXCHANGE IN
of mechanisms. One of the most impressive features of RENAL MICROCIRCULATION
endothelial cells lining the vasculature is their ability to
form fenestrations that serve as extracellular channels. Forces Governing Ultra ltration at the
This feature occurs predominantly in capillary struc- Glomerulus
tures having large rates of transcapillary volume ux. Bulk movement of uid across capillary membranes of the
Both glomerular and peritubular capillary systems have renal microcirculation is passive in nature, driven by physi-
fenestrations. The glomerular capillaries have abundant, cal forces. As blood ows from the afferent arterioles into the
well-rounded fenestrations that are 50 to 100 nm in di- glomerular capillary tufts, the high hydrostatic pressure pre-
ameter and lack a diaphragm. These fenestrations con- dominates over the counteracting forces caused by Bowman
stitute highly permeable pathways for the large volume space hydrostatic pressure and plasma colloid osmotic
of plasma ltrate that continuously traverses from the pressure. Therefore, uid is driven from the glomerular cap-
glomerular capillaries into the Bowman space. Although illaries through the endothelial fenestrations, across the base-
it is not clear whether subtle changes in the size of the ment membrane, and between the podocyte foot processes
fenestrations contribute to the regulation of the hydraulic into the Bowman space. This movement of uid can be de-
conductivity of the glomerular capillary barrier, it is ap- scribed quantitatively by the Starling ltration-reabsorption
parent that their integrity is essential for the maintenance principle, which is based on the premises that (1) water and
of glomerular ltration. 2,75 solutes ow through extracellular channels or pathways and
The fenestrations of the peritubular capillaries are (2) the diameters of these channels are large with respect to
bridged by a thin diaphragm and are smaller in diameter water molecules, hydrated ions, and solutes of low-molecular
(20 nm). Considering the total number of capillaries, there weight, such as urea, glucose, and amino acids. Thus, except
is much more peritubular than glomerular capillary sur- for the larger solutes, mainly plasma proteins that approach
face area. However, because the overall reabsorptive rate by or exceed the size of the channels, the ltrate is translocated
the peritubular capillaries is nearly equal to the GFR, the without substantive compositional alterations. Detailed con-
average hydraulic conductivity of the peritubular capillaries sideration of the structure and biochemical composition of
per unit of surface area is estimated to be less than that of the glomerular ltration barrier is provided in Chapter 1.
glomerular capillaries. Fenestrations also exist in the termi- The physical forces acting across the glomerular mem-
nal segments of afferent arterioles, and they may provide a brane are glomerular capillary pressure (Pg), Bowman space
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 885
5

pressure (PB), glomerular plasma colloid osmotic pressure relative to that of the afferent and efferent arterioles; thus,
( g), and colloid osmotic pressure of ltrate in the Bowman the hydrostatic pressure drop along the glomerular capil-
space ( B). The ltering capacity of the ltration barrier is laries is small as compared with the pressure drops across
expressed as the glomerular ltration coef cient (Kf), which the afferent and efferent arterioles. Computations based on
is the product of the hydraulic conductivity of the glomeru- the number and dimensions of the glomerular capillaries
lar membrane (Lp) and the total ltering surface area (Sf). yield estimates that are in the range of 1 to 4 mm Hg.
Because the net forces change as uid is ltered along the Thus, Pg is usually treated as a constant value. With these
length of the glomerular capillaries, total GFR can be ex- simplifying assumptions and the use of average values for
pressed by the equation: hydrostatic and colloid osmotic pressures in glomerular
capillaries, the more commonly used formulation for GFR
GFR Kf , 10 [(Pg(x) PB) ( g(x) B)] dx (1) results:

where x represents the normalized length of the glomerular GFR Kf (Pg PB g) (2)
capillaries, with 0 designating the afferent end and 1 desig-
nating the efferent end; (sigma) is the re ection coef cient, The net, or mean, effective ltration pressure (EFP) is cal-
which has a range of 0 to 1. When sigma is 1, proteins are culated as
completely “re ected” by the capillary wall, and the colloid
osmotic pressure is maximally effective. Normal glomerular EFP Pg PB g (3)
capillaries are extremely ef cient in restricting the passage
of macromolecules, and the amount of protein present in The increase in plasma protein concentration is a direct
the normal ltrate in the Bowman space is less than 0.1% of function of the ltration fraction, de ned as the quotient of
the plasma protein. For practical considerations, the effec- GFR and renal plasma ow. Because of the nonlinear rela-
tive colloid osmotic pressure is considered equivalent to that tionship between plasma protein concentration and colloid
of the plasma in the glomerular capillaries ( g). As is shown osmotic pressure, the rate of increase in colloid osmotic
in Figure 3.8, this value increases progressively along the pressure from the afferent to the efferent arteriole increases
length of the capillaries as a function of the relative volume progressively (Fig. 3.9). Empirically derived relationships
of protein-free uid that is ltered. Because colloid osmotic allow the prediction of colloid osmotic pressure ( ) from
pressure is the major force retarding glomerular ltration, the total plasma protein concentration (C) when the albu-
ltration is greatest in the initial segments of the glomerular min-to-globulin (A/G) ratio is known. The commonly used
capillaries and decreases progressively along the length of Landis-Pappenheimer relationship
the capillaries.1,2,77
The exact hydrostatic pressure drop along the glomeru- 2.1 C 0.16 C2 0.009 C3 (4)
lar capillaries is uncertain because experimental assessment
is not possible. Nevertheless, there are abundant parallel applies to an A/G ratio of about 1.2, which is considered
capillaries that collectively have a large cross-sectional area normal for humans.2

FIGURE 3.8 A schematic diagram of the


forces responsible for the ltration of uid
from the glomerular capillaries and the
reabsorption of uid into the peritubular
capillaries. The values are considered rep-
resentative of forces in humans. Ra , afferent
arteriole resistance; Re, efferent arteriole resis-
tance; Pg,B,c,i, pressure in glomerular capillaries,
Bowman’s space, peritubular capillaries, and
renal interstitium; IIg,B,C,i, colloid osmotic pres-
sure in glomerular capillaries, Bowman’s space,
peritubular capillaries, and renal interstitium;
EFP, effective ltration pressure; Kf, ltration
coef cient; GFR, glomerular ltration rate.
86
86 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Micropuncture studies in animals also indicate that glo-


merular pressure is 50 to 60 mm Hg and approximately
40 mm Hg greater than the opposing hydrostatic pressure
in the Bowman space. From this difference in transglomeru-
lar capillary hydrostatic pressure, it can be calculated that
EFP ranges from 15 mm Hg at the afferent end of the glo-
merular capillaries to about 3 mm Hg at the efferent end,
yielding an average EFP of 9 mm Hg (Fig. 3.8). Using this
value and one of 120 mL per minute for total GFR, a Kf
of 13 mL/min/mm Hg for the total nephron population is
calculated. Assuming there are 2 million nephrons in both
human kidneys, the Kf for a single glomerulus is approxi-
mately 6 to 7 nL/min/mm Hg. This value generally agrees
with micropuncture measurements, which indicates that Kf
for an individual glomerulus is 4 to 5 nL/min/mm Hg in
dogs and 2 to 5 nL/min/mm Hg in rats. The large varia-
tion in Kf among rats is due in part to differences observed
among different strains.2,77
The ltration process can operate under one of two
conditions. The rst condition is the case described previ-
ously, in which ltration continues throughout the entire
FIGURE 3.9 A nomogram relating the efferent arteriolar colloid length of the glomerular capillaries and a nite positive EFP
osmotic pressure to the initial plasma colloid osmotic pressure remains at the efferent end of the glomerular capillaries.
and the ltration fraction. Normal afferent arteriolar colloid os- This pattern of disequilibrium is shown by the solid lines
motic pressure, 25 mm Hg, is indicated by the thicker curve. An in the left panel of Figure 3.10. The second condition oc-
example of how to estimate efferent arteriolar colloid osmotic curs when the increase in colloid osmotic pressure is so
pressure for any given ltration fraction and plasma colloid os- rapid that the forces favoring and opposing ltration be-
motic pressure is shown by the dashed lines. come equal at some point within the capillary system, a
condition termed ltration pressure equilibrium (Fig. 3.10,
solid lines in right panel). Under equilibrium conditions,
the latter part of the available ltering surface area is not
The efferent arteriolar colloid osmotic pressure is deter- used and becomes a functional reserve. Studies in some
mined by the initial plasma value and the ltration fraction. strains of rats have suggested that the normal condition is
The nomogram in Figure 3.9 allows for the estimation of the one of ltration equilibrium. Data from other strains of rats
efferent arteriolar colloid osmotic pressure and is indepen- and dogs indicate that, under normal circumstances, glo-
dent of A/G ratios. For example, at a normal ltration frac- merular capillary hydrostatic pressure is suf ciently high
tion of 0.20 and normal plasma colloid osmotic pressure of and the Kf is suf ciently low to prevent the achievement
25 mm Hg, the predicted value for efferent colloid osmotic of ltration equilibrium within the glomerular capillaries,
pressure is 37 mm Hg. and thus ltration occurs along the entire length of the glo-
The hydrostatic pressure in Bowman space (PB) in hu- merular capillaries.2
mans has not been measured directly. In laboratory animals, A physiologic consequence of the equilibrium or dis-
PB is equal to proximal tubular pressure, which ranges from equilibrium of ltration pressures is the in uence of plasma
11 to 15 mm Hg in rats and from 18 to 22 mm Hg in dogs. ow on GFR. Using a mathematical model presented later,
Also, proximal tubular pressure is slightly higher than the the speci c effect of plasma ow can be predicted for both
pressure in adjacent peritubular capillaries. Peritubular conditions when the transcapillary hydrostatic pressure
capillary pressure has also not been measured directly in gradient is kept constant. As shown by the dashed line in
humans, but it can be estimated from intrarenal venous Figure 3.10 (right panel), an increase in plasma ow to a sys-
pressure measurements obtained by retrograde passage of tem in ltration equilibrium diminishes the rate of increase
a renal vein catheter. Values obtained in humans are 20 to of colloid osmotic pressure along the length of the glomer-
25 mm Hg and provide reasonable estimates of proximal ular capillaries. The EFP is not dissipated as quickly, and
tubular pressure. This pressure plus an average efferent col- the point of equilibration of hydrostatic and colloid osmotic
loid osmotic pressure of 37 mm Hg provides a minimal glo- forces is moved distally, which, in effect, results in the recruit-
merular pressure in humans in the range of 57 to 62 mm ment of additional ltering surface area (Sf) and an increase
Hg; actual values are higher to the extent that there is net in the functional Kf. Consequently, increases in plasma ow
ltration pressure at the terminal end of the glomerular can increase the GFR proportionately even when glomeru-
capillaries. lar capillary pressure is unchanged. In the case of ltration
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 887
7

FIGURE 3.10 A comparison of ltra-


tion dynamics in conditions of ltration
equilibrium (right) and disequilibrium
when ltration occurs throughout the
length of capillary (left). The lower pan-
els represent the changes in the trans-
capillary hydrostatic pressure gradient
(Pg PB) and the glomerular plasma
colloid osmotic pressure ( g), and the
upper panels represent the cumulative
GFR along the length of the glomeru-
lar capillary. The dashed lines indicate
the changes occurring in response to
doubling of plasma ow under both
conditions. Pg, glomerular capillary
hydrostatic pressure; PB, hydrostatic
pressure in Bowman’s space.

pressure disequilibrium, increases in plasma ow increase population of uid- lled cylindrical pores of about 5 nm in
GFR only modestly as a consequence of a reduced colloid radius, which totals approximately 5% of the total surface
osmotic pressure pro le, and there is no net recruitment of area. There may also be a very small population of much
previously unused surface area (see Fig. 3.10, dashed lines larger pores.2,75
in left panel). Thus, the magnitude of a selective plasma ow Studies involving quantitative consideration of macro-
effect is smaller during ltration pressure disequilibrium molecular passage through capillary membranes have relied
than during equilibrium. In humans, the low ltration frac- on the thermodynamic approach developed by Kedem and
tion and the relative lack of plasma ow dependence of GFR Katchelsky. Derivations for solute ux (Js) across a constrain-
suggest that the ltration process continues throughout the ing membrane include a convection term, which is the sol-
entire length of the glomerular capillaries (i.e., disequilib- ute ux that occurs as a consequence of the bulk volume
rium, as shown in the left panel of Fig. 3.9).1,2,77 ow (Jv), and a diffusion ux, which is a function of the
concentration gradient of the solute. In its most elementary
Glomerular Permeability to Macromolecules form, solute ux due to convection is
Experiments examining the lterability of test molecules
of different sizes, shapes, and charges have been used to Js JvCs (1 ) (5)
characterize the hydrodynamic properties of the ltration
barrier. A sieving coef cient ( ), or fractional clearance of and solute ux due to diffusion is
a test molecule, is obtained relative to that of a freely l-
tered reference molecule such as inulin. Accurate determi- Js PS ( Cs) (6)
nations can be made when both substances enter the urine
by means of ltration and are not subjected to tubular reab- where Jv is the volume ow (in this case the GFR), and Cs
sorption or secretion. Such data have been tted to various is the average concentration across the membrane; sigma
theoretic models based on limiting membrane structures, ( ) is the re ection coef cient previously discussed. Cs
consisting of an impermeable matrix that is perforated with is the concentration difference across the capillary wall,
cylindrical pores, rectangular slitlike openings, or a mesh- and PS is the diffusional, permeability surface-area product
work of brous or granular gel-like structures. An evalua- coef cient. With small uncharged molecules, such as glu-
tion of molecular sieving or steric restriction in each model, cose, sigma approaches zero and thus glucose ux is sim-
however, is based on the principle of geometric exclusion ply de ned by the product of GFR and the plasma glucose
of large solute molecules from a portion of the membrane concentration. For very large molecules that are restricted
that is accessible to water and small solutes. In essence, the with almost complete ef ciency, sigma approaches 1 and
larger molecules that approach or exceed the effective size thus solute ux due to convection is negligible. The most
of the channels are restricted or “sieved.” Conceptually, the relevant example is for plasma albumin. Using a value of
simplest model that is applicable to the glomerular barrier 1 to 3 mg per deciliter for albumin concentration in early
consists of a size-discriminating membrane with a large tubular uid and a systemic plasma albumin concentration
88
88 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

of 3,600 mg per deciliter, sigma is greater than 0.99. Fur-


thermore, the PS coef cient is so low (0.001 mL per min-
ute) that solute ux due to diffusion also approaches zero.
These quantitative considerations also highlight the dif -
culty in attempting to evaluate mechanisms of proteinuria.
Theoretically, protein passage across the glomerular mem-
brane could increase more than 100-fold, which could
be accounted for by a change in sigma from 0.99 to 0.95.
Such small changes in membrane permeability would not
be expected to be associated with discernible morphologic
changes.2,75
Passage of macromolecules across capillary membranes
is dependent on several factors in addition to the effective
radius. These factors include the electrical charge and the
structural conformation and rigidity of the molecule. As
shown in Figure 3.10, the glomerular sieving coef cient or
fractional clearance (usually determined as CD/CIN) of graded
sizes of electrically neutral dextran molecules declines pro-
gressively as effective radius and molecular weight increase. FIGURE 3.11 Representative sieving curves for several test
Water, electrolytes, and other small, uncharged solute mol- molecules in the glomerular circulation. The curve represent-
ecules with an effective Stokes-Einstein radius of less than ing neutral molecules is based on data obtained with the use
1.8 nm freely permeate. As the effective radius increases, of polyvinylpyrrolidone (PVP) and neutral dextran. The curves
there is a progressive restriction. The fractional clearance of for anionic and cationic molecules are based on studies with
macromolecules the size of immunoglobulin G (IgG) (5 nm) charged dextrans. Also shown are the sieving values for neu-
is essentially zero. For the same equivalent radius, the frac- tral horseradish peroxidase (neutral HRP) and for albumin. The
tional clearances of albumin (3.6 nm) and negatively charged smaller molecules are shown to have a sieving coef cient of 1.0.
dextran sulfate are considerably lower than the clearances See text for details.
of uncharged molecules. In addition, polycationic macro-
molecules are ltered more readily than neutral molecules.
These differences in transport of electrically charged mac-
romolecules are due to the membrane-bound polyanionic
glycoproteins that are rich in sialic acid and heparin sulfate channels. Data currently available indicate that the effective
residues, which set up a negative electrostatic eld that re- radius of the channels in the glomerular membrane is in the
pels polyanions. These are associated with the glycoprotein range of 4.5 to 6 nm.2,75
coat that covers the endothelial fenestrations, the basement Recent studies have challenged the generally held no-
membrane, and the podocytes. Partial loss of these anionic tions described previously regarding glomerular perme-
sites on the glomerular capillary wall can lead to albumin- ability and have suggested that much greater amounts of
uria in the absence of any gross structural abnormalities and albumin are ltered across the glomerular capillaries and
in cases of mild glomerulonephritis. Such a loss has been that most of it is then reabsorbed in the proximal tubules
induced experimentally by neutralization of the electrostatic by megalin and other macromolecule transporters. Stud-
barrier with the polycation protamine. In more severe glo- ies based on measurements of glomerular permeability of
merular injury–associated proteinuria, a larger fraction of uorescent labeled albumin suggest that even normal glo-
the ltrate appears to pass through a population of large di- merular capillaries allow passage of albumin that is avidly
ameter, nonselective pores. bound and retrieved by proximal tubules. This concept has
In addition to size and charge, molecular con gura- been met with substantial resistance and alternative studies
tion in uences the sieving coef cient (Fig. 3.11). Rigid or have failed to indicate such high levels of albumin passage
globular molecules such as horseradish peroxidase or coll across normal glomerular capillaries. Advanced multiphoton
have lower sieving coef cients for any given molecular size determinations have yielded a glomerular sieving coef cient
than neutral dextran polymers with highly deformable lin- of 0.001, a value consistent with the view that the primary
ear structures. Thus, it is likely that the curve for neutral determinant of albuminuria is of glomerular origin and that
dextrans in Figure 3.11 overestimates the true permeability charge does play a signi cant role.78–83
characteristics of more rigid, globular-structured macromol- The controversy about glomerular permselectivity high-
ecules such as plasma proteins. Because shape, exibility, lights important recent ndings regarding the extremely
and deformability contribute to the quantitative relationship complex nature of the glomerular barrier because it consists
between molecular size and transglomerular solute ux, it is of three distinct layers in series; each having its own selectiv-
dif cult to establish the true dimensions of the extracellular ity characteristics. Gene targeted mouse models illustrate the
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 889
9

important role that podocytes have in restricting macromol- 20 mm Hg. With regard to the interstitial compartment,
ecules. In particular, studies of the structural and molecu- i and Pi are about 6 to 8 mm Hg and tend to cancel each
lar characteristics of the slit diaphragms between adjacent other out. Thus, the mean effective reabsorption force is
podocytes reveal an extremely complex network of molecu- 15 mm Hg at the beginning of the peritubular capillary
lar interactions that serve to maintain the normal structure of bed. As uid is reabsorbed into the capillaries, plasma pro-
the podocytes and their close relationship with the basement teins are diluted and the colloid osmotic pressure progres-
membranes. The structure of the glomerular membrane is sively decreases to the original value entering the kidney.
described in detail in Chapter 1. With regard to macromo- There is also a small decline in capillary hydrostatic pres-
lecular permeability, the podocyte slit-diaphragm layer and sure along the peritubular capillaries. Thus, there is an
its complex array of proteins, including nephrin, podocin, effective reabsorptive force over the entire length of the
cadherin, and integrins, are clearly identi ed as the nal peritubular capillaries, which falls from about 15 mm Hg
barrier responsible for the extremely high degree of restric- to about 8 mm Hg (Fig. 3.8). The hydraulic reabsorptive
tion. Nevertheless, the emerging consensus is that all of the coef cient, Kr, for the peritubular capillaries is about 9 to
three major restriction sites make a contribution. In essence, 10 mL/min/mm Hg, which is slightly lower, overall, than
when the endothelial glycocalyx is compromised, protein- the glomerular Kf. This suggests a lower hydraulic conduc-
uria develops. Disorganization of the basement membrane tivity, which is compensated by the larger surface area of
also leads to proteinuria. Furthermore, disruption of the the peritubular capillaries.
nephrin-actin cytoskeleton complex of the podocyte layer With regard to macromolecular permeability, the situ-
and foot process effacement are associated with the greatest ation existing in the peritubular circulation contrasts with
degree of proteinuria. Conceptually, it is attractive to attri- that in the glomerulus because the convective component
bute a progressively greater re ection coef cient (sigma) to is directed inward in association with the continuous uid
the three layers in series.81,84 reabsorption. Thus, the loss of macromolecules from the
postglomerular capillaries occurs only as a consequence of
diffusion of macromolecules from the plasma into the in-
Hemodynamics in Peritubular Capillaries and terstitial compartment. Although signi cant amounts of
Its Role in Fluid Reabsorption protein accumulate in the interstitium, the actual permeabil-
Virtually all of the peritubular capillary network stems from ity is still quite low because of the low removal rate by the
efferent arterioles. About 85% of the postglomerular blood lymphatics in the renal cortex. Some studies indicate that
ow is distributed to peritubular capillaries in the cortex, the postglomerular circulation constrains molecules that can
and the remaining 15% goes to the medulla and papilla readily pass through the glomerular membrane. There also
(Fig. 3.5). The overall density of peritubular capillaries and may be a small population of pores with diameters greater
the total surface area are considerably greater than those of than 5 nm. Nevertheless, most of the channels have a high
glomerular capillaries. The peritubular capillary wall con- degree of ef ciency in restricting albumin and other plasma
sists of a fenestrated endothelial layer covered by a thin proteins, so their re ection coef cients are very close to 1.
basement membrane. Per unit of surface area, it has a lower This occurrence is due, in part, to an electrostatic barrier
hydraulic conductivity and a slightly higher permeability to similar to that found in the glomerular capillaries such that
large molecules than the glomerular wall. negatively charged macromolecules permeate more slowly
In a manner analogous to the process of ltration, the than neutral molecules of the same size. Thus, plasma pro-
peritubular capillary reabsorption (PR) of interstitial uid teins exert almost their full osmotic pressure across the
that is reabsorbed by the renal tubules is determined by the peritubular capillaries. In spite of these high re ection coef-
imbalance of hydrostatic and osmotic forces between the cients, the concentration of albumin in the renal lymph,
interstitial space and adjacent peritubular capillaries. If one and presumably in the interstitial uid, is about one-fourth
considers the forces responsible for reabsorption into the that in systemic plasma. Although this concentration seems
capillaries, then rather high, it should be noted that lymph ow is very low
and less than 1% of net protein is lost from the plasma ow-
PR Kr [( c i) (Pc Pi)] (7) ing through the peritubular capillaries.2
Maintaining the high density of peritubular capillaries is
where Kr is the reabsorptive coef cient, c and i represent of critical importance in providing adequate oxygenation to
the average colloid osmotic pressures in the capillaries and the surrounding tubules. Renal interstitial in ammation may
in the interstitial uid, and Pc and Pi represent the corre- lead to reduced peritubular capillary density and the impair-
sponding hydrostatic pressures. ment of renal function, resulting in salt-sensitive hyperten-
As plasma emerges from the glomerular capillaries, sion. Peritubular capillary loss in renal transplant patients is
it has a colloid osmotic pressure of 35 to 37 mm Hg associated with interstitial brosis and tubular atrophy, lead-
(Fig. 3.8). Furthermore, the hydrostatic pressure drops ing to reduced renal function.85,86
about 40 mm Hg along the efferent arteriole (Fig. 3.3), Lymphatic capillaries, primarily distributed throughout
yielding an initial peritubular capillary pressure of about the cortex, are very permeable to protein and uid. They
90
90 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

serve to return the proteins that leak out of the peritubular colloid osmotic pressure gradient provides an important re-
capillaries back to the circulation, and it is usually assumed absorptive force, favoring capillary uid uptake throughout
that the protein concentration in the lymph re ects the pro- these specialized capillaries. Importantly, the outer medul-
tein concentration in the interstitial uid. The normal renal lary descending vasa recta are encircled at points by contrac-
lymph ow in humans is estimated to be about 2 to 5 mL tile pericytes that provide a means to locally regulate blood
per minute, or less than 1% of the plasma ow. Lymph ow ow.2,15,19,89,90
is increased by elevations in interstitial hydrostatic pressure,
such as those accompanying diuretic states, ureteral obstruc- A Quantitative Analysis of Filtration and
tion, or increases in renal venous pressure.87,88 Reabsorption Dynamics
The mechanisms regulating GFR involve complex inter-
Capillary Uptake by the Vasa Recta actions among the individual determinants. To achieve a
Efferent arterioles of juxtamedullary nephrons provide the better understanding of the singular effects of each determi-
vascular supply to the medulla. These efferent arterioles nant, one can examine the theoretical in uence of selective
branch into long-looped capillaries, termed the vasa recta, changes in an idealized situation where the other determi-
which descend into the medulla in vascular bundles. The nants are held constant. Such theoretical predictions can
vasa recta bundles are intimately associated with and are sur- be made from the simple mathematical model shown in
rounded by concentric rings of loops of Henle and collecting Figure 3.12, which analyzes uid ow dynamics along the
ducts. The medullary circulation has the important function length of a single ltering capillary and the resistances of the
of removing water and solutes reabsorbed from descending afferent and efferent arterioles.
and ascending loops of Henle and collecting ducts without This model can be used to analyze the effects on GFR of
disrupting the large longitudinal osmotic gradients that exist singular perturbations, such as changes in the transcapillary
in the inner medulla during water conserving states. This hydrostatic pressure gradient, the systemic plasma protein
delicate balance is achieved by virtue of the low blood ow concentration, the glomerular plasma ow, and the ltration
and an ef cient countercurrent diffusion of uid and small coef cient. As shown in Figure 3.13 (panel A), changes in the
molecular-weight solutes, which occur because of the spe- transcapillary hydrostatic pressure difference produce strik-
cialized structures of the hairpin-shaped parallel loops of the ing responses in GFR. An increase of 10% causes a greater
descending and ascending vasa recta. The end result is pas- relative increase in EFP leading to an increase in GFR of 19%.
sive equilibration and shunting of uid across the vasa recta GFR is inversely related to plasma colloid osmotic pressure;
loops, from the descending to the ascending limbs, and the as can be seen in panel B, a 10% increase of the plasma pro-
trapping of solute at the bends. The descending vasa recta tein concentration reduces GFR by 25%. The in uence of
have a continuous thick endothelium but aquaporin-1 chan- changes in Kf and in plasma ow on GFR are more complex
nels allow for water ef ux. Because protein permeability is because they affect the rate of rise of plasma colloid osmotic
low, the high plasma protein concentration of the efferent pressure along the capillary bed and thus EFP. Panel C in
arteriolar blood is preserved. In contrast, the ascending vasa Figure 3.13 shows that GFR is affected more by decreases
recta have a highly fenestrated thin endothelium, which than by increases in Kf. The reduced effect of increases
greatly facilitates passive reabsorption. The ascending vasa in Kf above the normal values re ects the achievement of
recta also have a higher permeability to protein. However, the ltration–pressure equilibrium. Once ltration equilibrium
hydrostatic pressure in the ascending vasa recta is relatively is reached, further increases in Kf enhance ultra ltration in
low, about 10 mm Hg, and probably not much higher than early portions of the capillary, which causes protein concen-
interstitial hydrostatic pressure. In the face of a very small tration to increase more rapidly. However, this effect is offset
outward hydrostatic pressure gradient, the transcapillary because the colloid osmotic pressure equilibrates with the

FIGURE 3.12 A“single capillary”


model of glomerular ltration
dynamics and afferent arteriolar
(Ra) and efferent arteriolar (Re)
resistances. Pa, arterial pressure;
Pg, glomerular capillary pressure;
Pc, peritubular capillary pressure;
PB, Bowman’s space pressure; RBF,
renal blood ow; GFR, glomerular
ltration rate.
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 991
1

FIGURE 3.13 Theoretical effects of singular perturbations in (A) the transcapillary hydrostatic pressure gradient (Pg PB), (B) the
plasma protein concentration, (C) the ltration coef cient (Kf ), and (D) the plasma ow at entry to glomeruli. For these simulations,
control values (squares) estimated to be representative of single nephron function in humans were used: GFR 65 nL/min; plasma
ow 300 nL/min; (Pg PB) 40 mm Hg; plasma protein concentration 6.5 g/dL. GFR, glomerular ltration rate.

hydrostatic pressure difference at a more proximal site along and is strongly in uenced by changes in plasma ow. In con-
the capillary. Thus, the mean EFP and the total GFR remain trast, GFR is directly responsive to Kf and is less plasma ow
the same, although the locus of equilibrium is shifted to an dependent under disequilibrium conditions. In either situa-
earlier site. tion, glomerular capillary pressure is quantitatively a much
Panel D of Figure 3.13 demonstrates that the effects of more powerful determinant of GFR than plasma ow.
glomerular plasma ow on GFR are also nonlinear. In the A mathematical analysis that is of more physiologic
absence of changes in the other determinants, GFR increases relevance involves an integrated consideration of changes
only modestly with increases in plasma ow. On the other in preglomerular and efferent arteriolar resistance on glo-
hand, decreases in plasma ow below 200 nL per minute merular dynamics. The predicted effects of constriction and
produce roughly proportional decrements in GFR. This dilation of either afferent or efferent arterioles under ide-
increase in sensitivity is again due to the attainment of l- alized conditions are presented in Figure 3.14, when the
tration equilibrium at the lower values of plasma ow. Glo- other resistances as well as other inputs are maintained at
merular plasma ow exerts these effects by modifying the normal values. A selective increase in afferent resistance re-
intraglomerular pro le of colloid osmotic pressure and thus duces plasma ow and hydrostatic pressure in glomerular
mean EFP. The effects of increases in plasma ow during and peritubular capillaries. GFR decreases proportionately
ltration equilibrium and not in equilibrium were discussed more than plasma ow and thus the ltration fraction falls.
earlier and are shown in Figure 3.10. Changes in plasma In contrast, an increase in efferent arteriolar resistance re-
ow have a relatively small effect on the colloid osmotic duces plasma ow but increases glomerular pressure. GFR
pressure pro le during ltration disequilibrium (Fig. 3.10). initially increases slightly but then reaches a plateau. The
A decrease in plasma ow increases the fraction of plasma plateau region of mean EFP is due to the counteracting ef-
being ltered per unit of capillary length in proximal seg- fects of the increases in glomerular capillary and colloid os-
ments. As a result, the rate of rise of colloid osmotic pres- motic pressures. From these quantitative considerations, it is
sure is increased progressively. During ltration pressure apparent that the preglomerular resistance is ideally suited
equilibrium, GFR is highly plasma ow dependent. Thus, to control GFR. Efferent arteriolar resistance contributes to
there are two major functional consequences of ltration subtle alterations in GFR but exerts major effects on the dy-
pressure equilibrium. GFR is insensitive to increases in Kf namics of the postglomerular circulation. It is important to
92
92 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 3.14 Effects of increases and


decreases in afferent and efferent ar-
teriolar resistance on the glomerular
ltration rate (GFR), plasma ow (GPF),
the ltration fraction (FF), and the mean
effective ltration pressure (EFP).

emphasize that changes in ltration fraction alone cannot number of endothelial fenestrations, the thickness or per-
be used to determine the localization of resistance changes meability of the basement membrane, and the number or
to a speci c manipulation, condition, or drug. For example, structural con guration of the slit pores between the foot
combined increases in afferent and efferent resistances re- processes. Recent attention has been focused on the role of
duce the plasma ow proportionately more than the GFR, the podocytes in control of GFR. Changes in any of these
and the ltration fraction increases. Likewise, combined de- properties could be manifested as changes in Kf.91
creases in afferent and efferent arteriolar resistances increases Animal studies suggest that vasoconstrictor hormones
blood ow proportionately more than GFR and ltration and some vasodilators are capable of reducing Kf. Also, Kf
fraction falls. These changes in the ltration fraction have may be reduced drastically in disease states that involve scle-
often been interpreted incorrectly as being indicative of se- rosis of glomerular capillaries or thickening of the basement
lective change in efferent arteriolar resistance. membrane. Kf may be increased slightly in certain circum-
stances, such as increased plasma colloid osmotic pressure.
Differences in basal Kf are reported among different spe-
Control of Glomerular Dynamics by the cies and different strains and colonies of rats. Agents such
Filtration Coef cient as Ang II and catecholamines decrease Kf. Blockade of the
In addition to the regulation of capillary ow and pressure vascular effects of Ang II negates the Kf lowering effect of
by resistance changes of the preglomerular and postglo- prostaglandins E2 and I2 and parathyroid hormone (PTH).
merular vascular segments, Kf and Kr may be in uenced by In addition, several vasodilators (acetylcholine, bradykinin,
many factors. Alterations in the size of the capillaries or clo- and histamine) decrease Kf in rats through a mechanism that
sure of a fraction of the capillaries may reduce the available is not clear. Although the vasodilator actions are primarily
ltering surface area and thus in uence Kf. The hydraulic due to NO release, it is not apparent how this would decrease
conductivity may be altered by adjustments in the size and Kf. In contrast to the effects observed in rats, vasodilator
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 993
3

agents do not affect Kf appreciably in dogs. The reason for physiologically relevant arterial pressures, both above and
this apparent species difference is not known. Nevertheless, below normal. In response to reductions in arterial pressure,
it seems clear that a variety of paracrine agents, including which may occur during situations such as sleep or recum-
NO and ET-1, can in uence Kf and ltration dynamics.1,36,77 bence, intrarenal mechanisms decrease renal vascular resis-
With regard to the postglomerular vasculature, the tance (RVR) to maintain RBF and GFR at optimum levels.
major regulator of hydrostatic pressure in the peritubular Likewise, increases in arterial pressure, which might occur
capillaries is the efferent arteriolar tone. For a given ow, during exercise or acute episodes of stress, elicit intrarenal
an increase in efferent arteriolar resistance increases the signals that increase vascular resistance and thus maintain
pressure drop along this vessel and thus reduces the pres- RBF and GFR at or near control levels. In addition to RBF
sure in peritubular capillaries. An increase in downstream and GFR, the microvascular and tubular pressures exhibit
resistance due to venous obstruction or elevated tubular autoregulatory behavior. Because glomerular pressure and
pressure increases hydrostatic pressure in the capillaries. In- GFR are autoregulated, the predominant adjustments of vas-
terstitial hydrostatic pressure changes in the same direction cular resistance are localized to the preglomerular arterioles.
as pressure in the peritubular capillaries. Colloid osmotic Figure 3.15 shows representative relationships between the
pressure of blood entering the peritubular capillaries is pri- renal arterial pressure and RBF, GFR, and segmental vascular
marily regulated by the ltration fraction. A higher colloid resistances. The responses of vascular resistance to changes
osmotic pressure exerts a greater reabsorptive force in the in perfusion pressure represent the most commonly inves-
postglomerular circulation. The colloid osmotic pressure in tigated aspect of the renal autoregulatory mechanism, but
interstitial uid is determined by a balance of protein entry other stimuli, such as increases in ureteral or renal venous
from circulating plasma and protein exit by means of the pressure or changes in plasma colloid osmotic pressure, also
lymphatic circulation. In general, stimuli promoting effer- elicit autoregulatory responses. The response serves a nega-
ent arteriolar constriction reduce hydrostatic pressure in tive feedback function to counteract the effect of the distur-
peritubular capillaries and increase efferent arteriolar colloid bance and restore RBF or GFR back toward normal.1,2,92–94
osmotic pressure, changes that favor increased uid reab- Although complex interactions with multiple signaling
sorption from the renal interstitium into the peritubular cap- pathways are involved in the total response, the autoregu-
illaries. Vasodilating stimuli have the opposite effects and are latory component of the vasculature requires activation of
often accompanied by natriuretic and diuretic responses. In voltage-dependent Ca2 channels and is prevented by L-
all cases, however, there is a very intimate coupling between type Ca2 channel blockers. Much of the research oriented
the rate of uid reabsorption from the tubules into the inter- toward understanding this basic response is focused on the
stitium and uid reabsorption from the interstitial compart- mechanisms by which messages are initiated, transmitted to,
ment into peritubular capillaries.87 and received by the smooth muscle cells to affect the requi-
site alterations in vascular resistance. The two basic mecha-
nisms that contribute to the autoregulation phenomenon are
THE REGULATION OF RENAL the myogenic and the TGF mechanisms. A third possible
HEMODYNAMICS contributor has been identi ed recently, but the underlying
mechanism is not known.2,92–94
The high sensitivity of glomerular and peritubular capillary
The myogenic mechanism responds to a distending
dynamics to variations in the intrarenal pressures and ows
force on the vessel wall caused by an increase in arterial pres-
emphasizes the importance of regulatory mechanisms that
sure. The actual distending force can be calculated from the
maintain the intrarenal hemodynamic environment. Overall
law of Laplace that relates tangential wall tension (T) to the
control is shared by several mechanisms that exert speci c
inner radius of the vessel (r) and the transmural pressure
effects on various segments of the renal vasculature. Some of
difference:
these mechanisms are intrinsic to the kidney, whereas oth-
ers depend on extrarenal signals mediated by neural or hor-
T r (Pa Pi) (8)
monal stimuli.
where Pa is intra-arteriolar hydrostatic pressure, and Pi is the
Mechanisms of Autoregulation interstitial uid pressure. When the transmural pressure dif-
Intrinsic paracrine signals can adjust intrarenal vascular re- ference increases, wall tension is increased, which activates
sistance in response to a variety of extrarenal perturbations. Ca2 channels and leads to constriction and a reduction of
Alterations in vascular resistance serve to counter the effect the radius allowing the tension to return to normal. A myo-
of the extrarenal disturbance to stabilize RBF and GFR. The genic response occurs in preglomerular vessels but not
most widely studied manifestation of these intrinsic mech- postglomerular efferent arterioles. This may be due to the
anisms is the phenomenon of renal autoregulation. In re- differential activating mechanisms in these vessels because
sponse to alterations in renal arterial pressure over a wide efferent arterioles do not normally have functional L-type
range, the kidney adjusts its vascular resistance to main- Ca2 channels. In addition to the afferent arteriole, the arcu-
tain, or “autoregulate,” RBF. This range encompasses the ate and interlobular arteries also display myogenic responses.
94
94 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 3.15 Representative relationships between renal arterial pressure and renal blood ow (lower left panel), glomerular ltration
rate (lower right panel), and segmental vascular resistance (upper left panel) for afferent arteriolar resistance (RA), efferent arteriolar
resistance (RE), and hydrostatic pressure (upper right panel) in glomerular capillary (Pg ), proximal tubule (PPT), and peritubular capillary (PC).

Although a large vessel such as the arcuate artery responds the autoregulatory constrictor response to a pressure increase.
to pressure, its contribution to total resistance is quite small. Subconstrictor concentrations of either Ang II or ET-1 potenti-
Thus, the preglomerular arteriolar network has the ability to ate myogenic contraction of afferent arterioles. NO attenuates
respond to extrinsic physical or mechanical disturbances by the rate and strength of the myogenic response. Although en-
an intrinsic myogenic response of vascular smooth muscle. dothelial cells play a role in the rate of autoregulation, stretch-
The initial autoregulatory adjustments in vascular resistance induced, steady-state myogenic tone is observed in vessels
occur rapidly (a few seconds) as a result of a direct local without a functional endothelium. The actual mechanosensi-
vascular mechanism. Such a fast response is thought to buf- tive transducer on vascular smooth muscle cell membranes
fer the glomerular capillaries and the tubular network from has not been completely characterized. Cell surface integrins
sudden changes in arterial pressure and protect from high have been postulated to serve as mechanotransducers by re-
systolic pressures, especially at higher frequencies. Long- sponding to shear, stretch, or tension. Sodium channels have
standing glomerular hypertension is associated with protein- also been shown to respond to mechanical deformation via
uria and the development of glomerular sclerosis.1,93,95–97 a degenerin/epithelial sodium channel complex.2,14,92,93,98–101
A contraction caused by an increase in intraluminal Additional theories to explain the autoregulatory phe-
pressure is elicited by cell membrane depolarization and in- nomenon evolved because of the recognition that slower
creased Ca2 entry through voltage-gated L-type Ca2 chan- acting hemodynamic mechanisms are responsive to the
nels. L-type channel blockers inhibit the myogenic response metabolic demands of tubular transport. The existence of
and the stretch-activated Ca2 channels in the preglomeru- structures within the kidney that seem ideally suited to act
lar microcirculation. T-type Ca2 channels contribute to the as communication links between the distal tubular seg-
myogenic response as a blockade of T-type channels also at- ments and the vasculature provide the morphologic basis
tenuate autoregulatory responses. In afferent arterioles, the for the TGF hypothesis. Indeed, the unique con guration of
T-type channels may sense relatively small stimuli, which then the juxtaglomerular apparatus (Fig. 3.4) allows the macula
cause suf cient Ca2 entry and membrane depolarization to densa to sense some aspect of uid composition at the end
activate voltage-gated L-type channels to activate intracellular of the ascending loop of Henle and transmit a signal(s) to
signaling pathways and to elicit contraction. The increased the afferent arteriole of the parent glomerulus. Thus, the
[Ca2 ]i activates the inositol phosphate cascade to increase juxtaglomerular apparatus provides the anatomic basis for
IP3 and DAG and activate PKC. Inhibition of PKC attenuates a negative feedback mechanism, operating in each nephron,
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 995
5

FIGURE 3.16 The macula densa tubuloglomerular feedback hypothesis (left panel) and the myogenic response (right panel) as
mechanisms to explain renal autoregulation. Solid lines indicate direct relations; dashed lines indicate inverse effects.

that maintains balance between the hemodynamic inputs Conversely, a decrease in arterial pressure causes a reduction
that control GFR and the ltered load and the metabolically in tubular uid ow that elicits dilation of the afferent arte-
determined reabsorptive function of the tubules.1,2,102 rioles. The presence of primary cilia on the lumen of macula
Autoregulation mediated by the TGF mechanism is densa cells provides a ow sensing mechanism such that in-
shown in the left panel of Figure 3.16. For example, an in- creased ow, per se, activates macula densa signals that elicit
crease in arterial pressure initially increases RBF, glomerular afferent arteriolar constriction. The TGF mechanism helps
pressure, and GFR. The increased ltered load increases uid explain vascular responses that occur when the solute load
and solute delivery from the proximal convoluted tubule into to the distal nephron changes as a consequence of changes
the loop of Henle. Such an effect leads to ow-dependent in- in tubular reabsorptive function such as those pharmacologi-
creases in NaCl concentration and osmolality of the tubular cally induced in the proximal reabsorption rate.2,12,14,103–105
uid in the ascending loop of Henle. The macula densa cells Studies at the single nephron level indicate that the
sense the increased tubular uid NaCl or total solute concen- maintenance of ow to the distal nephron is a requisite for
tration and transmit a vasoconstrictor signal(s) to the afferent the full manifestation of autoregulation of GFR. As is shown
arteriole and thus restore RBF and GFR to preexisting levels. in Figure 3.17, autoregulation of single nephron GFR in

FIGURE 3.17 Responses of single nephron


glomerular ltration rate (GFR) and glomerular
pressure to changes in arterial pressure during
conditions of intact ow to the macula densa
(solid lines) and during interrupted ow condi-
tions (dashed lines).
96
96 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

response to acute changes in arterial pressure is highly ef-


cient when tubular uid ow to the distal nephron is
maintained, but it is signi cantly impaired when ow past
macula densa cells is interrupted. Similar responses have
been reported for glomerular capillary pressure. Neverthe-
less, the impairment in GFR autoregulation is not as great
as would be predicted for a fully passive mechanism, indi-
cating that the TGF mechanism works in concert with the
myogenic mechanism to yield the highly ef cient autoregu-
lation characteristic of renal circulation. Recent studies sug-
gest that the interactions are synergistic, in that the presence
of active macula densa signals augments the sensitivity of
the myogenic response. The afferent arteriole is the effector
limb of both mechanisms, and the blockade of Ca2 entry
through L-type channels inhibits both the myogenic and
TGF responses. The highly localized myogenic component
can respond very quickly to a pressure stimulus. The TGF
loop is more complex, involving multiple structures and cell
types, and its response to a pressure change being transmit-
ted along the tubule is slower, on the order of 15 seconds.
The relative importance of these two systems may vary ac-
cording to experimental conditions. Normally each con-
tributes about 45% to near perfect autoregulation initiated
by an acute, single step change in renal perfusion pressure.
A putative third component contributes roughly 10% to
the nal adjustments in RVR that occurs between 25 and
100 seconds.92,93,96,97,106
Increases in ow through the loop of Henle elicit the
constriction of the parent afferent arteriole with consequent
reductions in glomerular pressure and the ltration rate
of the same nephron. These responses are represented in
Figure 3.18. Note that the response is nonlinear, with the
most sensitive region in the physiologic range of tubular
ow. Another important feature is that the reactivity or sen-
sitivity of the TGF mechanism can be modi ed by a variety
of paracrine agents, hormones, and pharmacologic agents.
Increased sensitivity is generally associated with extracel-
FIGURE 3.18 The relationships between the perfusion rate into
lular uid volume contraction, and reduced responsiveness
a late proximal tubule and a single nephron glomerular ltration
has been observed during expansion of extracellular uid
rate (GFR). Dashed lines show responses during conditions of
volume.107
decreased and increased feedback sensitivity. The stippled area
The macula densa sensing segment is located at the end
shows the physiologic range of single nephron GFR as a func-
of the ascending loop of Henle, a nephron segment that is
tion of late proximal ow rate. The technique used to obtain the
virtually impermeable to water and has a powerful NaCl co-
feedback responses is illustrated in the top panel.
transport mechanism. Such transport characteristics result
in the delivery of a hypotonic uid to the macula densa cells.
The nature of the transport processes of the thick ascending
limb is discussed in detail in Chapter 4. In essence, increases The cellular mechanisms responsible for transmitting
in uid delivery from the proximal tubule lead to progres- signals have remained controversial and intriguing. Macula
sive increases in distal ow, sodium chloride concentration, densa cells, like those of the thick ascending limb of Henle,
and osmolality. This coupling between uid ow through possess a Na-K-2Cl cotransporter, which is sensitive to the
the ascending limb and tubular uid solute concentration diuretic furosemide. This cotransporter must be functional
at the macula densa provides the means by which volume for TGF signals to be transmitted, but it may not be the actual
delivery out of the proximal tubule is sensed and regulated. mechanism that activates intracellular signals, which appear
The signal sensed by macula densa cells may be a speci c to involve increases in Ca2 entry from the basolateral side of
constituent of tubular uid such as sodium or chloride or the macula densa cells and increased [Ca2 ]i in response to
total solute concentration.1,107,108 increases in tubular uid osmolarity or NaCl concentration
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 997
7

of the luminal uid. The mechanisms by which [Ca2 ]i is The neuronal NO synthase (NOS) isoform localized in
related to the formation and release of vasoactive media- macula densa cells produces NO in response to increased
tors of TGF signals remain unclear. Calcium changes can be tubular ow above the normal range, which modulates
counteracted by elevations in cAMP. These intracellular ionic TGF activity. A blockade of NO synthesis augments the
changes signal the macula densa cells to form and secrete strength of TGF responses, whereas enhanced NOS levels
constrictor and dilator substances that in uence vascular attenuate the vasoconstrictor response to increased distal
contraction as a function of macula densa transport. The nephron ow. Salt uptake across the luminal membrane by
actual nal effector messenger between the macula densa a furosemide-sensitive Na-K-2Cl transporter may link in-
cells and the afferent arteriole(s) remain under investigation. creases in cellular cAMP and [Ca2 ]i to NO production. O2
Early studies fostered the idea that the effector signal was radicals generated in the vicinity of the juxtaglomerular ap-
locally formed Ang II. However, it is now clearly established paratus can act to scavenge NO, limiting macula densa NO
that the changes in the activity of the renin–angiotensin sys- signaling and thereby producing vasoconstriction and en-
tem modulate the sensitivity of the feedback response but hancing TGF activity. Normal TGF is found in gene knock-
do not directly mediate TGF signaling. The effector agent out animals lacking neuronal NOS.2,113–115
is thought to operate primarily by activating voltage-depen- Arachidonic acid metabolites also modulate TGF activ-
dent Ca2 channels in the afferent arteriole and perhaps the ity and interact with other vasoactive modulators. Cyclooxy-
interlobular artery. Attractive candidates that have been con- genase 2 (COX-2) has been localized to macula densa cells
sidered recently include purinergic agents such as adeno- and the surrounding ascending loop of Henle cells; the re-
sine and ATP or arachidonic acid metabolites. Several recent lease of PGE2 from macula densa cells occurs in response to
studies link the secretion of ATP by macula densa cells to af- reduced luminal NaCl. A COX-2 metabolite attenuates the
ferent arteriolar constriction via the activation of P2 receptors vasoconstrictor autoregulatory and TGF-mediated response
and associated increases in Ca2 entry via voltage-dependent of the afferent arteriole to an increase in arterial pressure.
Ca2 channels. In support of this notion, a defective P2X re- Such a dilator agent also appears to contribute to the macula
ceptor function is associated with impaired TGF function of densa production of NO, which inhibits afferent arteriolar
juxtamedullary nephrons. responses to pressure. Thromboxane A2 and a cytochrome
An alternative view is that the ATP is metabolized to P450 metabolite such as 20-HETE are also involved in the
adenosine, which elicits afferent arteriolar vasoconstric- constrictor limb of TGF. However, gene targeting rendering
tion. Consistent with this proposal, mice lacking adenosine the thromboxane receptor nonfunctional has no effect on
A1 receptors do not exhibit TGF responses in super cial TGF activity.104,116–118
nephrons. Also, mice lacking the nucleotidase involved in Recent evidence supports the existence of a second TGF
degrading ATP to adenosine have impaired TGF. Another loop that links increases in connecting tubule sodium re-
possibility is that an arachidonic acid metabolite such as absorption via epithelial sodium channels to dilation of the
20-HETE participates in mediating TGF-dependent va- afferent arteriole of the parent glomerulus. It is noteworthy
soconstriction. Studies show that increased luminal NaCl that the response of this positive feedback loop is opposite
concentration leads to NO release, which counteracts the to the constrictor signal arising from macula densa cells in
constrictor response. In contrast, PGE2 release is increased response to increased salt delivery to the end of the thick
by reduced NaCl concentration.2,12,103,107,109–112 ascending limb of Henle. The connecting tubule signal
transmitted to the afferent arteriole to increase GFR involves
COX-derived prostaglandins and epoxygenase-derived
The Modulation of Tubuloglomerular EETs. The magnitude of this feedback response is enhanced
Feedback Activity by Vasoactive Agents by Ang II acting on AT1 receptors to stimulate epithelial so-
Macula densa cells also signal the juxtaglomerular cells to dium channel activity and sodium reabsorption. The con-
regulate renin synthesis and release. Mechanisms of renin necting tubule–glomerular feedback circuit may function to
release are discussed in the section on the renin–angiotensin dampen the effects of vasoconstrictor stimuli on the afferent
system. In brief, the directional changes in renin release and arteriole.119,120
Ang II formation are opposite to those that are required for
Ang II to mediate TGF. For example, high tubular ows and The Renin–Angiotensin System
elevated luminal NaCl concentrations are associated with
reduced renin release but TGF-mediated afferent arteriolar The Formation of Ang II
constriction. Nevertheless, Ang II exerts an important role in The renin–angiotensin system exerts control of renal he-
modulating TGF activity during changes in salt diet, extracel- modynamics via its major vasoactive metabolite Ang II,
lular uid volume, and renal perfusion pressure. Tubuloglo- which acts as both a circulating hormone and a locally gen-
merular feedback is absent in mice lacking AT1A receptors, erated paracrine agent. Renin is a proteolytic enzyme that
although the renal vasculature is capable of responding to cleaves Ang I from renin substrate (angiotensinogen) that is
Ang II. Tubuloglomerular feedback is nonresponsive in ani- formed primarily by the liver but also in the kidney. Renin
mals unable to produce Ang II when ACE is mutated.2,14,104 is synthesized primarily in epithelioid granular cells of the
98
98 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

juxtaglomerular apparatus and is secreted into the surround- The recently described enzyme ACE-2 forms Ang (1 to 9)
ing interstitium; renin is also formed in proximal tubule cells from Ang I and Ang (1 to 7) from Ang II. Thus, ACE-2 can
and principal cells of connecting tubule and collecting duct produce more Ang (1 to 7) from Ang II and, in this manner,
segments. Angiotensinogen availability in the plasma and reduces the available Ang II.124,126–128
intrarenal compartments is less than is required to produce Ang II is delivered to renal vascular receptors as a cir-
maximum reaction velocity, so alterations in substrate levels culating hormone or may be formed locally from systemi-
contribute to the regulation of Ang I production. The inac- cally delivered Ang I by endothelial ACE. About 20% of the
tive decapeptide Ang I is then cleaved by ACE to form the circulating Ang I is converted to vasoactive Ang II in the
active octapeptide Ang II. There are abundant amounts of kidney. Ang II is also formed in the interstitial uid from Ang
endothelium-bound ACE in the lungs and in almost all other I, generated as a consequence of enhanced renin release or
tissues. Most of the Ang II in the systemic arterial blood is from Ang I that diffuses from peritubular capillaries into the
formed in the lungs. The major sites of ACE localization in interstitium. The renal tissue Ang II levels are higher than
the kidney are on the luminal surface of endothelial cells that of arterial blood, indicating signi cant amounts of Ang
lining arteries and arterioles (in particular, the afferent II are generated intrarenally. In addition, Ang I and II may be
arterioles), but also the efferent arterioles and the glomerular formed within juxtaglomerular granular cells and coreleased
capillaries, and on the brush border and basolateral mem- with renin to act on adjacent glomerular arterioles. High
branes of the proximal tubule; extravascular ACE is also concentrations of Ang II also exist in proximal and distal tu-
present in the interstitial compartment and on the lumen of bular uid as a result of secretion by proximal tubular cells.
collecting duct segments.2,13,121–125 Ang II derived from proximal tubular cells also may have
Several peptidases act on angiotensinogen to form bio- vascular effects after traversing the interstitium.123,129,130
logically active peptides other than Ang II. Angiotensin with
amino acids 1 to 7 is formed by neutral endopeptidase, but Renin Production and Release
appears to have only slight effects on the renal vasculature As Figure 3.19 shows, renin is released in response to several
under physiologic conditions. Elevated levels of Ang 1 to 7 stimuli, including decreases in sodium intake, the contrac-
occur during ACE inhibition and may dilate renal and non- tion of extracellular uid volume or blood volume, increased
renal vascular beds. Ang III (angiotensin with amino acids sympathetic renal nerve activity, decreased sodium load to
2 to 8) has actions similar to that of Ang II, which can be the macula densa, and decreased renal arterial perfusion
blocked by Ang II receptor antagonists. Ang IV (angioten- pressure. Ang II, ET-1, NO, vasopressin, prostaglandins,
sin with amino acids 3 to 8), a hexapeptide, is reported to and potassium also in uence renin release, acting directly
produce vasodilation by the release of NO or prostanoids on juxtaglomerular cells. The nal effector mechanisms
from endothelial cells by acting on a speci c AT4 receptor. center on changes in the Ca2 and cAMP concentrations

Angiote ns inoge n

Re nin
NaCl Arte rial ECFV S tre s s
Intake Pre s s ure Vo lume Trauma
Aminope ptida s e s A,N,B,m
Angiote ns in I Angiote ns ina s e A, C
Dipe ptidyla minope ptida s e
Mac ula De ns a ACE-2 Ca the ps in
Me c hanis m Ang 1-9
Ca rboxype ptida s e
Baro re c e pto r
ACE Le ucyla minope ptida s e
Me c hanis m
S ympathe tic Ne rvo us
S ys te m
Angiote ns in II

Ang 1-7 ACE-2 Ang III [Ang (2-8)]


Juxtag lo me rular Ce lls Re nin
Re le as e
Cyto s o lic Ca ++ AT1 AT2 Ang IV [Ang (3-8)]
c AMP Re c e pto rs
Ma s
Re ce ptor AT4
Re ce ptor

FIGURE 3.19 A schematic representation of the renin–angiotensin system and the mechanisms of renin release.
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 999
9

in juxtaglomerular granular cells. The Ca2 mechanism is cells to increase renin secretion. Although the sensing mech-
unusual for exocytosis of secretory renin granules in that a anism involving furosemide-sensitive luminal uptake of
decrease in [Ca2 ]i functions as a stimulator of renin release, NaCl is not completely understood, evidence suggests that
in contrast to the opposite in most other secretory cell types. adenosine can inhibit renin release, and its precursor, ATP,
Renin exocytotic release is elicited by increases in cellular is secreted by macula densa cells in response to increases in
cAMP levels. Cytosolic cAMP concentration is controlled tubular NaCl concentration and is converted to adenosine
by synthesis via adenylate cyclase and hydrolysis by cyclic via ectonucleotidases. Circumstances that result in extra-
nucleotide phosphodiesterases (PDEs). Intracellular Ca2 cellular volume depletion or sodium deprivation stimulate
inhibits PDE to modulate the magnitude of cAMP-mediated renin release, at least in part, by the macula densa mecha-
renin release. PDE3 is the major isoform localized to afferent nism. Reduced tubular uid ow to the macula densa leads
arterioles, and recent evidence indicates that the pharma- to increased COX-2 activity and PGE2 release that directly
cologic inhibition of PDE3 increases cAMP and basal renin stimulates renin release via activation of EP4 receptors and
secretion and also enhances the renin secretory response to increased cAMP production in juxtaglomerular granular
-adrenergic and PGE2/EP4 receptor stimulation. cells.12,14,116,133
Exocytosis of secretory granules in the juxtaglomerular
cells is also stimulated by cell shrinkage due to high extra- Other Factors. Renin secretion is inhibited by elevated
cellular osmolality. Juxtaglomerular cells have Ca2 -sensitive plasma or local concentrations of Ang II, vasopressin, ade-
voltage-gated K channels (BKCa) that are activated by cAMP nosine, thromboxane A2, and potassium. The effects of Ang II
to hyperpolarize cells from -32 to -48 mV. Vasoconstric- and other vasoconstrictors appear to be a consequence of
tor G-protein coupled receptor agonists such as Ang II and an end-product inhibition due to increased [Ca2 ]i in juxta-
ET-1 inhibit renin release by activating PLC and mobilizing glomerular cells. PGE2 and PGI2 can stimulate renin release
Ca2 that activates Ca2 entry through store-operated cation through direct effects, which may be related to the stimula-
channels.2,13,121,131 tion of cellular cAMP levels. The atrial natriuretic peptide in-
There are several rst messenger systems that impact on creases cellular cGMP production and inhibits renin release.
cAMP or [Ca2 ]i in juxtaglomerular cells to regulate renin Endothelium-derived factors also modulate renin release,
release. These are discussed brie y in the following section with NO stimulating renin release.13,116,121,134
and in more detail in Chapter 9. COX-2, nNOS, and renin synthesis often change parallel
with changes in salt diet and alterations in tubular uid NaCl
Sympathetic Nervous System and Catecholamines. Jux- concentration, and their products may mutually determine
taglomerular granular cells are richly innervated and respond synthesis and activity of these enzymes. nNOS and COX-2
to renal sympathetic nerve stimulation and to circulating cat- are coexpressed in macula densa cells and the expression of
echolamines. There are both direct and indirect effects on both enzymes is stimulated in volume contraction and high
renin release. Subtle increases in renal nerve traf c or circu- renin states. Parallel changes are observed during chronic
lating epinephrine activate 1-adrenoreceptors on juxtaglo- changes in salt in the diet, with low salt stimulating nNOS,
merular cells, which activate G s proteins to increase cAMP, COX-2, and renin secretion as compared to the inhibition
thereby enhancing renin release. Strong renal nerve activity occurring during a chronic high salt diet. NO derived from
reduces RBF and GFR through activation of 1-adrenergic re- nNOS exerts a stimulatory role on COX-2 expression to pro-
ceptors, with subsequent indirect stimulation of renin release duce PGE2 / PGI2 and to stimulate renin release by acting on
secondary to afferent arteriolar vasoconstriction that reduces EP4 and IP receptors, respectively. PGE2 exerts short-loop
intrarenal baroreceptor and macula densa stimuli.13,132 feedback to inhibit nNOS expression. In some situations,
NO appears to play a more indirect permissive role, permit-
Renal Vascular Baroreceptor. Decreases in renal afferent ting the macula densa pathway of renin secretion to function
arteriolar pressure directly increase renin release indepen- normally. NO stimulates renin release by cGMP, inhibiting
dent of the renal nerves and the macula densa mechanism. PDE3 to increase cell cAMP concentration by reducing cAMP
The juxtaglomerular cells appear to be directly sensitive to breakdown in juxtaglomerular cells, with the activation of
intraluminal pressure and stretch such that decreased wall cAMP sensitive PKA.13,116,121,135
tension decreases cell Ca2 entry and [Ca2 ]i, whereas the Although many stimuli increase activity of the renin–
opposite occurs at elevated arterial pressures. Under some angiotensin system, most are related to circumstances
circumstances, the same extrinsic disturbance may in uence that compromise body uid volume homeostasis. Thus,
both the macula densa and the vascular baroreceptor mecha- the stimulation of renin release and the activation of Ang
nism to increase renin release, but the vascular receptor sys- II–dependent mechanisms help to minimize renal uid and
tem can act independently.2 sodium losses and to maintain extracellular uid volume
and arterial blood pressure. The myriad of actions exerted
Macula Densa. Macula densa cells detect decreases in the by Ang II all seem to be homeostatically appropriate to
NaCl load or concentration emerging from the ascending achieve this end. Only the renal vascular actions of Ang II
loop of Henle and send a signal(s) to the juxtaglomerular will be covered in this chapter, but it should be pointed out
100
10 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

that Ang II is also a potent stimulator of aldosterone release the renin–angiotensin system and the TGF mechanism are
and can directly enhance salt reabsorption by the proximal illustrated in Figure 3.20.1,2
tubule, the Henle loop, and the distal nephron segments. The precursor of renin, prorenin, is also released by jux-
It also has important effects on the central nervous system taglomerular granular cells and renin-producing tubular cells.
such as stimulating thirst, vasopressin release, and sympa- Although prorenin is inactive, it can bind to the prorenin recep-
thetic nerve activity.2,129 tor that has been recently discovered and characterized. Bind-
The macula densa plaque has distinct mechanisms for ing of prorenin to the prorenin receptor activates prorenin and
renin release and the TGF system. The TGF mechanism increases its catalytic ef ciency to generate Ang I from angioten-
involves the macula densa by sending a vasoconstrictor sinogen. Thus, increased tissue levels of prorenin or the prorenin
signal to the afferent arteriole in response to increases in receptor may in uence the local generation of Ang I, leading to
tubular ow and the accompanying increases in solute and increased Ang II formation. Prorenin also appears to signal via
sodium concentration from the Henle loop. Under these the MAP kinases extracellular signal regulated kinase (ERK) 1/2
conditions, the macula densa signals to decrease renin re- to upregulate pro brotic and COX-2 genes independent of Ang
lease and local Ang II activity, which is opposite to that re- II. Prorenin receptors are localized in vascular endothelial and
quired for Ang II to mediate a TGF-induced contraction of smooth muscle cells, glomerular mesangial cells and podocytes,
the afferent arteriole. Thus, Ang II clearly does not mediate and collecting duct segments of the nephron.136,137
TGF responses; it does, however, modulate the sensitivity
of the TGF mechanism to the macula densa vasoconstric- Angiotensin Receptors
tor signal(s). When tissue Ang II levels are increased, There are two major subtypes of receptors for Ang II in the
smooth muscle responsiveness is augmented. In contrast, renal circulation. In utero, animals have a larger population
when Ang II levels are suppressed, whether in response to of AT2 than AT1 receptors, which decreases progressively af-
physiologic manipulations or as a consequence of a phar- ter birth; in adult life, the major subtype on vascular smooth
macologic blockade, the sensitivity of the TGF system is muscle cells is the AT1 receptor. The AT1 receptor is pres-
attenuated. Interestingly, sensitivity can be restored by the ent on preglomerular arteries and arterioles, juxtaglomerular
administration of Ang II but not norepinephrine. In addi- granular cells, glomerular mesangial cells, efferent arteri-
tion to directly affecting the afferent arteriole, Ang II affects oles, and vasa recta bundles of the inner medullary stripe.
TGF activity by altering tubular reabsorption and uid Humans have one AT1 receptor, whereas rodents have two,
delivery to the macula densa. The vascular and tubular termed AT1A and AT1B, which are 94% homologous. Cur-
effects work in concert to reduce GFR when sodium excre- rently available pharmacologic antagonists of AT1 receptors,
tion is reduced as observed during volume contraction. As such as candesartan, losartan, and valsartan, do not distin-
previously mentioned (Fig. 3.18), changes in TGF respon- guish between the two AT1 subtypes in rodents. Almost all
siveness during altered volume states may be largely due to of the vasoconstrictor actions of Ang II on the renal vascula-
changes in tissue levels of Ang II. The interactions between ture under physiologic conditions are mediated by AT1. Both

FIGURE 3.20 The modulator hypothesis for postulated interactions between the tubuloglomerular feedback mechanism and the
intrarenal renin–angiotensin system. Flow-related changes in the tubular uid concentration can elicit signals from macula densa
(MD) cells to vascular contractile cells independent of angiotensin levels. Changes in the angiotensin II (A II) concentration in uence
the sensitivity or responsiveness of macula densa cells or of vascular smooth muscle cells to the signals coming from the macula
densa cells. A I, angiotensin I; A II, angiotensin II; Aff. Art., afferent arteriole; JGA, juxtaglomerular apparatus.
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1101
01

the AT1A and AT1B receptor mediate Ang II–induced Ca2 The AT2 receptor is characterized by a high af nity to
signaling in smooth muscle cells and renal vasoconstriction. the nonpeptide antagonists PD123319, CGP 42112, and
Evidence for the role of AT1B receptors derives from mice Compound 21, and has a sequence 33% identical to that of
lacking a functional AT1A receptor.36,138–141 the AT1 receptor. Its high expression in fetal tissue suggests a
The AT1 receptor is primarily coupled to the GTP- role in embryonic development. This receptor has a typical
binding protein G q11/12, whose activation leads to the trigger- pattern of seven transmembrane domains and is coupled to
ing of several signaling pathways, including the stimulation a G protein. Recent studies suggest that AT2 receptors exert
of PLC . The receptor activation of protein tyrosine kinases modulatory actions to partially counteract the effects caused
leads to somewhat slower stimulation of phospholipase C . by AT1 receptor activation. AT2 receptor activation increases
Phospholipases D and A2 may also be activated, favoring the bradykinin and NO levels, leading to increases in cGMP and
release of DAG and phosphatidic acid from phosphatidyl- vasodilation. AT2 receptor activity may be upregulated during
choline and arachidonic acid, respectively. The PLCs act on chronic salt deprivation. AT3 and AT4 receptors may be selec-
membrane-bound phosphoinositides to yield DAG and IP3. tive for angiotensin with amino acids 1 to 7 and Ang IV (angio-
DAG stimulates protein kinase C, whereas IP3 diffuses through tensin with amino acids 3 to 8), although they appear to play a
the cytosol to activate IP3-sensitive receptors/release channels minor role in regulating renal hemodynamics.107,126,138,144
on membranes of the sarcoplasmic reticulum, triggering Ca2 Ang II receptors are regulated in response to different
release to the cytosol. The mobilized Ca2 triggers a complex physiologic conditions. It is noteworthy that glomerular and
array of events. Calcium-sensitive chloride channels may be vascular receptors are regulated differently from proximal tu-
stimulated to promote chloride ef ux and depolarization of bular receptors. A low salt diet and high levels of Ang II lead
the plasma membrane. Such a signal will activate voltage- to the downregulation of arteriolar and mesangial cell AT1
gated L-type calcium channels to allow for in ux down a very receptors and the upregulation of tubular receptors.140,145,146
steep gradient, from 1 to 2 mM in the extracellular uid to
100 to 200 nM in the cytosol. Calcium release from inter- Actions of Ang II on the Renal Microvasculature
nal stores also signals store-operated cation channels to open Ang II elicits dose-dependent AT1-mediated decreases in RBF
and allow further Ca2 entry. AT1 receptor activation may and GFR. The decreases in GFR are often smaller than the de-
also trigger voltage-dependent Ca2 channels independent creases in RBF such that ltration fraction increases. The in-
of Ca2 release from intracellular stores. As discussed earlier, creased vascular resistance is due to both afferent and efferent
afferent arteriolar contractions induced by Ang II is depen- arteriolar constriction, and glomerular capillary pressure is
dent on Ca2 entry through voltage-gated channels, whereas well maintained. At high concentrations the glomerular ltra-
efferent arterioles are not affected by L-type channel blockers tion coef cient is reduced. Ang II produces more pronounced
but are in uenced by T-type channel blockers. Recent studies vasoconstriction when endogenous levels are low, presum-
indicate that AT1 receptors rapidly activate NADPH oxidase to ably because of receptor upregulation and when vasodilator
produce superoxide anion by the afferent arteriole. This leads prostaglandins are blocked by COX inhibitors. Larger Ang II
to Ca2 mobilization mediated by RyR through a direct action effects are also noted after the endothelial production of NO
and/or one mediated by ADP ribosyl cyclase and the produc- is blocked. As mentioned earlier, Ang II can potentiate TGF-
tion of cADP ribose that sensitizes RyR/release channels on the mediated changes in preglomerular vascular tone. The multi-
sarcoplasmic reticulum to increase [Ca2 ]i.1,2,36,107,142,143 ple effects of Ang II are illustrated in Figure 3.21. In addition

Angiote ns in II

AT1 type AT2 type


re ce ptor re ce ptor

FIGURE 3.21 Multiple actions of angiotensin


II on renal function mediated by AT1 and AT2
receptors. ET, endothelin; TXA2 , thromboxane;
ICAM1, intercellular adhesion molecule 1; MCP1,
monocyte chemotactic protein 1; IL-6, interleu-
kin 6; TGF , transforming growth factor ; PAI-1,
plasminogen activator inhibitor 1; NF- B, nuclear
factor kappa light chain enhancer of activated B
cells; VEGF, vascular endothelial growth factor.
102
10 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

to these effects, Ang II in uences the medullary circulation, interlobular arteries, afferent arterioles) and the efferent
notably at concentrations lower than those required to elicit arterioles. NO also appears to contribute to the increased
overall vasoconstriction in the cortex. This notion is support- or maintained glomerular ltration coef cient. Endothelial
ed by the nding that the Ang II receptor density is much factors mediate or modulate pressure-dependent responses
higher in the medullary vessels and in the interstitial cells of vascular segments exhibiting autoregulatory behavior.
than in the postglomerular cortical vasculature.1,2,36 Note however, that NO does not t a mediator role in this
The effects of endogenous Ang II are observed using scheme because increased pressure and shear stress cause
angiotensin receptor antagonists or ACE inhibitors in states increased production of NO, a vasodilator, at the same
when the prevailing Ang II levels are high. During sodium time the responsive vascular elements exhibit vasoconstric-
restriction, RBF is increased after the AT1 receptor blockade, tion. NO inhibition results in renal vasoconstriction, but
whereas the GFR responses are smaller and more variable; the steady-state autoregulatory RBF responses to changes
ltration fraction usually declines. Renin inhibition pro- in perfusion pressure are unaffected and remain highly
duces similar results, and combined renin and angiotensin ef cient.2,149,150
inhibition causes decreases in RVR and increases in GFR. NO-induced renal vasodilation is mediated by cGMP-
Activation of the AT1 receptor is primarily responsible for the dependent and perhaps a cGMP-independent system. The
renal vascular effects of the endogenous Ang II on afferent latter appears to involve cGMP cross-talk with the cAMP
and efferent arterioles and on Kf. Whole-kidney autoregula- pathway, with cGMP inhibiting cAMP breakdown by PDE3,
tion is not affected by AT1 receptor blockade or ACE inhibi- more so than the activation of cGMP-protein kinase activ-
tion, although the contribution of the TGF mechanism may ity. Part of the dilatory response to NO may be mediated
be reduced and the plateau of the autoregulation relation- through increased K channel activity and reduced produc-
ship is elevated.1,2,36 tion of 20-HETE.62,118,151
Although NO does not affect overall steady-state auto-
Endothelium-Derived Vasoactive Factors regulatory adjustments in RVR, NO attenuates the strength
and rapidity of the myogenic adjustments in resistance.
Nitric Oxide NO also attenuates the strength of TGF responses to in-
Endothelial cells release NO in response to many stimuli, creased distal tubular ow. The inhibition of NOS leads
including mechanical shear stress and hormone, paracrine, to greater relative decreases in medullary blood ow than
and autocrine factors, that increase [Ca2 ]i. NO is formed in cortical blood ow. A NO blockade usually reduces re-
by vascular endothelial cells from the amino acid L-argi- nin release and responsiveness to reductions in perfusion
nine via NOS, which is a soluble NADPH , and the Ca2 - pressure.92,93,152,153
calmodulin–dependent citrulline-forming enzyme that Endothelial NO synthase is localized to the endothelial
requires two cosubstrates (O2 and NADPH) and four cofac- cells all along the renal vasculature (the interlobular arteries,
tors (heme, avin mononucleotide, avin adenine dinucle- the afferent and efferent arterioles, the glomerular capillaries,
otide, and tetrahydrobiopterin). As shown in Figure 3.7, and the vasa recta) and the thick ascending limb of the Henle
several agents induce their vascular effects via endothelial loop. Targeting of eNOS to specialized plasma membrane
cell eNOS activation and the release of NO. Examples in- invaginations termed caveolae is required for maximal eNOS
clude factors that stimulate the M1-muscarinic receptor activity. Neuronal NO synthase (nNOS) is present primarily
(acetylcholine), the B2-kinin receptor, the 2-adrenoceptor, in epithelial cells (thick ascending limb of the Henle loop,
the purinergic receptors (ATP, ADP), and the ETB receptor. the macula densa, the collecting duct). NO inhibits tubular
The rate of renal NO production is higher in the medulla transport along the nephron. An inducible form of NO
than in the cortex. NO and nitrosamines have short bio- synthase is normally quiescent but is capable of producing
logic half-lives of less than 10 seconds, which is especially large amounts of NO in vascular smooth cells and mesangial
shortened by hemoglobin scavenging and in oxygenated cells during in ammation. An endogenous inhibitor of L-
solutions in the presence of the superoxide anion ( O 2 ), arginine cellular uptake and NOS activity is asymmetric
by a mechanism involving the production of peroxynitrite dimethylarginine (ADMA) that is normally inactivated by
(NO3 ) from NO. In the same manner, NO acts to scav- NG-NG-dimethylarginine dimethylaminohydrolase (DDAH)
enge O2 .147–149 to form L-citrulline. DDAH expression is found at the same
The basal release of NO tonically maintains a low RVR sites as NO synthase. ADMA concentrations are elevated
at rest, acting to buffer vasoconstriction produced by Ang II, during states of oxidative stress and disease.154–156
ET-1, catecholamines, and other endogenous factors. Acute
inhibition of basal NO production causes renal vasocon- Endothelin
striction, with decreases of 25% to 35% in RBF and reduc- Endothelin refers to a family of long lasting vasoconstrictor
tions in cGMP levels in the interstitium and the urine. GFR peptides that act locally as paracrine hormones. ET-1, -2, and
responses are smaller and tend to be unchanged or decrease -3, each a 21 amino-acid peptide, are constitutively released.
about 10%. Thus, the ltration fraction is increased. Basal ET-1 is the major form synthesized and secreted by endothe-
NO dilates both the preglomerular vasculature (arcuate and lial cells of the preglomerular vasculature and the vasa recta
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1103
03

and by the medullary collecting ducts. Endothelin-convert- equal and opposite. The ETA receptor blockade produces re-
ing enzyme (ECE) is the main enzyme responsible for the nal vasodilation due to a 5% to 10% increase in RBF, whereas
genesis of ET-1 from prepro-ET-1 ( 200 amino acids); chy- ETB receptor antagonism leads to constriction of 5% to 10%.
mase and matrix metalloproteinase II are also involved in the Although ET-1 contributes to basal vascular tone, it does not
production of ET intermediates. Ang II is a potent stimulus interfere with renal autoregulatory mechanisms.2,150,160,161
for ET-1 production. Other simulants include bradykinin, ETA and ETB receptors stimulate [Ca2 ]i in smooth
ATP, platelet activating factor, thrombin, and shear stress. muscle cells of preglomerular arteries/arterioles through a
Neutral endopeptidase 24-11 degrades and inactivates ET-1. combination of mobilization and entry pathways. Low con-
Cytokines such as interleukin-1-beta (IL-1 ) stimulate ET-1 centrations of ET-1 activate Ca2 entry channels, with higher
production. The two known receptor types, ETA and ETB, concentrations mobilizing intracellular Ca2 via the PLC-
are G-protein coupled and lead to IP3 and cyclic ADP ribose IP3R and NADPH oxidase/cADPR/RyR pathways. Calcium
formation, PKC activation, and Ca2 mobilization, in addi- channel blockers reduce the magnitude and duration of
tion to Ca2 entry. ETA receptors are predominantly found ET-1–induced renal vasoconstriction. The afferent arterio-
on vascular smooth muscle cells. ETB receptors are localized lar constriction produced by ET-1, similar to that caused by
on endothelial and tubular cells as well as vascular smooth Ang II, is dependent in part on Ca2 entry through voltage-
muscle cells. The afferent arteriolar constriction produced dependent L-type channels as well as mobilization in intra-
by ET-1, like that caused by Ang II, is dependent in part cellular Ca2 stores. The action of ET-1 on efferent arterioles
on Ca2 entry through voltage-dependent L-type channels. appears to depend exclusively on the mobilization of intra-
The action of ET-1 on efferent arterioles appears to depend cellular Ca2 . ET-1 inhibits renin synthesis and release by
exclusively on the mobilization of intracellular Ca2 and/ ETA and ETB receptor stimulation of [Ca2 ]i in juxtaglomeru-
or entry through voltage-independent cation channels. The lar granular cells.13,20,162
highest concentration of ET-1 in the body exists in the renal ET-1 acts on glomerular mesangial cells to cause the
medulla, where it is synthesized by collecting duct cells and contraction and stimulation of mitogenesis. High concentra-
acts in a paracrine/autocrine manner to cause natriuretic and tions of ET-1 that reduce sodium excretion increase plasma
diuretic effects through ETB receptors via the stimulation of renin activity, presumably via the macula densa mechanism.
NO. In addition, ET-1 has inotropic, chemotactic, and mito- In addition, ET-1 may affect neurotransmission, eicosanoid
genic properties. Overall, ET-1 increases blood pressure and synthesis, and ANP synthesis and release. The activation of
vascular tone.156–158 endothelin receptors causes the release of eicosanoids and
The vascular actions of ET-1 re ect a combination of NO from endothelial cells and ANP from myocytes.157,158,163
vasoconstrictor ETA and ETB receptors on smooth muscle
cells and ETB receptors on endothelial cells, which cause va-
sodilation mediated by NO. Concurrent ET-1 stimulation of Heme Oxygenase and Carbon Monoxide
ETA ETB receptors causes net renal vasoconstriction, and Heme oxygenases (HO) are microsomal enzymes that cata-
the inhibition of ETB receptor activation leads to more pro- lyze the degradation of heme to form iron, biliverdin, and
nounced vasoconstriction. Thus, endothelial ETB receptors carbon monoxide (CO). The vascular actions of CO include
buffer the constriction caused by the stimulation of both ETA the direct relaxation of vascular smooth muscle cells and the
and ETB receptors on the smooth muscle cells. Nevertheless, indirect contraction through the inhibition of NOS. Similar
selective stimulation of vascular ETB receptors using a phar- to NO, CO produces vasodilation by stimulating soluble
macologic agonist elicits renal vasoconstriction. Thus, there guanylyl cyclase in smooth muscle cells to signal through
appears to be a complex interaction between receptor signal- the cGMP/PKG pathway. CO also may bind directly to Ca2 -
ing. Endothelial ETB receptors in the kidney and lung also activated BK channels to depolarize the plasma membrane.
seem to function as nonsignaling clearance receptors, effec- A primary action of CO is to attenuate vasoconstriction pro-
tively reducing the local concentration of ET-1, and thereby duced by agents such as Ang II and catecholamines, with
attenuating vasoconstriction.150,159 greater buffering effects in the absence rather than in the
Exogenous ET-1 reduces RBF by stimulating both ETA presence of NO. Nevertheless, CO can dose-dependently in-
and ETB receptors to cause the constriction of the arcuate crease NADPH oxidase-dependent O2 production and con-
and interlobular arteries and the afferent and efferent arte- strict interlobar and interlobular arteries via thromboxane
rioles. Glomerular capillary pressure is relatively well main- thromboxane prostanoid (TP) receptor activation, perhaps
tained in the presence of reduced RBF. The decrease in GFR involving isoprostanes, effects inhibited by the O2 scaven-
is primarily mediated by reductions in plasma ow and Kf. ger biliverdin. Thus, the vascular response to CO is mixed
The reduction in Kf appears to be mediated by a second- in that CO can elicit signaling leading to vasodilation and
ary release of Ang II, eicosanoids, or neurotransmitters. The vasoconstriction, the net effect depending on experimental
preglomerular vasculature has equal proportions of ETA and conditions.164,165
ETB receptors as compared to a ratio of 2:1 on their smooth HO-2 is constitutively expressed in the kidney, mainly
muscle cells devoid of endothelial cells. Under basal condi- in proximal tubules with relatively weak presence in the
tions, ET-1 exerts dual actions on the vasculature that are renal vasculature. HO-1 is inducible during in ammation
104
10 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

and oxidative stress. The in uence of endogenous HO-2 me- classi cation). Cystathionine- -lyase and - -synthetase are
tabolites on renal hemodynamics appears to be minor un- the main enzymes forming H2S from L-cysteine or L-homo-
der physiologic conditions. Pharmacologic inhibition of HO cysteine in endothelial and smooth muscle cells of the vas-
(chromium mesoporphyrin [CrMP]) reduces urinary excre- culature wall and erythrocytes, physiologically activated by
tion of sodium and water even under conditions where there Ca2 -calmodulin. H2S is inactivated by binding to hemoglo-
are no effects on arterial pressure, RBF, GFR, or plasma renin bin to form sulfhemoglobin, As an endothelial-derived relax-
activity, both in control animals and after NOS inhibition, ing factor, H2S produces concentration-dependent dilation
suggesting a primary tubular effect of endogenous CO. Like- of large conduit arteries and small resistance arterioles, pri-
wise, HO inhibition with CrMP does not alter afferent arte- marily acting directly to open KATP channels to hyperpolarize
riolar diameter of juxtamedullary nephrons of normal rats. vascular smooth muscle cells. In contrast to NO and CO,
However, other studies using SnMP to inhibit HO report H2S does not stimulate soluble guanylate cyclase. Being a re-
decreases in RBF with variable responses in GFR. Another ducing agent, H2S appears to alter cellular redox status. Low
study shows that acute inhibition of renal medullary HO ac- concentrations of H2S may cause vasoconstriction reducing
tivity and CO production reduces medullary blood ow and NO availability by reacting with NO to form a nitrosothiol
sodium excretion and blunts pressure natriuresis. Chronic compound and inhibit eNOS. H2S and the H2S donor NaHS
HO inhibition produces hypertension that is salt sensitive. downregulate cAMP production in vascular smooth muscle
Mice de cient in HO-2 are normotensive with normal RBF cells, producing vasoconstriction, and in juxtaglomerular
during basal conditions or during NOS inhibition, whereas granular cells, inhibiting renin release. Physiologic levels of
Ang II paradoxically produces less pronounced renal vaso- H2S are also angiogenic, antiproliferative, and anti-in am-
constriction in the absence of HO-2. Other investigators re- matory. Mice de cient in a synthetic enzyme have reduced
port that the pressor and renal vasoconstrictor responses to H2S levels, reduced endothelium-mediated vasodilation,
low levels of Ang II are magni ed by inhibition of HO activity and develop hypertension.173–176
(tin mesoporphyrin) in normal euvolemic animals.164,166–169 Little is known about H2S effects on the renal micro-
Both HO-1 and HO-2 mRNA are expressed in macula circulation. H2S is produced in the kidney and the exog-
densa cells, and HO metabolites inhibit TGF. Increased HO enous H2S donor NaHS exerts diuretic, natriuretic, and
activity attenuates TGF-induced vasoconstriction through kaliuretic effects in association with increases in GFR and
macula densa release of CO and biliverdin during increased RBF. Conversely, acute intrarenal inhibition of H2S produc-
salt reabsorption. The CO effect on TGF is linked to cGMP tion reduces GFR and sodium excretion without affecting
pathway and biliverdin to reduce O2 levels. Renal HO-1 RBF. Chronic inhibition of H2S synthesis reduces RBF but
induction (hemin, SnCl2) dilates afferent arteriolar diameter not GFR because sodium excretion decreases in association
and attenuates juxtamedullary afferent arteriolar autoregula- with the development of hypertension. The actions of H2S
tory responses to increases in renal perfusion pressure, an on afferent and efferent arterioles and TGF activity await in-
effect mimicked by CO acting through cGMP/PKG signaling vestigation.177,178
but not biliverdin. These results are consistent with ndings Because H2S is oxidized in mitochondria in pO2-
that increasing CO levels directly or with hemin administra- dependent manner and ambient pO2 is lower in the renal
tion increase RBF, urine ow, and sodium excretion. Whether medulla than the cortex, H2S accumulates in medullary re-
the effects involve myogenic and/or TGF mechanisms await gions. High H2S concentrations in the relatively hypoxic re-
investigation.164,170,171 nal medulla function as an oxygen sensor that acts to increase
Hypoxia-induced HO upregulation protects renal tis- local O2 by increasing medullary blood ow in combination
sue from acute and chronic injury. Activation of HO has an with the direct inhibition of mitochondrial respiration.179
antioxidant effect by degrading the heme moiety of heme-
containing enzymes such as NOS, COX-2, and cytochrome Arachidonic Acid Metabolites
P450 monooxygenase. HO can attenuate the production of
reactive oxygen species through its heme degradation and Renal prostaglandins, or eicosanoids, are biologically active
production of CO, biliverdin/bilirubin, and free iron. Excess fatty acid products of arachidonic acid that contribute to
free heme catalyzes ROS formation, which may lead to endo- the regulation of renal hemodynamics. They are synthe-
thelial cell dysfunction, vasoconstriction, and tissue damage sized intracellularly and immediately released to act locally
commonly associated with pathologic cardiovascular–renal on the renal vasculature as paracrine/autocrine agents.
conditions. Increased HO-1 activity and the metabolite Free intracellular arachidonic acid can be metabolized on
bilirubin suppress NADPH oxidase activity and O2 produc- demand via one of three major enzymatic pathways: cyclo-
tion in vascular smooth muscle cells.164,172 oxygenase, lipoxygenase, or cytochrome P-450 monooxy-
genase. Depending on cell types, the net production of the
various metabolites may cause vasoconstriction under some
Hydrogen Sul de conditions and vasodilation during others. The multiple
Endogenous hydrogen sul de (H2S) is a recently discov- products of this cascade are depicted in Figure 3.22. Phos-
ered vasoactive gas transmitter (joining NO and CO in this pholipase A2 (PLA2) catalyzes the formation of arachidonic
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1105
05

FIGURE 3.22 Three major pathways of eicosanoid synthesis from arachidonic acid involving cyclooxygenase, lipoxygenase, and cyto-
chrome P-450 monooxygenase enzyme systems. Note that only the con guration of the cyclopentane ring is shown. LT, leukotriene;
PLA2, phospholipase A2; PG, prostaglandin; TX, thromboxane; EET, epoxy-eicosatrienoic acid; HETE, hydroxyeicosatetraenoic acid.

acid, an unsaturated 20-carbon fatty acid, from membrane lipoxygenase. Vasoactive metabolites are also formed via
phospholipids.2 the cytochrome P-450 monooxygenase pathway. Medul-
A key regulatory, rate-limiting step for prostaglandin lary tubular and interstitial cells have a larger synthetic ca-
synthesis is the conversion of arachidonic acid to prostaglan- pacity than the vasculature in the cortex. Endothelial cells
din (PG) G2/H2 by COX. There are two major COX enzymes. produce PGE2 and PGI2, whereas thromboxane A2 seems
COX-1, a constitutive enzyme, is found in renal arteries to derive from vascular smooth muscle cells and mesangial
and arterioles, glomeruli, cortical and medullary collecting cells.118,180–184
ducts, and medullary interstitial cells. COX-2 is a regulated
constitutive enzyme that is important in development, and Prostaglandins
in adult animals is commonly activated by growth factors, General stimuli for renal prostaglandin synthesis are renal
in ammatory states, glucocorticoids, and low salt diet. Renal vasoconstriction and states of volume depletion and hypo-
COX-2 mRNA is localized primarily to epithelial cells of the perfusion. Anesthesia, surgery, and associated stress may
cortical thick ascending limb that includes the macula densa exacerbate prostaglandin production. The diuretics etha-
region and medullary interstitial cells, with greater amounts crynic acid and furosemide also stimulate renal release of
in the papilla than the cortex. COX-2 expression is notice- prostaglandins. Many vasoactive receptor agonists stimulate
ably absent from arterioles, glomeruli, and cortical or med- phospholipases that promote the release of arachidonic acid
ullary collecting ducts, sites of COX-1 expression; COX-2 is from membrane phospholipids. The stimulation of PGE2
also expressed in endothelial cells of medullary vasa recta. and PGI2 production by Ang II is well characterized. In turn,
COX-2 expression in thick ascending limb cells and macula the vasodilatory PGE2 and PGI2 usually buffer the vasocon-
densa cells varies with salt diet, increasing during chronic striction elicited by Ang II and stimulate renin release from
sodium restriction and decreasing during sodium loading. juxtaglomerular cells. Other stimulants include ETA recep-
COX-2 expression in medullary interstitial cells is induced tor agonists. Vasodilators such as acetylcholine and brady-
by water deprivation and high interstitial osmolality, high kinin stimulate the production of PGE2 or PGI2 as well as
levels of Ang II and AVP, as well as growth factors and cyto- NO. In addition, acetylcholine may stimulate the produc-
kines through MAP kinase pathways. Leukotrienes are syn- tion of thromboxane A2. The vasoactive peptides Ang II, ET-
thesized by another major pathway involving the enzyme 1, vasopressin, acetylcholine, and bradykinin increase the
106
10 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

availability of the substrate, arachidonic acid, secondary to protective function and homeostatically balance the hemo-
membrane receptor–mediated Ca2 in ux and the activation dynamic effects of vasoconstrictor substances. Studies of in-
of phospholipase A2. Phospholipase A2 is activated by in- hibition of COX activity indicate that the major function of
creased activity of the Ca2 -calmodulin complex, increased COX-derived prostaglandins is to attenuate the in uence of
production of DAG, or phospholipase C-mediated phos- vasoconstrictor substances during the activation of the re-
phorylation of lipocortin, a membrane-bound enzyme that nin–angiotensin system, the sympathetic nervous systems, or
normally inhibits phospholipase A2. Thus, there is a common both. These counteracting effects provide a balance between
pathway by which many vasoconstrictor agents can increase the vascular effects of Ang II and those of prostaglandins
the production of COX-derived prostaglandins, primarily during variations in plasma volume and sodium intake. As
vasodilatory PGE2 and PGI2, which in turn can counteract is shown in Figure 3.23, blockade of the compensatory dila-
vasoconstriction. Vasodilatory prostaglandins produced by tor action of prostaglandins promotes vasoconstriction when
the endothelium of glomerular arterioles and mesangial the renin–angiotensin and sympathetic nervous systems are
cells exert net effects to stimulate adenylate cyclase and the
formation of cAMP and the activation of protein kinase A.
-Adrenergic neurotransmission can be inhibited prejunc-
tionally and postjunctionally by prostaglandins.71,185,186
Four forms of PGE2 receptors (termed EP1 through EP4)
have been identi ed and cloned. The EP4 receptor, coupled
via G s-proteins to generate cAMP and activate protein ki-
nase A, predominates along the preglomerular vasculature
and mediates the principal vasodilator actions of PGE2. Sim-
ilar actions are exerted by the single IP receptor for PGI2
(prostacyclin). Low concentrations of the prostaglandins
PGE2 and PGI2 normally formed by the arterial vasculature
exert part of their physiologic effects by attenuating the ac-
tions of vasoconstrictors, with larger amounts acting as vaso-
dilators that increase RBF. The buffering action of PGE2 and
PGI2 in the preglomerular vasculature is primarily mediated
by the ability of cAMP and protein kinase A activation to
inhibit IP3-induced release of Ca2 from internal stores. A
low density of a vasoconstrictor receptor (EP1 or EP3) may
counteract some of the net dilation. The EP1 receptor in-
creases Ca2 mobilization. The EP3 receptor inhibits the pro-
duction of cAMP via a pertussis-toxin-sensitive G i protein.
EP2 receptors, which act through a G s protein to increase
cAMP in tubules, appear to be absent from the renal vascu-
lature under normal conditions. Arachidonic acid dilates the
isolated interlobular artery and afferent arteriole; a smaller
response occurs in efferent arterioles. Intrarenal infusions of
arachidonic acid, PGE2 or PGI2 increase RBF and reduce re-
nal resistance without affecting GFR. PGF2 has little or no
effect on the renal circulation. Local administration of small
amounts of PGE2 or its stable analog cause renal vasodila-
tion and attenuate the constrictor effects of Ang II, throm-
boxane A2, and norepinephrine. PGE2 and PGI2 dilate both
afferent and efferent arterioles such that glomerular capil-
lary pressure is constant when the stimulatory effects of the
prostaglandins on Ang II formation are blocked. However,
the application of PGE2 from the interstitial side causes va- FIGURE 3.23 Renal plasma ow (RPF), glomerular ltration rate
soconstriction of juxtamedullary nephrons, a response due (GFR), cortical blood ow (CBF), and medullary blood ow (MBF)
to subsequent metabolism to a vasoconstrictor agent or the responses to the inhibition of cyclooxygenase (Cox-2, nimesu-
activation of EP1 or EP3 receptors.60,185,187,188 lide) in anesthetized rats maintained on a normal salt diet ( )
Endogenous prostaglandins regulate RBF and GFR or on a low sodium diet ( ). * P .05. (Data from Green T,
by direct effects on vascular smooth muscle and indirectly Rodriguez J, Navar LG. Augmented cyclooxygenase-2 effects on
by modi cation of the action of other hormones or neural renal function during varying states of angiotensin II.
stimuli. The vasodilator prostaglandins serve an important Am J Physiol Renal Physiol. 2010;299:F954–F962.)
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1107
07

stimulated, such as during sodium depletion. Prostaglandins may be mediated by thromboxane A2 in pathophysiologic
also antagonize the tendency for high concentrations of va- settings. Administration of a stable thromboxane-receptor
sopressin to produce renal vasoconstriction. In contrast, in agonist reduces RBF and GFR by causing afferent and ef-
a healthy, unstressed individual with a normal plasma vol- ferent arteriolar constriction. Kf appears to be unaffected,
ume, the inhibition of prostaglandin synthesis has little or although it is noteworthy that thromboxane A2 causes the
no effect on RBF and GFR (Fig. 3.23). Basic autoregulatory contraction of isolated glomeruli and cultured mesangial
ef ciency of whole-kidney RBF and GFR remains high even cells. As mentioned in the section on TGF, although very
during the blockade of renal prostaglandin synthesis via the little thromboxane A2 is produced under normal condi-
COX pathway. The inhibition of COX-2 produces renal va- tions, thromboxane plays a modulatory role in the preglo-
soconstriction that may be greater in the medulla than in merular vasoconstriction associated with the activation of
the cortex. During afferent arteriolar constriction induced by the TGF mechanism and the vasoconstrictor response to
TGF, COX-2 generates vasodilatory metabolites in response chronic Ang II infusion. The thromboxane A2-PGH2 recep-
to increased nNOS activity that attenuate the strength of tor is coupled by the G q protein to the activation of phos-
feedback-mediated vasoconstriction.2,116,182 pholipase C, increased [Ca2 ]i, and possible inhibition of
Renin release is increased by arachidonic acid, endoper- adenylate cyclase. The renal vasoconstriction produced
oxides, PGE2, and PGI2. PGE2 and PGI2 act on their respec- by thromboxane A2 is primarily mediated by Ca2 in ux.
tive EP4 and IP receptors on juxtaglomerular granular cells Chronic activation of PKC appears to reduce the number of
to stimulate renin release by stimulating adenylate cyclase thromboxane A2 TP receptors. In Ang II–dependent hyper-
activity and producing cAMP. COX inhibitors reduce renin tension, the administration of COX-2 inhibitors decreases
release under basal as well as during stimulated conditions. RBF and GFR due to increased RVR, re ecting a renal va-
-Adrenergic neurotransmission can be inhibited prejunc- sodilator action of intrarenal COX-2 metabolites; however,
tionally and postjunctionally by prostaglandins.13,116,121,133 systemic vascular resistance is decreased, re ecting a va-
The COX-2 metabolite PGE2 plays a key role in linking soconstrictor role of COX-2 metabolites on the systemic
NaCl control of macula densa signaling to juxtaglomerular circulation.116,184,190
cells to regulate renin release and to afferent arterioles to reg-
ulate TGF. COX-2 activity in the macula densa increases in Leukotrienes
response to a salt de cient diet and to a loop diuretic, such Lipoxygenase enzymes convert arachidonic acid to leuko-
as furosemide, with increases in PGE2 production and renin trienes (LTs), HETEs, and lipoxins (see Fig. 3.22). The ma-
release. nNOS in the same cells may serve as an upstream jor lipoxygenase products are monohydroxyeicosatetraenoic
intermediate responding to salt transport. High levels of acids—12-HETE and 15-HETE—generated by glomeruli,
nNOS and COX-2 parallel each other, and the inhibition of mesangial cells, renal cortical tubules, and vascular tissues.
nNOS causes a fall in COX-2 expression and an uncoupling LTs (LTB4, LTC4, LTD4, and LTE4) are hydroperoxy fatty acid
to transport. On the other hand, high renin states suppress products of the intermediate 5-hydroperoxyeicosatetraenoic
COX-2 expression in the cortical thick ascending limb and acid (HPETE). Stereoisomers of 12-HETE are produced by
macula densa cells via Ang II and AT1 receptors; ACE in- different enzymatic pathways. The 12(S)-HETE isomer is
hibition has the opposite effect. The recruitment of renin- predominantly produced in the cortex; however, the 12(R)-
containing cells retrograde along the afferent arteriole dur- HETE is a primary biologically active metabolite of the cy-
ing chronic salt restriction or ACE inhibition appears to be tochrome P-450 pathway. The renal vasculature constricts
COX-2 dependent as renin is restricted to juxtaglomerular in response to 12(S)-HETE and 15-HETE due to the depo-
cells during these stimuli in COX-2–de cient mice. Plasma larization of smooth muscle cells, increased Ca2 entry, and
renin activity is reduced in COX-2–null mice and also in the activation of PKC. Vascular generation of 12-HETE is
nNOS-de cient mice. The inhibition of COX-2 produces increased in pathologic conditions associated with oxidative
renal vasoconstriction that may be greater in the medulla stress and in ammation.2,180,183,191
than the cortex.117,182,189 Receptors for LTC4 and LTD4 have been identi ed on
In disease states, endogenous vasodilator prostaglan- preglomerular arteries, glomeruli and cultured mesangial
dins serve a protective role in maintaining renal function cells, and efferent arteriole. The infusion of LTC4 or LTD4
by means of their effects on vascular resistance, GFR, and causes renal vasoconstriction, reduces GFR and ltration
renin release. The administration of nonsteroidal anti- fraction, and activates the renin–angiotensin system. Leu-
in ammatory drugs to patients with clinical disorders kotriene C4 receptors are linked to ion channels, but specif-
such as advanced hepatic cirrhosis, severe congestive heart ic mechanisms are not known. Some of the LT actions may
failure, and sodium depletion often produces deleteri- be mediated by endothelial cells. LTD4 is known to release
ous effects with reductions in renal perfusion and GFR. NO in addition to activating a pertussis-toxin–sensitive G
In other pathophysiologic conditions, there may be en- protein. Lipoxin A4 increases renal plasma ow and GFR,
hanced production of the vasoconstrictor thromboxane A2, with a small reduction in Kf. Interestingly, the effects are
which may contribute to the deterioration of renal func- COX dependent and can be completely reversed by the in-
tion. Some of the vasoconstriction produced by Ang II hibition of this enzyme. On the other hand, the ability of
108
10 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

lipoxin B4 to decrease RBF and GFR is independent of COX Isoprostanes such as 8-epi-PGF2 are vasoconstrictor
activity.180,183 metabolites related to prostaglandins. They are stable prod-
ucts of nonenzymatic lipid peroxidation of arachidonic acid,
Cytochrome P-450 Metabolites formed in and released from cell membranes and excreted
Vasoactive metabolites of the cytochrome P-450 pathway in urine. Isoprostane production is stimulated by peroxyni-
are produced by vascular smooth muscle cells, endothelial trate, a reactive oxygen species resulting from NO scaveng-
cells, renal tubular cells, and glomeruli. Arachidonic acid is ing by superoxide anion.114,198,199
oxygenated via NADPH-dependent microsomal monooxy-
genases. As is shown in Figure 3.22, epoxygenase enzymes Kallikrein–Kinin System
are responsible for the production of epoxides or epoxyeico- Plasma and glandular/tissue kallikreins are distinct ser-
satrienoic acids (EETs) (e.g., 11,12-EET and 14,15-EET). A ine protease enzymes acting on kininogens (inactive 2-
second cytochrome P-450 pathway involves the -hydrox- glycoproteins) to form the biologically active nonapeptide
ylase formation of 19- and 20-HETEs. The major metabo- bradykinin and also the decapeptide lysyl-bradykinin
lites of this pathway are EETs in the cortex and HETE in the (kallidin). It is unlikely that circulating kinins affect the
medulla. Salt diet, Ang II and other hormones, and various renal microcirculation because they are rapidly inacti-
pathophysiologic settings alter the renal cytochrome P-450 vated enzymatically by endothelial-bound kininase II
metabolism.118,183,192–195 (ACE) and neutral endopeptidase. Within the kidney,
Cytochrome P-450 -hydroxylase metabolites, in par- tissue kallikrein and its substrate, kininogen, are located
ticular 20-HETE, participate in regulation of cortical and predominantly in the distal convoluted and cortical col-
whole-kidney blood ow. Although rodent studies report lecting tubules. The synthesis and the release of kallikrein
effects of 20-HETE, studies in dogs and rabbits have failed into the tubular uid and interstitium are stimulated by
to show an effect of cytochrome P-450 inhibition on au- prostaglandins, mineralocorticoids, Ang II, increased renal
toregulation and pressure natriuresis. In isolated rat ves- perfusion pressure, and several diuretic drugs. The renal
sels, transmural pressure stimulates -hydroxylase activity vasculature and tubules contain the constitutive B2 recep-
to produce 20-HETE, which is thought to participate in tor and the inducible B1 receptor. Bradykinin B2 receptors
myogenic vasoconstriction. Cytochrome P450-4A–derived on vascular endothelial cells cause renal vasodilation as a
eicosanoids may also participate in the renal hemodynamic result of stimulation of NO, EET, and prostanoid produc-
effects of Ang II and endothelin. Products of the cytochrome tion. The B2 receptor density in glomeruli is reduced by
P-450 pathway potentiate control of preglomerular vaso- a low sodium diet and water deprivation. B2 receptors on
motor tone by the juxtaglomerular apparatus. Inhibition of tubular epithelial cells inhibit sodium reabsorption. The
P-450 metabolites blunts TGF activity, whereas the luminal bradykinin B1 receptor is not normally expressed and is si-
perfusion of 20-HETE restores TGF responses. 20-HETE lent under physiologic conditions. Its expression is highly
causes vasoconstriction by the inhibition of tonically active inducible by in ammatory mediators and tissue damage,
K channels, thereby causing depolarization and activation chronic ACE (kininase II) inhibition, or genetic deletion of
of voltage-gated Ca2 channels and increases intracellular B2 receptors.200–203
Ca2 .118,181,183 The infusion of bradykinin elicits renal vasodilation
Epoxygenase metabolites elicit variable vascular re- characterized by a larger increase in RBF than GFR, and a
sponses. 11,12-EET is an endothelial-derived factor dis- natriuresis and diuresis. Exogenous kinins also produce an
tinct from NO that produces vasodilation in interlobular independent stimulation of prostaglandin formation (PGE2
arteries and afferent arterioles mediated by membrane and PGI2) and renin release. Kinin-induced vasodilation,
hyperpolarization following the activation of cAMP/ however, may be similar in the presence and absence of COX
protein kinase A and of K channels, actions independent inhibition of prostaglandin synthesis owing to major ac-
of the vascular endothelium or COX activity. 5,6-EET or tions of NO and EETs. As a result of vasodilatorlike actions,
8,9-EET induces vasoconstriction with a decrease in GFR, exogenous bradykinin reduces the vasoconstrictor responses
mediated in part by thromboxane TP receptor activation. to Ang II and norepinephrine. The observed decline in glo-
However, COX inhibition changes the renal response to merular Kf contrasts with the well-known effect of kinins to
these EETs to vasodilation and an increase in GFR. In- increase capillary permeability in other tissues. The mecha-
hibition of epoxygenase activity enhances afferent arte- nisms responsible for the reduction in Kf are not known.
riolar autoregulation. Not clear is whether epoxygenase/ Additional effects include the enhanced conversion of inac-
EET actions blunt the pressure-induced myogenic re- tive to active renin and the presynaptic inhibition of adren-
sponse, or TGF, or both. EETs, in particular 11,12-EET, ergic neurotransmitter release. Isolated vessels and cultured
have now been identi ed as being an endothelial-derived mesangial cells devoid of endothelium exhibit B2-receptor–
hyperpolarizing factor and activate potassium channels, dependent constrictor responses to bradykinin. Signal trans-
thus increasing the membrane potential, leading to renal duction appears to involve a pertussis-toxin–insensitive G
vasodilation and attenuated responses to vasoconstrictor protein, a PKC pathway, increased [Ca2 ]i, and arachidonic
in uences.183,196,197 acid metabolites.1,2
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1109
09

B2 receptors reside on endothelial and vascular smooth Animal studies on the role of the renal kallikrein–
muscle cells. Vasodilator effects of bradykinin are observed kinin system using kininogen-de cient rats and also B2
in isolated preparations of afferent and efferent arterioles. receptor knockout mice indicate that this system primar-
In the isolated perfused afferent arteriole, low concen- ily functions to promote a natriuresis that impacts on the
trations of bradykinin ( 10 10 M) vasodilate by acting pressure–natriuresis relation when there is excess sodium
on endothelial B2 receptors to produce NO and prosta- intake or high plasma aldosterone concentration. Genetic
glandins. Epoxygenase-dependent EETs also contribute. dysfunction of the renal kallikrein–kinin system leads to
Higher concentrations ( 10 9 M) vasoconstrict by acting altered functional maturation of the kidney and the devel-
on B2 receptors to produce COX-derived thromboxane. In opment of salt-sensitive hypertension. Rats with a genetic
contrast, the vasodilator effect of bradykinin in efferent reduction in urinary kallikrein excretion have an altered
arterioles (perfused in retrograde direction) via B2 recep- pressure–natriuresis relationship, with this defect being cor-
tors is mediated by cytochrome P450 metabolites (prob- rected by an infusion of puri ed tissue kallikrein. Knockout
ably EETs), but not by NO or COX products. Perfusion mice lacking the B2-receptor gene have elevated arterial pres-
of bradykinin through glomeruli releases prostaglandins sure under basal conditions, reduced RBF, increased renin
that dilate the efferent arteriole. Bradykinin relaxes peri- mRNA, and enhanced blood pressure sensitivity to salt.210
cytes surrounding the outer medullary descending vasa
recta. Endogenous bradykinin dilates afferent and effer- Purinergic Actions on Renal Microcirculation
ent arterioles in vivo to a greater extent in the deep versus
Adenosine and ATP
super cial cortical glomeruli with primary mediation by
NO. EETs also participate in vasodilation of afferent ar- Adenosine nucleosides and nucleotides have received con-
terioles of juxtamedullary nephrons. Isolated vessels and siderable attention as regulators of renal hemodynamics
cultured mesangial cells devoid of endothelium exhibit B2- and renin release. It is proposed that GFR and ltered so-
receptor–dependent constrictor responses to bradykinin. dium load are coupled to tubular transport capacity and O2
Signal transduction appears to involve a pertussis-toxin– consumption via the hydrolysis of ATP and the resultant
insensitive G protein, a PKC pathway, increased [Ca2 ]i, adenosine production. Adenosine and other adenine nucleo-
and arachidonic acid metabolites. Endogenous bradykinin, tides are secreted into the interstitial uid in the extracellular
potentiated during ACE inhibition in animals fed a low so- compartment. ATP is released from cells through membrane
dium diet, dilates the renal vasculature with predominant channels and coreleased with transmitters from nerve ter-
actions in the medulla. During extracellular uid volume minals. In this fashion, purine-based substances act as para-
expansion, bradykinin promotes sodium excretion and re- crine agents to in uence renal microcirculation.1,2,121
duces regional autoregulatory ef ciency to increase med-
ullary blood ow, effects largely mediated by B2 receptor Purinergic Receptors
stimulation of NO.15,204–209 Renal purinoceptors display different sensitivities for
Early studies evaluated endogenous kinin activity us- ATP, ADP, AMP, and adenosine. P1 purinoceptors respond
ing an infusion of bradykinin-binding antibodies, the sup- primarily to adenosine and sometimes to AMP, but are
pression of renal kallikrein activity with the serine protease relatively insensitive to ADP and ATP. Extracellular ATP
inhibitor aprotinin, and the pharmacologic inhibition of predominantly activates P2 purinoceptors. There are at least
kininase II. The results suggest that locally formed kinins two subtypes of adenosine-responsive P1 receptors in the
attenuate renal vasoconstriction. Further understand- renal vasculature. P1-vasoconstrictor A1 receptors are cou-
ing has been gained by employing more speci c receptor pled to a pertussis-toxin–sensitive G i protein that inhibits
antagonists. Use of a speci c B2 receptor antagonist reveals adenylate cyclase and cAMP production. The stimulation of
that basal kinin levels do not contribute appreciably to the A1 receptors decreases GFR and RBF. A local application of
regulation of renal function during normal conditions. How- an A1 receptor agonist constricts both preglomerular and
ever, when their levels are elevated as during sodium restric- postglomerular microvessels. A2 receptors cause vasodila-
tion and volume depletion, kinins act as vasodilators in a tion of afferent and efferent arterioles, stimulating adenylate
manner similar to the prostanoids, which buffer the renal cyclase through a G s protein, and also increase EETs and
vasoconstriction associated with elevated local levels of Ang NO, which contribute to the vasodilation. There are two
II, norepinephrine, and vasopressin. A combined blockade subtypes (A2a and A2b), with A2b being prevalent in afferent
of both degrading enzymes (ACE or kininase II and neu- arterioles.
tral endopeptidase) leads to increases in RBF and GFR in P2 receptors, present on endothelial and vascular
association with an increased urinary excretion of kinins. smooth muscle cells, have a greater af nity for ATP and ADP
Kinins may participate in the autoregulation of GFR and the than for adenosine or AMP. Interstitial uid ATP concentra-
TGF mechanism, although overall steady-state RBF auto- tions are suf ciently high to play a role in regulating the vas-
regulation is not affected by kinin receptor blockade. Kinins cular resistance changes responsible for autoregulation and
exert larger vasodilatory effects in the medulla than in the TGF. The receptor subtype P2X increases [Ca2 ]i by increas-
cortex.208 ing Ca2 in ux through voltage-gated and receptor-operated
110
11 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Ca2 channels. The low frequency stimulation of renal the myogenic response of the preglomerular vasculature. In
sympathetic nerves causes renal vasoconstriction that is me- addition, mice lacking the enzyme NTPDase/CD39, which
diated, in part, by a nonadrenergic component involving reduces the formation of adenosine, have impaired TGF
ATP corelease with the neurotransmitter acting on P2X recep- activity.1,104,213–217
tors. P2Y receptor activation increases [Ca2 ]i via the PLC-IP3 Although adenosine and Ang II can act independently
cascade. Endothelial cells have a high concentration of P2Y of each other on glomerular arterioles, they also act syner-
receptors, and their activation by ATP leads to vasorelaxation gistically. Adenosine enhances afferent arteriolar reactivity
mediated by NO, or PGI2, or both. However, ATP causes to Ang II by increasing Ca2 sensitivity. On the other hand,
a rapid, marked constriction of the afferent arteriole when Ang II ampli es afferent arteriolar constriction to adenosine
NO synthesis is inhibited. The P2U purinoceptor is termed by a different mechanism, one due to increased [Ca2 ]i. In
a nucleotide or 5 -nucleotide receptor because it responds this manner, adenosine can amplify vasoconstriction during
to all nucleotides with a similar potency of ATP and uridine high renin states and increased endogenous Ang II. Adeno-
triphosphate. P2U receptor activation increases [Ca2 ]i by a sine causes larger long-term decreases in GFR and the l-
G-protein stimulating the PLC pathway.1,2,211,212 tration fraction in sodium-depleted animals with increased
renin-angiotensin activity than in animals consuming a nor-
Adenosine mal salt diet.218,219
The intrarenal administration of adenosine produces a bi- Adenosine participates in the regulation of renin secre-
phasic renal response. Resistance vessels respond initially tion. Adenosine infusion inhibits renin release in vivo. The
with transient vasoconstriction, mediated by P1-A1 recep- mechanism involves adenosine receptors on juxtaglomerular
tors, followed by gradual vasodilation, mediated in part cells, with A1 and A2 receptors having opposite effects. Renin
by P1-A2 receptors. In isolated preparations, adenosine A1 release is inhibited by A1 purinergic receptors coupled to a
receptor stimulation constricts both afferent and efferent G i protein and stimulated by A2 receptors. A tonic inhibi-
arterioles. Endogenous adenosine serves as an important tory effect of adenosine is observed in isolated afferent arte-
regulator of renal hemodynamics. The blockade of A1 recep- rioles with and without macula densa cells attached.220–222
tors decreases afferent arteriolar resistance and Kf in anes-
thetized animals. ATP
Evidence about the importance of adenosine in intrinsic P2 receptors are also involved in mediating autoregulatory
autoregulatory mechanisms is mixed. One set of results sug- adjustments in RVR involving TGF responses. Extracellular
gests that adenosine is not essential for autoregulation. Inter- ATP, at high levels that saturate P2 purinoceptors, causes re-
stitial uid concentrations of adenosine are unchanged over nal vasoconstriction and impairs the autoregulatory ability of
the autoregulatory range of arterial pressure, and the admin- the vasculature to dilate in response to decreases in arterial
istration of adenosine-receptor antagonists does not impair pressure. The effect on autoregulation appears to be speci c
autoregulatory capability. Other results, however, implicate a to ATP because similar studies show that RBF autoregulation
role of adenosine in the regulation of afferent arteriolar tone is not impaired when the vasoconstrictor is norepinephrine
by TGF. Salt transport by the ascending limb of the Henle or Ang II.
loop or macula densa cells requires metabolic energy and The renal arterial infusion of ATP increases RBF. The
the use of ATP that is linked to control of preglomerular vas- renal vasodilation is mediated by activation of endothe-
cular resistance through macula densa signaling. As trans- lial receptors to release NO. The ATP effect is converted
port increases, more adenosine is liberated within cells and to vasoconstriction during NO synthesis inhibition. P2 re-
is available to diffuse to the afferent arteriole, where it elicits ceptors are present on vascular endothelium, with P2Y re-
vasoconstriction to reduce RBF and GFR. Luminal perfu- ceptors being responsible for NO-dependent vasodilation.
sion of A1 adenosine-receptor agonists cause TGF-induced P2X receptors on vascular smooth muscle cells produce va-
vasoconstrictor changes in glomerular capillary pressure. soconstriction. In considering a paracrine role for ATP, it
Attenuated TGF-mediated responses of the afferent arteri- seems likely that endogenous ATP reaches vascular smooth
ole are observed when an adenosine antagonist is added to muscle cells from the interstitium. The vasoconstriction is
either the luminal uid or peritubular capillary blood. The mediated by Ca2 mobilization and entry through L-type
effects are insensitive to the salt transport inhibitor furose- Ca2 channels.1,2
mide, indicating P1-A1 adenosine-receptor–mediated effects ATP acts on P2 purinoceptors to selectively constrict
by acting on either the macula densa or vascular smooth afferent arterioles in a sustained manner, whereas ATP does
muscle cells. Moreover, an adenosine-receptor blockade not elicit any response in efferent arterioles. This is con-
reduces TGF control of preglomerular resistance, and mice sistent with autoradiographic and immunohistochemical
without functional A1 receptors lack TGF activity. However, evidence of abundant P2x receptors limited to the preglo-
unopposed A2 receptor activation may result in substantial merular vasculature. Pressure-induced TGF–mediated af-
vasodilation. Dynamic RBF studies on gene-targeted mice ferent arteriolar constriction is prevented by P2 receptor
are consistent with A1 receptors mediating pressure-induced saturation or desensitization by high arterial concentrations
TGF responses participating in RBF autoregulation but not of ATP. Tubuloglomerular responses also are markedly
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1111
11

blunted during peritubular capillary perfusion with satu- abolished by 1-adrenergic receptor antagonists and attenu-
rating doses of ATP or slowly metabolizable analogs. P2(x)1 ated by NPY Y1 receptor blockade. Very low levels of nerve
receptor blockade prevents TGF-initiated responses of af- stimulation affect renin release and tubular sodium reabsorp-
ferent arterioles. tion without causing major changes in renal hemodynamics.
Renal interstitial uid concentrations of ATP measured Intermediate nerve activity elicits renal vasoconstrictor
by microdialysis are closely associated with autoregulatory responses in the medullary as well as the cortical circula-
and TGF-mediated changes in RVR. A direct relationship tion, with more pronounced reductions in cortical blood
is observed when TGF is stimulated by increased distal ow. The increased vascular resistance in the cortex is due to
uid delivery due to a carbonic anhydrase inhibition of the constriction of preglomerular and efferent arteriolar seg-
proximal tubular reabsorption and when TGF is inhibited ments. As is shown in Figure 3.24, low levels of stimulation
by the transport inhibitor furosemide. Mice lacking func- cause an equivalent constriction of afferent and efferent ar-
tional P2x receptors have partially blunted whole kidney terioles such that glomerular pressure is unchanged. Similar
RBF autoregulation and a loss of TGF. Macula densa cells results are obtained in isolated afferent and efferent arterioles
secrete ATP through a basolateral maxi-chloride channel when 1-adrenoceptors are stimulated. Higher frequencies
in response to increases in luminal NaCl concentration. of nerve stimulation produce predominant constriction of
Although macula densa cells have abundant mitochondria, preglomerular vessels and thus reduce glomerular capillary
they have low levels of Na -K -ATPase, making them a pressure and GFR. Intense stimulation at 10 Hz produces
good candidate for a source of extracellular ATP. The ac- glomerular ischemia.226
tivation of macula densa signals trigger the rapid propa- The renal medullary circulation is less sensitive than
gation of Ca2 waves and associated afferent arteriolar the cortical vasculature to renal nerve stimulation, par-
constriction, which are prevented by blocking P2 receptor ticularly at low stimulus intensities. This is largely due to
activation or increasing ATP hydrolysis. Collectively, the more effective blunting of vasoconstriction by NO and eico-
data support an important role for the ATP-dependent ac- sanoids. The electrical stimulation of renal nerves causes
tivation of P2 receptors in the mediation of autoregulation parallel reductions in RBF and cortical blood ow, whereas
and TGF.1,2,12,107,211,223,224 the decrease in medullary perfusion is approximately 50%
less. Low frequency nerve stimulation ( 2 Hz) increases
NO production, which buffers the vasoconstriction in all
Sympathetic Nervous System and regions of the kidney. More severe renal vasoconstriction
Catecholamines produced by high frequency nerve stimulation ( 4 Hz)
The renal vasculature is richly innervated with postgangli- leads to the activation of AT1 receptors that magni es nerve-
onic adrenergic bers originating from sympathetic ce- induced constriction in both the renal cortex and the me-
liac and aorticorenal plexuses that receive inputs from the dulla. NO derived from nNOS is critical to protecting the
sixth thoracic through the second lumbar spinal nerves. medullary regions.227,228
All arterial segments of the renal vasculature and the large Infusions of norepinephrine, epinephrine, or 1-
veins are extensively innervated with neuroeffector junctions adrenergic agonists produce similar dose-related vasocon-
containing norepinephrine as the primary neurotransmitter. strictor effects on the renal microcirculation. Studies on
A heavy concentration appears in subadventitial layers of ar- individual vessels indicate that norepinephrine constricts
cuate arteries, with notable innervation of smooth muscle the interlobular artery and the afferent and efferent arte-
cells of both the afferent and efferent arterioles as well as rioles. Total RVR responses to norepinephrine are sub-
the juxtaglomerular granular cells and the outer medullary stantially attenuated by Ca2 channel blockers and by the
descending vasa recta. The incoming efferent innervation is inhibition of IP3-mediated release of stored Ca2 . Ca2 re-
predominantly adrenergic, although some nerve endings are sponses of the afferent arteriole to -adrenoceptor stimula-
reported to contain dopamine and neuropeptide Y (NPY) as tion are mediated by the mobilization from internal stores in
well as ATP. Alpha1-adrenoceptors, primarily 1A and 1D, in- combination with Ca2 entry through voltage-gated L-type
crease [Ca2 ]i and Ca2 sensitivity, and mediate nerve stim- channels and channels insensitive to dihydropyridine Ca2
ulation-induced vasoconstriction of glomerular arterioles in channel blockers. The latter includes voltage-independent
the renal cortex and medullary pericytes, an action opposed receptor-operated Ca2 channels. Ca2 sensitivity is also
in part by concurrent activation of 2-adrenoceptors. Beta- increased. Recent evidence implicates superoxide anion
adrenoceptors stimulate renin release from juxtaglomerular generation and downstream signaling involving cyclic ADP
granular cells via cAMP/PKA signaling.132,225 ribose, ryanodine receptors on the sarcoplasmic reticulum,
Renal nerves are divided into two types. Type I almost and Ca2 -induced Ca2 release. Efferent arteriolar responses
exclusively innervate afferent arterioles, whereas type II in- to norepinephrine appear to be independent of L-type
nervate both afferent and efferent arterioles. Type II nerves channels.20,58,143
contain NPY, whereas type I terminals do not. The electrical The renal nerves are not essential for ef cient steady-state
stimulation of the greater splanchnic or renal efferent nerves autoregulation of RBF and GFR or for the operation of either
produces frequency-dependent renal vasoconstriction that is the myogenic or the TGF mechanism. These conclusions are
112
11 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 3.24 The effect of electrical stimulation of


renal efferent nerves at 1 to 5 Hz on a single nephron
glomerular ltration rate (GFR), plasma ow, and the re-
sistance of preglomerular vessels and efferent arterioles.

reinforced by a frequency analysis of RBF dynamics indicat- associated with an increase in PCO2 from 25 to 70 mm Hg
ing that barore ex-mediated changes in renal sympathetic activates the renal efferent nerves but the neurally mediated
nerve activity and acute denervation have little impact on renal vasoconstriction is partially counteracted by enhanced
intrinsic autoregulatory mechanisms stabilizing renal perfu- synthesis of vasodilatory prostaglandins. Neurally induced
sion in normotensive animals. Under unstressed conditions, vasoconstriction is more readily demonstrable during more
while basal efferent sympathetic tone is low, neither acute stressful states, such as during dehydration, blood-volume
nor chronic renal denervation affects RBF or GFR. Physio- contraction, hemorrhage, and congestive heart failure.
logic changes in renal efferent nerve activity during sleeping, The pronounced activation of renal efferent nerve activity
grooming, and movement cause inverse changes in RBF in ( 500%) produced by auditory or emotional stimuli causes
conscious animals. In anesthetized animals, moderate hy- intense renal vasoconstriction.1,132,229
poxia and re exively induced increases in renal sympathetic In addition to a direct effect on vascular smooth muscle,
nerve activity reduce RBF more than GFR with increases in the renal efferent nerves exert secondary hemodynamic ef-
glomerular capillary pressure as a result of greater constric- fects as a consequence of an intrarenal stimulation of the
tion of efferent over afferent arterioles. More severe hypoxia constrictors Ang II and ET-1 and the formation of dilator
reduces RBF and GFR equally as afferent and efferent arte- prostaglandins and NO. Renal nerve stimulation elicits an
riolar resistances were increased to the same extent. Moder- increase in renin release from juxtaglomerular cells primar-
ate increases of about 50% in renal nerve activity that are ily by the activation of 1-adrenoceptors signaling through
induced re exively by high- or low-pressure receptors fail to the cAMP/PKA pathway. Prostaglandin synthesis is enhanced
alter renal hemodynamics, although this level of stimulation by the activation of phospholipase A2 and the augmented
is capable of affecting tubular sodium reabsorption and renal availability of arachidonic acid. Ang II formation accentu-
release of renin and prostaglandins. Hypercapnic acidosis ates norepinephrine release and renal vasoconstriction
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1113
13

produced by increased renal nerve activity. 1-Adrenocep- mediated by a combination of NO as well as prostanoids
tor activation enhances Ang II–mediated afferent arteriolar and EETs.132
vasoconstriction by increasing calcium sensitivity. The vaso- Dopamine, a sympathomimetic amine precursor of nor-
constriction caused by moderate renal nerve stimulation is epinephrine, is a neurotransmitter capable of regulating re-
reduced in the presence of the Ang II/AT1 receptor blockade, nal hemodynamics, renin secretion, and sodium excretion.
suggesting that part of the effect is mediated by increased Histo uorescent evidence suggests dopaminergic innerva-
intrarenal Ang II formation due to the neural stimulation of tion of the cortical vessels, primarily the glomerular vascu-
renin. On the other hand, vasodilatory prostaglandins buf- lar poles. However, dopamine synthesized from L-dopa via
fer a signi cant fraction of the vasoconstriction elicited by L-dopa decarboxylase in proximal tubular cells is clearly the
nerve stimulation as evidenced by much larger changes in major source of urinary dopamine and its metabolites, with
GFR, RBF, and vascular resistance during COX inhibition. the greatest production during high salt diet and lowest syn-
Moreover, endothelium-dependent NO release, possibly via thesis during sodium restriction. There are two major recep-
the activation of 3-adrenergic receptors, functions as an in- tor families: D1-like (D1 and D5 receptors) and D2-like (D2,
hibitory modulator of vasoconstrictor responses to the sym- D3, D4 receptors). D1 receptors are postsynaptic receptors
pathetic transmitters norepinephrine and NPY. The effect of located on vascular smooth muscle and tubular cells, but are
renal nerve stimulation on glomerular Kf is controversial. absent from glomeruli. They cause vasodilation by cAMP/
Some investigators report a reduction; others nd that Kf is PKA signaling. D2 receptors are either presynaptic or post-
normally unaffected by nerve stimulation but that a reduc- synaptic and are located in glomeruli as well as vessels and
tion of Kf is evident during the inhibition of prostaglandin tubules in both the cortex and medulla. The presynaptic D2
synthesis.2 receptor indirectly dilates vessels by the inhibition of norepi-
Afferent renal nerves serve important functions in the nephrine release. Dopamine is commonly used to evaluate
neurohumoral control of arterial pressure, vasopressin re- the renal functional reserve—the ability to dilate the renal
lease, and renal excretion of sodium and water. Afferent nerve vasculature—in various pathologic conditions.232–236
endings contain neuropeptides such as calcitonin gene-relat- Although receptor blockers abolish renal effects pro-
ed peptide, substance P, and vasoactive intestinal peptide. duced by exogenous dopamine, such antagonists do not con-
Afferent input from intrarenal sensory receptors in the pelvic sistently affect basal renal hemodynamics, questioning the
wall participates in renorenal re exes that modulate effer- role of physiologic levels of dopamine. Further, the prejunc-
ent nerve activity to the contralateral kidney. Baroreceptors tional stimulation of dopamine receptors during moderate
or mechanoreceptors are activated by high venous or inter- levels of renal efferent nerve activity has little in uence on
stitial pressure. High renal pelvic pressure reduces efferent RVR. Small amounts of exogenous dopamine or a D1 recep-
renal nerve traf c to the opposite kidney, which results in tor agonist dilate the renal vasculature. Responses include
compensatory increases in RBF, GFR, and sodium excretion. an increase in RBF and a decrease in ltration fraction; GFR
Similar functional changes in the opposite kidney are elic- and glomerular capillary pressure are unaffected. Most of
ited by ischemia-sensitive chemoreceptors responding to the dopamine-induced renal vasodilation is mediated by D1
increased urinary potassium or very high sodium concentra- receptors, which are coupled to cAMP/PKA signaling. Do-
tion. Outgoing afferent nerve activity is in uenced also by pamine and D1 receptor agonists relax afferent and efferent
incoming efferent nerve activity such that increased efferent arterioles and interlobular and arcuate arteries. D1 receptor
input usually increases afferent nerve activity. The norepi- stimulation attenuates the constriction of both afferent and
nephrine release from efferent nerves activates adrenocep- efferent arterioles produced by either Ang II or ET-1. The
tors on sensory nerves, with 1-adrenoceptor activation D1-induced dilation of the preglomerular vasculature does
increasing afferent nerve activity and 2 receptor stimulation not impair autoregulatory adjustments in vascular resistance
reducing afferent ring.230,231 to changes in perfusion pressure. D1-receptor stimulation
Although cholinesterase-containing bers and - attenuates tubuloglomerular feedback responsiveness. The
adrenergic sites have been identi ed histologically in the vasodilation elicited by dopamine is more pronounced after
renal cortex, there is little functional evidence for neu- denervation or pharmacologic blockade of -adrenoceptors,
rogenic renal vasodilation mediated by acetylcholine or suggesting that presynaptic D2-dopamine receptors augment
-adrenoceptors. The neural release of acetylcholine, how- norepinephrine release from nerve terminals. D2-receptor
ever, may exert a small indirect effect at presynaptic sites stimulation is associated with increases in GFR and the at-
to inhibit norepinephrine release. Infusions of exogenous tenuation of Ang II–induced contraction of glomerular me-
acetylcholine increase RBF and reduce RVR, whereas GFR is sangial cells. D1 receptors also stimulate a renin release from
unaffected. The decline in total resistance is due to parallel juxtaglomerular cells. D1 agonists inhibit uid and electro-
reductions in afferent and efferent arteriolar resistance. Ace- lyte transport indirectly via hemodynamic mechanisms and
tylcholine relaxes isolated preparations of the interlobular directly by the occupation of D1 receptors in the proximal
artery and the afferent and efferent arterioles. As pointed tubule, the thick ascending limb of Henle, and the collecting
out earlier, however, the full vasodilatory effect of acetyl- duct. D1 receptor null mice have elevated proximal tubular
choline requires an intact endothelium and is primarily salt transport and hypertension.2,132
114
11 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

NPY is another neurotransmitter. Nerve endings con- inhibit tubular reabsorption without altering RBF, GFR, or
taining immunoreactive neuropeptides are localized along the ltered sodium load. Medullary blood ow is increased
the interlobular and arcuate arteries extending to glomerular during the administration of ANP, but the effect seems to
arterioles, innervating both afferent and efferent arterioles. be secondary to the ANP-induced natriuresis. Several tu-
NPY is a cotransmitter of the renal sympathetic nerves that is bular sites have been evaluated, including a Na-ATPase
coreleased with and may potentiate the vascular pressor ef- of the proximal tubule and an amiloride-sensitive sodium
fects of norepinephrine and ATP. NPYis less potent than nor- channel in the collecting duct and an angiotensin-sensitive
epinephrine. The kidney expresses NPY receptors that can exchanger in the proximal tubule. ANP has a direct inhibi-
also be activated by peptide YY (PYY), a circulating hormone tory action on renin release in cultured juxtaglomerular cells
released from gastrointestinal cells. NPY and PYY produce that is mediated by cGMP.243,244
renal vasoconstriction via the Y1 receptor. Administered NPY The renal vascular effects of ANP are mediated by NPR-
constricts both the afferent and efferent arterioles and in- A and NPR-B receptors. Activation of NPR-A receptors di-
hibits renin release. Nerves innervating the efferent arteriole lates preglomerular resistance vessels, including arcuate and
contain more immunoreactive NPY than those to the afferent interlobular arteries, afferent arterioles, and efferent arteri-
arteriole. NPY acts prejunctionally to inhibit norepinephrine oles. The NPR-B receptor contributes to the dilation of the
release via Y2 receptors. Despite marked reductions in RBF, preglomerular vasculature. The reported effect of ANP on
systemic NPY infusion elicits a diuresis and natriuresis that Kf is variable. Although glomerular capillary pressure is in-
is mediated in part by bradykinin. NPY antagonists increase creased, GFR is usually unchanged, presumably because of
basal RBF but do not alter basal urinary excretion.226,228,237 a decrease in Kf. ANP infusions increase glomerular perme-
ability to macromolecules. The preglomerular vasodilation
Other Vasoactive Agents produced by ANP does not impair autoregulation of either
RBF or GFR. ANP exerts both indirect and direct effects on
Atrial Natriuretic Peptide the TGF mechanism. ANP markedly inhibits TGF control of
ANP is a 28-amino acid peptide family involved in the phys- glomerular hemodynamics when feedback activity is evalu-
iologic regulation of renal function and sodium excretion. ated by the perfusion of the Henle loop with arti cial uid.
A high molecular weight precursor of ANP is constitutively Studies on mice with underexpression and overexpression
synthesized in cardiocytes of the atria. ANP is released into of the NPR-A receptor highlight the important role ANP has
the circulation as a function of atrial volume or distention in mediating the renal vascular and tubular changes result-
in association with changes in sodium and water balance. ing from blood volume expansion. Overexpression of NPR-
In addition to ANP, related peptides include ventricular A receptors is associated with enhanced renal vasodilatory
brain natriuretic peptide (BNP) and renal urodilatin. Studies and natriuretic responses, whereas the responses are mark-
implicate renal conversion of ANP to urodilatin, which edly attenuated in mice lacking NPR-A receptors. Synthetic
is closely related to sodium excretion under a variety of ANP (M-ANP), being tested for use in hypertension and con-
conditions.238,239 gestive heart failure, are potent agents for lowering blood
ANP receptors are concentrated in glomerular capil- pressure and systemic vascular resistance, while increasing
laries and the collecting duct; they are also present along RBF, GFR, and sodium excretion. Transgenic mice with an
the cortical arterioles and the medullary arterioles and the overexpression of natriuretic peptide receptor A respond
vasa recta; cell types include the collecting duct, the vas- to blood volume expansion with marked natriuresis, along
cular smooth muscle, and the endothelial and mesangial with increases in RBF and GFR, whereas mice de cient in
cells. There are three subtypes of natriuretic peptide re- the NPR-A fail to show the increases in RBF and GFR or
ceptors (NPR), termed NPR-A, NPR-B, and NPR-C. The sodium excretion. These data demonstrate the importance
A- and B-type receptors include a cytoplasmic catalytic of ANP in regulating renal function in response to volume
domain and the active particulate guanylate cyclase, ex- expansion.78,107,244–247
erting physiologic effects by increasing cell cGMP. ANP
may buffer the action of vasoconstrictors such as Ang II Vasopressin
and norepinephrine via an interaction of cGMP with in- In addition to its antidiuretic properties, AVP is a vasocon-
tracellular Ca2 , perhaps secondary to the inhibition of strictor, with predominant actions in the renal medulla. The
Ca2 mobilization and the stimulation of Ca2 ef ux. plasma AVP concentration is directly related to plasma os-
The renal vasculature also has biologically silent, NPR-C molality and inversely with blood volume and pressure. AVP
“clearance” receptors, that remove ANP from the circula- produces effects on renal function by binding to V1a or V2
tion with no role in signal transduction or guanylate cy- membrane receptors. The well-known osmoregulatory func-
clase activity. 240–242 tion of low plasma AVP concentrations is mediated through
ANP is a rapid-acting, potent natriuretic and diuretic a V2 tubular membrane receptor that signals through cAMP/
peptide that is also capable of lowering arterial pressure by PKA to alter water permeability and sodium reabsorption.
direct vasodilatory effects on the systemic vasculature and Vasopressin also stimulates the production of medullary NO
reducing cardiac output. Physiologic concentrations of ANP via the activation of V2 receptors, which serves a protective
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1115
15

function to prevent excessive vasoconstriction. AVP affects by the inhibition of cytochrome P450 epoxide activity and
renal hemodynamics by acting on V1a receptors localized to EET production.107,253
cortical arteries/arterioles (interlobar, arcuate and interlobu- In normal, unstressed conscious animals, the an-
lar arteries, afferent and efferent arterioles, glomeruli) and tagonism of V1 receptors has no effect on basal RBF or
outer medullary vasa recta. V1a mRNA and protein in preglo- arterial pressure, indicating minimal pressor activity of
merular resistance arteries/arterioles are upregulated in re- AVP when its plasma concentration is low. Physiologic
sponse to reduced plasma AVP concentration associated with increases in plasma AVP concentration during 48-hour
water loading and downregulated as plasma AVP is increased water restriction and maximum urine osmolality reduce
during water deprivation. Desensitization of V1 receptors blood flow to the inner medulla via V1 receptors while
and reduced receptor density are related to PKC-mediated maintaining a constancy of blood flow to the outer me-
phosphorylation. AVP activates vascular V1a receptors to dulla. V2 receptor antagonism produces a diuresis. In-
exert pressor effects that contribute to the maintenance of creases in plasma AVP concentrations up to 8 pg per
arterial pressure during conditions of chronic water depriva- millimeter reduce medullary perfusion selectively and
tion, graded hemorrhage, and possibly in various forms of greatly attenuate the arterial pressure–blood flow and
hypertension.107,248,249 pressure–natriuresis relations without affecting total RBF
The vascular effects of AVP are due to the activation or renal cortical blood flow. AVP-induced vasoconstric-
of V1a receptors on vascular smooth muscle cells, which is tion in the renal medulla is normally modulated by AVP-
coupled to G q protein that leads to increased IP3 produc- stimulated local release of NO, perhaps mediated by V2
tion and PKC activation. In afferent arterioles, [Ca2 ]i is receptors, reflecting a compensatory response that buf-
increased by a combination of Ca2 release from the sar- fers the magnitude of the vasoconstriction and stabilizes
coplasmic reticulum and Ca2 entry through voltage-gated medullary perfusion. 15
L-type Ca2 channels and voltage-insensitive store-operat-
ed cation channels. The contraction of efferent arterioles Adrenomedullin
is more dependent on Ca2 release from sarcoplasmic re- Adrenomedullin, - and -calcitonin gene-related peptide
ticular stores with little Ca2 entry through L-type chan- (CGRP), calcitonin, and amylin are homologous polypep-
nels. In addition to increasing [Ca2 ]i, AVP increases Ca2 tides with overlapping biologic actions such as vasodila-
sensitivity via PKC and Rho-kinase signaling. The Kf low- tion, inhibition of bone resorption, and antiproliferative
ering effect of AVP may be mediated by a direct action on activity. Adrenomedullin is a potent renal vasodilating and
mesangial cells. It was found, however, that Kf could be natriuretic peptide (52 amino acids with a disul de ring),
normalized by a combined blockade of the vascular action which is made in the kidney as well as the adrenal gland.
of both AVP and Ang II, whereas the selective blockade of mRNA for adrenomedullin and its receptor are colocalized
either peptide was ineffective. The V1A receptor on endo- to renal vessels, glomeruli, and inner medullary collecting
thelial cells mediates AVP-induced NO release. Also, AVP ducts. The proximal tubule is abundant in adrenomedullin
interacts with the renal arachidonic acid and the renin–an- mRNA, whereas the greatest amounts of receptor mRNA
giotensin systems. AVP stimulates renal release of PGE2 in are in the papilla. Changes in the salt diet do not appear
vivo. Prostaglandin production is increased by AVP-recep- to change the expression of the peptide or its receptor in
tor interaction with either V1 or V2 receptors on medul- either the cortex or the medulla. This hormone/paracrine
lary interstitial cells or glomerular mesangial cells. Vascular agent increases RBF and sodium excretion without affect-
smooth muscle V1A receptor activation stimulates phos- ing GFR. Natriuretic potency increases during the inhi-
pholipase A2 and prostaglandin production. Mice lacking bition of neutral endopeptidase, an enzyme that cleaves
the V1A receptor are hypotensive and have decreased blood endogenous peptides with a disul de ring such as adreno-
volume and sympathetic activity. They also have decreased medullin and ANP.
activity of the renin–angiotensin system and lower aldo- Adrenomedullin produces vasodilation by cAMP/PKA
sterone levels. The V1A receptor is expressed in macula signaling and by stimulating endothelial NO production,
densa cells and colocalizes with COX-2 and nNOS. The leading to the activation of ATP-sensitive K channels and
reduced RBF and GFR are due, in part, to reduced blood Ca2 -dependent K channels and the hyperpolarization of
pressure.1,55,107,142,248,250–252 vascular smooth muscle cells. Adrenomedullin increases re-
The administration of exogenous AVP produces renal nin release via increasing cAMP in juxtaglomerular granular
vasoconstriction that is buffered more by NO than COX- cells. CGRP exerts NO-dependent as well as cAMP/PKA va-
dependent prostaglandins, effects mediated by V1 but not sodilation. The administration of adrenomedullin or CGRP
V2 receptors. Vasoconstriction in the renal medulla is more increases RBF and to a lesser extent GFR; both are natriuretic
prominent than in the renal cortex. AVP-induced cortical and diuretic. However, the renal vasodilator effects of adre-
vasoconstriction is effectively buffered by the cytochrome nomedullin are not dependent on CGRP. Adrenomedullin
P450 epoxide production of vasodilator EETs such that cor- and CGRP dilate afferent arterioles and blunt Ang II- and
tical blood ow is basically unchanged by AVP. In contrast, norepinephrine-induced renal vasoconstriction. Increased
the medullary vasoconstriction elicited by AVP is unaffected adrenomedullin levels in diabetic rats are associated with
116
11 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

the early hyper ltration and may contribute to the afferent signaling and participates in renal vasoconstriction elicited
arteriolar vasodilation.107,254–257 by Ang II, catecholamines, and ET-1 activation of ETA and
ETB receptors.199,262
• O2 exerts tonic renal vasoconstriction in normoten-
Reactive Oxygen Species sive rats, an action that becomes more pronounced when
Small amounts of ROS constantly produced by aerobic me- NO production is inhibited, indicating an interaction
tabolism have important roles in signal transduction in the between • O2 and NO that have opposing actions on vas-
vasculature under physiologic conditions. In pathophysi- cular resistance. This is evident because the administration
ologic states, ROS initiate and amplify deleterious events of superoxide dismutase and NOX inhibition increases RBF.
such as lipid oxidation and tissue/DNA damage associated Moreover, NOX2-de cient mice have an increased basal RBF
with glomerular in ammation and proteinuria, and vascular with normal GFR and arterial pressure, with less renal vaso-
hypertrophy in addition to vasoconstriction. ROS are prod- constriction in response to NOS inhibition than observed
ucts of partial reduction of oxygen, generated by enzymatic in wild-type mice that are able to produce the vasoconstric-
and nonenzymatic reactions. Common oxidative enzymes tor via NOX2. NOX-derived • O2 , largely independent of
include nicotinamide adenine dinucleotide(NADH)/reduced H2O2, contributes to GPCR signaling and participates in re-
NADPH oxidases (NOX), COX, cytochrome P450, and xan- nal vasoconstriction elicited by Ang II, catecholamines, and
thine and glucose oxidases. The reduction of molecular oxy- ET-1 activation of ETA and ETB receptors. Ang II–induced
gen (O2 e ) produces superoxide anion (• O2 ), which renal vasoconstriction and reduced GFR are magni ed dur-
is normally balanced by its degradation. Superoxide dis- ing NOS inhibition and weakened during administration of
mutases (SOD) catalyze the conversion of • O2 to hydrogen superoxide dismutase to scavenge • O2 . Increased endog-
peroxide (H2O2) that has oxidizing potential. H2O2 is subse- enous • O2 activity produced by acute pharmacologic in-
quently neutralized by glutathione peroxidases and catalase. hibition of superoxide dismutase (diethyldithiocarbamate)
An alternative pathway for • O2 degradation is to rapidly re- reduces total renal, as well as both cortical and medullary
act with NO to form peroxynitrite (ONOO ), which clearly blood ow. The vasoconstriction is greater when the buffer-
limits the half-life, the diffusion distance, and the biologic ing afforded by NO is removed by NOS inhibition. This is
activity of NO as well as • O2 . Vitamins A, E, and C and also the case for renal vasoconstriction produced by norepi-
bilirubin are scavengers of ROS.199,258–261 nephrine, phenylephrine, and ET-1. Other studies show that
As intracellular signals, ROS may activate or inactivate increased • O2 production stimulated by the acute infusion
redox-sensitive protein kinases and phosphatases to modu- of Ang II produces more pronounced renal vasoconstriction
late GPCR phosphorylation and transcription factors. Reac- when NADPH oxidase is intact than when NADPH oxidase
tive nitrogen species such as S-nitrosothiols can modulate is rendered nonfunctional in transgenic animals with NOX2
GPCR signaling and internalization through S-nitrosylation mutated. Ang II elicits less pronounced renal vasoconstric-
of -arrestin and GRK. Normally there is a ne balance tion in the absence of NOX2. Extracellular superoxide dis-
between activities of oxidative and antioxidant enzymes, mutase inactivation of the • O2 plays an important role in
optimizing NO activity and minimizing superoxide anion buffering acute Ang II–induced constriction of the afferent
generation. • O2 functions as the counterpart to NO and arteriole and increased RVR produced by chronic Ang II
its antiproliferative and vasodilatory actions. In the kid- infusion.198,263–266
ney, ROS are formed in arteries and arterioles, glomeruli, The mechanism by which • O2 causes or modulates
and juxtaglomerular cells endowed with oxidases such as renal vasoconstriction in normal kidneys is still unresolved.
NOX, NOS, and COX. NOX1, NOX2, and NOX4 are ex- Ang II stimulates NADPH oxidase activity and • O2 produc-
pressed in the kidney. Predominant isoforms in the renal tion in the renal cortex and medulla. Ang II and ET-1 recep-
vasculature are NOX1 and NOX2 that primarily produce tor activation rapidly stimulates NOX2-mediated O2- release
• O2 ; epithelial NOX4 mainly generates H2O2. • O2 is de- from afferent arterioles that increases cytosolic Ca2 con-
graded by superoxide dismutases, of which the cytosolic or centration in smooth muscle cells by increasing ADP ribosyl
intracellular isoform accounts for ~70% of the enzyme in cyclase activity. The metabolite cyclic ADP ribose sensi-
the kidney, with lesser amounts in extracellular and mito- tizes ryanodine receptors on the sarcoplasmic reticulum of
chondrial compartments. In the juxtaglomerular apparatus, smooth muscle to release Ca2 . • O2 may also scavenge NO
endothelial cells produce NO via eNOS and macula densa and reduce bioavailability of this vasodilator, a major fac-
cells via nNOS. Stimulants of • O2 production include va- tor contributing to endothelial dysfunction in disease states.
soconstrictor agents (e.g., Ang II, ET-1, norepinephrine), Accordingly, • O2 activity is enhanced during NOS inhibi-
growth factors, and stretch. Chronic high Ang II and a high tion. It should be appreciated that • O2 -induced acute renal
salt diet are potent stimulants, increasing the expression of vasoconstriction is observed during NOS inhibition, high-
NOX subunits and reducing superoxide dismutase isoforms. lighting a principal direct action on vascular smooth muscle
Overall renal actions of • O2 are vasoconstriction and the independent of NO.20,113,160,198,263,264,266–269
enhancement of tubular sodium reabsorption. NOX-derived Increased intrarenal • O2 produced by infusions of hy-
• O2 , largely independent of H2O2, contributes to GPCR poxanthine and xanthine oxidase increases sodium excretion,
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1117
17

while GFR is reduced more than RBF and arterial pressure is by an endothelial-derived and COX-derived vasoconstric-
unchanged. In these rats, • O2 did not affect the ef ciency of tor that acts on TP receptors on vascular smooth muscle
pressure-induced steady-state RBF autoregulation. However, cells.279
other evidence convincingly indicates that • O2 regulates Endogenous H2O 2 appears to have little effect on pre-
both myogenic and TGF mechanisms responsible for renal glomerular resistance vessels in the renal cortex because
autoregulation. The pressure-induced myogenic constrictor the combination of catalase with a superoxide dismutase
response of isolated afferent arterioles is mediated or en- mimetic has no additional effect as compared to super-
hanced by • O2 independent of H2O2 and of eNOS produc- oxide dismutase quenching O 2 alone. As mentioned in
tion of NO. In addition, • O2 produced by NOX2 in macula the following, increases in H2O 2 above basal levels of 100
densa cells plays a role in TGF-induced afferent arteriolar to 500 nmol per liter cause vasoconstriction in the re-
vasoconstriction via direct action on afferent arterioles. This nal medulla. H2O 2, at apparent pharmacologic concen-
direct action complements quenching NO availability. The trations in micrometer per millimeter range in vitro, is
effects of • O2 are attenuated by increased levels of superox- reported to have biphasic effects on nonrenal resistance
ide dismutase. Thus, ROS are important signaling molecules arterioles, dilating at low concentrations, perhaps due to
that participate in intrinsic autoregulatory responses of the NO, and constricting at high concentrations ( 50 mol
preglomerular vasculature to changes in renal perfusion pres- per liter), possibly due to an isoprostane. H2O 2 inhibits
sure. Local production of NO and ROS modulates reactiv- Ang II increases in [Ca2 ]i in afferent arterioles when the
ity of descending vasa recta pericytes that control medullary concentration is 1 m but not less. Electrophysiologic
perfusion.114,171,270–274 studies of endothelium of freshly isolated porcine renal
eNOS and NADPH oxidase are both expressed in tu- arteries indicates that a large conductance (300 pS) Ca2 -
bular epithelial cells within the renal medulla, particularly activated K channel (BKCa) is inhibited dose depend-
the thick ascending limb of the Henle loop, with the pro- ently by ROS in nanomole per liter range and H2O 2 in
duction of NO and • O2 participating in the regulation of the micromole per liter range. If tonically active, closure
medullary blood ow and in uencing the set point of the of BKCa channels causes depolarization and leads to vaso-
pressure–natriuresis relation. Sodium retention and hyper- constriction. Consistent with this view, H2O 2 was found
tension result when the balance of production of these free to reduce bradykinin-induced dilation of isolated renal
radicals favors • O2 in conditions such as in activation of arteries.272,280,281
the renin–angiotensin system, NOS inhibition, and diabetes.
For example, during chronic NOS inhibition, renal vascular
actions of • O2 are largely unopposed. Intrarenal infusion
MECHANISMS REGULATING
of the superoxide dismutase tempol in hypertensive L-nitro- MEDULLARY MICROCIRCULATION
arginine-methyl-ester (L-NAME) treated rats increases total Blood ow to the renal medulla is only about 20% of the
RBF and blood ow to both the renal cortex and the me- total RBF; however, it is of major importance in regulating
dulla, and GFR, while urinary excretion of 8-isoprostane is sodium homeostasis and in the maintenance of the medul-
reduced.275–277 lary hypertonic environment. Expressed per gram of tissue,
Peroxynitrite (ONOO ) is formed endogenously by medullary blood ow is lower than cortical blood ow. The
NO reacting with • O2 , exerting NO-like biologic activ- unique architecture of the renal medullary microcircula-
ity as well as nitrating proteins during oxidative stress. The tion preserves the axial osmotic gradient generated by the
administration of low OONO concentrations causes renal countercurrent exchange of water and solutes between the
vasodilation as RBF and GFR increase in parallel in a NO- descending and ascending vasa recta while allowing for
dependent manner, reverting to the constriction during NOS the removal of the water and solutes reabsorbed from the
inhibition. High ONOO concentrations produce renal va- descending limb of the Henle loop and the medullary col-
soconstriction with larger reductions in GFR than in RBF. lecting ducts. Blood ow to the renal medulla is supplied
The reductions in RBF and GFR are magni ed during NOS from efferent arterioles of juxtamedullary nephrons, which
inhibition.278 give rise to the vascular bundles located in the outer me-
ONOO can oxidize arachidonic acid to form the va- dulla. The descending vasa recta (DVR) gradually transform
soconstrictor 8-iso-PGF2 (F2-isoprostane), which acti- until the smooth muscle layer, present in the outer medulla,
vates a thromboxane TP receptor to elicit vasoconstriction. is replaced by the discontinuous rings of pericytes. Thus, the
Isoprostanes may increase ET-1 release from endothelial vascular smooth muscle cells of the juxtamedullary neph-
cells. Chronic exposure to high levels of Ang II stimulates ron arterioles and of the outer medullary DVR are primarily
isoprostane production by the kidney. Oxidative stress responsible for the differential regulation of the medullary
and exaggerated isoprostane levels acting on TP receptors circulation. The outer zone of the inner medulla is parti-
enhances the strength of TGF in some models of hyper- tioned into two distinct compartments with intercluster re-
tension. Such signaling may explain the afferent arteriolar gions, one consisting of the ascending and the descending
vasoconstriction associated with oxidative stress in Ang vasa recta and another consisting of the ascending vasa recta
II–induced hypertension, which appears to be mediated (AVR) and collecting ducts. Efferent arterioles and the DVR
118
11 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

are the main medullary structures with sympathetic innerva- perfusion pressure changes. As mentioned previously, how-
tion, which terminates when pericytes replace the smooth ever, the renal medulla is largely perfused by the efferent
muscle cells. Extravascular renomedullary interstitial cells arterioles from juxtamedullary nephrons, which have very
also exhibit contractile properties, but their role in regulating high autoregulatory ef ciency. Although a small population
medullary blood ow is unclear. It has been more dif cult of shunt vessels bypass glomeruli, and may escape the auto-
to study the medullary circulation due to its inaccessibility regulatory adjustments, the extent of periglomerular shunt-
and the fact that it is positioned in series with the glomeru- ing is very small and unlikely to contribute signi cantly to
lar circulation of juxtamedullary nephrons. Although there overall medullary blood ow responses. Both cortical and
is considerable discrepancy regarding the absolute values of medullary blood ows are ef ciently autoregulated in the
medullary blood ow obtained using various techniques, dog. This holds for both outer and inner medullary per-
most of them provide a reasonable index of relative changes fusion. The same is true for cortical blood ow in the rat.
in blood ow.1,15,18,89,282,283 Inner and outer medullary blood ows are also autoregu-
lated ef ciently in hydropenic rats, but not during marked
Hematocrit volume expansion. Thus, it appears that loss of medullary
The hematocrit of renal medullary blood is lower than that blood ow autoregulation occurs primarily during marked
of systemic blood or blood derived from the renal cortex. volume expansion in rats subjected to increases in perfu-
Red blood cell (RBC) transit time is shorter than for plasma, sion pressure. Recruitment of ow through the previously
and tissue hematocrit varies inversely with the medullary unperfused vasa recta may also contribute to that process, in
axis. Studies using videomicroscopic techniques and com- particular because individual medullary vessels also exhibit
plementary direct measurements of hematocrit with micro- autoregulatory responses. Volume-expanded sodium-replete
puncture have demonstrated low microvessel hematocrit in dogs and rabbits exhibit ef cient intact medullary blood
the renal medulla. Shrinkage of RBCs in the hypertonic me- ow autoregulation. The in vitro juxtamedullary nephron
dulla also shifts water from the interior of RBCs to plasma preparation that evaluates blood ow through nephrons that
and also contribute to the lower medullary microvessel give rise to the vasa recta clearly exhibits normal autoregula-
hematocrit.2,15 tory behavior of the preglomerular vasculature. Thus, the
In contrast to the peritubular capillary plexus that extent of medullary blood ow autoregulation ef ciency as
arises from efferent arterioles in the cortex to reabsorb well as the role of medullary perfusion in the generation of
massive volumes of tubular uid, the vasa recta serve pressure natriuresis varies with species and degree of vol-
different needs speci c to the medulla. Through their ume expansion. Nevertheless, paracrine factors act within
countercurrent arrangement, the DVR and the AVR trap the medulla to exert local control of medullary blood ow.
NaCl and urea deposited to the interstitium by collecting The pressurization of in vitro perfused DVR increases endo-
ducts and the loops of Henle and maintain corticomedul- thelial [Ca2 ]i and the generation of NO. The release of NO
lary osmotic gradients. However, metabolic substrates, by the vasa recta could increase local blood ow as well as
including O 2, that enter the DVR diffuse to the AVR to inhibit salt reabsorption by adjacent tubules. A role for NO
be shunted back to the cortex, leading to lower O 2 levels to provide a diffusible signal between the vasculature and
in the medulla than in the cortex. Paracrine agents reg- nephrons seems likely.1,2,15,18,107,153,277,284–286
ulate the renal medulla perfusion in a complex manner
involving tubular-to-vascular and vascular-to-tubular Vasopressin
paracrine signaling cross-talk. Protection of the medulla
from hypoxia by these agents involves the local genera- Increases in medullary blood ow reduce the ef ciency of
tion of paracrine agents by tubular and vascular struc- passive countercurrent exchange, leading to “solute wash-
tures and trapping by countercurrent exchange to yield out” and reductions in the corticomedullary gradients for
axial concentration gradients that exert variable effects on NaCl and urea. Vasopressin exerts an important role in
the medullary vessels.90 reducing medullary blood ow which contributes to in-
creased urinary concentrating ability during antidiuresis.
Homozygous Brattleboro rats that lack vasopressin have
Autoregulation central diabetes insipidus and elevated medullary plasma
As already discussed, overall RBF is autoregulated with very ow. Vasopressin exerts its actions via subtype speci c V1
high ef ciency over a wide range of systemic perfusion pres- (vasoconstrictor) and V2 (antidiuretic) receptors and reduces
sure. Although cortical blood ow is well autoregulated vasa recta blood ow through the activation of both V1 or
within the physiologic range, the extent to which medullary V2, indicating roles for both vasoactive and reabsorptive
blood ow is autoregulated is more controversial, especially mechanisms. A selective V1 receptor agonist reduces inner
in the rat. Some studies suggest that medullary blood ow medullary more than outer medullary blood ow. Similar-
autoregulation is not as ef cient as in the cortex and that ly, elevated circulating vasopressin in water-deprived rats
changes in medullary blood ow contribute to pressure na- reduces inner medullary blood ow with weaker effects in
triuresis and the regulation of salt and water excretion as the cortex and the outer medulla; these effects are blocked
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1119
19

by a V1 antagonist, supporting V1-mediated vascular effects blood ow and reduces blood pressure in hypertensive
of vasopressin to modulate inner medullary blood ow and Dahl rats and spontaneously hypertensive rats (SHR). DVR
to promote antidiuresis by maintaining the elevated osmolal- is dilated by NO donors and endothelium-dependent vaso-
ity in the medullary interstitium. Vasopressin reduces med- dilators. Similarly, NOS inhibition increases DVR vasomo-
ullary perfusion by constricting juxtamedullary afferent and tor tone, producing as much as a 35% decrease in blood
efferent arterioles. Afferent arteriolar vasopressin–mediated ow and blunts the dilation of preconstricted DVR by the
constriction is dependent upon voltage-gated Ca2 entry via endothelium-dependent vasodilators, acetylcholine and
voltage-gated L-type and store-operated channels, whereas bradykinin. NO has a particularly important role to protect
efferent constriction may be primarily related to Ca2 mobi- against the excessive reduction of medullary perfusion as-
lization from stores. Vasopressin also constricts outer med- sociated with hypoxia and oxidative stress, and NO levels in
ullary DVR. A vasopressin V1 agonist reduces medullary the medullary interstitium rise in response to Ang II, norepi-
blood ow without constricting either afferent or efferent nephrine, and vasopressin. Conversely, medullary inhibition
arterioles, suggesting a greater sensitivity of the vasa recta to of NOS and NO production increases vascular sensitivity so
vasopressin.15,252,287,288 that infusion of otherwise ineffective doses of vasoconstric-
In addition to V1 mediated constrictor effects of vaso- tors reduce perfusion and generate tissue hypoxia.284,293
pressin, vascular V2 receptors may cause vasodilation via the NO has both vasodilatory and natriuretic effects and is
stimulation of NO release. V2 agonists dilate preconstricted synthesized by epithelial as well as endothelial cells, sug-
afferent arterioles and the outer medullary DVR in vitro. The gesting critical tubular–vascular interactions. NO generated
V2 agonist, dDAVP, stimulates medullary NO release and in- by the medullary thick ascending limb in uences DVR tone.
creases medullary blood ow. AVP-induced V2 receptor ac- Conversely, NO generated by the DVR inhibits sodium reab-
tivation in collecting ducts stimulates the phosphoinositide sorption by adjacent nephron segments.15,284,294
pathway and causes the mobilization of Ca2 to increase NO The bioavailability and actions of NO are modi ed to a
production in the medulla and to protect the outer medulla great extent by opposing the generation of ROS. NO levels
from excessive vasoconstriction and ischemia.1,15,248,286 in the renal medulla are modulated through reactions with
oxygen radicals. ROS generation plays a role in the agonist-
Angiotensin induced constriction of renal microvessels in the cortex and
In addition to its actions on the afferent and efferent the medulla and in Na reabsorption by the ascending limb
arterioles, Ang II tonically constricts the medullary micro- of the loop of Henle. In the DVR, ROS are generated upon
circulation via effects on the vasa recta. A NOS blockade stimulation with Ang II and PKC agonists; superoxide anion
constricts DVR and intensi es the constriction by Ang II. generation by the medullary thick ascending limb may limit
Similar to NO, PGE2 and adenosine blunt Ang II constriction availability of NO delivery and the vasodilation of DVR. The
of DVR.15,275,289 SOD inhibitor, diethyldithiocarbamate, reduces medullary
AT2 receptor activation elicits vasodilation via the gen- blood ow, indicating a vasoconstrictor action of superox-
eration of NO and the synthesis of vasodilatory cytochrome ide anion. Infusion of the SOD mimetic, tempol, increases
P450 epoxygenase-derived EETs. AT2 activation vasodilates medullary blood ow and sodium excretion, an effect that is
DVR where it inhibits reactive oxygen species formation and more pronounced when H2O2 is simultaneously eliminated
facilitates endothelium-dependent [Ca2 ]i signaling that with catalase.228,270,275,294–296
leads to the release of vasodilators. There are also mecha-
nisms leading to the Ang II–induced enhancement of med- Arachidonic Acid Metabolites
ullary perfusion via AT1 receptors due to the generation of Prostaglandins, generated from arachidonic acid by COX-1
compensatory vasodilators, particularly NO and vasodilator and COX-2 enzymes, alter intrarenal hemodynamics and
prostaglandins. Interstitial Ang II concentrations in the renal increase blood ow toward the juxtamedullary cortex.
medulla are higher than in the cortex and Ang II receptor Medullary blood ow is protected from excessive vaso-
density is also higher in this region.15,129,228,290–292 constrictors by prostaglandins as well as NO. Nonselective
COX blockade decreases vasa recta blood ow by up to
Nitric Oxide 50% and potentiates medullary hypoxia with a relative
NO is particularly important in defending the renal medulla sparing of cortical perfusion. PGE2 is generated in large
against hypoxia and ischemia. Chronic and acute systemic quantities in the renal medulla and blunts Ang II–induced
or intrarenal NOS inhibition reduces medullary blood ow constriction of isolated perfused DVR. Both COX-1 and
more than cortical blood ow and elicits hypertension. Per COX-2 isoforms contribute to renal prostaglandin synthe-
volume of tissue, NO production in the renal medulla ex- sis and are predominantly expressed in the renal medulla;
ceeds that in the cortex. NO production is closely coupled to COX-2 is subject to greater regulation. Renomedullary in-
L-arginine availability and cellular uptake via an amino acid terstitial cells express receptors and release paracrine sub-
transporter. Accordingly, dietary L-arginine supplementa- stances including PGE2, EETs, and medullipin, and express
tion increases renal medullary interstitial NO and medullary both COX-1 and COX-2. Medullary COX-2 expression is
120
12 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

stimulated by tonicity. Genetic de ciency of COX-2, or its During hypoxia, the medullary thick ascending limb of
chronic inhibition, reduces medullary blood ow and en- Henle synthesizes adenosine, which serves a paracrine va-
hances vasoconstrictor responses to Ang II. COX-2–derived sodilator function to preserve medullary perfusion and to
prostanoids serve to maintain medullary blood ow under augment medullary pO2.15,61,212,217,301,302
most conditions.15,180,290,297,298
Products of arachidonic acid are also generated by cy- Endothelin
tochrome P450 isoforms to yield EETs, the HETEs, and
Medullary ET receptors are present on medullary vascular
their products, dihydroxyeicosatetraenoic acids (DHETs).
bundles, medullary interstitial cells, and adjoining collecting
20-HETE reduces medullary blood ow and the inhibition
duct cells. ET1 concentrations in the renal medulla are regu-
of 20-HETE with HET0016 enhances medullary blood
lated by osmolality. ET1 binds to and stimulates both ETA
ow. In contrast, EETs have been linked to endothelial-
and ETB receptors, thus inducing vasoconstriction. Isolated
derived hyperpolarizing factors and cause vasodila-
ETB receptor stimulation, however, mediates vasodilation.
tion directly and also oppose vasoconstrictor actions of
Preglomerular smooth muscle cells show [Ca2 ]i responses
vasopressin.118,253,299
to ETA and ETB agonists. The DVR from the outer medulla
constrict in response to ET1, but medullary perfusion is not
Kallikrein–Kinin System greatly affected due to the enhanced production of NO from
Kallikreins release kinins from kininogens and are expressed ETB-receptor stimulation, counteracting the vasoconstric-
by the outer and inner medullary collecting ducts. Kinins tion. ETB-receptor stimulation of NO production plays a
exert their actions by activating B1 and B2 receptors. B2 re- signi cant role in protecting the medullary circulation from
ceptors are expressed throughout the outer and inner me- excessive vasoconstriction.15,150,156,162,303,304
dulla, and modulate blood ow and sodium reabsorption in ET1 treatment selectively reduces cortical blood ow
the renal medulla. The infusion of a kinin antagonist causes while transiently increasing medullary blood ow. Medul-
a 20% reduction of papillary blood ow. The enhancement lary vasodilation and natriuresis are prevented by blocking
of kinin activity through the infusion of bradykinin or the ETB receptors or NO synthesis. Renal hypoxia stimulates
inhibition of kininases with enalaprilat or phosphoramidon ET1 production. The effects of endothelins on medullary
increases both medullary blood ow and sodium excretion. blood ow vary with dietary salt intake. Chronic Ang II in-
Blocking bradykinin receptors decreases medullary blood fusion, combined with salt loading, increases cortical and
ow, an effect that is blocked by NOS inhibition. ACE in- medullary immunoreactive ET. The administration of ET1
hibition in the presence of an AT1 receptor blockade causes into ETB-receptor de cient rats or wild-type rats in which
greater increases in MBF than cortical blood ow, and the the ETB receptor is blocked fails to increase sodium excre-
effects are blocked by the B2 receptor antagonist, icatibant. tion. Mice with the collecting duct-speci c knockout of the
Low sodium diets augment the kinin-mediated component ET1 gene have impaired sodium excretion in response to so-
elicited by ACE inhibition. In volume-expanded anesthe- dium loading and have salt-dependent hypertension.156,304
tized rats, a B2 receptor blockade with icatibant reduces pap-
illary blood ow. Bradykinin, acting through B2 receptors, Reactive Oxygen Species
generates robust endothelial cytoplasmic calcium [Ca2 ]i re-
The production of ROS in the kidney is greatest in the outer
sponses in isolated DVR leading to marked NO production
medulla. eNOS and NADPH oxidase are both expressed in
and EETs leading to vasodilation of Ang II preconstricted
tubular epithelial cells within the renal medulla, particularly
vessels.15,208,300
the thick ascending limb of the Henle loop, with the pro-
duction of NO and • O2 participating in the regulation of
Adenosine medullary blood ow by acting on pericytes encircling the
As discussed earlier, adenosine exerts its actions on the DVR. • O2 and H2O2 cause vasoconstriction, effects that
renal vasculature predominantly through A1 and A2 recep- are partially offset by the vasodilator NO. The inhibition of
tor subtypes. Adenosine A1 receptor activation transiently superoxide dismutase in the renal medulla increases local
reduces cortical and medullary blood ow, whereas A2 re- • O2 levels and reduces medullary perfusion. This is also
ceptor stimulation leads to medullary vasodilation and na- the case for increased H2O2. Chronically increased oxida-
triuresis. Both A1 and A2 receptors are expressed by DVR, tive stress such as in the activation of the renin –angiotensin
and their respective stimulation induces constriction or system and diabetes, reduces medullary blood ow and so-
dilation. Interstitial adenosine concentrations are near the dium excretion, resetting pressure–natriuresis to produce
af nity for the A2 receptor so that changes should modulate hypertension.270,275,277,305
vasodilatory and natriuretic effects. A1-induced constriction Collectively, the various vasoactive agents selectively
is mediated by the pertussis-toxin –sensitive G i protein regulate medullary blood ow through their actions on
and phospholipase C activation. A2-mediated dilation is the pericytes that surround the vasa recta. Interestingly,
mediated by the stimulation of the G s protein and the ac- the pericytes respond relatively weakly to vasoconstrictors
tivation of KATP channels via enhanced levels of 11,12-EET. such as Ang II, ET1, norepinephrine, and vasopressin, but
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1121
21

more robustly to the vasodilators including NO, CO, PGE2, inhibition with aliskiren in humans on a low salt diet reveal
and adenosine. Furthermore, endogenous Ang II enhances, larger increases in RBF and GFR than in inhibition of the
whereas NO reduces, the impact of increased renal sym- renin –angiotensin system using an ACE inhibitor or AT1 re-
pathetic activity on medullary blood ow. The continuous ceptor antagonist.308
interactions among these components regulate medullary A high salt diet increases the urinary excretion of salt to
blood ow and local oxygen supply.15,18,208,306 maintain a steady-state balance. There is a moderate increase
in extracellular uid volume, whereas the arterial pressure
and the RBF and GFR usually remain within the normal
ADAPTATION OF RENAL range in individuals who are not salt sensitive. The stability
HEMODYNAMICS TO ALTERED of renal hemodynamics is achieved by a balance between
PHYSIOLOGIC CONDITIONS vasoconstrictor and dilator systems. The vasoconstrictor
effects of the renin –angiotensin system are reduced due to
Changes in Salt Intake multiple mechanisms, including the suppression of renin
In healthy young adults, chronic changes in salt in the diet release and Ang II production. Macula densa nNOS and
cause proportional changes in extracellular uid volume and COX-2 expression and activity are reduced during sodium
sodium excretion while having a subtle or minor in uence loading. Reduced PGE2 production by macula densa cells
on renal hemodynamics and arterial pressure. RBF and GFR contributes to a reduced renin release along with reduced
are usually maintained within normal limits as a result of -adrenergic stimulation by sympathetic nerves and in-
basically parallel adjustments in vasoconstrictor and dilator creased baroreceptor inhibition in juxtaglomerular granular
systems; this most often is also the case for arterial pressure. cells. Accordingly, inhibition of the renin –angiotensin sys-
Some people, especially the elderly with compromised renal tem has little in uence on renal hemodynamics and arterial
functions, develop salt-sensitive hypertension when con- pressure in animals fed a high salt diet. This also is the case
suming a high sodium diet. for the AT1-receptor antagonism and for ACE inhibition.
As discussed earlier, the sodium diet and the extracellu- Renal efferent sympathetic nerve activity is also suppressed,
lar uid volume are major regulators of renin synthesis and as is the case for opposing actions of the major vasodila-
release from juxtaglomerular granular cells. Renin secretion tor systems of NO and prostaglandins, which are reduced
and Ang II production are stimulated by volume contraction in parallel. TGF activity is reduced, whereas distal tubular
due to macula densa signaling and increased sympathetic ac- ow is increased during volume expansion. The underlying
tivity. A reduced delivery of salt to the macula densa stimu- mechanisms are not known, but low ambient levels of Ang
lates COX-2 activity and PGE2 production and, thereby, renin II may contribute to reduced reactivity.2
release. Macula densa COX-2 and microsomal PGE synthase Renal vascular reactivity to exogenous Ang II is en-
mRNA are induced to increase PGE2 production during a hanced during sodium loading and attenuated during a low
low sodium diet, whereas the EP4 receptor expression is in- salt diet, being related to AT1-receptor density that is recip-
creased in glomeruli and in renin-secreting juxtaglomerular rocally associated with endogenous Ang II levels. Thus, Ang
granular cells. Other important stimuli to renin-containing II–induced vasoconstriction is less pronounced when Ang
cells are sympathetic nerve activity and baroreceptor input. II is chronically elevated because of the downregulation of
High salt intake inhibits renin release and Ang II production vascular AT1 receptors. This is not the case for norepineph-
by these three major mechanisms of regulating renin. During rine as glomerular hemodynamic responses to renal nerve
sodium restriction, the elevated circulating and intrarenal stimulation are similar whether animals are maintained on
Ang II levels constrict both afferent and efferent arterioles to a low, normal, or high salt diet.146 Intrarenal blood ow
either maintain or increase glomerular capillary pressure. Kf varies during changes in the sodium diet, with cortical and
tends to be reduced. GFR is maintained in the normal range outer medullary but not inner medullary blood ow paral-
or is slightly reduced, whereas RBF declines to a greater ex- leling sodium intake. A high salt diet and reductions in the
tent, thus increasing the ltration fraction. Single nephron plasma Ang II concentration lead to increased cortical blood
studies indicate that the TGF regulation of the afferent arte- ow but not outer or inner medullary perfusion. Medul-
riolar tone is augmented during chronic salt restriction and lary blood ow is relatively well maintained during sodium
high endogenous Ang II.13,121,131,182,307 restriction.111,309
The adaptive renal vasoconstrictor component is pri- The renal hemodynamic in uence of vasodilator sys-
marily the elevated Ang II acting on vascular AT1 receptors tems such as eNOS/NO, COX/PGE2-PGI2, and epoxygen-
because the Ang II receptor blockade increases RBF and GFR ase 11,12-EET are upregulated by a low salt diet, effectively
and reduces arterial pressure, with predominant effects in counteracting the exaggerated vasoconstrictor in uence
reducing afferent arteriolar resistance. Intrarenal ACE inhi- of high Ang II and sympathetic nerve activity. In this set-
bition increases RBF and GFR in sodium-restricted dogs as ting, COX inhibition reduces RBF, renal cortical blood
a result of equal dilation of afferent and efferent arterioles ow, and GFR, along with PGE2 production as compared
in the presence of unchanged arterial pressure; Kf increases to no effect on renal hemodynamics in sodium-replete ani-
slightly. It is noteworthy that recent studies of direct renin mals (Fig. 3.25). Whether the prostanoids are produced
122
12 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

for cortical and medullary blood ow. Macula densa nNOS


is downregulated during a high salt diet. In contrast, pro-
tein expression of eNOS, iNOS, and nNOS is increased in
the inner medulla. Afferent arteriolar vasodilatory responses
to acetylcholine and sodium nitroprusside were unaffected
by chronic high salt diets.316–318
Sodium restriction upregulates the kallikrein–kinin as
well as the renin–Ang II system. The AT1-receptor antago-
nism and ACE inhibition in dogs maintained on a low salt
diet increase RBF and cortical blood ow. ACE inhibition
causes larger increases in medullary ow than AT1 receptor
antagonism, an effect normalized by the blockade of brady-
kinin B2 receptors. The vasodilator kallikrein–kinin system
also participates in the renal adaptation to salt loading. Mice
lacking vasodilator B2 receptors respond to a high salt diet
by becoming hypertensive with reduced RBF and increased
RVR. In contrast, wild-type mice are normotensive with ten-
dencies toward increased RBF and reduced RVR.206
Renal vascular reactivity to exogenous ET-1 is reduced
by a low salt diet. ETA receptor blockade has little effect on
RBF during volume contraction. Sodium restriction upregu-
FIGURE 3.25 A comparison of renal hemodynamic and arterial
lates pre-pro-ET-mRNA in the renal cortex, whereas ETA and
pressure responses to angiotensin converting enzyme (ACE)
ETB receptor density is unchanged. Low sodium intake and
inhibition in sodium-replete (solid lines) and sodium-depleted
elevated Ang II acting on AT1 receptors increase renal med-
(dashed lines) anesthetized dogs. GFR, glomerular ltration rate.
ullary ET-1 and ETA and ETB receptor mRNA, but not the
sensitivity of renal medullary perfusion or sodium excretion
to the ETA or ETB receptor blockade.319,320
by COX-1 or COX-2 is uncertain. One study nds that the Endogenous ET-1 levels are increased in the renal cortex
renal hemodynamic changes are dependent on COX-2 ac- and the medulla of animals fed a high salt diet, and renal
tivity, whereas another study reports that selective COX-2 vascular reactivity to exogenous ET-1 at the whole kidney
inhibition is without effect on whole kidney hemodynamics level is enhanced. ET-1 reduces renal cortical blood ow
during sodium depletion. COX-2 inhibition decreases med- and increases medullary perfusion in rats on a high salt diet
ullary blood ow equally in animals on normal and low salt as compared to cortical vasoconstriction, and has no effect
diets.71,116,310,311 on medullary blood ow in animals on a normal salt diet.
COX activity and renal cortical production of vasodi- Juxtamedullary-afferent arteriolar vasoconstriction produced
latory prostanoids are low during volume expansion. COX by ET-1 and the ETB receptor agonist are reduced. Arteriolar
inhibition in high salt animals has essentially no effect on ETB receptors, but not ETA receptors, are upregulated pre-
whole kidney RBF or GFR. On the other hand, a high salt sumably on endothelial cells during salt loading, and their
diet increases COX-2 expression in the renal medulla. Selec- vasodilation appears to buffer ET-1–induced vasoconstric-
tive medullary COX-2 inhibition reduces medullary blood tion mediated by ETA receptors. The ETA-receptor blockade
ow, which is associated with sodium retention and hyper- has no effect on whole kidney renal hemodynamics in rats
tension in rats consuming a high salt diet.312–315 placed on a high salt diet.158,321
Macula densa nNOS activity and NO productivity are Sodium loading increases renal ET-1 content and NO
inversely related to the sodium diet. Both endothelial eNOS production that is in part mediated by ETB receptors that are
and macula densa nNOS are upregulated during a low salt responsible for increasing medullary blood ow. A genetic
diet. NO generation in the kidney from L-arginine partici- de ciency of ETB receptors leads to salt-induced hyperten-
pates in adapting renal function to changes in salt intake. sion. The more speci c deletion of ET-1 or ETB receptors in
Dietary salt loading in animals enhances eNOS expression the collecting duct also causes salt-sensitive hypertension.
and activity and NO generation in the renal vasculature. In contrast, the speci c mutation of ETB receptors in vas-
Urinary nitrate nitrite excretion, an index of NO produc- cular endothelial cells does not affect the AP sensitivity to
tion, increases during high sodium intake. NOS inhibition salt.156,322
produces greater increases in arterial pressure (AP) and RVR Normal kidneys, however, exhibit highly ef cient whole
and greater reductions in RBF in animals and humans on kidney steady-state autoregulation of RBF whether animals
high versus low salt diets. NOS inhibition reduces medul- are fed either a low or high salt diet and thus is indepen-
lary more than cortical perfusion during low and normal dent of endogenous Ang II. Sodium depleted dogs have a
sodium diets. During sodium loading, NO control is similar normal RBF and GFR, with ef cient autoregulation of RBF,
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1123
23

GFR, and single nephron GFR to changes in renal perfusion in the kidney. Salt-sensitive hypertension can be induced by
pressure. RBF is also effectively autoregulated during a low pharmacologic NOS inhibition or genetic deletion of eNOS;
salt diet whether or not ACE is inhibited or AT1 receptors are the pressor response is reversed by the superoxide dismutase
antagonized.2 A high salt diet has little impact on dynamic mimetic tempol. Enhanced renal vasoconstriction is associ-
RBF autoregulation, with either no change or an increased ated with stronger than normal actions of Ang II and am-
responsiveness of the myogenic mechanism in Sprague- pli cation by oxidative stress and NADPH oxidase–derived
Dawley or Long-Evans rats, respectively. Normal myogenic ROS as well as isoprostane. A chronic high salt diet leads
responses of afferent arterioles to increased luminal pres- to oxidative stress and in ammation in the kidney and vas-
sure is observed in mice fed a high salt diet for 3 months. A cular tissues and, eventually, hypertension, which is inde-
slight attenuation of pressure-induced myogenic responses pendent of Ang II activation of AT1 receptors. Salt loading
is noted during high salt versus low salt diets for isolated increases renal cortical NADPH oxidase subunit expression
interlobular arteries and afferent arterioles of Dahl salt- (NOX2 and p47phox) and activity, increases superoxide gen-
resistant rats. In hypertensive states, ef cient autoregulatory eration, and increases urinary H2O2, 8-isoprostane, malon-
responses to an increased arterial pressure play an important dialdehyde, and thromboxane B2 excretion, and decreases
role in protecting sensitive glomerular capillaries and other plasma NO end products. This is accompanied by the re-
renal structures from severe barotrauma. In some individu- duced expression of antioxidant superoxide dismutase iso-
als, high salt intake may increase RBF and impair autoregu- forms. Such changes are limited more to the renal cortex
latory mechanisms such that the increased arterial pressure than the renal medulla. It is noteworthy that the functional
is transmitted to glomeruli, with the eventual development changes associated with oxidative stress and high salt intake
of proteinuria and glomerular sclerosis, along with intersti- are counteracted by oral L-arginine supplementation and im-
tial in ammation.93,323–325 proved NO production.262,329–335
Renal cortical cytochrome P450-2 epoxygenase pro- Sodium restriction increases plasma renin activity and
tein levels in renal microvessels and urinary metabolite AT1-receptor dependent oxidative stress in the kidneys as
EET excretion are increased during the consumption of a urinary excretion of 8-isoprostane is increased. Chronic
high salt diet. Epoxygenase products such as 11,12-EET activation of the renin–angiotensin system induces oxida-
exert antihypertensive effects as a result of vasodilator and tive stress in the kidney. The administration of Ang II act-
natriuretic actions. Epoxygenase inhibition leads to salt- ing on AT1 receptors enhances renal cortical NOX1, p22phox
sensitive hypertension. In contrast, cytochrome P450-4A expression, superoxide production, and urinary excretion
-hydroxylase protein levels are reduced in the renal cortex of 8-isoPGF2 (isoprostane). mRNA is decreased for NOX4
and in the renal vasculature. Nevertheless, a high salt diet and extracellular superoxide dismutase. Although weaker,
increases overall renal excretion of 20-HETE in an adap- AT2 receptors tend to blunt the actions of AT1 receptors. The
tive response that contributes to increased sodium excre- administration of the superoxide dismutase tempol increases
tion. The pharmacologic inhibition of 20-HETE formation RBF and arterial pressure, whereas GFR and sodium excre-
reduces sodium excretion and leads to salt-sensitive hyper- tion are unchanged, suggesting predominant actions of
tension in rats.183,318,326 superoxide on the renal vasculature.262,336,337
Adenosine produces more pronounced A1 receptor–
mediated renal vasoconstriction in high renin/Ang II states Changes in Protein Intake
such as during sodium restriction. In contrast, adenosine The Western-style diet is characterized by highly processed
produces dilation mediated by A2 receptors in animals on a and re ned foods with a high content of sugars, salt, and
high salt diet, with increases observed in both the renal cor- fat, and high protein from meat that is chronically associated
tex and the outer and inner medulla. The reduction in RVR with dyslipidemia, oxidative stress, and in ammation. It is a
is due in part to enhanced cytochrome P450 activity and major contributor to metabolic disturbances and the devel-
EET production during high salt intake. The pharmacologic opment of obesity-related diseases, including type 2 diabe-
stimulation of A1 receptors decreases RBF and perfusion to tes, hypertension, and cardiovascular and renal disease.
the cortex and both the outer and inner medulla. In contrast, It has long been recognized that variations in the pro-
an A1 receptor agonist causes reductions in RBF and corti- tein diet and plasma amino acid concentrations can have
cal and outer medullary blood ow, but not inner medullar signi cant effects on the renal circulation. The consump-
perfusion in low salt animals.218,327,328 tion of protein in excess of 1 g/kg/day is usually associated
Conditions that shift the balance to favor increased with renal vasodilation in animals and humans. In dogs, the
vasoconstrictor actions of superoxide and other reactive consumption of a high protein meal leads to increases in
oxygen species over the vasodilator buffering effects of NO RBF and GFR, which are maximal at 3 to 6 hours and then
promote salt-sensitive hypertension that is characterized by progressively return to normal by 24 hours. The effect of
renal vasoconstriction and enhanced salt retention. A hall- protein feeding on renal function in humans is less marked
mark of endothelial dysfunction is reduced NO produc- than that in dogs. A short-term intravenous infusion of ca-
tion that can result from a reduced expression of eNOS and sein produces renal vasodilation that is sustained for up to
nNOS and impaired eNOS activation and NO production 8 hours even though the blood amino acid concentrations
124
12 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

rapidly return to preexisting levels after the infusion is consumption of large amounts of protein may exacerbate
stopped. Various combinations of amino acids usually pro- or cause kidney damage. A long-term high protein intake
duce renal vasodilation and increase GFR; the changes are often leads to proteinuria and increased GFR with renal hy-
rapid in onset and reversible. It has been noted that only pertrophy that is accompanied with larger glomeruli and
amino acids that are metabolized dilate the renal vascula- more glomerulosclerosis and tubulointerstitial brosis. The
ture, whereas nonmetabolized amino acids do not affect glomerular hypertrophy is due in part to increased vascular
RBF.338–341 endothelial growth factor production.344,345
The postprandial response to a protein-rich meal or a During a low protein diet, arterial pressure is normal,
response to metabolizable amino acid infusion involves re- whereas plasma renin, Ang II, and aldosterone levels are
nal hyperemia and hyper ltration because both RBF and reduced. Nevertheless, renal renin content is increased and
GFR increase in both humans and animals. The ability of the exerts tonic actions on RVR. RBF and GFR are markedly re-
kidneys to vasodilate and increase RBF and GFR in response duced by protein restriction. The reduction in RBF is due
to an acute protein or amino acid load is used clinically as a to equal increases in both afferent and efferent arteriolar re-
diagnostic index of vascular adaptability of “renal functional sistance. GFR is reduced due to reductions in plasma ow
reserve.” The greater the response, the more adaptable and and the ultra ltration coef cient. ACE inhibition increases
thus the healthier the reserve; a weak or absent response is RBF and GFR while reducing RVR, which is consistent
associated with severe nephron loss and aging. The mecha- with a prominent vasoconstrictor role of intrarenal Ang II.
nisms responsible for the renal vasodilation are multiple, in- Glomerular AT1 receptor density in the renal cortex and the
volving blood-borne vasoactive agents, including pancreatic medulla is increased during low protein feeding. Dietary
glucagon and the intrarenal release of COX-derived vasodi- protein restriction lowers plasma renin activity as a result of
lator prostanoids, NO, as well as reduced renin–Ang II and reduced PGE2 production.346
reduced TGF activity. Plasma renin activity and Ang II levels In general, high protein intake aggravates renal inju-
are unchanged and ACE inhibition does not impact on the ry, whereas restriction of dietary protein (~1 g protein per
protein-induced renal vasodilation.154 kilogram of body weight per day) while avoiding malnutri-
In healthy humans and laboratory animals, a high pro- tion ameliorates the progressive development of glomeru-
tein diet or the infusion of amino acids exert proportional lar disease in various models. Based on animal studies, a
effects on RBF, GFR, and urinary urea nitrogen excretion. low protein diet tends to be renoprotective in that it delays
Renal vascular resistance is reciprocally related to protein damage and proteinuria associated with hypertension, dia-
intake, with larger changes in RBF than in GFR. Arterial betes, obesity, or reduced renal mass and chronic renal dis-
pressure remains stable and is independent of protein in- ease, thus reducing GFR, glomerular growth, and interstitial
take. Some reports, however, nd that vegetarians have a in ltrate. A restricted protein diet reduces uremia, the pro-
reduced GFR relative to omnivores. The high protein diet gression of proteinuria, and the decline in GFR in adult pa-
leads to renal vasodilation and increased RBF and GFR in tients with moderate-to-severe chronic renal disease. Dietary
the face of normal arterial pressure and normal levels of protein restriction reduces the glomerular permselective
plasma renin activity. Increased RBF and GFR are mediated defect responsible for proteinuria in human renal disease.
by COX-dependent prostanoids and are largely prevented Implicated mechanisms include a hemodynamic basis, with
by COX inhibition. Glomerular COX and PLA2 activities are reduced blood ow and glomerular capillary pressure, and
increased, and the glomerular (but not renal papillary) pro- nonhemodynamic factors related to reduced glomerular
duction of PGE2, PGF2 , and TxA2 are increased under basal growth and less oxidative stress and in ammation. A low
conditions and in response to Ang II. The urinary excretion protein diet may reduce RBF by increasing TxA2 production.
of PGE2 and PGF2 metabolites are increased while plasma Local Ang II levels may also participate. A protein restric-
renin activity is elevated. Renal vasodilation is little affected tive diet lowers plasma renin activity by attenuating renin
by Ang II, as protein-induced changes in renal hemodynam- release, mediated in part by reduced PGE2 stimulation, and
ics are unaffected by ACE inhibition. Nevertheless, vascular increases AT1 receptor density in the renal cortex and the
reactivity to Ang II is reduced during high protein feeding, medulla.347–351
whereas that of norepinephrine is normal. One mechanism
contributing to renal vasodilation and increased GFR on a
high protein diet is suppressed TGF control of preglomeru- CONCLUSION
lar vascular resistance. Nevertheless, glomerular hyper l- As is evident from the previous discussion, there are many
tration is observed in adenosine A1 receptor–de cient mice exciting issues concerning the area of renal hemodynamics
lacking TGF.2,111,342,343 and the multiple control mechanisms that are under active
Athletes and exercisers often use high-protein diets to investigation. Modern technologic developments have al-
enhance strength and muscle hypertrophy and recovery lowed a more detailed and a more direct evaluation of the
from intense exercise or injury. High protein diets combined characteristics of speci c segments of the renal microvascu-
with carbohydrate or fat restriction are also advocated for lature and of the various membrane and cellular mechanisms
weight loss. However, it should be appreciated that chronic mediating differential responses. The direct assessment of
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1125
25

responses of individual arterioles has allowed for the clari- 16. Yuan J, Pannabecker
19. Pannabecker TL, DantzlerTL. Architecture o inner medullary
WH. Three-dimensional descending
architecture and
of collect-
ascending
ing ducts, vasaloopsrecta: pathways
of Henle, or countercurrent
and blood vessels in the exchange. Am J Physiol
renal papilla. Renal
Am J Physiol
cation of long-standing controversies. In addition, devel- Physiol. 2010;299(1):F265–F272.
Renal Physiol. 2007;293(3):F696–F704.
opments related to interactions between endothelial cells 17. Cowley AW Jr.
20. Arendshorst WJ, Renal
Thaimedullary oxidative
TL. Regulation of stress, pressure
the renal natriuresis, and
microcirculation by
and vascular smooth muscle cells are now receiving greater hypertension.
ryanodine receptors Hypertension. 2008;52(5):777–786.
and calcium-induced calcium release. Cur Opin Nephrol
http://www.ncbi.nlm.nih.gov/pubmed/18852392
Hypertens. 2009;18(1):40–49.
attention from investigators studying the renal circulation, 18. Evans G,
21. Bastin RG,Heximer
Eppel GA, Anderson WP
SP. Intracellular , et al. Mechanisms
regulation of heterotrimeric underlying
G-proteinthe
as has been the case for interactions between distal tubular di erentialmodulates
signaling control ovascular
blood fsmoothow in the renalcell
muscle medulla and cortex.
contraction. J Hypertens.
Arch Biochem Bio-
macula densa cells and vascular cells of the afferent arteri- 2004;22(8):1439–1451.
phys. 2011;510(2):182–189.
19. Pannabecker
22. Brinks TL, Dantzler
HL, Eckhart WH. Threeofdimensional
AD. Regulation GPCR signaling architecture o collect
in hypertension.
ole and glomerulus. This has led to exciting new concepts ing ducts,
Biochim loopsActa.
Biophys o Henle, and blood vessels in the renal papilla. Am J Physiol
2010;1802(12):1268–1275.
with far-reaching implications. Additionally, many of these Renal
23. Physiol.
Deupi X,2007;293(3):F696–F704.
Kobilka BK. Energy landscapes as a tool to integrate GPCR struc-
integrative mechanisms are now being addressed in terms 20. dynamics,
ture, Arendshorst andWJ, Thai TL.
function. Regulation
Physiology o the renal
(Bethesda). microcirculation by ry
2010;25(5):293–303.
anodine receptors
24. Kobilka BK. and calcium
Structural induced
insights intocalcium release.
adrenergic Cur Opin
receptor Nephrol
function andHyper-
phar-
of dynamic as well as steady-state characteristics. This com- tens. 2009;18(1):40–49.
macology. Trends Pharmacol Sci. 2011;32(4):213–218.
bination of new developments has provided the impetus for 21. Bastin G,JH
25. Chidlow Heximer
Jr, SessaSP . Intracellular
WC. Caveolae, regulation
caveolins, ando heterotrimeric
cavins: complex G protein
control
renewed interest in the area of renal hemodynamics and the signaling
of cellular signalling and in ammation. Cardiovasc Res. 2010;86(2):219–225.Bio-
modulates vascular smooth muscle cell contraction. Arch Biochem
phys.
26. 2011;510(2):182–189.
Cohen AW, Hnasko R, Schubert W, et al. Role of caveolae and caveolins in
interactions with other intrarenal systems. These new inves- http://www.ncbi.nlm.nih.gov/pubmed/21616051
health and disease. Physiol Rev. 2004;84(4):1341–1379.
tigations should result in a much better appreciation of the 22. Brinks
27. Insel PA,HL,
PatelEckhart AD. Regulation
HH. Membrane rafts ando GPCR
caveolae signaling in hypertension.
in cardiovascular signal-
exact mechanisms that regulate renal microvascular contrac- Biochim Biophys Acta. 2010;1802(12):1268–1275.
ing. Curr Opin Nephrol Hypertens. 2009;18(1):50–56.
http://www.ncbi.nlm.nih.gov/pubmed/20060896
28. Ushio-Fukai M, Alexander RW. Caveolin-dependent angiotensin II type
tility and reactivity and how disruptions of these mecha- 23. Deupisignaling
X, KobilkainBK. Energysmooth
landscapes as a tool to integrate GPCR struc
1 receptor vascular muscle. Hypertension. 2006;48(5):
nisms can lead to or predispose the kidneys to dysfunction ture, dynamics, and unction. Physiology (Bethesda). 2010;25(5):293–303.
797–803.
and increased injury. http://www.ncbi.nlm.nih.gov/pubmed/20940434
29. Luttrell LM, Gesty-Palmer D. Beyond desensitization: physiologi-
24.relevance
cal Kobilka of BK.arrestin-dependent
Structural insights signaling.
into adrenergicPharmacolreceptor
Rev. unction
2010;62(2):and
phar macology. Trends Pharmacol Sci. 2011;32(4):213–218.
305–330.
REFERENCES 25. Chidlow
30. Penela JH Jr, Sessa
P, Murga C, RibasWC. C,Caveolae, caveolins,Gand
et al. The complex cavins: complex
protein-coupled con
receptor
trol o cellular
kinase 2 (GRK2) signalling and inf
interactome ammation.
unveils Cardiovasc Res. 2010;86(2):219–225.
new physiopathological targets. Br J Phar-
Navar LG,
1. Navar LG, Inscho
Inscho EW, EW, Majid
Majid SA,SA, et
et al.
al. Paracrine
Paracrine regulation
regulation ofo the
the renal 26. Cohen
macol. AW, Hnasko R, Schubert W, et al. Role o caveolae and caveolins in
2010;160(4):821–832.
microcirculation. Physiol Rev. 1996;76(2):425–436. health and disease.
31. Reiter E, AhnPhysiol
S, ShuklaRev. 2004;84(4):1341–1379.
AK, et al. Molecular mechanism of -arrestin-
http://www.ncbi.nlm.nih.gov/pubmed/8618962
2. Navar LG, Arendshorst WJ, Pallone TL, et al. The renal microcirculation. http://www.ncbi.nlm.nih.gov/pubmed/15383654
biased agonism at seven-transmembrane receptors. Annu Rev Pharmacol Toxicol.
In:2. Navar
Tuma LG, Arendshorst
R, Duran WN, Ley N,WJ, eds.Pallone TL, et al.ofThe
APS Handbook renal microcirculation.
Physiology, Microcirculation. 27. Insel PA, Patel HH. Membrane ra ts and caveolae in cardiovascular signal
2012;52:179–197.
In: Tuma
2nd R, Duran
ed. Boston: WN, Ley
Academic N, eds.
Press APS Handbook
(Elsevier); of Physiology, Microcirculation.
2008;550–683. ing.
32.Curr
BansalOpinG,Nephrol
Druey Hypertens.
KM, Xie Z.2009;18(1):50–56.
R4 RGS proteins: regulation of G-protein sig-
2nd 3.ed. Boston:
Deng Academic
A, Miracle CM,Press (Elsevier);
Lortie M, et al.2008;550–683.
Kidney oxygen consumption, car- http://www.ncbi.nlm.nih.gov/pubmed/19077689
naling and beyond. Pharmacol Ther. 2007;116(3):473–495.
3. anhydrase,
bonic Deng A, Miracle CM, Lortie
and proton M, et
secretion. Amal.J Physiol
KidneyRenal
oxygen consumption,
Physiol. car
2006;290(5): 28. Ushio Fukai
33. Francis SH, BuschM, Alexander
JL, Corbin RW.JD,Caveolin dependent angiotensin
et al. cGMP-dependent II type
protein kinases
bonic anhydrase, and proton secretion. Am J Physiol Renal Physiol. 2006;290(5):
F1009–F1015. 1andreceptor signaling in vascular
cGMP phosphodiesterases smooth
in nitric oxide muscle.
and cGMP Hypertension. 2006;48(5):
action. Pharmacol Rev.
F1009–F1015.
4. Evans RG, Goddard D, Eppel GA, et al. Factors that render the kidney sus- 797–803.
2010;62(3):525–563.
http://www.ncbi.nlm.nih.gov/pubmed/16339960
ceptible to tissue hypoxia in hypoxemia. Am J Physiol Regul Integr Comp Physiol. http://www.ncbi.nlm.nih.gov/pubmed/17015782
34. Gurley SB, Grif ths RC, Mendelsohn ME, et al. Renal actions of RGS2
4. Evans RG, Goddard D, Eppel GA, et al. Factors that render the kidney sus
2011;300(4):R931–R940. 29. Luttrell
control LM, Gesty
blood pressure. J AmPalmer D. Beyond
Soc Nephrol. desensitization: physiological rel
2010;21(11):1847–1851.
ceptible to tissue
5. Evans hypoxia in
RG, Goddard D, hypoxemia.
Eppel GA, etAm al.JStability
Physiol Regul Integr
of tissue Comp
PO2 Physiol.
in the face evance o arrestin
35. Somlyo AP, dependent
Somlyo AVsignaling. Pharmacol of
. Ca2 sensitivity Rev.smooth
2010;62(2):
muscle 305–330.
and non-
2011;300(4):R931–R940.
of altered perfusion: a phenomenon speci c to the renal cortex and independent 30. Penela
muscle myosin P, Murga C, Ribasby
II: modulated C, G
et proteins,
al. The complex
kinases,Gand protein
myosin coupled receptor
phosphatase.
of 5. Evans
resting RG,oxygen
renal Goddard D, Eppel GA,Clin
consumption. et al.
ExpStability
Pharmacolo tissue PO22011;38(4):
Physiol. in the ace kinase
Physiol 2Rev.(GRK2) interactome unveils new physiopathological targets. Br J Phar-
2003;83(4):1325–1358.
o altered per usion: a phenomenon speci c to the renal cortex and independent
247–254. macol. 2010;160(4):821–832.
36. Arendshorst WJ, Brännström K, Ruan X. Actions of angiotensin II on the
o 6.resting renal BS,
Gardiner oxygen
Smithconsumption.
DW, O’Connor ClinPM,ExpetPharmacol Physiol. 2011;38(4):
al. A mathematical model of 31. Reiter E,
renal microvasculature.Ahn S, JShukla
Am SocAK, et al.1999;10(Suppl
Nephrol. Molecular mechanism o b -arrestin-
11):S149–S161.
247–254. shunting of oxygen from arteries to veins in the kidney. Am J Physiol
diffusional biased agonism at
37. Facemire CS,seven transmembrane
Arendshorst receptors. Annu
WJ. Calmodulin Rev Pharmacol
mediates Toxicol.
norepinephrine-
6. Physiol.
Renal Gardiner BS, Smith DW, O’Connor PM, et al. A mathematical model o
2011;300(6):F1339–F1352. 2012;52:179–197.
induced receptor-operated calcium entry in preglomerular resistance arteries.
di 7.usional
Palm shunting
F, Nordquist o oxygen
L. Renalrom arteries
oxidative to veins
stress, in the kidney.
oxygenation Am J Physiol
and hypertension. Am32.J Physiol
BansalRenal
G, Druey KM,2005;289(1):F127–F136.
Physiol. Xie Z. R4 RGS proteins: regulation o G protein sig
Renal
Am Physiol.Regul
J Physiol 2011;300(6):F1339–F1352.
Integr Comp Physiol. 2011;301(5):R1229–R1241. naling and beyond. K,
38. Loutzenhiser Pharmacol
LoutzenhiserTher. 2007;116(3):473–495.
RD. Angiotensin II-induced Ca(2 ) in ux
7. Palm F, Nordquist
8. Welch WJ. Intrarenal L. Renal
oxygen oxidative stress, oxygenation
and hypertension. and hypertension.
Clin Exp Pharmacol Physiol. http://www.ncbi.nlm.nih.gov/pubmed/18006065
in renal afferent and efferent arterioles: differing roles of voltage-gated and store-
Am J Physiol Regul Integr Comp Physiol. 2011;301(5):R1229–R1241.
2006;33(10):1002–1005. 33. Francis
operated Ca(2 SH, BuschCirc
) entry. JL, Res.
Corbin JD, et al. cGMP dependent protein kinases
2000;87(7):551–557.
8. Jelkmann
9. Welch WJ.W. Intrarenal
Regulationoxygen and hypertension.
of erythropoietin Clin Exp Pharmacol
production. Physi-
J Physiol. 2011; and39.cGMP
Cahalan phosphodiesterases
MD. Stimulating in store-operated
nitric oxide andCa(2 cGMP) action.
entry. Pharmacol Rev.
Nat Cell Biol.
ol. 2006;33(10):1002–1005.
589(Pt 6):1251–1258. 2010;62(3):525–563.
2009;11(6):669–677.
http://www.ncbi.nlm.nih.gov/pubmed/17002680
10. Lee FS, Percy MJ. The HIF pathway and erythrocytosis. Annu Rev Pathol. http://www.ncbi.nlm.nih.gov/pubmed/20716671
40. Dietrich A, Kalwa H, Gudermann T. TRPC channels in vascular cell func-
9. Jelkmann W. Regulation o erythropoietin production. J Physiol. 2011;
2011;6:165–192. 34. Gurley Haemost.
tion. Thromb SB, Gri 2010;103(2):262–270.
ths RC, Mendelsohn ME, et al. Renal actions o RGS2
589(Pt 6):1251–1258.
11. Pruijm M, Hofmann L, Maillard M, et al. Effect of sodium loading/depletion control bloodSE,
41. Dryer pressure.
Reiser J.J Am TRPC6Soc Nephrol.
channels2010;21(11):1847–1851.
and their binding partners in podo-
http://www.ncbi.nlm.nih.gov/pubmed/21078592
on renal oxygenation in young normotensive and hypertensive men. Hypertension. http://www.ncbi.nlm.nih.gov/pubmed/20847141
cytes: role in glomerular ltration and pathophysiology. Am J Physiol Renal
10. Lee FS, Percy MJ. The HIF pathway and erythrocytosis. Annu Rev Pathol.
2010;55(5):1116–1122. 35. Somlyo
Physiol. AP, Somlyo AV Ca2+ sensitivity o smooth muscle and non muscle
2010;299(4):F689–F701.
2011;6:165–192.
12. Bell PD, Komlosi P, Zhang ZR. ATP as a mediator of macula densa cell myosin II: modulated
42. Facemire CS, Mohlerby G PJ, proteins, kinases,WJ.
Arendshorst andExpression
myosin phosphatase.
and relative Physiol
abun-
http://www.ncbi.nlm.nih.gov/pubmed/20939709
signalling. Purinergic Signal. 2009;5(4):461–471. Rev.
dance2003;83(4):1325–1358.
of short transient receptor potential channels in the rat renal microcircula-
11. Kurtz
13. PruijmA.M, Ho mann
Renin release:L, sites,
Maillard M, et al. Eand
mechanisms, ect control.
o sodium loading/deple
Annu Rev Physiol. 36. Arendshorst
tion. Am J Physiol Renal WJ, Physiol.
Brännström K, Ruan X. Actions o angiotensin II on the
2004;286(3):F546–F551.
tion on renal
2011;73:377–399. oxygenation in young normotensive and hypertensive men. Hyper- renal
43. microvasculature.
Fellner SK, Arendshorst J Am Soc WJ.Nephrol. 1999;10(Suppl
Angiotensin II-stimulated11):S149–S161.
Ca2 entry mech-
tension. 2010;55(5):1116–1122.
14. Schnermann J, Levine DZ. Paracrine factors in tubuloglomerular feedback: 37. Facemire CS, Arendshorst WJ. Calmodulin mediates
anism in afferent arterioles: role of transient receptor potential canonical chan- norepinephrine in
http://www.ncbi.nlm.nih.gov/pubmed/20308608
adenosine, ATP, and nitric oxide. Annu Rev Physiol. 2003;65:501–529. duced
nels and receptor
reverseoperated
Na /Ca2 calcium entry inAm
exchange. preglomerular
J Phyisol Renal resistance arteries. Am J
Physiol. 2008;294(1):
12. Pallone
15. Bell PD,TL, Komlosi
ZhangP, Z,Zhang ZR. ATP
Rhinehart K. as a mediator
Physiology of othemacula densa cell
renal medullary Physiol
F212–F219.Renal Physiol. 2005;289(1):F127–F136.
signalling. Purinergic
microcirculation. AmSignal. 2009;5(4):461–471.
J Physiol Renal Physiol. 2003;284(2):F253–F266. 38. Loutzenhiser K,
44. Salomonsson M, Loutzenhiser
Braunstein TH, RD. Holstein-Rathlou
Angiotensin II induced NH, etCa(2+) inf ux
al. Na( )-
http://www.ncbi.nlm.nih.gov/pubmed/19330465
16. Yuan J, Pannabecker TL. Architecture of inner medullary descending and in renal a erent and e erent arterioles: di ering roles
independent, nifedipine-resistant rat afferent arteriolar Ca(2 ) responses too voltage gated and store
13. Kurtz
ascending A. recta:
vasa Renin pathways
release: sites, mechanisms, and
for countercurrent control.Am
exchange. Annu Rev Physiol.
J Physiol Renal operated Ca(2+)possible
noradrenaline: entry. CircroleRes. 2000;87(7):551–557.
of TRPC channels. Acta Physiol (Oxf). 2010;200(3):
2011;73:377–399.
Physiol. 2010;299(1):F265–F272. http://www.ncbi.nlm.nih.gov/pubmed/11009559
265–278.
http://www.ncbi.nlm.nih.gov/pubmed/20936939
17. Cowley AW Jr. Renal medullary oxidative stress, pressure-natriuresis, and 39. CahalanJT,MD.
45. Smyth Hwang Stimulating
SY, Tomitastore T, etoperated Ca(2+)
al. Activation andentry. Nat Cell
regulation Biol.
of store-
14. Schnermann
hypertension. J, Levine2008;52(5):777–786.
Hypertension. DZ. Paracrine actors in tubuloglomerular eedback: 2009;11(6):669–677.
operated calcium entry. J Cell Mol Med. 2010;14(10):2337–2349.
adenosine,
18. EvansATP RG,, and nitric
Eppel oxide.
GA, Annu Rev
Anderson WPPhysiol.
, et al. 2003;65:501–529.
Mechanisms underlying the http://www.ncbi.nlm.nih.gov/pubmed/19488056
46. Takenaka T, Suzuki H, Okada H, et al. Transient receptor potential chan-
http://www.ncbi.nlm.nih.gov/pubmed/12208992
differential control of blood ow in the renal medulla and cortex. J Hypertens. nels inDietrich
40. rat renal A, microcirculation:
Kalwa H, Gudermann actions T. TRPC channels in
of angiotensin II. vascular
Kidney Int.cell2002;
unc
15. Pallone TL, Zhang Z, Rhinehart K. Physiology o the renal medullary mi
2004;22(8):1439–1451. tion. Thromb Haemost. 2010;103(2):262–270.
62(2):558–565.
crocirculation. Am J Physiol Renal Physiol. 2003;284(2):F253–F266. http://www.ncbi.nlm.nih.gov/pubmed/20126834
126
12 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

41. Varnai
47. Dryer PSE, Reiser J.L,TRPC6
, Hunyady Balla T.channels
STIM andand Orai:their
thebinding
long-awaited partners in podo
constituents 64. Rosivall
76. Hansen L, PB,Mirzahosseini
Poulsen CB, Walter S, TomaS, I,etetal.al.Functional
Fluid owimportance o L and
in the juxtaglomer-
cytes:
of role in glomerular
store-operated calcium entry.ltration and pathophysiology.
Trends Am J Physiol Renal Physi-
Pharmacol Sci. 2009;30(3):118–128. P/Q type
ular voltage gated
interstitium calcium
visualized channels
in vivo. Am in human Renal
J Physiol renal vasculature. Hyperten-
Physiol. 2006;291(6):
ol.48.
2010;299(4):F689–F701.
Blaustein MP, Hamlyn JM. Signaling mechanisms that link salt retention sion. 2011;58(3):464–470.
F1241–F1247.
to42. Facemire CS,
hypertension: Mohler PJ,ouabain,
endogenous Arendshorst the Na(WJ. Expression
) pump, the andNa( relative abun)
)/Ca(2 http://www.ncbi.nlm.nih.gov/pubmed/21788606
77. Arendshorst WJ, Gottschalk CW. Glomerular ultra ltration dynamics:
dance o short
exchanger and transient
TRPC proteins. receptor potential
Biochim channels
Biophys in the rat renal microcircula
Acta. 2010;1802(12):1219–1229. 65. Hayashi
historical K, Wakino
perspective. Am JS,Physiol.
Sugano1985;248(2
N, et al. Ca2+ channel subtypes and phar
Pt 2):F163–F174.
tion.
49. Am J Physiol
Schweda Renal Physiol.
F, Kramer BK, Kurtz2004;286(3):F546–F551.
A. Differential roles of the sodium-calcium macology
78. Axelssonin theJ,kidney.
Rippe Circ Res. 2007;100(3):342–353.
A, Rippe B. Transient and sustained increases in glo-
43. Fellner
exchanger SK, Arendshorst
in renin secretion and WJ.renal
Angiotensin
vascular IIresistance.
stimulated P Ca2+
ugers entry
Arch. mech
2001; http://www.ncbi.nlm.nih.gov/pubmed/17307972
merular permeability following ANP infusion in rats. Am J Physiol Renal Physiol.
anism in a erent arterioles: role o transient receptor potential canonical chan
442(5):693–699. 66. Poulsen CB, Al Mashhadi RH, Cribbs LL, et al. T type voltage gated
2011;300(1):F24–F30.
nels
50. and
Zhao reverse
D, Zhang Na+/Ca2+ exchange.
J, Blaustein MP, etAm al. JAttenuated
Phyisol Renal renal Physiol.
vascular 2008;294(1):
responses calcium
79. Comper channels WD,regulateRusso LM. the tone o mouse e lter:
The glomerular erentanarterioles.
imperfectKidney
barrierInt.is
F212–F219.
to acute angiotensin II infusion in smooth muscle-speci c Na /Ca2 exchanger 2010;79(4):
required for 443–451.
perfect renal function. Curr Opin Nephrol Hypertens. 2009;18(4):
44. Salomonsson
knockout mice. Am JM, Braunstein
Physiol TH, Holstein
Renal Physiol. Rathlou NH, et al. Na(+) inde
2011;301(3):F574–F579. http://www.ncbi.nlm.nih.gov/pubmed/21068717
336–342.
pendent,
51. Chilton ni edipine resistant
L, Smirnov SV,rat a erent arteriolar
Loutzenhiser K, et al. Ca(2+) responses to
Segment-speci noradrena
c differences 67. Hausmann
80. Zhang Z, LinR,H,Kuppe Cao C,C,et Egger
al. Voltage
H, etgated divalent currents
al. Electrical in descending
forces determine glo-
line:
in thepossible
inward role rectio er TRPC
K channels.
current along Acta thePhysiol
renal (Ox ). 2010;200(3):
interlobular artery.265–278.
Cardiovasc vasa recta
merular pericytes. Am
permeability. J Physiol
J Am Renal Physiol.
Soc Nephrol. 2010;299(4):F862–F871.
2010;21(12):2053–2058.
45.2011;92(1):169–177.
Res. Smyth JT, Hwang SY, Tomita T, et al. Activation and regulation o store 68. Navar
81. HerreraLG. GA,Glomerular
Turbat Herrera EA, Teng J.a Mesangial
permeability: never-ending homeostasis
saga. Am and patho
J Physiol
operated
52. Flagg calcium entry. J Cell Mol
TP, Enkvetchakul Med. 2010;14(10):2337–2349.
D, Koster JC, et al. Muscle KATP channels: recent biology:
Renal their 2009;296(6):F1266–F1268.
Physiol. role in health and disease. Contrib Nephrol. 2011;169:6–22.
http://www.ncbi.nlm.nih.gov/pubmed/20807283
insights to energy sensing and myoprotection. Physiol Rev. 2010;90(3):799–829. 69. Peti-Peterdi
82. Ma R, Pluznick JL, Sansom
J, Sipos SC. Ion channels
A. A high-powered view of in the
mesangial
ltrationcells: unction,
barrier. J Am
46. Troncoso
53. Takenaka Brindeiro
T, Suzuki CM, H, OkadaFalletH,RW, et al.
LaneTransient
PH, etreceptor potential
al. Potassium chan
channel malNephrol.
Soc unction,2010;21(11):1835–1841.
or ction. Physiology (Bethesda). 2005;20:102–111.
nels in rat renal
contributions microcirculation:
to afferent arteriolar tone actions o angiotensin
in normal and diabetic II. Kidney Int. 2002;
rat kidney. Am J 70. Tanner
83. Schlondor GA, D, Banas
Rippe C, B.Shao
The Y mesangial cell revisited:
, et al. Glomerular no cell is an
permeability to island.
macro-J
62(2):558–565.
Physiol Renal Physiol. 2008;295(1):F171–F178. Am Soc Nephrol.
molecules in the2009;20(6):1179–1187.
Necturus kidney. Am J Physiol Renal Physiol. 2009;296(6):
http://www.ncbi.nlm.nih.gov/pubmed/12110018
54. Wu RS, Marx SO. The BK potassium channel in the vascular smooth mus- http://www.ncbi.nlm.nih.gov/pubmed/19470685
F1269–F1278.
47.
cle andVarnai
kidney: P, Hunyady L, Balla T. STIM
- and -subunits. Kidneyand Int.Orai: the long awaited constituents
2010;78(10):963–974. 71. Patrakka
84. Hao CM, J, Breyer MD. Physiological
Tryggvason regulation
K. New insights intoo theprostaglandins in the kid
role of podocytes in
o55.store operated
Bauer calcium
J, Parekh entry. Trends
N. Variations in Pharmacol
cell signaling Sci. pathways
2009;30(3):118–128.
for different va- ney. Annu Rev
proteinuria. NatPhysiol. 2008;70:357–377.
Rev Nephrol. 2009;5(8):463–468.
48. Blaustein
soconstrictor MP, Hamlyn
agonists in renal JM.circulation
Signaling of mechanisms
the rat. Kidney that link Int. salt retention
2003;63(6): http://www.ncbi.nlm.nih.gov/pubmed/17988207
85. Rodriguez-Iturbe B, Johnson RJ. The role of renal microvascular disease
to hypertension: endogenous ouabain, the Na(+) pump, the Na(+)/Ca(2+) ex
2178–2186. and72. interstitial
Sudhahar V in, Shaw S, Imigin
ammation JD.salt-sensitive
Epoxyeicosatrienoic acid analogs
hypertension. and vascu
Hypertens Res.
changer and TRPC
56. Cavarape proteins.
A, Endlich N,Biochim
AssaloniBiophys
R, et al.Acta. 2010;1802(12):1219–1229.
Rho-kinase inhibition blunts renal lar unction. Curr Med Chem. 2010;17(12):1181–1190.
2010;33(10):975–980.
49. Schweda Finduced
vasoconstriction , KramerbyBK, Kurtzsignaling
distinct A. Di erentialpathways rolesin ovivo.theJsodium calcium
Am Soc Nephrol. http://www.ncbi.nlm.nih.gov/pubmed/20158473
86. Steegh FM, Gelens MA, Nieman FH, et al. Early loss of peritubular capil-
exchanger in renin secretion and renal vascular resistance. Pf ugers Arch. 2001;
2003;14(1):37–45. 73. after
laries Segalkidney
MS, Baylis C, JohnsonJ Am
transplantation. RJ. Soc
Endothelial health and diversity in the
Nephrol. 2011;22(6):1024–1029.
442(5):693–699.
57. Inscho EW, Cook AK, Webb RC, et al. Rho-kinase inhibition reduces pres- kidney. J Am Soc
87. Levick JR, Nephrol.
Michel CC. 2006;17(2):323–324.
Microvascular uid exchange and the revised Star-
http://www.ncbi.nlm.nih.gov/pubmed/11512025
sure-mediated autoregulatory adjustments in afferent arteriolar diameter. Am J http://www.ncbi.nlm.nih.gov/pubmed/16434502
ling principle. Cardiovasc Res. 2010;87(2):198–210.
50. Zhao
Physiol RenalD, Zhang2009;296(3):F590–F597.
Physiol. J, Blaustein MP, et al. Attenuated renal vascular responses 74. Oliver
88. MichelG, T, Alitalo
Vanhoutte K. ThePM.lymphatic
Cellular signaling
vasculature: andrecent
NO production.
progress andPfpara-
ugers
to58.
acute
Laiangiotensin
EY, FählingIIM,inMa usion
Z, etinal.smooth muscle speci
Norepinephrine c Na+/Ca2+
increases calciumexchanger
sensitiv- Arch. 2010;459(6):807–816.
digms. Annu Rev Cell Dev Biol. 2005;21:457–483.
knockout
ity of mouse mice. Am Jarteriole,
afferent Physiol Renal Physiol.
thereby 2011;301(3):F574–F579.
enhancing angiotensin II-mediated vaso- http://www.ncbi.nlm.nih.gov/pubmed/20082095
89. Cowley AW Jr, Roman RR. Countercurrent exchange in the renal medulla.
51. ChiltonKidney
constriction. L, Smirnov SV Loutzenhiser K, et al. Segment speci c di erences
Int. 2009;76(9):953–959. Am 75.J Physiol
Haraldsson B, Nyström
Regul Integr J, Deen 2003;284(5):R1151.
Comp Physiol. WM. Properties o the glomerular barrier
in59.
thePatzak
inwardA,recti Lai EY er, K+ current
Fähling M, along the renal interlobular
et al. Adenosine enhances long artery.
termCardiovasc
the con- and
90.mechanisms
Pallone TL, oTurner proteinuria.
MR, Edwards Physiol A,
Revet2008;88(2):451–487.
al. Countercurrent exchange in the
Res. 2011;92(1):169–177.
tractile response to angiotensin II in afferent arterioles. Am J Physiol Regul Integr http://www.ncbi.nlm.nih.gov/pubmed/18391170
renal medulla. Am J Physiol Regul Integr Comp Physiol. 2003;284(5):R1153–R1175.
http://www.ncbi.nlm.nih.gov/pubmed/21697146
Comp Physiol. 2007;293(6):R2232–R2242. 76. Morton
91. RosivallMJ, L, Mirzahosseini
Hutchinson K, S, Toma I, PW,
Mathieson et al.et Fluid f ow podocytes
al. Human in the juxtaglom
possess
52. Purdy
60. Flagg TP KE, , Enkvetchakul
Arendshorst WJ. D, Koster
Iloprost JC,inhibits
et al. Muscle KATP channels: recent
inositol-1,4,5-trisphosphate- aerstretch-sensitive,
ular interstitium Ca2 visualized in vivo.
-activated K Am J Physiol
channel: Renal Physiol.
potential 2006;291(6):
implications for the
insights tocalcium
mediated energy sensing
mobilizationand myoprotection.
stimulated by angiotensinPhysiol Rev. 2010;90(3):799–829.
II in cultured preglo- F1241–F1247.
control of glomerular ltration. J Am Soc Nephrol. 2004;15(12):2981–2987.
http://www.ncbi.nlm.nih.gov/pubmed/20664073
merular vascular smooth muscle cells. J Am Soc Nephrol. 2001;12(1):19–28. 77. Cupples
92. Arendshorst WA,WJ, Gottschalk
Braam CW Glomerular
B. Assessment ultra ltration dynamics:
of renal autoregulation. Am J Physiolhis
53. Hansen
61. Troncoso PB,Brindeiro
Castrop H, CM, Fallet
Briggs J, etRW, Lane PH, et
al. Adenosine al. Potassium
induces channel
vasoconstriction toricalPhysiol.
Renal perspective. Am J Physiol. 1985;248(2 Pt 2):F163–F174.
2007;292(4):F1105–F1123.
contributions
through to a erentactivation
Gi-dependent arteriolaroftone in normal and
phospholipase C indiabetic
isolatedrat kidney.affer-
perfused Am J 78. Just
93. Axelsson J, Rippe A,
A. Mechanisms Rippeblood
of renal B. Transient and sustained
ow autoregulation: increases
dynamics andin con-
glo
Physiol
ent Renal Physiol.
arterioles of mice.2008;295(1):F171–F178.
J Am Soc Nephrol. 2003;14(10):2457–2465. merular permeability
tributions. Am J PhysiolollowingRegul Integr ANP in usion
Comp in rats.
Physiol. Am J Physiol Renal Physiol.
2007;292(1):R1–R17.
54. Fellner
62. Wu RS, SK, MarxArendshorst
SO. The BK WJ. potassium
Complex channel in the vascular
interactions smooth mus
of NO/cGMP/PKG 2011;300(1):F24–F30.
94. Loutzenhiser RD, Grif n KA, Williamson GA, et al. Renal autoregulation:
cle and kidney:
systems on Ca2 a - signaling
and b -subunits.
in afferent Kidney Int. 2010;78(10):963–974.
arteriolar vascular smooth muscle. Am J new79. perspectives
Comper WD, Russo the
regarding LM.protective
The glomerular
and regulatory lter: anrolesimper ectunderlying
of the barrier is
http://www.ncbi.nlm.nih.gov/pubmed/20861815
Physiol Heart Circ Physiol. 2010;298(1):H144–H151. required or per
mechanisms. Am Ject renalRegul
Physiol unction.
Integr Curr
CompOpin Nephrol
Physiol. Hypertens. 2009;18(4):
2006;290(5):R1153–R1167.
55. Feng
63. BauerMG, J, Parekh
Navar N. LG.Variations
Nitric oxide in cell signaling
synthase pathways
inhibition or di L-erent
activates and va
T- 336–342.
95. Bidani AK, Grif n KA, Williamson G, et al. Protective importance of the
soconstrictor
type Ca2 channels agonists in in renaland
afferent circulation o the rat.Am
efferent arterioles. Kidney
J PhysiolInt.Renal
2003;63(6):
Physiol. 80. Hausmann
myogenic responseR,inKuppe the renal C, circulation.
Egger H, etHypertension.
al. Electrical2009;54(2):393–398.
orces determine glo
2178–2186.
2006;290(4):F873–F879. merular permeability.
96. Loutzenhiser RD,J Am
Grif SocnNephrol.
KA, Bidani2010;21(12):2053–2058.
AK. Systolic blood pressure as the
http://www.ncbi.nlm.nih.gov/pubmed/12753305
64. Hansen PB, Poulsen CB, Walter S, et al. Functional importance of L- and http://www.ncbi.nlm.nih.gov/pubmed/20947631
trigger for the renal myogenic response: protective or autoregulatory? Curr Opin
56. Cavarape
P/Q-type A, Endlich
voltage-gated calcium N, channels
Assaloni R, et al. Rho
in human renal kinase inhibition
vasculature. blunts
Hyperten- 81. Navar
Nephrol LG. Glomerular
Hypertens. permeability: a never ending saga. Am J Physiol Re-
2006;15(1):41–49.
renal2011;58(3):464–470.
sion. vasoconstriction induced by distinct signaling pathways in vivo. J Am Soc nal
97.Physiol.
Walker 2009;296(6):F1266–F1268.
M III, Harrison-Bernard LM, Cook AK, et al. Dynamic interaction
Nephrol.
65. Hayashi2003;14(1):37–45.
K, Wakino S, Sugano N, et al. Ca2 channel subtypes and phar- 82. Petimyogenic
between Peterdi J,and Sipos
TGF A. mechanisms
A high powered view o arteriolar
in afferent the ltrationbloodbarrier. J Am
ow auto-
http://www.ncbi.nlm.nih.gov/pubmed/12506136
macology in the kidney. Circ Res. 2007;100(3):342–353. Soc Nephrol.Am
regulation. 2010;21(11):1835–1841.
J Physiol Renal Physiol. 2000;279(5):F858–F865.
57. Poulsen
66. Inscho EW, Cook AK, Webb
CB, Al-Mashhadi RH,RC, Cribbset al.LL,Rho kinase
et al. T-type inhibition
voltage-gatedreduces pres
calcium http://www.ncbi.nlm.nih.gov/pubmed/20576805
98. Balasubramanian L, Ahmed A, Lo CM, et al. Integrin-mediated mechano-
sure mediated
channels regulate autoregulatory
the tone of mouse adjustmentsefferentin arterioles.
a erent arteriolar
Kidney Int. diameter.
2010;79(4):Am J 83. TannerinGA,
transduction renal Rippe C, Shao
vascular smooth Y, et al. Glomerular
muscle permeability
cells: activation of calcium to sparks.
macro
Physiol Renal Physiol. 2009;296(3):F590–F597.
443–451. molecules
Am J PhysiolinRegulthe Integr
NecturusCompkidney.
Physiol.Am J Physiol Renal Physiol. 2009;296(6):
2007;293(4):R1586–R1594.
58. Zhang
67. Lai EY,Z,Fähling
Lin H, M, CaoMa C, Z, et al.
et al. Norepinephrine
Voltage-gated divalentincreases
currents calcium sensitiv
in descending F1269–F1278.
99. Jernigan NL, Drummond HA. Myogenic vasoconstriction in mouse renal
ity o recta
vasa mouse a erentAm
pericytes. arteriole,
J Physiol thereby enhancing
Renal Physiol. angiotensin II mediated vaso
2010;299(4):F862–F871. http://www.ncbi.nlm.nih.gov/pubmed/19339627
interlobar arteries: role of endogenous beta and gamma ENaC. Am J Physiol Renal
constriction.
68. HerreraKidney Int. 2009;76(9):953–959.
GA, Turbat-Herrera EA, Teng J. Mesangial homeostasis and patho- 84. Patrakka
Physiol. J, Tryggvason K. New insights into the role o podocytes in pro
2006;291(6):F1184–F1191.
http://www.ncbi.nlm.nih.gov/pubmed/19625991
biology: their role in health and disease. Contrib Nephrol. 2011;169:6–22. teinuria.
100. GuanNatZ,Rev Nephrol.
Pollock JS,2009;5(8):463–468.
Cook AK, et al. Effect of epithelial sodium channel
59. Ma
69. Patzak A, Lai EY
R, Pluznick , Fähling
JL, SansomM,SC. et al.
IonAdenosine
channels in enhances
mesangial longcells:
termfunction,
the con 85. Rodriguez
blockade on the Iturbe myogenic B, Johnson
responseRJ. Thejuxtamedullary
of rat role o renal microvascular
afferent arterioles.dis
tractile response
malfunction, or to angiotensin
ction. Physiology II (Bethesda).
in a erent 2005;20:102–111.
arterioles. Am J Physiol Regul Integr ease and interstitial
Hypertension. inf ammation in salt sensitive hypertension. Hypertens Res.
2009;54(5):1062–1069.
Comp Physiol. 2007;293(6):R2232–R2242.
70. Schlöndorff D, Banas B. The mesangial cell revisited: no cell is an island. 2010;33(10):975–980.
101. Wang X, Takeya K, Aaronson PI, et al. Effects of amiloride, benzamil, and
60. Soc
J Am Purdy KE, Arendshorst
Nephrol. WJ. Iloprost inhibits inositol 1,4,5 trisphosphate
2009;20(6):1179–1187. http://www.ncbi.nlm.nih.gov/pubmed/20686485
alterations in extracellular Na on the rat afferent arteriole and its myogenic
mediated
71. Hao calcium
CM, Breyer mobilization stimulated by
MD. Physiological angiotensin
regulation II in cultured preglo
of prostaglandins in the 86. Steegh
response. Am FM, Gelens
J Physiol MA,
Renal Nieman
Physiol. FH, et al. Early loss o peritubular capil
2008;295(1):F272–F282.
merularAnnu
kidney. vascular smooth 2008;70:357–377.
Rev Physiol. muscle cells. J Am Soc Nephrol. 2001;12(1):19–28. laries aDaniels
102. ter kidney transplantation.
FH, Arendshorst WJ, JRoberds
Am Soc Nephrol. 2011;22(6):1024–1029.
RG. Tubuloglomerular feedback and
61. Sudhahar
72. Hansen PB, Castrop
V, Shaw H, Briggs
S, Imig J, et al. Adenosine induces
JD. Epoxyeicosatrienoic acid analogs vasoconstriction
and vascu- http://www.ncbi.nlm.nih.gov/pubmed/21566051
autoregulation in spontaneously hypertensive rats. Am J Physiol. 1990;258(6 Pt 2):
through
lar Gi dependent
function. Curr Med Chem. activation o phospholipase C in isolated per used a erent
2010;17(12):1181–1190. 87. Levick JR, Michel CC. Microvascular f uid exchange and the revised Star
F1479–F1489.
arterioles
73. Segalo MS, mice.Baylis
J Am C, SocJohnson
Nephrol. RJ. 2003;14(10):2457–2465.
Endothelial health and diversity in the ling principle.
103. CardiovascJYRes.
Bell PD, Lapointe 2010;87(2):198–210.
, Peti-Peterdi J. Macula densa cell signaling. Annu Rev
62. Fellner
kidney. J Am Soc SK, Arendshorst
Nephrol. WJ. Complex interactions o NO/cGMP/PKG
2006;17(2):323–324. http://www.ncbi.nlm.nih.gov/pubmed/20200043
Physiol. 2003;65:481–500.
systems
74. Michel on Ca2+ signalingPM.
T, Vanhoutte in aCellular
erent arteriolar
signalingvascular
and NOsmooth production.muscle. Am J
P ugers 88. Schnermann
104. Oliver G, Alitalo K. The
J, Briggs lymphatic vasculature:
JP. Tubuloglomerular recentmechanistic
feedback: progress and para
insights
Physiol
Arch. Heart Circ Physiol. 2010;298(1):H144–H151.
2010;459(6):807–816. digms.
from Annu Rev Cell Dev
gene-manipulated Biol. Kidney
mice. 2005;21:457–483.
Int. 2008;74(4):418–426.
63. Haraldsson
75. Feng MG, Navar B, NyströmLG. Nitric
J, Deenoxide WM. synthase
Properties inhibition activates L barrier
of the glomerular and T http://www.ncbi.nlm.nih.gov/pubmed/16212503
105. Sipos A, Vargas S, Peti-Peterdi J. Direct demonstration of tubular uid ow
typemechanisms
and Ca2+ channels in a erent and
of proteinuria. e erent
Physiol arterioles. Am J Physiol Renal Physiol.
Rev 2008;88(2):451–487. 89. Cowley
sensing by macula AWdensa Jr, Roman
cells. AmRR.J Physiol
Countercurrent
Renal Physiol. exchange in the renal medulla.
2010;299(5):F1087–F1093.
2006;290(4):F873–F879. Am J Physiol Regul Integr Comp Physiol. 2003;284(5):R1151.
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1127
27

90. Sosnovtseva
106. Pallone TL, OV Turner
, PavlovMR,AN, Edwards
Mosekilde A, E,et etal. al.Countercurrent
Synchronization exchange
among 115. Vallon V
134. Salazar , Traynor
FJ, Llinas MT. T, Barajas L, et al. Feedback
Renal hemodynamic control
effects o glomerular
elicited by acute cyclo- vas
in the renal ofmedulla.
mechanisms Am J Physiolis Regul
renal autoregulation reduced Integr Comp Physiol.
in hypertensive rats.2003;284(5):
Am J Physiol cular tone ininhibition
oxygenase-2 neuronal are nitric
notoxide
related synthase knockout
to angiotensin mice. Am
II levels. J Am Soc Nephrol.
J Physiol Renal
R1153–R1175.
Renal Physiol. 2007;293(5):F1545–F1555. 2001;12(8):1599–1606.
Physiol. 2010;299(5):F952–F953.
91. Braam
107. MortonB,MJ, Hutchinson
Mitchell K, Mathieson
KD, Koomans HA, et PW,al.etRelevance
al. Human of podocytes
the tubuloglo-pos 116. Green RC.
135. Harris T, Gonzalez
An update AA,on Mitchell KD, et al. The
cyclooxygenase-2 complexand
expression interplay between
metabolites in
sess a stretch
merular sensitive,
feedback Ca2+ activated
mechanism K+ channel:Jpotential
in pathophysiology. implications
Am Soc Nephrol. or the
1993;4(6): cyclooxygenase
the kidney. Curr 2Opin andNephrol
angiotensin II in 2008;17(1):64–69.
Hypertens. regulating kidney unction. Curr Opin
control o glomerular ltration. J Am Soc Nephrol. 2004;15(12):2981–2987.
1257–1274. Nephrol Hypertens.
136. Nguyen 2012;21(1):7–14.
G, Muller DN. The biology of the (pro)renin receptor. J Am Soc
92. Schnermann
108. Cupples WA,J.Braam HomerB.W. Assessment
Smith Award o renal autoregulation.
lecture. The juxtaglomerularAm J Physiol
ap- 117.
Nephrol.Ichihara A, Imig JD, Navar LG. Cyclooxygenase 2 modulates a erent arte
2010;21(1):18–23.
Renal Physiol.
paratus: from 2007;292(4):F1105–F1123.
anatomical peculiarity to physiological relevance. J Am Soc Nephrol. riolar
137. responses
Ichihara A,to SakodaincreasesM,inKurauchi-Mito
pressure. Hypertension.A, et al.1999;34(4 Pt 2):843–847.
Renin, prorenin and the
93. Just A. Mechanisms o renal blood f ow autoregulation: dynamics and con
2003;14(6):1681–1694. http://www.ncbi.nlm.nih.gov/pubmed/10523371
kidney: a new chapter in an old saga. J Nephrol. 2009;22(3):306–311.
tributions.
109. Castrop AmH, J Physiol
HuangRegul Integr Comp
Y, Hashimoto Physiol.
S, et 2007;292(1):R1–R17.
al. Impairment of tubuloglomerular 118. Roman RJ.
138. Bosnyak S, PJones
450 ES,
metabolites o arachidonic
Christopoulos A, et al. acid in the
Relative af control
nity of oangioten-
cardio
94. Loutzenhiser
feedback regulation RD, of GFRGri inn ecto-5
KA, Williamson GA, et al. Renal
-nucleotidase/CD73-de autoregulation:
cient mice. J Clin vascular
sin peptidesunction.
and novel Physiol Rev. 2002;82(1):131–185.
ligands at AT1 and AT2 receptors. Clin Sci (Lond). 2011;
new perspectives
Invest. regarding the protective and regulatory roles o the underlying
2004;114(5):634–642. http://www.ncbi.nlm.nih.gov/pubmed/11773611
121(7):297–303.
mechanisms.
110. Inscho EW, Am JCook PhysiolAK, RegulImigIntegr
JD,Comp
et al.Physiol. 2006;290(5):R1153–R1167.
Physiological role for P2X1 recep- 119.
139. de RenGasparo
Y, D’Ambrosio
M, Catt MA, Garvin T,
KJ, Inagami JL,etetal.al.International
Possible mediatorsunion of o pharmacol-
connecting
95.inBidani
tors AK, Gri n KA,
renal microvascular Williamson G,
autoregulatory et al. Protective
behavior. J Clin Invest. importance o the
2003;112(12): tubule glomerular
ogy. XXIII. eedback.IIHypertension.
The angiotensin 2009;53(2):319–323.
receptors. Pharmacol Rev. 2000;52(3):415–472.
myogenic response in the renal circulation. Hypertension. 2009;54(2):393–398.
1895–1905. http://www.ncbi.nlm.nih.gov/pubmed/19047578
140. Harrison-Bernard LM, Zhuo J, Kobori H, et al. Intrarenal AT(1) recep-
http://www.ncbi.nlm.nih.gov/pubmed/19546375
111. Singh P, Thomson SC. Renal homeostasis and tubuloglomerular feedback. 120.
tor andWangACEH, GarvininJL,ANG
binding D’Ambrosio
II-induced MA, et al. Connecting
hypertensive rats. Am tubule
J Physiolglomeru
Renal
96. Opin
Curr Loutzenhiser RD, Gri 2010;19(1):59–64.
Nephrol Hypertens. n KA, Bidani AK. Systolic blood pressure as the lar eedback
Physiol. antagonizes tubuloglomerular eedback in vivo. Am J Physiol Renal
2002;282(1):F19–F25.
triggerSun
112. or D,
theSamuelson
renal myogenic LC, Yresponse:
ang T, et al. protective
Mediation or ofautoregulatory?
tubuloglomerular Curr feed-
Opin Physiol.
141. Ruan 2010;299(6):F1374–F1378.
X, Oliverio MI, Coffman TM, et al. Renal vascular reactivity in mice:
Nephrol
back by Hypertens.
adenosine:2006;15(1):41–49.
evidence from mice lacking adenosine 1 receptors. Proc Natl 121. Castrop H,vasoconstriction
Ang II-induced Höcherl K, Kurtz A, et al.
in AT1A Physiology
receptor o kidney
null mice. J Amrenin. Physiol
Soc Nephrol.
http://www.ncbi.nlm.nih.gov/pubmed/16340665
Acad Sci USA. 2001;98(17):9983–9988. Rev. 2010;90(2):607–673.
1999;10(12):2620–2630.
97. Araujo
113. WalkerM, M Welch
III, Harrison Bernard stress
WJ. Oxidative LM, Cook AK, etoxide
and nitric al. Dynamic
in kidneyinteraction
function. 122. KomlosiSK,
142. Fellner P, Arendshorst
Fuson AL, Fintha A, et al. Angiotensin
WJ. Angiotensin II Ca2 signaling I conversion to an
in rat afferent
between
Curr Opinmyogenic and TGF mechanisms
Nephrol Hypertens. 2006;15(1):72–77. in a erent arteriolar blood f ow auto giotensin
arterioles: IIstimulation
stimulatesofcortical
cyclic ADP collecting
riboseductand IP3 sodium transport.
pathways. Am JHypertension.
Physiol Renal
regulation.
114. Wilcox AmCS. J Physiol
RedoxRenal Physiol.
regulation 2000;279(5):F858–F865.
of the afferent arteriole and tubuloglomerular 2003;42(2):195–199.
Physiol. 2005;288(4):F785–F791.
98. Balasubramanian
feedback. Acta Physiol Scand. L, Ahmed A, Lo CM, et al. Integrin mediated mechano
2003;179(3):217–223. 123. Navar LG, Prieto
143. Salomonsson MC, SatouCM,
M, Sorensen R, etArendshorst
al. Intrarenal WJ,angiotensin
et al. CalciumII and its contri
handling in
transduction
115. Vallon V in, Traynor
renal vascular smooth
T, Barajas L, etmuscle cells: activation
al. Feedback control of o glomerular
calcium sparks.vas- bution
afferenttoarterioles.
the genesis Actao Physiol
chronicScand.
hypertension. Curr Opin Pharmacol. 2011;11(2):
2004;181(4):421–429.
Am J Physiol
cular tone inRegul neuronalIntegrnitric
CompoxidePhysiol. 2007;293(4):R1586–R1594.
synthase knockout mice. J Am Soc Nephrol. 180–186.
144. Carey RM, Padia SH. Angiotensin AT2 receptors: control of renal sodium
99. Jernigan NL, Drummond HA. Myogenic vasoconstriction in mouse renal
2001;12(8):1599–1606. 124. Prieto
excretion andMC, blood Gonzalez
pressure. Villalobos RA, Botros
Trends Endocrinol FT, et
Metab. al. Reciprocal changes
2008;19(3):84–87.
interlobar
116. Green arteries:
T, Gonzalezrole o AA, endogenous
Mitchell KD, beta etandal.gamma ENaC. interplay
The complex Am J Physiol Renal
between in renal
145. ACE/ANG
Harris RC, Becker II andBN, ACE2/ANG
Cheng HF1–7 are and
. Acute associated
chronicwith enhancedfor
mechanisms collect
regu-
Physiol. 2006;291(6):F1184–F1191.
cyclooxygenase-2 and angiotensin II in regulating kidney function. Curr Opin ing duct
lating renin intubule
proximal Goldblatt hypertensive
angiotensin rats. Am
II receptor J Physiol Renal
expression. J AmerPhysiol. 2011;
Soc Nephrol.
100. Guan
Nephrol Z, Pollock
Hypertens. JS, Cook AK, et al. E ect o epithelial sodium channel
2012;21(1):7–14. 300(3):F749–F755.
1997;8(2):306–313.
blockade
117. on the
Ichihara A,myogenic
Imig JD, Navarresponse LG.o Cyclooxygenase-2
rat juxtamedullarymodulates a erent arterioles. Hy-
afferent arte- 125. Zhuo X,
146. Ruan JL,Wagner
Li XC.C,New insights andC,perspectives
Chatziantoniou et al. Regulation on intrarenal
of angiotensin reninII anre-
pertension.
riolar 2009;54(5):1062–1069.
responses to increases in pressure. Hypertension. 1999;34(4 Pt 2):843–847. gioten
ceptor ATsin1 subtypes
system: in ocus
renalonafferent
intracrine/intracellular
arterioles during chronic angiotensin
changes II.inPeptides.
sodium
101. Roman
118. Wang X,RJ.Takeya P-450K, Aaronson of
metabolites PI,arachidonic
et al. E ectsacid o amiloride,
in the control benzamil, and
of cardio- 2011;32(7): 1551–1565.
diet. J Clin Invest. 1997;99(5):1072–1081.
alterations
vascular in extracellular
function. Physiol Rev. Na+2002;82(1):131–185.
on the rat a erent arteriole and its myogenic re http://www.ncbi.nlm.nih.gov/pubmed/21699940
147. Baylis C. Nitric oxide de ciency in chronic kidney disease. Am J Physiol
sponse.
119. RenAmY,JD’Ambrosio
Physiol RenalMA, Physiol. 2008;295(1):F272–F282.
Garvin JL, et al. Possible mediators of connecting 126. Ferrario2008;294(1):F1–F9.
Renal Physiol. CM. ACE2: more o Ang (1–7) or less Ang II? Curr Opin Nephrol
102. Daniels
tubule glomerular FH, Arendshorst WJ, Roberds
feedback. Hypertension. RG. Tubuloglomerular eedback and
2009;53(2):319–323. Hypertens.
148. Kone2011;20(1):1–6.
BC. Nitric oxide synthesis in the kidney: isoforms, biosynthesis, and
autoregulation
120. Wang H, in spontaneously
Garvin JL, D’Ambrosio hypertensive
MA, etrats. Am J Physiol.
al. Connecting 1990;258(6
tubule glomeru-Pt 127. Navar
functions LG, Prieto
in health. SeminCarrasquero MC, Kobori H. Molecular aspects o the
Nephrol. 2004;24(4):299–315.
2): feedback
lar F1479–F1489. antagonizes tubuloglomerular feedback in vivo. Am J Physiol Renal renal
149. renin
Singh angiotensin
P, Deng A, system. Weir MR, In: Re R, Dipette
et al. The balance DJ, Schi rin EL, Sowers
of angiotensin II andJR,
http://www.ncbi.nlm.nih.gov/pubmed/2360648
Physiol. 2010;299(6):F1374–F1378. eds.
nitricMolecular
oxide inMechanisms in Hypertension.
kidney diseases. Curr Opin London,
Nephrol England:
Hypertens. Taylor & Francis
2008;17(1):
103. Castrop
121. Bell PD, H, Lapointe
Höcherl JY,K,Peti
KurtzPeterdi
A, etJ.al.
Macula densa of
Physiology cellkidney
signaling.
renin.Annu Rev
Physiol Group;
51–56. 2006:3–13.
Physiol.
Rev. 2003;65:481–500.
2010;90(2):607–673. 128. Tikellis
150. Just A, C,OlsonBernardi S, Burns
AJ, Falck JR, WC.
et al.Angiotensin converting enzyme
NO and NO-independent 2 is a
mecha-
http://www.ncbi.nlm.nih.gov/pubmed/12524458
122. Komlosi P, Fuson AL, Fintha A, et al. Angiotensin I conversion to an- key
nismsmodulator
mediate ETB o thereceptor
renin angiotensin
buffering of system in cardiovascular
ET-1-induced and renal dis
renal vasoconstriction
104. Schnermann
giotensin II stimulates J, Briggs JP. Tubuloglomerular
cortical collecting duct sodium eedback: mechanistic
transport. insights
Hypertension. ease.
in theCurr
rat. Opin
Am J Nephrol
Physiol RegulHypertens.
Integr2011;20(1):62–68.
Comp Physiol. 2005;288(5):R1168–R1177.
rom gene manipulated mice. Kidney Int. 2008;74(4):418–426.
2003;42(2):195–199. 129. Kobori H,
151. Sandner NangakuM,
P, Kornfeld M,RuanNavar X,LG,
et al.etNitric
al. The intrarenal interactions
oxide/cAMP renin angiotensin in the
105. Navar
123. Sipos LG,A, Vargas
Prieto MC, S, Peti
Satou Peterdi J. Direct
R, et al. Intrarenal demonstration
angiotensin II o andtubular f uid
its contri- system:
control of romrat physiology
renal vascular to the pathobiology
resistance. Circ Res.o hypertension
1999;84(2):186–192.and kidney disease.
f ow sensing
bution by macula
to the genesis densahypertension.
of chronic cells. AmJ Physiol RenalPharmacol.
Curr Opin Physiol. 2010;299(5):
2011;11(2): Pharmacol
152. MajidRev. DSA,2007;59(3):251–287.
Navar LG. Nitric oxide in the control of renal hemodynamics
F1087–F1093.
180–186. 130. Navar LG,
and excretory KoboriAm
function. H, JPrieto MC, et2001;14(6
Hypertens. al. Intratubular renin angiotensin sys
Pt 2):74S–82S.
106. Prieto
124. Sosnovtseva OV, Pavlov AN, Mosekilde
MC, González-Villalobos RA, Botros E, et
FT,al.etSynchronization
al. Reciprocal changes among tem
153.inMattson
hypertension. Hypertension.
DL. Importance 2011;57(3):355–362.
of the renal medullary circulation in the control
mechanisms
in renal ACE/ANG o renal II autoregulation
and ACE2/ANGis 1–7 reduced in hypertensive
are associated rats. Am Jcollect-
with enhanced Physiol http://www.ncbi.nlm.nih.gov/pubmed/21282552
of sodium excretion and blood pressure. Am J Physiol Regul Integr Comp Physiol.
Renal
ing Physiol.
duct renin2007;293(5):F1545–F1555.
in Goldblatt hypertensive rats. Am J Physiol Renal Physiol. 2011; 131. Beierwaltes WH. The role o calcium in the regulation o renin secretion.
2003;284(1):R13–R27.
107. Braam B, Mitchell KD, Koomans HA, et al. Relevance o the tubuloglo
300(3):F749–F755. Am
154.J Physiol
Baylis C.Renal Physiol.arginine
Arginine, 2010;298(1):F1–F11.
analogs and nitric oxide production in chronic
merular
125. Zhuo eedback
JL, Li XC. mechanism
New insights in pathophysiology.
and perspectives JonAm Soc Nephrol.
intrarenal 1993;4(6):
renin-angioten- 132.
kidneyDiBona
disease.GF Nat. Physiology in perspective:
Clin Pract Nephrol. the wisdom o the body. Neu
2006;2(4):209–220.
1257–1274.
sin system: focus on intracrine/intracellular angiotensin II. Peptides. 2011;32(7): ral
155.conDeng
trol oA, the
Miraclekidney.
CM,Am J Physiol
Suarez JM, etRegul Integr Comp
al. Oxygen Physiol. in
consumption 2005;289(3):
the kidney:
108. Schnermann J. Homer W. Smith Award lecture. The juxtaglomerular ap
1551–1565. R633–R641.
effects of nitric oxide synthase isoforms and angiotensin II. Kidney Int. 2005;
paratus:
126. rom anatomical
Ferrario CM. ACE2:peculiarity to physiological
more of Ang-(1–7) or lessrelevance.
Ang II? Curr J AmOpin
Soc Nephrol.
Nephrol 133. Peti Peterdi J, Harris RC. Macula densa sensing and signaling mechanisms
68(2):723–730.
2003;14(6):1681–1694.
Hypertens. 2011;20(1):1–6. o156.
renin release.
Kohan DE,J Am RossiSocNF Nephrol.
, Inscho2010;21(7):1093–1096.
EW, et al. Regulation of blood pressure and
109. Navar
127. CastropLG, H, Prieto-Carrasquero
Huang Y, HashimotoMC, S, etKobori
al. Impairment
H. Molecular o tubuloglomerular
aspects of the http://www.ncbi.nlm.nih.gov/pubmed/20360309
salt homeostasis by endothelin. Physiol Rev. 2011;91(1):1–77.
eedback
renal regulation o GFR
renin-angiotensin in ecto
system. In: 5’
Renucleotidase/CD73
R, Dipette DJ, Schiffrin de cient EL, mice.
Sowers J Clin
JR, 134. Salazar N,
157. Dhaun FJ, Goddard
Llinas MT.J, RenalKohanhemodynamic
DE, et al. Role e ects elicited by acute
of endothelin-1 cyclo
in clinical
Invest.
eds. 2004;114(5):634–642.
Molecular Mechanisms in Hypertension. London, England: Taylor & Francis oxygenase
hypertension: 2 inhibition
20 years on. are not related to 2008;52(3):452–459.
Hypertension. angiotensin II levels. Am J Physiol Renal
110. Inscho
Group; EW, Cook AK, Imig JD, et al. Physiological role or P2X1 recep
2006:3–13. Physiol. 2010;299(5):F952–F953.
158. Schneider MP, Boesen EI, Pollock DM. Contrasting actions of endothelin
tors inTikellis
128. renal microvascular
C, Bernardi S,autoregulatory behavior. J Clin Invest. 2003;112(12):
Burns WC. Angiotensin-converting enzyme 2 is a 135.
ET(A) Harris
and ET(B) RC. receptors
An updateinon cyclooxygenase
cardiovascular 2 expression
disease. Annu Revand metabolites
Pharmacol Toxicol.in
1895–1905.
key modulator of the renin-angiotensin system in cardiovascular and renal dis- the kidney. Curr Opin Nephrol Hypertens. 2008;17(1):64–69.
2007;47:731–759.
http://www.ncbi.nlm.nih.gov/pubmed/14679185
ease. Curr Opin Nephrol Hypertens. 2011;20(1):62–68. 136. Nguyen G, AP
159. Davenport Muller DN. The
, Maguire biology o theof(pro)renin
JJ. Pharmacology receptor.receptors.
renal endothelin J Am Soc
111. Kobori
129. Singh P,H, Thomson
NangakuSC. M,Renal
Navarhomeostasis
LG, et al. The and intrarenal
tubuloglomerular eedback.
renin-angiotensin Nephrol. 2010;21(1):18–23.
Contrib Nephrol. 2011;172:1–17.
Curr Opin
system: fromNephrol
physiologyHypertens.
to the2010;19(1):59–64.
pathobiology of hypertension and kidney disease. http://www.ncbi.nlm.nih.gov/pubmed/19917780
160. Just A, Whitten CL, Arendshorst WJ. Reactive oxygen species participate in
http://www.ncbi.nlm.nih.gov/pubmed/19864948
Pharmacol Rev. 2007;59(3):251–287. 137. Ichihara
acute renal A, Sakoda M,
vasoconstrictor Kurauchi
responses Mito A,
induced byetETA-al. Renin, prorenin andAm
and ETB-receptors. theJ
112. Navar
130. Sun D,LG, Samuelson
Kobori H, LC, YangMC,
Prieto T, etetal.al.Mediation
Intratubular o tubuloglomerular
renin-angiotensin eed sys- kidney: a newPhysiol.
Physiol Renal chapter2008;294(4):F719–F728.
in an old saga. J Nephrol. 2009;22(3):306–311.
backinbyhypertension.
tem adenosine: evidence Hypertension.rom mice lacking adenosine 1 receptors. Proc Natl
2011;57(3):355–362. http://www.ncbi.nlm.nih.gov/pubmed/19557706
161. Masaki T. Historical review: Endothelin. Trends Pharmacol Sci. 2004;25(4):
Acad Sci
131. USA. 2001;98(17):9983–9988.
Beierwaltes WH. The role of calcium in the regulation of renin secretion. 138.
219–224.Bosnyak S, Jones ES, Christopoulos A, et al. Relative a nity o angioten
http://www.ncbi.nlm.nih.gov/pubmed/11504952
Am J Physiol Renal Physiol. 2010;298(1):F1–F11. sin
162.peptides
Fellnerand SK, novel ligands WJ.
Arendshorst at AT1 and AT2 receptors.
Endothelin-A Clin Sci (Lond).
and -B receptors, 2011;
superoxide,
113. Araujo M,
132. DiBona GF.Welch WJ. Oxidative
Physiology stressthe
in perspective: andwisdom
nitric oxide
of theinbody.
kidney unction.
Neural con- 121(7):297–303.
and Ca2 signaling in afferent arterioles. Am J Physiol Renal Physiol. 2007;292(1):
CurrofOpin
trol NephrolAm
the kidney. Hypertens.
J Physiol 2006;15(1):72–77.
Regul Integr Comp Physiol. 2005;289(3):R633–R641. 139. de Gasparo M, Catt KJ, Inagami T, et al. International union o pharmacol
F175–F184.
114. Peti-Peterdi
133. Wilcox CS. Redox J, Harris regulation
RC. Macula o the a erent
densa arteriole
sensing and tubuloglomerular
and signaling mechanisms ogy.
163.XXIII.
Sorokin TheA.angiotensin
EndothelinIIsignaling
receptors. andPharmacol
actions in Rev.the2000;52(3):415–472.
renal mesangium. Con-
eedback.
of Acta Physiol
renin release. J Am Soc Scand. 2003;179(3):217–223.
Nephrol. 2010;21(7):1093–1096. http://www.ncbi.nlm.nih.gov/pubmed/10977869
trib Nephrol. 2011;172:50–62.
128
12 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

140. Botros
164. Harrison FT,Bernard
Navar LG. LM,HemeZhuooxygenase
J, Kobori in H,regulation
et al. Intrarenal
of renalAT(1) recepand
function tor 192. Jackson KE,
166. Capdevila JH. Jackson
Regulation DW,of Quadri S, et and
ion transport al. Inhibition
blood pressure o heme oxygenase
by cytochrome
and ACE
blood binding
pressue. in ANG
Current II induced
Hyperten hypertensive rats. Am J Physiol Renal Physiol.
Rev. 2009;5:13–23. augments
p450 tubular sodium
monooxygenases. Currreabsorption.
Opin NephrolAm J Physiol2007;16(5):465–470.
Hypertens. Renal Physiol. 2011;300(4):
2002;282(1):F19–F25.
165. Lamon BD, Zhang FF, Puri N, et al. Dual pathways of carbon monoxide- F941–F946.
193. Ferreri NR, McGiff JC, Carroll MA, et al. Renal COX-2, cytokines and 20-
141. Ruan
mediated X, Oliverio MI,
vasoregulation: Co manby
modulation TM, et al.
redox Renal vascular
mechanisms. Circreactivity in mice:
Res. 2009;105(8): 167. Litubular
HETE: N, Yi Fand , dosvascular
Santos EA, et al. RoleCurr
mechanisms. o renal
Pharm medullary heme oxygenase in
Des. 2004;10(6):613–626.
Ang II induced vasoconstriction in AT1A receptor null mice. J Am Soc Nephrol.
775–783. the regulation
194. Imig JD. oCardiovascular
pressure natriuresis and arterial
therapeutic aspects blood pressure.
of soluble epoxide Hypertension.
hydrolase
1999;10(12):2620–2630.
166. Jackson KE, Jackson DW, Quadri S, et al. Inhibition of heme oxygenase 2007;49(1):148–154.
inhibitors. Cardiovasc Drug Rev. 2006;24(2):169–188.
http://www.ncbi.nlm.nih.gov/pubmed/10589703
augments tubular sodium reabsorption. Am J Physiol Renal Physiol. 2011;300(4): 168. Kompanowska-Jezierska
195. Nath KA, Hernandez MC,E,Croatt AJ, et al. M.
Kuczeriszka Heme oxygenaseP-450
Cytochrome activitymetabo-
as a de
142. Fellner SK, Arendshorst WJ. Angiotensin II Ca2+ signaling in rat a erent
F941–F946. terminant
lites in renalo the renal hemodynamic
circulation response to lowwith
and excretion—interaction dosethe ANG II. Am
nitric oxideJ Physiol
(NO)
arterioles:
167. Li N,stimulation o cyclic
Yi F, dos Santos EA, ADP
et al. ribose
Role ofand IP3medullary
renal pathways.heme Am Joxygenase
Physiol Renalin Regul Integr
system. CompPharmacol.
J Physiol Physiol. 2010;299(5):R1183–R1191.
2008;59 Suppl 9:137–149.
Physiol.
the 2005;288(4):F785–F791.
regulation of pressure natriuresis and arterial blood pressure. Hypertension. 169. Campbell
196. Stec DE, Vera WB, T, Storm MV
Gauthier KM. , etWhat
al. Blood
is newpressure and renal blow f hyper-
in endothelium-derived ow re
143. Salomonsson M, Sorensen CM, Arendshorst WJ, et al. Calcium handling in
2007;49(1):148–154. sponses infactors?
polarizing heme oxygenase 2 knockout
Curr Opin Nephrol mice. 2002;11(2):177–183.
Hypertens. Am J Physiol Regul Integr Comp
a erent
168. arterioles.
Nath Acta Physiol
KA, Hernandez MC,Scand.
Croatt2004;181(4):421–429.
AJ, et al. Heme oxygenase activity as a de- Physiol.
197. Imig2009;297(6):R1822–R1828.
JD, Navar LG, Roman RJ, et al. Actions of epoxygenase metabolites on
http://www.ncbi.nlm.nih.gov/pubmed/15283754
terminant of the renal hemodynamic response to low-dose ANG II. Am J Physiol 170.preglomerular
the Ren Y, D’Ambrosio MA, JWang
vasculature. Amer H, SocetNephrol.
al. Heme oxygenase metabolites in
1996;7(11):2364–2370.
144. Integr
Regul CareyCompRM, Padia
Physiol.SH. Angiotensin AT2 receptors: control o renal sodium
2010;299(5):R1183–R1191. hibit tubuloglomerular
198. Majid DSA, Kopkan eedback
L. Nitric(TGF).
oxideAm andJ superoxide
Physiol Renalinteractions
Physiol. 2008;295(4):
in the kid-
excretion
169. StecandDE,blood
Vera pressure.
T, Storm Trends
MV, etEndocrinol
al. BloodMetab.pressure 2008;19(3):84–87.
and renal blow ow F1207–F1212.
ney and their implication in the development of salt-sensitive hypertension. Clin
145. Harris
responses RC, Becker
in heme BN, Cheng
oxygenase-2 HF. Acute
knockout mice.andAm chronic
J Physiolmechanisms
Regul IntegrorComp
regu http://www.ncbi.nlm.nih.gov/pubmed/18715939
Exp Pharmacol Physiol. 2007;34(9):946–952.
lating proximal
Physiol. tubule angiotensin II receptor expression. J Amer Soc Nephrol.
2009;297(6):R1822–R1828. 171. Wilcox
199. Wang H, CS.Garvin
OxidativeJL, D’Ambrosio MA, etoxide
stress and nitric al. Heme oxygenase
de ciency in themetabolites
kidney: a
1997;8(2):306–313.
170. Ren Y, D’Ambrosio MA, Wang H, et al. Heme oxygenase metabolites in- inhibit link
critical tubuloglomerular
to hypertension? eedback in vivo.
Am J Physiol AmIntegr
Regul J Physiol
Comp Heart Circ Physiol.
Physiol. 2011;
2005;289(4):
146. tubuloglomerular
hibit Ruan X, Wagner feedback C, Chatziantoniou
(TGF). AmC, et al. Regulation
J Physiol Renal Physiol.o angiotensin
2008;295(4):II 300(4):H1320–H1326.
R913–R935.
receptor AT1 subtypes in renal a erent arterioles during chronic changes in so
F1207–F1212. 172.
200. DukaAbraham NG, Cao J, E,
I, Kintsurashvili Sacerdoti
Gavras I,D, et et
al.al. Heme oxygenase:
Vasoactive potential ofthe thekey
b(1)tobrady-
renal
dium Wang
171. diet. J H,
ClinGarvin
Invest. JL,
1997;99(5):1072–1081.
D’Ambrosio MA, et al. Heme oxygenase metabolites unction
kinin regulation.
receptor Am J Physiol
in normotension andRenal Physiol. 2009;297(5):F1137–F1152.
hypertension. Circ Res. 2001;88(3):275–281.
147. Baylis
inhibit C. Nitric oxide
tubuloglomerular de ciency
feedback in vivo.in Am
chronic kidney
J Physiol Heartdisease. Am J Physiol
Circ Physiol. 2011; 173. Marin-Castaño
201. Lu M, Liu YH, Goh HS, et al. Hydrogen
ME, Schanstra JP, Neau E,sul deInduction
et al. inhibits plasma renin activ
of functional bra-
Renal Physiol. 2008;294(1):F1–F9.
300(4):H1320–H1326. ity. J Am b(1)-receptors
dykinin Soc Nephrol. 2010;21(6):993–1002.
in normotensive rats and mice under chronic angioten-
148. Abraham
172. Kone BC.NG, NitricCaooxide synthesisD,inetthe
J, Sacerdoti al.kidney: iso orms, biosynthesis,
Heme oxygenase: and
the key to renal http://www.ncbi.nlm.nih.gov/pubmed/20360313
sin-converting enzyme inhibitor treatment. Circulation. 2002;105(5):627–632.
unctionsregulation.
function in health. Semin Nephrol.
Am J Physiol 2004;24(4):299–315.
Renal Physiol. 2009;297(5):F1137–F1152. 174. Moreau
202. Skovgaard ME,N,Garbacki
GouliaevN, A, Molinaro
Aalling M,G,etetal.al.The Therole o endogenoussystem:
kallikrein-kinin H(2)S
149. Lu
173. Singh P, Deng
M, Liu YH, GohA, Weir
HS, MR,
et al. etHydrogen
al. The balance o angiotensin
sul de inhibits plasma IIrenin
and activ-
nitric in cardiovascular
current and futurephysiology.
pharmacologicalCurr Pharmtargets.Biotechnol.
J Pharmacol2011 Jan2005;99(1):6–38.
Sci. 11 [Epub ahead o
oxide
ity. J Am in Soc
kidney diseases.
Nephrol. Curr Opin Nephrol Hypertens. 2008;17(1): 51–56.
2010;21(6):993–1002. print].Tang SC, Leung JC, Lai KN. The kallikrein-kinin system. Contrib Nephrol.
203.
http://www.ncbi.nlm.nih.gov/pubmed/18090670
174. Skovgaard N, Gouliaev A, Aalling M, et al. The role of endogenous H(2)S 175. Wang R. Signaling pathways or the vascular e ects o hydrogen sul de.
2011;170:145–155.
150.
in Just A, Olson
cardiovascular AJ, FalckCurr
physiology. JR, Pharm
et al. NO and NO
Biotechnol. 2011independent
Jan 11 [Epub mecha nisms
ahead of Curr Opin
204. ImigNephrol
JD, Falck Hypertens.
JR, Wei 2011;20(2):107–112.
S, et al. Epoxygenase metabolites contribute to ni-
mediate ETB receptor bu ering o ET 1 induced renal vasoconstriction in the rat.
print]. 176.oxide-independent
tric Yang G, Wu L, afferent Jiang B,arteriolar
et al. H2S as a physiologic
vasodilation in response vasorelaxant:
to bradykinin.hy
Am J Physiol
175. Wang R. Regul Integr Comp
Signaling pathwaysPhysiol.
for2005;288(5):R1168–R1177.
the vascular effects of hydrogen sul de. Jpertension in mice with deletion o cystathionine gamma lyase. Science.
Vasc Res. 2001;38(3):247–255.
151. Opin
Curr Sandner
NephrolP, Korn eld M,2011;20(2):107–112.
Hypertens. Ruan X, et al. Nitric oxide/cAMP interactions in the 2008;322(5901):
205. Matsuda H, 587–590. Hayashi K, Wakino S, et al. Role of endothelium-derived hy-
control
176. o rat
Yang G, renal
Wu L,vascular
Jiang B, etresistance.
al. H2S asCirc Res. 1999;84(2):186–192.
a physiologic vasorelaxant: hypertension 177. Roy A, Khan
perpolarizing factor AH, in ACEIslam MT, et al. Interdependency
inhibitor-induced renal vasodilationo cystathione
in vivo. g-lyase
Hyper-
http://www.ncbi.nlm.nih.gov/pubmed/9933250
in mice with deletion of cystathionine gamma-lyase. Science. 2008;322(5901): and cystathione
tension. b a synthase in hydrogen sul de induced blood pressure regula
2004;43(3):603–609.
152. Majid DSA, Navar LG. Nitric oxide in the control o renal hemodynamics
587–590. tion inOmoro
206. rats. Am SA,J Hypertens.
Majid DSA,2012;25(1):74–81.
el Dahr SS, et al. Roles of ANG II and bradykinin
and excretory
177. Roy A, Khan unction. Am J Hypertens.
AH, Islam MT, et al. 2001;14(6
InterdependencyPt 2):74S 82S.
of cystathione -lyase 178.
in Zhangregional
the renal C, Hu blood JJ, Xia M, owetresponses
al. Redox to signaling
ACE inhibitionvia lipid ra t clustering in
in sodium-depleted
153.cystathione
and Mattson DL.a-synthase
Importance o the renal
in hydrogen sulmedullary
de-induced circulation in the regula-
blood pressure control homocysteine
dogs. Am J Physiol. induced injury o podocytes. Biochim BIophys Acta. 2010;1803(4):
2000;279(2):F289–F293.
o sodium
tion in rats.excretion and blood
Am J Hypertens. pressure. Am J Physiol Regul Integr Comp Physiol.
2012;25(1):74–81. 482–491.
207. Ren Y, Garvin JL, Falck JR, et al. Glomerular autacoids stimulated by bra-
2003;284(1):R13–R27.
178. Zhang C, Hu JJ, Xia M, et al. Redox signaling via lipid raft clustering in http://www.ncbi.nlm.nih.gov/pubmed/20036696
dykinin regulate efferent arteriole tone. Kidney Int. 2003;63(3):987–993.
154. Baylis C. Arginine,
homocysteine-induced arginine
injury analogs and
of podocytes. nitricBIophys
Biochim oxide production in chronic
Acta. 2010;1803(4): 179. Sadowski
208. BeltowskiJ,J.Badzynska
Hypoxia in the renal medulla:
B. Intrarenal vasodilator implications
systems: NO, orprostaglandins
hydrogen sul
kidney disease. Nat Clin Pract Nephrol. 2006;2(4):209–220.
482–491. andde bradykinin.
signaling. J Pharmacol
An integrative Exp Ther. 2010;334(2):358–363.
approach. J Physiol Pharmacol. 2008;59 Suppl 9:
155. Beltowski
179. Deng A, Miracle CM, in
J. Hypoxia Suarez JM, etmedulla:
the renal al. Oxygen consumption
implications in the kidney:
for hydrogen sul- http://www.ncbi.nlm.nih.gov/pubmed/20427475
105–119.
edeects o nitricJ Pharmacol
signaling. oxide synthase iso orms
Exp Ther. and angiotensin II. Kidney Int. 2005;
2010;334(2):358–363. 180. Wang
209. Campean V, Theilig
X, Trottier G, F , Paliege A, etRD.
Loutzenhiser al. Key enzymes or
Determinants ofrenal
renalprostaglandin
afferent arte-
68(2):723–730.
180. Câmpean V, Theilig F, Paliege A, et al. Key enzymes for renal prostaglandin synthesis:
riolar actionssiteofspeci c expression
bradykinin: evidence in that
rodent kidneypathways
multiple (rat, mouse).mediate Amresponses
J Physiol
156. Kohan
synthesis: DE, Rossi
site-speci NF, InschoinEW,
c expression et al.kidney
rodent Regulation
(rat, omouse).
blood pressure and
Am J Physiol Renal Physiol.
attributed 2003;285(1):F19–F32.
to EDHF . Am J Physiol Renal Physiol. 2003;285(3):F540–F549.
salt homeostasis
Renal by endothelin. Physiol Rev. 2011;91(1):1–77.
Physiol. 2003;285(1):F19–F32. 181. Trabold
210. Carroll MA, F, Pons McGi JC, Ferreri
S, Hagege AA, etNR. Products o arachidonic
al. Cardiovascular phenotypes acid metabo
of kinin B2
http://www.ncbi.nlm.nih.gov/pubmed/21248162
181. Carroll MA, McGiff JC, Ferreri NR. Products of arachidonic acid metabo- lism. Methods
receptor- Mol Med.
and tissue 2003;86:385–397.
kallikrein-de cient mice. Hypertension. 2002;40(1):90–95.
157. Methods
lism. Dhaun Mol N, Goddard J, Kohan DE, et al. Role o endothelin 1 in clinical
Med. 2003;86:385–397. http://www.ncbi.nlm.nih.gov/pubmed/12886783
211. Guan Z, Inscho EW. Role of adenosine 5 -triphosphate in regulating renal
hypertension:
182. Harris RC, 20 Zhang
years on.MZ,Hypertension. 2008;52(3):452–459.
Cheng HF. Cyclooxygenase-2 and the renal renin- 182. Harris RC,
microvascular Zhangand
function MZ, in Cheng HF. Cyclooxygenase
hypertension. Hypertension. 22011;58(3):333–340.
and the renal renin
http://www.ncbi.nlm.nih.gov/pubmed/18678788
angiotensin system. Acta Physiol Scand. 2004;181(4):543–547. angiotensin
212. Nishiyama system. Acta Physiol
A, Jackson KE, Scand.
Majid DSA,2004;181(4):543–547.
et al. Renal interstitial uid ATP re-
158. Imig
183. Schneider MP, Boesen
JD. Eicosanoids andEI,renal
Pollock DM.function
vascular Contrasting actionsClin
in diseases. o endothelin
Sci (Lond). http://www.ncbi.nlm.nih.gov/pubmed/15283769
sponses to arterial pressure and tubuloglomerular feedback activation during cal-
ET(A) and ET(B) receptors in cardiovascular disease. Annu Rev Pharmacol Toxicol.
2006;111(1):21–34. 183. channel
cium ImigJD.blockade.Eicosanoids Am JandPhysiolrenal
Heart vascular unction
Circ Physiol. in diseases. Clin Sci
2006;290(2):H772–H777.
2007;47:731–759.
184. Wilcox CS, Welch WJ. Thromboxane synthase and TP receptor mRNA in (Lond).Carroll
213. 2006;111(1):21–34.
MA, Doumad AB, Li J, et al. Adenosine2A receptor vasodilation of
http://www.ncbi.nlm.nih.gov/pubmed/17002597
rat kidney and brain: effects of salt intake and ANG II. Am J Physiol Renal Physiol. http://www.ncbi.nlm.nih.gov/pubmed/16764555
rat preglomerular microvessels is mediated by EETs that activate the cAMP/PKA
159. Davenport AP, Maguire JJ. Pharmacology o renal endothelin receptors.
2003;284(3):F525–F531. 184. Wilcox
pathway. Am JCS, Welch
Physiol RenalWJ. Thromboxane
Physiol. synthase and TP receptor mRNA in
2006;291(1):F155–F161.
Contrib
185. Nephrol.
Breyer MD,2011;172:1–17.
Breyer RM. G protein-coupled prostanoid receptors and the rat kidney
214. Fengand MG,brain: Navare LG. ectsAdenosine
o salt intake A2 and ANGactivation
receptor II. Am J Physiol Renalafferent
attenuates Physiol.
http://www.ncbi.nlm.nih.gov/pubmed/21893985
kidney. Annu Rev Physiol. 2001;63:579–605. 2003;284(3):F525–F531.
arteriolar autoregulation during adenosine receptor saturation in rats. Hyperten-
160. Carroll
186. Just A, MA,
Whitten CL,NR,
Ferreri Arendshorst
Escalante WJ. BA, etReactive oxygen species
al. Cytochrome participate
P450 arachidon- 185. 2007;50(4):744–749.
sion. Breyer MD, Breyer RM. G protein coupled prostanoid receptors and the
in acid
ic acutemetabolites
renal vasoconstrictor
modulate responses
renal tubular induced by ETA
function. Adv and
ExpETBMedreceptors. Am
Biol. 2003; kidney.Just
215. Annu Rev Physiol. 2001;63:579–605.
A, Arendshorst WJ. A novel mechanism of renal blood ow autoregu-
J Physiol Renal Physiol. 2008;294(4):F719–F728.
525:67–69. http://www.ncbi.nlm.nih.gov/pubmed/11181968
lation and the autoregulatory role of A1 adenosine receptors in mice. Am J Physiol
161. Chatziantoniou
187. Masaki T. Historical review:
C, Ruan Endothelin.WJ.
X, Arendshorst Trends Pharmacol
Defective Sci. 2004;25(4):
G protein activation 186. Physiol.
Renal Carroll 2007;293(5):F1489–F1500.
MA, Ferreri NR, Escalante BA, et al. Cytochrome P450 arachidon
219–224.
of the cAMP pathway in rat kidney during genetic hypertension. Proc Natl Acad ic acidOppermann
216. metabolitesM,modulate Friedmanrenal tubular unction.R,Adv
DJ, Faulhaber-Walter Exp
et al. Med Biol. 2003;
Tubuloglomerular
162.
Sci USA. Fellner SK, Arendshorst WJ. Endothelin A and B receptors, superoxide,
1995;92(7):2924–2928. 525:67–69.
feedback and renin secretion in NTPDase1/CD39-de cient mice. Am J Physiol
and Ca2+
188. Purdy signaling in a erentWJ.
KE, Arendshorst arterioles.
EP(1) and AmEP(4)
J Physiol Renal Physiol.
receptors mediate2007;292(1):
prostaglan- http://www.ncbi.nlm.nih.gov/pubmed/12751738
Renal Physiol. 2008;294(4):F965–F970.
F175–F184.
din E(2) actions in the microcirculation of rat kidney. Am J Physiol Renal Physiol. 187. V
217. Chatziantoniou
allon V, Mühlbauer C, Ruan X, Arendshorst
B, Osswald H. AdenosineWJ. De and ective
kidney G protein
function.activation
Physiol
http://www.ncbi.nlm.nih.gov/pubmed/16788136
2000;279(4):F755–F764. o the
Rev. cAMP pathway in rat kidney during genetic hypertension. Proc Natl Acad
2006;86(3):901–940.
163. Paliege
189. SorokinA,A.MizelEndothelin
D, Medina signaling
C, et and actions in of
al. Inhibition thenNOS
renal expression
mesangium.inCon- the Sci USA.
218. Lai1995;92(7):2924–2928.
EY, Patzak A, Persson AE, et al. Angiotensin II enhances the afferent
trib Nephrol.
macula densa2011;172:50–62.
by COX-2-derived prostaglandin E(2). Am J Physiol Renal Physiol. 188. Purdy
arteriolar KE, Arendshorst
response to adenosine WJ. EP(1)
through andincreases
EP(4) receptors mediate
in cytosolic prostaglan
calcium. Acta
164. Botros FT, Navar LG. Heme oxygenase in regulation o renal unction and
2004;287(1):F152–F159. din E(2)(Oxf).
Physiol actions in the microcirculation o rat kidney. Am J Physiol Renal Physiol.
2009;196(4):435–445.
blood Brännström
190. pressue. Current Hyperten Rev.WJ.
K, Arendshorst 2009;5:13–23.
Thromboxane A2 contributes to the 2000;279(4):F755–F764.
219. Nishiyama A, Navar LG. ATP mediates tubuloglomerular feedback. Am J
165. Lamon
enhanced BD, Zhang FF, feedback
tubuloglomerular Puri N, etactivity
al. Dual in pathways
young SHR. o carbon
Am J monox
Physiol. 189. Paliege
Physiol A, Mizel
Regul Integr Comp D,Physiol.
Medina2002;283:R273–R275.
C, et al. Inhibition o nNOS expression in the
ide mediatedPt vasoregulation:
1999;276(5 2):F758–F766. modulation by redox mechanisms. Circ Res. macula
220. densa by COX
Al-Mashhadi RH, 2Skøtt
derived prostaglandin
O, Vanhoutte PM, E(2).
et al. Am J PhysiolofRenal
Activation A(2) Physiol.
adeno-
2009;105(8):
191. Hao CM,775–783.
Breyer MD. Physiologic and pathophysiologic roles of lipid me- 2004;287(1):F152–F159.
sine receptors dilates cortical efferent arterioles in mouse. Kidney Int. 2009;75(8):
diators in the kidney. Kidney Int. 2007;71(11):1105–1115. 793–799.
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1129
29

190. Awad
221. BrännströmAS, Rouse K, M,Arendshorst
Liu L, et al. WJ. Thromboxane
Activation A2 contributes
of adenosine 2A receptors to pre-
the 213.
249. V Carroll
agnes OB, MA,Hansen
Doumad FH,AB, Li J,JJ,etetal.al.Adenosine2A
Feng Enhanced Ca2 receptor vasodilation
response to AVP in o
enhanced
serves tubuloglomerular
structure and function ofeedbackpodocytes. activity in Nephrol.
J Am Soc young SHR. Am J Physiol.
2008;19(1):59–68. rat preglomerular
preglomerular microvessels
vessels from rats is mediated
with genetic by EETs that activateduring
hypertension the cAMP/PKA
different
1999;276(5
222. Ren Y, Pt 2):F758–F766.
Arima S, Carretero OA, et al. Possible role of adenosine in macula pathway.
hydrationAm J Physiol
states. Am JRenal
Physiol Physiol. 2006;291(1):F155–F161.
Renal Physiol. 2005;288(6):F1249–F1256.
191. control
densa Hao CM, ofBreyer
glomerularMD. hemodynamics.
Physiologic and Kidney pathophysiologic roles o lipid me
Int. 2002;61(1):169–176. 214. Feng MG,
250. Aoyagi Navar LG.
T, Izumi AdenosineM,A2etreceptor
Y, Hiroyama activationregulates
al. Vasopressin attenuates thea renin-
erent
diatorsOsmond
223. in the kidney. KidneyEW.
DA, Inscho Int. 2007;71(11):1105–1115.
P2X(1) receptor blockade inhibits whole kid- arteriolar autoregulation system
angiotensin-aldosterone during adenosine
via V1a receptorsreceptorinsaturation
macula densain rats.cells.
Hyperten-
Am J
http://www.ncbi.nlm.nih.gov/pubmed/17361113
ney autoregulation of renal blood ow in vivo. Am J Physiol Renal Physiol. 2010; sion. 2007;50(4):744–749.
Physiol Renal Physiol. 2008;295(1):F100–F107.
192. Capdevila JH. Regulation o ion transport and blood pressure by cyto
298(6):F1360–F1368. 215. Just A, Arendshorst
251. Cavarape A, Bauer J,WJ. A novel
Bartoli E, etmechanism
al. Effects of o angiotensin
renal blood II, f ow autoregu
arginine va-
chrome
224. p450 monooxygenases.
Peti-Peterdi J. Calcium wave Curr Opin Nephrol Hypertens.
of tubuloglomerular feedback. Am 2007;16(5):
J Physiol lation andand
sopressin the autoregulatory
thromboxane A2 role
inorenal
A1 adenosine
vascular bed: receptors
role ofinrho-kinase.
mice. Am JNephrol
Physiol
465–470.
Renal Physiol. 2006;291(2):F473–F480. Renal Physiol. 2007;293(5):F1489–F1500.
Dial Transplant. 2003;18(9):1764–1769.
http://www.ncbi.nlm.nih.gov/pubmed/17693763
225. Eppel GA, Lee LL, Evans RG. alpha-Adrenoceptor subtypes mediating re- 216. Oppermann
252. Fallet RW, IkenagaM, Friedman
H, Bast DJ,JP, etFaulhaber
al. Relative Walter R, et al. Tubuloglomerular
contributions of Ca2 mobili-
193. Ferreri
gional kidneyNR, bloodMcGi ow JC, Carroll to
responses MA, et al.nerve
renal Renalstimulation.
COX 2, cytokines and 20
Auton Neurosci. eedback
zation andand in ux renin secretion
in renal in NTPDase1/CD39
arteriolar contractile responses de cient mice. Am
to arginine J Physiol
vasopressin.
HETE: tubular and vascular mechanisms. Curr Pharm Des. 2004;10(6):613–626.
2004;112(1–2):15–24. Renal Physiol.Renal
Am J Physiol 2008;294(4):F965–F970.
Physiol. 2005;288(3):F545–F551.
http://www.ncbi.nlm.nih.gov/pubmed/14965324
226. Denton KM, Luff SE, Shweta A, et al. Differential neural control of glo- 217. Vallon V Mühlbauer
253. Rajapakse NW, Roman B, RJ,
Osswald
Falck H. JR,Adenosine and kidney
et al. Modulation unction. Physiol
of V1-receptor-medi-
194. Imig
merular ultra JD.ltration.
Cardiovascular
Clin Exptherapeutic
Pharmacol Physiol.aspects2004;31(5–6):380–386.
o soluble epoxide hydrolase Rev.
ated 2006;86(3):901–940.
renal vasoconstriction by epoxyeicosatrienoic acids. Am J Physiol Regul Integr
inhibitors.
227. BadzynskaCardiovasc Drug Rev.J.2006;24(2):169–188.
B, Sadowski Moderate intrarenal vasoconstriction after high http://www.ncbi.nlm.nih.gov/pubmed/16816141
Comp Physiol. 2004;287(1):R181–R187.
195. Kompanowska
pressor doses of norepinephrineJezierska E,in Kuczeriszka M. Cytochrome
the rat: comparison with effectsP 450 metabo
of angioten- 218. Lai EY
254. Ando K,, Patzak
Shimosawa A, Persson
T, FujitaAE,T. et al. AngiotensininIIvascular
Adrenomedullin enhances the a erent
diseases. Curr
litesII.inKidney
sin renal circulation
Blood Press Res.and 2011;34(5):307–310.
excretion—interaction with the nitric oxide (NO) arteriolar
Hypertens response to adenosine through increases in cytosolic calcium. Acta
Rep. 2004;6(1):55–59.
system.
228. J Physiol
Evans RG, Pharmacol.
Head GA, 2008;59
Eppel GA, Suppl
et al.9:137–149.
Angiotensin II and neurohumoral Physiol
255. Bell(Oxf).
D, 2009;196(4):435–445.
McDermott BJ. Intermedin (adrenomedullin-2): a novel counter-
196. Campbell
control of the renal WB, Gauthiercirculation.
medullary KM. WhatClin is new
Expin endothelium
Pharmacol derived
Physiol. hyper
2010;37(2): http://www.ncbi.nlm.nih.gov/pubmed/19141138
regulatory peptide in the cardiovascular and renal systems. Br J Pharmacol.
polarizing actors? Curr Opin Nephrol Hypertens. 2002;11(2):177–183.
e58–e69. 219.
2008;153Nishiyama A, Navar LG. ATP mediates tubuloglomerular eedback. Am J
Suppl 1:S247–S262.
http://www.ncbi.nlm.nih.gov/pubmed/11856910
229. Denton KM, Shweta A, Anderson WP. Preglomerular and postglomerular Physiol RegulSD,
256. Brain Integr Comp
Grant AD.Physiol.
Vascular2002;283:R273–R275.
actions of calcitonin gene-related peptide
197. Imigresponses
resistance JD, NavartoLG, Romanlevels
different RJ, etofal.sympathetic
Actions o epoxygenase
activation bymetabolites
hypoxia. J Am on 220. Al Mashhadi RH,
and adrenomedullin. SkøttRev.
Physiol O, 2004;84(3):903–934.
Vanhoutte PM, et al. Activation o A(2) adeno
the Nephrol.
Soc preglomerular vasculature. J Amer Soc Nephrol. 1996;7(11):2364–2370.
2002;13(1):27–34. sine
257.receptors
Nishikimi dilates cortical e erent arterioles
T. Adrenomedullin in mouse. Kidney
in the kidney—renal Int. 2009;75(8):
physiological and
http://www.ncbi.nlm.nih.gov/pubmed/8959626
230. Genovesi S, Pieruzzi F, Camisasca P, et al. Renal afferents responsive to chemi- 793–799.
pathophysiological roles. Curr Med Chem. 2007;14(15):1689–1699.
198.
cal andMajid
mechanicalDSA, Kopkan L. Nitric
pelvic stimuli in theoxide andClin
rabbit. superoxide
Sci (Lond).interactions in the kid
1997;92(5):505–510. http://www.ncbi.nlm.nih.gov/pubmed/19165174
258. Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxi-
ney and
231. Kopp theirUC, implication
Cicha MZ, in the
Smithdevelopment o salt sensitive
LA, et al. Dietary sodium hypertension.
modulates theClin in- 221.
dases: Awad
physiologyAS, Rouse M, Liu L, et al. Activation
and pathophysiology. Physiol Rev. o 2007;87(1):245–313.
adenosine 2A receptors pre
Exp Pharmacol
teraction between Physiol. 2007;34(9):946–952.
efferent and afferent renal nerve activity by altering activation serves structureRE,
259. Clempus andGriendling
unction o KK. podocytes.
ReactiveJoxygen Am Soc species
Nephrol.signaling
2008;19(1):59–68.
in vascular
http://www.ncbi.nlm.nih.gov/pubmed/17645645
of alpha2-adrenoceptors on renal sensory nerves. Am J Physiol Regul Integr Comp 222.
smoothRen Y, Arima
muscle cells.S,Cardiovasc
Carretero Res.OA,2006;71(2):216–225.
et al. Possible role o adenosine in macula
199. Wilcox
Physiol. CS. Oxidative stress and nitric oxide de ciency in the kidney: a
2011;300(2):R298–R310. densa control o glomerular
260. Reckelhoff JF, Romero hemodynamics.
JC. Role of oxidative Kidneystress
Int. 2002;61(1):169–176.
in angiotensin-induced
criticalCarey
232. link toRM. hypertension?
Theodore Am J Physiol
Cooper Regul Integr
Lecture: RenalComp Physiol.system:
dopamine 2005;289(4):
para- 223. OsmondAm
hypertension. DA, InschoRegul
J Physiol EW. Integr
P2X(1) Compreceptor blockade
Physiol. inhibits whole kid
2003;284(4):R893–R912.
R913–R935.
crine regulator of sodium homeostasis and blood pressure. Hypertension. 2001; ney
261.autoregulation
Ushio-Fukai M. o renal
Vascularblood f ow in through
signaling vivo. AmGJ Physiol Renal Physiol.
protein-coupled 2010;
receptors:
http://www.ncbi.nlm.nih.gov/pubmed/16183628
38(3):297–302. 298(6):F1360–F1368.
new concepts. Curr Opin Nephrol Hypertens. 2009;18(2):153–159.
200. Jose
233. DukaPA,I,Eisner Kintsurashvili
GM, Felder E, RA.
Gavras I, et al.
Regulation Vasoactive
of blood pressure potential o the
by dopamine 224. Peti PS,
262. Gill PeterdiWilcox J. Calcium
CS. NADPH waveoxidases
o tubuloglomerular
in the kidney. eedback. Am J Physiol
Antioxid Redox Signal.
b(1) brady
receptors. kinin Physiol.
Nephron receptor2003;95(2):19–27.
in normotension and hypertension. Circ Res. Renal Physiol. 2006;291(2):F473–F480.
2006;8(9–10):1597–1607.
2001;88(3):275–281.
234. Tayebati SK, Lokhandwala MF, Amenta F. Dopamine and vascular dynam- 225. Eppel GA,M,
263. Carlström LeeLaiLL,
EYEvans
, Ma Z,RG. alpha
et al. RoleAdrenoceptor
of NOX2 in the subtypes
regulation mediating re
of afferent
http://www.ncbi.nlm.nih.gov/pubmed/11179194
ics control: present status and future perspectives. Curr Neurovasc Res. 2011;8(3): gional
arteriolekidney blood f ow Am
responsiveness. responses
J Physiol to Regul
renal Integr
nerve Comp
stimulation.
Physiol.Auton Neurosci.
2009;296(1):
201. Marin Castaño ME, Schanstra JP, Neau E, et al. Induction o unctional
246–257. 2004;112(1–2):15–24.
R72–R79.
bra dykinin
235. Wang X, b(1) receptors
Villar in normotensive
VA, Armando rats and mice
I, et al. Dopamine, under
kidney, andchronic
hyperten-an 226. Denton
264. Just KM, LuAJ, SE,
A, Olson ShwetaCL,
Whitten A, et et al. Di erential neural
Superoxide mediates control
acuteo renal
glo
giotenstudies
sion: sin converting
in dopamine enzyme
receptor inhibitor
knockout treatment.
mice. PediatrCirculation. 2002;105(5):
Nephrol. 2008;23(12): merular ultra ltration.
vasoconstriction producedClin Exp Pharmacol Physiol.
by angiotensin II and 2004;31(5–6):380–386.
catecholamines by a mecha-
627–632.
2131–2146. http://www.ncbi.nlm.nih.gov/pubmed/15191417
nism independent of nitric oxide. Am J Physiol Heart Circ Physiol. 2007;292(1):
http://www.ncbi.nlm.nih.gov/pubmed/11827930
236. Zeng C, Jose PA. Dopamine receptors: important antihypertensive counter- 227.
H83–H92.Badzynska B, Sadowski J. Moderate intrarenal vasoconstriction a ter high
202. Moreau
balance againstME, Garbacki factors.
hypertensive N, Molinaro G, et al.2011;57(1):11–17.
Hypertension. The kallikrein kinin system: pressor
265. Haque dosesMZ, o norepinephrine
Majid DSA. Reduced in the rat: renal comparison
responses with e ects
to nitric o angioten
oxide synthase
current
237. and uture
Winaver pharmacological
J, Abassi targets. J Pharmacol
Z. Role of neuropeptide Y in theSci.regulation
2005;99(1):6–38.
of kidney sin II. Kidney
inhibition Bloodlacking
in mice Press Res.the2011;34(5):307–310.
gene for gp91phox subunit of NAD(P)H oxidase.
203. Tang
function. EXS. SC,2006;(95):123–132.
Leung JC, Lai KN. The kallikrein kinin system. Contrib Nephrol. http://www.ncbi.nlm.nih.gov/pubmed/21606652
Am J Physiol Renal Physiol. 2008;295(3):F758–F764.
2011;170:145–155.
238. Clerico A, Giannoni A, Vittorini S, et al. Thirty years of the heart as an 228. EvansMA,
266. Khan RG, Islam
Head MT, GA, Castillo
Eppel GA, A, et al. Attenuation
Angiotensin of II and
renalneurohumoral
excretory re-
http://www.ncbi.nlm.nih.gov/pubmed/21659767
endocrine organ: physiological role and clinical utility of cardiac natriuretic hor- control
sponsesotothe ANG renalIImedullary circulation.
during inhibition of Clin Exp Pharmacol
superoxide dismutase Physiol. 2010;37(2):
in anesthetized
204. Imig
mones. Am JD, FalckHeart
J Physiol JR, Wei
CircS,Physiol.
et al. Epoxygenase metabolites contribute to ni
2011;301(1):H12–H20. e58–e69.
rats. Am J Physiol Renal Physiol. 2010;298(2):F401–F407.
tric
239.oxidede Boldindependent
AJ. Thirty ayears erent arteriolaronvasodilation
of research in response
atrial natriuretic to bradykinin.
factor: historical back- http://www.ncbi.nlm.nih.gov/pubmed/19566838
267. Carlström M, Lai EY, Ma Z, et al. Superoxide dismutase 1 limits renal mi-
J Vasc Res.
ground and2001;38(3):247–255.
emerging concepts. Can J Physiol Pharmacol. 2011;89(8):527–531. 229. Denton
crovascular KM, Shweta
remodeling andA,attenuates
Anderson arteriole
WP. Preglomerular
and blood and postglomerular
pressure responses
http://www.ncbi.nlm.nih.gov/pubmed/11399897
240. Misono KS, Philo JS, Arakawa T, et al. Structure, signaling mechanism and resistance
to angiotensinresponses II viato modulation
di erent levels of onitric
sympathetic activation by hypoxia.
oxide bioavailability. J Am
Hypertension.
205. Matsuda
regulation of the H,natriuretic
Hayashi K,peptide
Wakinoreceptor
S, et al. guanylate
Role o endothelium
cyclase. FEBS derived hy
J. 2011; Soc Nephrol. 2002;13(1):27–34.
2010;56(5):907–913.
perpolarizing actor in ACE inhibitor induced renal vasodilation in vivo. Hyper-
278(11):1818–1829. 230. Genovesi
268. Ozawa S, Pieruzzi
Y, Hayashi F, Camisasca
K, Wakino S, et al.P, Freeet al. Renalactivity
radical a erentsdependsresponon
tension.
241. 2004;43(3):603–609.
Pandey KN. Guanylyl cyclase/atrial natriuretic peptide receptor-A: role in sive to chemi
underlying cal and mechanical
vasoconstrictors in renalpelvic stimuli in the
microcirculation. Clin rabbit. Clin Sci (Lond).
Exp Hypertens. 2004;
http://www.ncbi.nlm.nih.gov/pubmed/14769805
the pathophysiology of cardiovascular regulation. Can J Physiol Pharmacol. 2011; 1997;92(5):505–510.
26(3):219–229.
206. Omoro SA, Majid DSA, el Dahr SS, et al. Roles o ANG II and bradykinin
89(8):557–573. http://www.ncbi.nlm.nih.gov/pubmed/9176025
269. Welch WJ, Chabrashvili T, Solis G, et al. Role of extracellular superoxide
in the Potter
242. renal regional
LR, Yoder blood
AR, fFlora
ow responses
DR, et al. to ACE inhibition
Natriuretic peptides:in sodium depleted
their structures, 231. KoppinUC,
dismutase Cicha MZ,
the mouse Smith LA,
angiotensin slow et pressor
al. Dietary sodiumHypertension.
response. modulates the in
2006;
dogs. Am Jphysiologic
receptors, Physiol. 2000;279(2):F289–F293.
functions and therapeutic applications. Handb Exp Phar- teraction between e erent and a erent renal nerve activity by altering activation
48(5):934–941.
207. Ren
macol. Y, Garvin JL, Falck JR, et al. Glomerular autacoids stimulated by bra
2009;(191):341–366. o270.
alpha2
Cao adrenoceptors
C, Edwards A,on renal sensory
Sendeski M, et al. nerves. Am Jnitric
Intrinsic Physiol Reguland
oxide Integr Comp
superox-
dykinin
243. regulate
Vives e erent
D, Farage arteriole
S, Motta R, ettone. Kidneynatriuretic
al. Atrial Int. 2003;63(3):987–993.
peptides and urodilatin Physiol. 2011;300(2):R298–R310.
ide production regulates descending vasa recta contraction. Am J Physiol Renal
208. SadowskiJ,
modulate proximalBadzynska
tubule Na(B. )-ATPase
Intrarenalactivity
vasodilator
throughsystems: NO, of
activation prostaglan
the NPR- 232.
Physiol.Carey RM. Theodore Cooper Lecture: Renal dopamine system: para
2010;299(5):F1056–F1064.
dins and bradykinin.
A/cGMP/PKG pathway. AnPeptides.
integrative approach. J Physiol Pharmacol. 2008;59 Sup-
2010;31(5):903–908. crine
271. regulator
Fu Y, Zhang o sodium
R, Lu D,homeostasis
et al. NOX2and is theblood pressure.
primary source Hypertension.
of angiotensin 2001;II-
pl 9: 105–119.
244. Zhao D, Pandey KN, Navar LG. ANP-mediated inhibition of distal nephron 38(3):297–302.
induced superoxide in the macula densa. Am J Physiol Regul Integr Comp Physiol.
209. Wang
fractional X, Trottier
sodium G, Loutzenhiser
reabsorption in wild-type RD.and Determinants o renal a natriuretic
mice overexpressing erent arte http://www.ncbi.nlm.nih.gov/pubmed/11566894
2010;298(3):R707–R712.
riolar actions
peptide receptor.o bradykinin:
Am J Physiolevidence that multiple
Renal Physiol. pathways mediate responses
2010;298(1):F103–F108. 233.
272. LaiJoseEY PA, Eisner GM,
, Wellstein FelderWJ,
A, Welch RA.etRegulation
al. Superoxideo blood pressuremyogenic
modulates by dopamine con-
attributed
245. to EDHF. Am J Physiol
Antunes-Rodrigues RenalM,
J, de Castro Physiol.
Elias 2003;285(3):F540–F549.
LL, et al. Neuroendocrine control receptors.
tractions ofNephron Physiol. 2003;95(2):19–27.
mouse afferent arterioles. Hypertension. 2011;58(4):650–656.
210.
of body Trabold F, Pons S, Hagege
uid metabolism. PhysiolAA,Rev.et2004;84(1):169–208.
al. Cardiovascular phenotypes o kinin B2 http://www.ncbi.nlm.nih.gov/pubmed/14610323
273. Liu R, Ren Y, Garvin JL, et al. Superoxide enhances tubuloglomerular feed-
receptor
246. McKie andPM, tissue kallikrein
Cataliotti de cient mice.
A, Boerrigter G, etHypertension. 2002;40(1):90–95.
al. A novel atrial natriuretic pep- 234.
back by Tayebati
constricting SK, Lokhandwala MF, Amenta
the afferent arteriole. F. Dopamine
Kidney and vascular dynam
Int. 2004;66(1):268–274.
http://www.ncbi.nlm.nih.gov/pubmed/12105144
tide based therapeutic in experimental angiotensin II mediated acute hyperten- ics
274.control:
Ren Ypresent
, D’Ambrosio status and
MA, uture
Liu R,perspectives.
et al. Enhanced Currmyogenic
Neurovascresponse
Res. 2011;8(3):
in the
211. Hypertension.
sion. Guan Z, Inscho EW Role o adenosine 5’ triphosphate in regulating renal
2010;56(6):1152–1159. 246–257.
afferent arteriole of spontaneously hypertensive rats. Am J Physiol Heart Circ
microvascular
247. unction and
Shi SJ, Vellaichamy E, in hypertension.
Chin Hypertension.
SY, et al. Natriuretic 2011;58(3):333–340.
peptide receptor A medi- 235.
Physiol.Wang X, Villar VA, Armando I, et al. Dopamine, kidney, and hyperten
2010;298(6):H1769–H1775.
212.renal
ates Nishiyama
sodium A, Jacksonresponses
excretory KE, MajidtoDSA, bloodetvolume
al. Renal interstitial
expansion. Amf uid ATP
J Physiol sion:
275. studies
Evans in RG, dopamine
Fitzgerald receptor knockout
SM. Nitric oxidemice.and Pediatr
superoxide Nephrol.
in the2008;23(12):
renal me-
responses
Renal Physiol.to 2003;285(4):F694–F702.
arterial pressure and tubuloglomerular eedback activation dur 2131–2146.
dulla: a delicate balancing act. Curr Opin Nephrol Hypertens. 2005;14(1):9–15.
ing calCowley
248. cium channel blockade.
AW Jr. Control Am renal
of the J Physiol Heart Circ
medullary Physiol.by2006;290(2):
circulation vasopressin 236. Zeng C, Jose
276. Kopkan PA. Dopamine
L, Majid receptors:superoxide
DSA. Enhanced important antihypertensive
activity modulates counter
renal
H772–H777.
V1 and V2 receptors in the rat. Exp Physiol. 2000;85 Spec No.:223S–231S. balance
functionagainst
in NO-de hypertensive actors. Hypertension.
cient hypertensive 2011;57(1):11–17.
rats. Hypertension. 2006;47(3):568–572.
130
13 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

237. O’Connor
277. Winaver J,PM, Abassi Z. Role
Cowley AWoJr.neuropeptide
Modulation of Y in the regulation o kidney
pressure-natriuresis by re- 302. Carlström
263. Jackson EK,M,Zhu Lai C,
EY,Tofovic
Ma Z, etSPal. Role o NOX2
. Expression in the regulation
of adenosine receptorso ina theer
unction.
nal medullaryEXS. 2006;(95):123–132.
reactive oxygen species and nitric oxide. Curr Hypertens Rep. ent arteriole responsiveness.
preglomerular microcirculation. AmAm J Physiol
J PhysiolRegul Integr
Renal Comp
Physiol. Physiol. 2009;296(1):
2002;283(1):F41–F51.
http://www.ncbi.nlm.nih.gov/pubmed/16383002
2010;12(2):86–92. R72–R79.
303. Denton KM, Shweta A, Finkelstein L, et al. Effect of endothelin-1 on
238. Matavelli
278. Clerico A,LC, Giannoni
KadowitzA,PJ, Vittorini
Navar LG, S, etet al. Renal
Thirtyhemodynamic
years o the heart as an
and excre- http://www.ncbi.nlm.nih.gov/pubmed/18987286
regional kidney blood ow and renal arteriole calibre in rabbits. Clin Exp Phar-
endocrine
tory responsesorgan: physiologicalinfusion
to intra-arterial role and of clinical utility oincardiac
peroxynitrite natriuretic
anesthetized rats. hor
Am 264. Physiol.
macol Just A, 2004;31(8):494–501.
Olson AJ, Whitten CL, et al. Superoxide mediates acute renal
Jmones.
PhysiolAm RenalJ Physiol
Physiol. Heart Circ Physiol. 2011;301(1):H12–H20.
2009;296(1):F170–F176. vasoconstriction
304. Pollock JS, produced
Pollock DM. by Endothelin
angiotensin and II and catecholamines
NOS1/nitric by a mecha
oxide signaling and
239. Wang
279. de BoldD,AJ.Chabrashvili
Thirty years oT, research
Wilcox on CS.atrial natriuretic
Enhanced actor: historical
contractility of renal back
af- nism independent
regulation of sodiumohomeostasis.
nitric oxide.Curr AmOpinJ Physiol
NephrolHeart Circ Physiol.
Hypertens. 2007;292(1):
2008;17(1):70–75.
groundarterioles
ferent and emerging fromconcepts. Can J Physiol Pharmacol.
angiotensin-infused 2011;89(8):527–531.
rabbits: roles of oxidative stress, H83–H92.
305. Ahmeda AF, Johns EJ. The regulation of blood perfusion in the renal cortex
http://www.ncbi.nlm.nih.gov/pubmed/21671768
thromboxane prostanoid receptors, and endothelium. Circ Res. 2004;94(11): http://www.ncbi.nlm.nih.gov/pubmed/16951043
and medulla by reactive oxygen species and nitric oxide in the anaesthetised rat.
240. Misono KS, Philo JS, Arakawa T, et al. Structure, signaling mechanism and
1436–1442. 265. Physiol
Acta Haque(Oxf).MZ, 2012;204(3):443–450.
Majid DSA. Reduced renal responses to nitric oxide synthase
regulation
280. o the S,
Brakemeier natriuretic
Eichler I,peptide
Knorr A,receptor guanylate of
et al. Modulation cyclase. FEBS J. 2011;
Ca2 -activated K inhibition
306. in mice
Rajapakse NW,lacking the gene
Sampson AK, or Eppelgp91phox
GA, et subunit o NAD(P)H
al. Angiotensin II andoxidase.
nitric
278(11):1818–1829.
channel in renal artery endothelium in situ by nitric oxide and reactive oxygen Am J Physiol
oxide in neural Renal Physiol.
control of 2008;295(3):F758–F764.
intrarenal blood ow. Am J Physiol Regul Integr Comp
http://www.ncbi.nlm.nih.gov/pubmed/21375693
species. Kidney Int. 2003;64(1):199–207. 266. Khan
Physiol. MA, Islam MT, Castillo A, et al. Attenuation o renal excretory re
2005;289(3):R745–R754.
241. Fellner
281. PandeySK, KN.Arendshorst
Guanylyl cyclase/atrial
WJ. Angiotensin natriuretic peptideoxygen
II, reactive receptor A: role
species in
and sponses
307. to ANG
Fuson II duringP,inhibition
AL, Komlosi Unlap TM, o etsuperoxide dismutase in anesthetized
al. Immunolocalization of a micro-
the pathophysiology
Ca2 signaling in afferent o cardiovascular
arterioles. regulation.
Am J Physiol Can J Physiol
Renal Pharmacol.
Physiol. 2011;
2005;289(5): rats. Am
somal J Physiol RenalEPhysiol.
prostaglandin synthase 2010;298(2):F401–F407.
in rabbit kidney. Am J Physiol Renal Physiol.
89(8):557–573.
F1012–F1019. 267. Carlström M, Lai EY, Ma Z, et al. Superoxide dismutase 1 limits renal mi
2003;285(3):F558–F564.
242. Hughes
282. Potter LR, AK,Yoder
Kohan AR,DE.Flora DR, et al.ofNatriuretic
Mechanism peptides: their
vasopressin-induced structures,
contraction of crovascular
308. Hollenbergremodeling
NK. Directand attenuates
renin inhibition arteriole
andand theblood
kidney.pressure
Nat Revresponses
Nephrol.
receptors,
renal physiologic
medullary unctions
interstitial and therapeutic
cells. Nephron applications. Handb Exp Phar-
Physiol. 2006;103(3):119–124. to angiotensin II via modulation o nitric oxide bioavailability. Hypertension.
2010;6(1):49–55.
macol.Kim
283. 2009;(191):341–366.
J, Pannabecker TL. Two-compartment model of inner medullary vas- 2010;56(5):907–913.
309. Gross V, Kurth TM, Skelton MM, et al. Effects of daily sodium intake
243. Vives
culature D, Farage
supports dualS,modes
Motta of R, vasopressin-regulated
et al. Atrial natriuretic innerpeptides and urodilatin
medullary blood 268.ANG
and Ozawa II onY,cortical
Hayashiand K, medullary
Wakino S,renal et al.blood
Free radical activity depends
ow in conscious rats. AmonJ
modulate
ow. Am J proximal
Physiol Renal tubule Na(+)2010;299(1):F273–F279.
Physiol. ATPase activity through activation o the NPR underlying
Physiol. vasoconstrictors
1998;274(5 in renal microcirculation. Clin Exp Hypertens. 2004;
Pt 2):R1317–R1323.
A/cGMP/PKG
284. Cowley AW pathway. Peptides.
Jr, Mori 2010;31(5):903–908.
T, Mattson DL, et al. Role of renal NO production in 26(3):219–229.
310. Rodríguez F, Llinás MT, González JD, et al. Renal changes induced by a
244.regulation
the Zhao D, of Pandey KN, Navar
medullary bloodLG.ow. ANP Ammediated
J Physiolinhibition
Regul Integr o distal
Compnephron
Physiol. http://www.ncbi.nlm.nih.gov/pubmed/15132300
cyclooxygenase-2 inhibitor during normal and low sodium intake. Hypertension.
ractional sodium reabsorption in wild type and mice overexpressing natriuretic
2003;284(6):R1355–R1369. 269. Welch WJ, Chabrashvili T, Solis G, et al. Role o extracellular superoxide
2000;36(2):276–281.
peptide
285. receptor.
Majid DSA, Am SaidJ Physiol
KE, Omoro RenalSA, Physiol.
et al.2010;298(1):F103–F108.
Nitric oxide dependency of arterial dismutase
311. Tojo in A, the mouse
Kimoto M,angiotensin
Wilcox CS. slow Renalpressor response.
expression Hypertension.
of constitutive NOS2006;
and
245. Antunes Rodrigues
pressure-induced changes J,inderenal
Castro M, Eliashydrostatic
interstitial LL, et al. Neuroendocrine
pressure in dogs. control
Circ 48(5):934–941.
DDAH: separate effects of salt intake and angiotensin. Kidney Int. 2000;58(5):
o body
Res. f uid metabolism. Physiol Rev. 2004;84(1):169–208.
2001;88(3):347–351. http://www.ncbi.nlm.nih.gov/pubmed/17015770
2075–2083.
http://www.ncbi.nlm.nih.gov/pubmed/14715914
286. O’Connor PM, Cowley AW Jr. Vasopressin-induced nitric oxide produc- 270. Bergström
312. Cao C, Edwards G, Evans A, Sendeski M, et al. Intrinsic
RG. Mechanisms underlying nitricthe
oxide and superox
antihypertensive
246.inMcKie
tion rat innerPM,medullary
Cataliotti collecting
A, Boerrigter ductG,isetdependent
al. A novelonatrial natriuretic
V2 receptor pep
activa- ide production
functions of the regulates
renal medulla.descending vasa recta
Acta Physiol Scand.contraction. Am J Physiol Renal
2004;181(4):475–486.
tide based
tion of the therapeutic
phosphoinositide in experimental
pathway. Am angiotensin
J Physiol II mediated
Renal Physiol.acute hyperten
2007;293(2): Physiol.
313. 2010;299(5):F1056–F1064.
Chen J, Zhao M, He W, et al. Increased dietary NaCl induces renal medul-
sion. Hypertension. 2010;56(6):1152–1159.
F526–F532. 271.PGE2
lary Fu Y,production
Zhang R, Lu andD, natriuresis
et al. NOX2viais the the EP2
primary sourceAm
receptor. o Jangiotensin
Physiol Renal II
http://www.ncbi.nlm.nih.gov/pubmed/20975033
287. Bankir L. Antidiuretic action of vasopressin: quantitative aspects and in- induced2008;295(3):F818–F825.
Physiol. superoxide in the macula densa. Am J Physiol Regul Integr Comp Physiol.
247. Shibetween
teraction SJ, Vellaichamy
V1a andE,V2 Chin SY, et al. Natriuretic
receptor-mediated peptide
effects. receptor
Cardiovasc Res.A2001;
medi 2010;298(3):R707–R712.
314. Yang T, Singh I, Pham H, et al. Regulation of cyclooxygenase expres-
ates renal sodium excretory responses to blood volume expansion. Am J Physiol
51(3):372–390. 272. inLaithe
sion EY,kidney
Wellstein byA,dietary
Welch salt WJ, et al. Superoxide
intake. Am J Physiol.modulates myogenic
1998;274(3 Ptcon
2):
Renal Correia
288. Physiol. 2003;285(4):F694–F702.
AG, Denton KM, Evans RG. Effects of activation of vasopressin- tractions o mouse a erent arterioles. Hypertension. 2011;58(4):650–656.
F481–F489.
248. CowleyonAW
V1-receptors Jr. Control
regional kidney o blood
the renal owmedullary circulation
and glomerular by vasopressin
arteriole diameters. 273. Y
315. Liu
e W,R,Zhang
Ren Y,H, Garvin
HillasJL, E, et
et al.
al. Superoxide
Expression enhances
and function tubuloglomerular
of COX isoforms eedin
JV1 and V2 receptors
Hypertens. 2001;19(3 in Pt
the2):649–657.
rat. Exp Physiol. 2000;85 Spec No.:223S 231S. back by
renal constricting
medulla: evidence the for
a erent arteriole.
regulation Kidney
of salt Int. 2004;66(1):268–274.
sensitivity and blood pressure. Am
249. Matsuda
289. Vagnes OB, Hansen K,
H, Hayashi FH,Arakawa
Feng JJ,K,et etal.al.Enhanced Ca2+ response
Distinct modulation to AVP
of super - J274. Ren
Physiol Y, D’Ambrosio
Renal MA, Liu R, et al. Enhanced myogenic response in the
Physiol. 2006;290(2):F542–F549.
in preglomerular
cial and juxtamedullary vesselsarterioles
rom rats bywith genetic hypertension
prostaglandin during
in vivo. Hypertens di 2002;
Res. erent a erent
316. arteriole
Bech o spontaneously
JN, Nielsen CB, Ivarsenhypertensive
P, et al. Dietaryrats.sodium
Am J Physiol Heart
affects Circ Phys-
systemic and
hydration states. Am J Physiol Renal Physiol. 2005;288(6):F1249–F1256.
25(6):901–910. iol. 2010;298(6):H1769–H1775.
renal hemodynamic response to NO inhibition in healthy humans. Am J Phyisol.
250. Badzynska
290. Aoyagi T, Izumi Y, Hiroyama
B, Sadowski J. OpposedM, et effects
al. Vasopressin regulatesE2theonrenin
of prostaglandin per- 275. Evans Pt
1998;274(5 RG, Fitzgerald SM. Nitric oxide and superoxide in the renal me
2):F914–F923.
angiotensin
fusion of rat aldosterone
renal cortexsystem via V1ainteractions
and medulla: receptors inwith macula densa cells. Am J
the renin-angiotensin dulla: Sanders
317. a delicate PW.balancing
Vascularact. Curr Opin Nephrol
consequences Hypertens.
of dietary 2005;14(1):9–15.
salt intake. Am J Physiol Re-
Physiol Renal
system. Physiol.2008;93(12):1292–1302.
Exp Physiol. 2008;295(1):F100–F107. 276.Physiol.
nal Kopkan L, Majid DSA. Enhanced superoxide activity modulates renal
2009;297(2):F237–F243.
251. Carey
291. Cavarape RM.A,UpdateBauer J,onBartoli
the roleE, etofal.the
E ects
AT2 oreceptor.
angiotensinCurrII,Opin
arginine
Nephrolva unction
318. Zhao in X,
NOPollock
de cient DM,hypertensive
Inscho EW,rats. et al.Hypertension.
Decreased renal 2006;47(3):568–572.
cytochrome P450
sopressin and
Hypertens. thromboxane A2 in renal vascular bed: role o rho kinase. Nephrol
2005;14(1):67–71. http://www.ncbi.nlm.nih.gov/pubmed/16401762
2C enzymes and impaired vasodilation are associated with angiotensin salt-
Dial Transplant.
292. 2003;18(9):1764–1769.
Zhang Z, Rhinehart K, Kwon W, et al. ANG II signaling in vasa recta peri- 277. O’Connor
sensitive PM, Cowley
hypertension. AW Jr.2003;41(3
Hypertension. Modulation Pt o2):709–714.
pressure natriuresis by re
252. by
cytes Fallet
PKCRW, andIkenaga
reactiveH, Bast JPspecies.
oxygen , et al. Relative contributions
Am J Physiol Heart Circ o Physiol.
Ca2 + mobili
2004; nal medullary
319. Fujihara CK, reactive oxygen
De Nucci species
G, Zatz and nitric role
R. Physiologic oxide. Currreceptors
of ETA HypertensinRep.the
zation and inf ux in renal arteriolar contractile responses to arginine vasopressin.
287(2):H773–H781. 2010;12(2):86–92.
regulation of renal hemodynamics in normal and salt-depleted rats. J Cardiovasc
Am J Physiol
293. Rajapakse Renal NW,Physiol. 2005;288(3):F545–F551.
Mattson DL. Role of L-arginine in nitric oxide production http://www.ncbi.nlm.nih.gov/pubmed/20424940
Pharmacol. 1995;26 Suppl 3:S466–S469.
253.
in Rajapakse
health NW, RomanClin
and hypertension. RJ,ExpFalck JR, et al.Physiol.
Pharmacol Modulation o V1 receptor me
2009;36(3):249–255. 278.
320. V Matavelli LC, Kadowitz
anni S, Polidori G, Cecioni PJ,I,Navar LG, etreceptor
et al. ET(B) al. Renal hemodynamic
in renal medulla is and en-
di atedMori
294. renal vasoconstriction
T, O’Connor PM, AbebyM,epoxyeicosatrienoic
et al. Enhanced superoxideacids. Am J Physiol in
production Regul
re- excretory
hanced byresponses
local sodium to intra
during arterial
low saltin intake.
usion oHypertension.
peroxynitrite in anesthetized rats.
2002;40(2):179–185.
Integr
nal Comp
outer Physiol.
medulla of2004;287(1):R181–R187.
Dahl salt-sensitive rats reduces nitric oxide tubular-vascular Am J Physiol
321. Fattal I,Renal
AbassiPhysiol. 2009;296(1):F170–F176.
Z, Ovcharenko E, et al. Effect of dietary sodium intake on
254. Ando
cross-talk. K, Shimosawa
Hypertension. T, Fujita T. Adrenomedullin in vascular diseases. Curr
2007;49(6):1336–1341. 279.expression
the Wang D,ofChabrashvili T, Wilcox CS.
endothelin-converting enzymeEnhanced
in the contractility
renal medulla. o renal
Nephrona
Hypertens
295. Abe Rep. 2004;6(1):55–59.
M, O’Connor P, Kaldunski M, et al. Effect of sodium delivery on super- erent arterioles
Physiol. rom angiotensin in used rabbits: roles o oxidative stress,
2004;98(4):89–96.
http://www.ncbi.nlm.nih.gov/pubmed/14972095
oxide and nitric oxide in the medullary thick ascending limb. Am J Physiol Renal thromboxane
322. Taylor TA, prostanoid
Gariepy CE, receptors,
Pollockand DM,endothelium.
et al. GenderCirc Res. 2004;94(11):
differences in ET and
255. Bell
Physiol. D, McDermott BJ. Intermedin (adrenomedullin 2): a novel counter
2006;291(2):F350–F357. 1436–1442.
NOS systems in ETB receptor-de cient rats: effect of a high salt diet. Hyperten-
regulatory
296. peptide inCG.
Schnackenberg thePhysiological
cardiovascular andand renal systems. Br
pathophysiological J Pharmacol.
roles of oxygen http://www.ncbi.nlm.nih.gov/pubmed/15117817
sion. 2003;41(3 Pt 2):657–662.
2008;153
radicals in Suppl
the renal1:S247–S262.
microvasculature. Am J Physiol Regul Integr Comp Physiol. 280. Lai
323. Brakemeier
EY, Onozato S, Eichler
ML, SolisI, Knorr
G, etA, al.etMyogenic
al. Modulation
responses o Ca2+ activated
of mouse K+
isolated
256. Brain SD, Grant AD. Vascular actions o calcitonin gene related peptide
2002;282(2):R335–R342. channel inrenal
perfused renalafferent
artery arterioles:
endothelium in situ
effects by nitric
of salt intakeoxide and reactive
and reduced renaloxygen
mass.
and adrenomedullin.
297. Vera T, Stec DE. Physiol Moderate Rev.hyperbilirubinemia
2004;84(3):903–934. improves renal hemodynam- species. Kidney
Hypertension. Int. 2003;64(1):199–207.
2010;55(4):983–989.
257.
ics Nishikimi
in ANG T. Adrenomedullin
II-dependent hypertension.in Am theJ kidney—renal
Physiol Regul Integr physiological
Comp Physiol.and http://www.ncbi.nlm.nih.gov/pubmed/12787410
324. Lau C, Sudbury I, Thomson M, et al. Salt-resistant blood pressure and salt-
pathophysiological roles. Curr Med Chem. 2007;14(15):1689–1699.
2010;299(4):R1044–R1049. 281. Fellner
sensitive renalSK, Arendshorst in
autoregulation WJ. Angiotensin
chronic II, reactive
streptozotocin oxygenAm
diabetes. species and
J Physiol
258. Oliver
298. Bedard JJ, K, Eppel
Krause GA,KH. Rajapakse
The NOX NW, amilyeto al. ROS generating NADPH
Lipoxygenase and cyclo-oxi Ca2+ Integr
Regul signalingCompinPhysiol.
a erent2009;296(6):R1761–R1770.
arterioles. Am J Physiol Renal Physiol. 2005;289(5):
dases: physiology
oxygenase products andin pathophysiology.
the control of regional Physiolkidney
Rev. 2007;87(1):245–313.
blood ow in rabbits. Clin F1012–F1019.
325. Saeed A, Dibona GF, Marcussen N, et al. High NaCl intake impairs dy-
259.Pharmacol
Exp ClempusPhysiol. RE, Griendling KK. Reactive oxygen species signaling in vascular
2003;30(11):812–819. 282. Hughes
namic AK, Kohan
autoregulation DE. Mechanism
of renal blood owoinvasopressinangiotensininduced contraction
II-infused rats. AmoJ
smooth
299. muscle cells.
Kuczeriszka M,Cardiovasc
Badzynska Res.B,2006;71(2):216–225.
Kompanowska-Jezierska E. Cytochrome renal medullary
Physiol Regul Integr interstitial cells. Nephron
Comp Physiol. Physiol. 2006;103(3):119–124.
2010;299(5):R1142–R1149.
http://www.ncbi.nlm.nih.gov/pubmed/16616906
P-450 monooxygenases in control of renal haemodynamics and arterial pressure 283. Hoagland
326. Kim J, Pannabecker
KM, Flasch TL.AK, TwoRoman
compartment model of
RJ. Inhibitors o inner
20-HETEmedullary vas
formation
260.
in Reckelho rats.
anaesthetized JF, Romero
J PhysiolJC. Role o oxidative
Pharmacol. 2006;57 Supplstress in angiotensin induced
11:179–185. culature salt-sensitive
promote supports dualhypertension
modes o vasopressin regulated inner
in rats. Hypertension. medullary blood
2003;42(4):669–673.
hypertension.
300. Badzynska AmB,J Physiol
Sadowski RegulJ. Integr Comp action
Differential Physiol.of2003;284(4):R893–R912.
bradykinin on intrarenal f ow. Am
327. J Physiol Renal
Dobrowolski Physiol. 2010;299(1):F273–F279.
L, Kompanowska-Jezierska E, Walkowska A, et al. Sodium
261. Ushio
regional FukaiinM.theVascular
perfusion rat: waningsignaling
effectthrough G protein
in the cortex coupled
and major receptors:
impact in the 284. Cowley
intake determines AW the Jr, Mori
role of T, adenosine
Mattson DL, A2etreceptors
al. Role oin renal
control NOofproduction
renal medul- in
new concepts.
medulla. Curr2009;587(Pt
J Physiol. Opin Nephrol Hypertens. 2009;18(2):153–159.
15):3943–3953. the regulation
lary perfusion in o the
medullary bloodDial
rat. Nephrol f ow. Am J Physiol
Transplant. Regul Integr Comp Physiol.
2007;22(10):2805–2809.
http://www.ncbi.nlm.nih.gov/pubmed/19434053
301. Cheng MK, Doumad AB, Jiang H, et al. Epoxyeicosatrienoic acids mediate 2003;284(6):R1355–R1369.
328. Walkowska A, Dobrowolski L, Kompanowska-Jezierska E, et al. Role of
262. Gill PS, Wilcox
adenosine-induced CS. NADPH
vasodilation in oxidases in the kidney.
rat preglomerular Antioxid Redox
microvessels (PGMV) Signal.
via 285.and
NO Majid
COX DSA, Said KE,
pathways in Omoro
mediation SA, of et al. Nitric oxide
adenosine dependency o arterial
A1 receptor-induced renal
2006;8(9–10):1597–1607.
A2A receptors. Br J Pharmacol. 2004;141(3):441–448. pressure induced Exp
vasoconstriction. changes
Biol Medin renal interstitial
(Maywood). hydrostatic pressure in dogs. Circ
2007;232(5):690–694.
http://www.ncbi.nlm.nih.gov/pubmed/16987014 Res. 2001;88(3):347–351.
CHAPTER 3 RENAL CIRCULATION AND GLOMERULAR HEMODYNAMICS 1131
31

286. O’Connor
329. Fujii S, ZhangPM,L, Cowley
Igarashi AW Jr. L-arginine
J, et al. Vasopressin induced
reverses nitric and
p47phox oxide produc
gp91phox 308. Hollenberg NK.
341. Gonin-Jmaa D, Direct
Senior renin
DF. Theinhibition
hyper and the kidney.
ltration theory:Nat Rev Nephrol.
progression of
tion in rat induced
expression inner medullary collecting
by high salt ductHypertension.
in Dahl rats. is dependent2003;42(5):1014–1020.
on V2 receptor activa 2010;6(1):49–55.
chronic renal failure and the effects of diet in dogs. J Am Vet Med Assoc. 1995;
tion oJohns
330. the phosphoinositide
EJ, O’Shaughnessy pathway. Am J Physiol
B, O’Neill S, et al.Renal Physiol.
Impact of 2007;293(2):
elevated di- http://www.ncbi.nlm.nih.gov/pubmed/19935744
207(11):1411–1415.
F526–F532.
etary sodium intake on NAD(P)H oxidase and SOD in the cortex and me- 309. Gross V, E,
342. Brändle Kurth TM, Skelton
Sieberth MM, et al.
HG, Hautmann RE.EEffect
ects oofdaily sodium
chronic intake
dietary and
protein
287. of
dulla Bankir L. Antidiuretic
the rat kidney. Am JactionPhysiol o Regul
vasopressin: quantitative
Integr Comp Physiol.aspects and in
2010;299(1): ANG
intakeIIonon the
cortical
renaland medullary
function renal blood
in healthy f ow in
subjects. Eurconscious rats.1996;50(11):
J Clin Nutr. Am J Physiol.
teraction between V1a and V2 receptor mediated e ects. Cardiovasc Res. 2001;
R234–R240. 1998;274(5
734–740. Pt 2):R1317–R1323.
51(3):372–390.
331. Lara LS, McCormack M, Semprum-Prieto LC, et al. AT1 receptor-mediated 310. RodríguezJ,F,Carlström
343. Sallström Llinás MT, M,González
Olerud J,JD, et High-protein-induced
et al. al. Renal changes induced by a
glomeru-
http://www.ncbi.nlm.nih.gov/pubmed/11476728
augmentation of angiotensinogen, oxidative stress, and in ammation in ANG cyclooxygenase
lar hyper ltration 2 inhibitor duringof
is independent normal and low sodiumfeedback
the tubuloglomerular intake. Hypertension.
mechanism
288. Correia
II-salt AG, Denton
hypertension. KM, Evans
Am J Physiol RenalRG. E ects
Physiol. o activation o vasopressin V1
2012;302(1):F85–F94. 2000;36(2):276–281.
and nitric oxide synthases. Am J Physiol Regul Integr Comp Physiol. 2010;299(5):
receptors
332. on regional
Kitiyakara kidney blood
C, Chabrashvili f owY,and
T, Chen et al.glomerular
Salt intake,arteriole
oxidativediameters.
stress, andJ http://www.ncbi.nlm.nih.gov/pubmed/10948090
R1263–R1268.
Hypertens.
renal 2001;19(3
expression Pt 2):649–657.
of NADPH oxidase and superoxide dismutase. J Am Soc Nephrol. 311. Tojo A, Kimoto
344. Schrijvers BF, RaschM, Wilcox
R, Tilton CS.RG,
Renal expression
et al. o constitutiveglomerular
High protein-induced NOS and
http://www.ncbi.nlm.nih.gov/pubmed/11327642
2003;14(11):2775–2782. DDAH:
hypertrophyseparate e ects oendothelial
is vascular salt intake and angiotensin.
growth factor-dependent.KidneyKidney
Int. 2000;58(5):
Int. 2002;
289. Schulman
333. Matsuda H, IH,Hayashi
Zhou MS, K, Raij
Arakawa K, et al. between
L. Interaction Distinct modulation o super
nitric oxide and angio- 2075–2083.
61(5):1600–1604.
cial andII juxtamedullary
tensin in the endothelium: arterioles
role inbyatherosclerosis
prostaglandinand in vivo. HypertensJRes.
hypertension. 2002;
Hypertens 312. Bergström
345. Wake eld APG,, House
EvansJD, RG. Mechanisms
Ogborn MR, et al.underlying
A diet withthe 35% antihypertensive
of energy from
25(6):901–910.
Suppl. 2006;24(1):S45–S50. unctions
protein o the
leads to renal
kidney medulla.
damageActa PhysiolSprague-Dawley
in female Scand. 2004;181(4):475–486.
rats. Br J Nutr. 2011;
http://www.ncbi.nlm.nih.gov/pubmed/12484515
334. Toda N, Arakawa K. Salt-induced hemodynamic regulation mediated by http://www.ncbi.nlm.nih.gov/pubmed/15283761
106(5):656–663.
290. oxide.
nitric Badzynska B, Sadowski
J Hypertens. J. Opposed e ects o prostaglandin E2 on per
2011;29(3):415–424. 313. Chen J, JE,
346. Benabe ZhaoWangM, He W, et al.JN,
S, Wilcox Increased dietary NaCl
et al. Modulation of induces renal medul
ANG II receptor and
usionKopkan
335. o rat renal
L, Hesscortex and medulla:
A, Husková Z, et al.interactions with the
High-salt intake renin angiotensin
enhances superoxide lary PGE2 inproduction
its mRNA normal ratand natriuresis via
by low-protein the EP2
feeding. Amreceptor.
J Physiol. Am J Physiol Pt
1993;265(5 Renal
2):
system. in
activity ExpeNOS
Physiol. 2008;93(12):1292–1302.
knockout mice leading to the development of salt sensitivity. Physiol.
F660–F669.2008;295(3):F818–F825.
291.J Physiol
Am Carey Renal
RM. Update
Physiol.on the role o the AT2 receptor. Curr Opin Nephrol Hy-
2010;299(3):F656–F663. 314. Yang T, Singh
347. Brenner I, Pham H,
BM. Retarding et progression
the al. Regulation of orenal
cyclooxygenase
disease. Kidney expres
Int.sion
2003;in
pertens.
336. 2005;14(1):67–71.
Chabrashvili T, Kitiyakara C, Blau J, et al. Effects of ANG II type 1 and 2 the kidney by dietary salt intake. Am J Physiol. 1998;274(3 Pt 2): F481–F489.
64(1):370–378.
http://www.ncbi.nlm.nih.gov/pubmed/15586018
receptors on oxidative stress, renal NADPH oxidase, and SOD expression. Am J 315.
348. De Ye W, Zhang
Santo NG,H,Perna
HillasA,E,Cirillo
et al. Expression
M. Low proteinand unction
diets areomainstay
COX isofor orms
man-in
292. Zhang
Physiol Z, Rhinehart
Regul Integr K, Kwon
Comp Physiol. W, et al. ANG II signaling in vasa recta peri
2003;285(1):R117–R124. renal
agementmedulla: evidence
of chronic kidney or disease.
regulation o Biosci
Front salt sensitivity
(Schol Ed). and blood pressure. Am
2011;3:1432–1442.
cytes by
337. PKCUK,
Dutta andLane
reactive oxygenLJ,species.
J, Roberts Am J Physiolformation
et al. Superoxide Heart Circ Physiol.
and 2004;
interaction J349.
Physiol Renal
Levey AS,Physiol.
Greene 2006;290(2):F542–F549.
T, Sarnak MJ, et al. Effect of dietary protein restriction
287(2):H773–H781.
with nitric oxide modulate systemic arterial pressure and renal function in salt- 316.
on theBech JN, Nielsen
progression CB, Ivarsen
of kidney disease:P,long-term
et al. Dietary sodiumofathe
follow-up ects
Modisystemic
cationandof
293. Rajapakse
depleted dogs. Exp NW,BiolMattson DL. Role 2006;231(3):269–276.
Med (Maywood). o L arginine in nitric oxide production renal
Diet inhemodynamic
Renal Diseaseresponse(MDRD)toStudy.NO inhibition
Am J Kidney in Dis.
healthy humans. Am J Phyisol.
2006;48(6):879–888.
in health
338. FrankandH,hypertension. Clin Exp Pharmacol
Graf J, Amann-Gassner Physiol.
U, et al. Effect of2009;36(3):249–255.
short-term high-protein 1998;274(5
350. Mandayam Pt 2):F914–F923.
S, Mitch WE. Dietary protein restriction bene ts patients with
http://www.ncbi.nlm.nih.gov/pubmed/19076168
compared with normal-protein diets on renal hemodynamics and associated vari- 317.
chronic Sanders
kidneyPW. Vascular
disease. consequences
Nephrology o dietary
(Carlton). salt intake. Am J Physiol Re-
2006;11(1):53–57.
294. inMori
ables T, O’Connor
healthy young men. PM,AmAbe M,Nutr.
J Clin et al. Enhanced superoxide production
2009;90(6):1509–1516. nal
351.Physiol. 2009;297(2):F237–F243.
Phisitkul S, Hacker C, Simoni J, et al. Dietary protein causes a decline
in renal
339. outer medulla
Slomowitz o Dahl
LA, Gabbai FB,salt sensitive
Khang SJ, etrats
al. reduces
Protein nitric
intakeoxide tubular
regulates the 318.
in the Zhao X, Pollock
glomerular DM, Inscho
ltration rate of EW, et al. Decreased
the remnant kidney renal cytochrome
mediated P450
by metabolic
vascular crossfunction
vasodilatory talk. Hypertension.
of the kidney 2007;49(6):1336–1341.
and NMDA receptor expression. Am J Physiol 2C enzymes
acidosis and and impaired
endothelin vasodilation
receptors. KidneyareInt.
associated with angiotensin salt sen
2008;73(2):192–199.
http://www.ncbi.nlm.nih.gov/pubmed/17470722
Regul Integr Comp Physiol. 2004;287(5):R1184–R1189. sitive
352. hypertension.
Green T, Rodriguez Hypertension.
J, Navar2003;41(3
LG. AugmentedPt 2):709–714.
cyclooxygenase-2 effects on
295. Pecoits-Filho
340. Abe M, O’Connor P, Kaldunski
R. Dietary proteinM, et al.and
intake E ect o sodium
kidney diseasedelivery on super
in Western diet. http://www.ncbi.nlm.nih.gov/pubmed/12623984
renal function during varying states of angiotensin II. Am J Physiol Renal Physiol.
oxide and
Contrib nitric 2007;155:102–112.
Nephrol. oxide in the medullary thick ascending limb. Am J Physiol Renal 319. Fujihara CK, De Nucci G, Zatz R. Physiologic role o ETA receptors in the
2010;299:F954–F962.
Physiol. 2006;291(2):F350–F357. regulation o renal hemodynamics in normal and salt depleted rats. J Cardiovasc
296. Schnackenberg CG. Physiological and pathophysiological roles o oxygen Pharmacol. 1995;26 Suppl 3:S466–S469.
radicals in the renal microvasculature. Am J Physiol Regul Integr Comp Physiol. 320. Vanni S, Polidori G, Cecioni I, et al. ET(B) receptor in renal medulla is en
2002;282(2):R335–R342. hanced by local sodium during low salt intake. Hypertension. 2002;40(2):179–185.
297. Vera T, Stec DE. Moderate hyperbilirubinemia improves renal hemody 321. Fattal I, Abassi Z, Ovcharenko E, et al. E ect o dietary sodium intake on
nam ics in ANG II dependent hypertension. Am J Physiol Regul Integr Comp Phys- the expression o endothelin converting enzyme in the renal medulla. Nephron
iol. 2010;299(4):R1044–R1049. Physiol. 2004;98(4):89–96.
298. Oliver JJ, Eppel GA, Rajapakse NW, et al. Lipoxygenase and cyclo oxyge 322. Taylor TA, Gariepy CE, Pollock DM, et al. Gender di erences in ET and
nase products in the control o regional kidney blood f ow in rabbits. Clin Exp NOS systems in ETB receptor de cient rats: e ect o a high salt diet. Hyperten-
Pharmacol Physiol. 2003;30(11):812–819. sion. 2003;41(3 Pt 2):657–662.
http://www.ncbi.nlm.nih.gov/pubmed/14678242 323. Lai EY, Onozato ML, Solis G, et al. Myogenic responses o mouse isolated
299. Kuczeriszka M, Badzynska B, Kompanowska Jezierska E. Cytochrome per used renal a erent arterioles: e ects o salt intake and reduced renal mass.
P 450 monooxygenases in control o renal haemodynamics and arterial pressure Hypertension. 2010;55(4):983–989.
in anaesthetized rats. J Physiol Pharmacol. 2006;57 Suppl 11:179–185. 324. Lau C, Sudbury I, Thomson M, et al. Salt resistant blood pressure and
http://www.ncbi.nlm.nih.gov/pubmed/17244949 salt sensitive renal autoregulation in chronic streptozotocin diabetes. Am J Physiol
300. Badzynska B, Sadowski J. Di erential action o bradykinin on intrarenal Regul Integr Comp Physiol. 2009;296(6):R1761–R1770.
regional per usion in the rat: waning e ect in the cortex and major impact in the 325. Saeed A, Dibona GF, Marcussen N, et al. High NaCl intake impairs dy
medulla. J Physiol. 2009;587(Pt 15):3943–3953. namic autoregulation o renal blood f ow in angiotensin II in used rats. Am J
301. Cheng MK, Doumad AB, Jiang H, et al. Epoxyeicosatrienoic acids mediate Physiol Regul Integr Comp Physiol. 2010;299(5):R1142–R1149.
adenosine induced vasodilation in rat preglomerular microvessels (PGMV) via 326. Hoagland KM, Flasch AK, Roman RJ. Inhibitors o 20 HETE ormation
A2A receptors. Br J Pharmacol. 2004;141(3):441–448. promote salt sensitive hypertension in rats. Hypertension. 2003;42(4):669–673.
302. Jackson EK, Zhu C, To ovic SP. Expression o adenosine receptors in http://www.ncbi.nlm.nih.gov/pubmed/12874093
the preglomerular microcirculation. Am J Physiol Renal Physiol. 2002;283(1): 327. Dobrowolski L, Kompanowska Jezierska E, Walkowska A, et al. Sodium
F41–F51. intake determines the role o adenosine A2 receptors in control o renal medul
303. Denton KM, Shweta A, Finkelstein L, et al. E ect o endothelin 1 on re lary per usion in the rat. Nephrol Dial Transplant. 2007;22(10):2805–2809.
gional kidney blood f ow and renal arteriole calibre in rabbits. Clin Exp Phar macol http://www.ncbi.nlm.nih.gov/pubmed/17556415
Physiol. 2004;31(8):494–501. 328. Walkowska A, Dobrowolski L, Kompanowska Jezierska E, et al. Role o
http://www.ncbi.nlm.nih.gov/pubmed/15298540 NO and COX pathways in mediation o adenosine A1 receptor induced renal
304. Pollock JS, Pollock DM. Endothelin and NOS1/nitric oxide signaling and vasoconstriction. Exp Biol Med (Maywood). 2007;232(5):690–694.
regulation o sodium homeostasis. Curr Opin Nephrol Hypertens. 2008;17(1):70–75. http://www.ncbi.nlm.nih.gov/pubmed/17463166
http://www.ncbi.nlm.nih.gov/pubmed/18090673 329. Fujii S, Zhang L, Igarashi J, et al. L arginine reverses p47phox and gp
305. Ahmeda AF, Johns EJ. The regulation o blood per usion in the renal cortex 91phox expression induced by high salt in Dahl rats. Hypertension. 2003;42(5):
and medulla by reactive oxygen species and nitric oxide in the anaesthetised rat. 1014–1020.
Acta Physiol (Oxf). 2012;204(3):443–450. http://www.ncbi.nlm.nih.gov/pubmed/14504257
http://www.ncbi.nlm.nih.gov/pubmed/21827636 330. Johns EJ, O’Shaughnessy B, O’Neill S, et al. Impact o elevated dietary
306. Rajapakse NW, Sampson AK, Eppel GA, et al. Angiotensin II and nitric sodium intake on NAD(P)H oxidase and SOD in the cortex and medulla o the
oxide in neural control o intrarenal blood f ow. Am J Physiol Regul Integr Comp rat kidney. Am J Physiol Regul Integr Comp Physiol. 2010;299(1): R234–R240.
Physiol. 2005;289(3):R745–R754. http://www.ncbi.nlm.nih.gov/pubmed/20427726
307. Fuson AL, Komlosi P, Unlap TM, et al. Immunolocalization o a micro 331. Lara LS, McCormack M, Semprum Prieto LC, et al. AT1 receptor mediated
somal prostaglandin E synthase in rabbit kidney. Am J Physiol Renal Physiol. augmentation o angiotensinogen, oxidative stress, and inf ammation in ANG II
2003;285(3):F558–F564. salt hypertension. Am J Physiol Renal Physiol. 2012;302(1):F85–F94.
(contd.)
132
131
13 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

332. O’Connor
277. Kitiyakara PM, C, Chabrashvili
Cowley AW T, Jr. Chen Y, et al.ofSalt
Modulation intake, oxidative by
pressure-natriuresis stress,
re- 343. Jackson
302. SallströmEK, J, Carlström M, Olerud
Zhu C, Tofovic J, et al. High
SP. Expression protein induced
of adenosine glomeru
receptors in the
and medullary
nal renal expression
reactiveo oxygen
NADPHspeciesoxidaseand andnitric
superoxide dismutase.
oxide. Curr J AmRep.
Hypertens Soc lar hyperf ltration
preglomerular is independent
microcirculation. AmoJ Physiol
the tubuloglomerular eedback mechanism
Renal Physiol. 2002;283(1):F41–F51.
Nephrol. 2003;14(11):2775–2782.
2010;12(2):86–92. and nitric
303. DentonoxideKM,synthases.
Shweta Am J Physiol Regul
A, Finkelstein L, Integr
et al. Comp
EffectPhysiol. 2010;299(5):
of endothelin-1 on
333. Matavelli
278. SchulmanLC, IH, Zhou MS,
Kadowitz Raij L.LG,Interaction
PJ, Navar et al. Renalbetween
hemodynamicnitric and
oxide and
excre- R1263–R1268.
regional kidney blood ow and renal arteriole calibre in rabbits. Clin Exp Phar-
angioresponses
tory tensin IItoinintra-arterial
the endothelium:
infusion role
ofin atherosclerosis
peroxynitrite and hypertension.
in anesthetized rats. AmJ http://www.ncbi.nlm.nih.gov/pubmed/20357023
macol Physiol. 2004;31(8):494–501.
JHypertens Suppl.
Physiol Renal 2006;24(1):S45–S50.
Physiol. 2009;296(1):F170–F176. 344. Pollock
304. SchrijversJS, BF , RaschDM.
Pollock R, Tilton RG, et and
Endothelin al. High protein oxide
NOS1/nitric induced glomerular
signaling and
334. Wang
279. Toda N, D, Arakawa
ChabrashviliK. Salt inducedCS.
T, Wilcox hemodynamic regulation mediated
Enhanced contractility by
of renal af- hypertrophy
regulation is vascular
of sodium endothelial
homeostasis. Currgrowth actor dependent.
Opin Nephrol Kidney Int. 2002;
Hypertens. 2008;17(1):70–75.
nitric oxide.
ferent J Hypertens.
arterioles 2011;29(3):415–424.
from angiotensin-infused rabbits: roles of oxidative stress, 61(5):1600–1604.
305. Ahmeda AF, Johns EJ. The regulation of blood perfusion in the renal cortex
http://www.ncbi.nlm.nih.gov/pubmed/21150639
thromboxane prostanoid receptors, and endothelium. Circ Res. 2004;94(11): http://www.ncbi.nlm.nih.gov/pubmed/11967009
and medulla by reactive oxygen species and nitric oxide in the anaesthetised rat.
335. Kopkan L, Hess A, Huskova Z, et al. High salt intake enhances superoxide
1436–1442. 345. Physiol
Acta Wakef(Oxf).
eld AP , House JD, Ogborn MR, et al. A diet with 35% o energy rom
2012;204(3):443–450.
activity
280. in eNOS knockout
Brakemeier S, Eichlermice leading
I, Knorr A, ettoal.
theModulation
development of oCa2
salt -activated
sensitivity.KAm proteinRajapakse
306. leads to kidney damage AK,
NW, Sampson in emale
EppelSprague
GA, et al.Dawley rats. Br II
Angiotensin J Nutr.
and 2011;
nitric
J Physiol in
channel Renal Physiol.
renal artery2010;299(3):F656–F663.
endothelium in situ by nitric oxide and reactive oxygen 106(5):656–663.
oxide in neural control of intrarenal blood ow. Am J Physiol Regul Integr Comp
336. Chabrashvili
species. T, Kitiyakara C, Blau J, et al. E ects o ANG II type 1 and 2
Kidney Int. 2003;64(1):199–207. http://www.ncbi.nlm.nih.gov/pubmed/21535903
Physiol. 2005;289(3):R745–R754.
receptors
281. on oxidative
Fellner stress, renal
SK, Arendshorst NADPH oxidase,
WJ. Angiotensin and SOD
II, reactive expression.
oxygen species Am
andJ 346. Fuson
307. BenabeAL, JE, Komlosi
Wang S, P Wilcox
, UnlapJN, TM,et al. Modulation
et al. o ANG II receptor
Immunolocalization and
of a micro-
Physiolsignaling
Ca2 Regul Integr in Comp Physiol.
afferent 2003;285(1):R117–R124.
arterioles. Am J Physiol Renal Physiol. 2005;289(5): its mRNA
somal in normal rat
prostaglandin by low protein
E synthase in rabbit eeding. Am Am
kidney. J Physiol. 1993;265(5
J Physiol Pt 2):
Renal Physiol.
337. Dutta UK, Lane J, Roberts LJ, et al. Superoxide ormation and interaction
F1012–F1019. F660–F669.
2003;285(3):F558–F564.
with nitric
282. Hughes oxide
AK,modulate
Kohan DE. systemic
Mechanism arterial pressure and renal unction
of vasopressin-induced in salt
contraction of 347. Hollenberg
308. Brenner BM.NK. Retarding the progression
Direct renin inhibition oandrenal the disease.
kidney. Kidney
Nat RevInt. 2003;
Nephrol.
depleted
renal dogs. Exp
medullary Biol Medcells.
interstitial (Maywood).
Nephron2006;231(3):269–276.
Physiol. 2006;103(3):119–124. 64(1):370–378.
2010;6(1):49–55.
http://www.ncbi.nlm.nih.gov/pubmed/16514172
283. Kim J, Pannabecker TL. Two-compartment model of inner medullary vas- http://www.ncbi.nlm.nih.gov/pubmed/12787440
309. Gross V, Kurth TM, Skelton MM, et al. Effects of daily sodium intake
338. Frank
culature H, Gradual
supports J, Amann
modes Gassner U, et al. E ect o short
of vasopressin-regulated inner term high protein
medullary blood 348.ANG
and De Santo NG, Perna
II on cortical andA,medullary
Cirillo M.renal
Low protein
blood dietsow inare mainstayrats.
conscious or man
Am J
compared with normal
ow. Am J Physiol protein2010;299(1):F273–F279.
Renal Physiol. diets on renal hemodynamics and associated agement1998;274(5
Physiol. o chronic kidney disease. Front Biosci (Schol Ed). 2011;3:1432–1442.
Pt 2):R1317–R1323.
variables
284. in healthy
Cowley AW Jr,young
Mori T,men. Am J Clin
Mattson DL, Nutr.
et al. 2009;90(6):1509–1516.
Role of renal NO production in http://www.ncbi.nlm.nih.gov/pubmed/21622279
310. Rodríguez F, Llinás MT, González JD, et al. Renal changes induced by a
http://www.ncbi.nlm.nih.gov/pubmed/19812175
the regulation of medullary blood ow. Am J Physiol Regul Integr Comp Physiol. 349. Levey AS, Greene
cyclooxygenase-2 inhibitor T, during
Sarnak normal
MJ, et al. andE low
ect sodium
o dietary protein
intake. restriction
Hypertension.
339. Slomowitz LA, Gabbai FB, Khang SJ, et al. Protein intake regulates the
2003;284(6):R1355–R1369. on the progression o kidney disease: long term ollow up o the Modif cation o
2000;36(2):276–281.
vasodilatory
285. Majid DSA,unction
SaidoKE,theOmoro
kidneySA,andetNMDA receptor
al. Nitric oxideexpression.
dependencyAm ofJarterial
Physiol Diet inTojo
311. RenalA, Disease
Kimoto (MDRD)
M, Wilcox Study.
CS. Am RenalJ Kidney Dis. 2006;48(6):879–888.
expression of constitutive NOS and
Regul Integr Comp changes
pressure-induced Physiol. 2004;287(5):R1184–R1189.
in renal interstitial hydrostatic pressure in dogs. Circ http://www.ncbi.nlm.nih.gov/pubmed/17162142
DDAH: separate effects of salt intake and angiotensin. Kidney Int. 2000;58(5):
340.2001;88(3):347–351.
Res. Pecoits Filho R. Dietary protein intake and kidney disease in Western diet. 350. Mandayam S, Mitch WE. Dietary protein restriction benef ts patients with
2075–2083.
Contrib
286. Nephrol. 2007;155:102–112.
O’Connor PM, Cowley AW Jr. Vasopressin-induced nitric oxide produc- chronic
312. kidney disease.
Bergström G, Evans Nephrology (Carlton). 2006;11(1):53–57.
RG. Mechanisms underlying the antihypertensive
http://www.ncbi.nlm.nih.gov/pubmed/17369718
tion in rat inner medullary collecting duct is dependent on V2 receptor activa- 351. Phisitkul
functions of the S, Hacker
renal C, Simoni
medulla. J, et Scand.
Acta Physiol al. Dietary protein causes a decline
2004;181(4):475–486.
341.ofGonin
tion Jmaa D, Seniorpathway.
the phosphoinositide DF. TheAm hyperf ltration
J Physiol Renaltheory:
Physiol.progression
2007;293(2): o in theChen
313. glomerular
J, Zhaof M, ltration
He W,rate o Increased
et al. the remnant kidney
dietary NaClmediated
induces by metabolic
renal medul-
chronic renal ailure and the e ects o diet in dogs. J Am Vet Med Assoc. 1995;
F526–F532. acidosis
lary PGE2 andproduction
endothelinand receptors. Kidney
natriuresis viaInt.
the2008;73(2):192–199.
EP2 receptor. Am J Physiol Renal
207(11):1411–1415.
287. Bankir L. Antidiuretic action of vasopressin: quantitative aspects and in- 352. Green
Physiol. T, Rodriguez J, Navar LG. Augmented cyclooxygenase 2 e ects on
2008;295(3):F818–F825.
http://www.ncbi.nlm.nih.gov/pubmed/7493867
teraction between V1a and V2 receptor-mediated effects. Cardiovasc Res. 2001; renal Y
314. unction duringI,varying
ang T, Singh Pham states
H, et oal.angiotensin
RegulationII.ofAm J Physiol Renalexpres-
cyclooxygenase Physiol.
342. Brändle E, Sieberth HG, Hautmann RE. E ect o chronic dietary protein
51(3):372–390. 2010;299:F954–F962.
sion in the kidney by dietary salt intake. Am J Physiol. 1998;274(3 Pt 2):
intakeCorreia
288. on the AG,renalDenton
unction KM,in Evans
healthyRG. subjects.
Effects Eur J Clin Nutr.
of activation of 1996;50(11):
vasopressin- F481–F489.
734–740. on regional kidney blood ow and glomerular arteriole diameters.
V1-receptors 315. Ye W, Zhang H, Hillas E, et al. Expression and function of COX isoforms in
J Hypertens. 2001;19(3 Pt 2):649–657. renal medulla: evidence for regulation of salt sensitivity and blood pressure. Am
289. Matsuda H, Hayashi K, Arakawa K, et al. Distinct modulation of super - J Physiol Renal Physiol. 2006;290(2):F542–F549.
cial and juxtamedullary arterioles by prostaglandin in vivo. Hypertens Res. 2002; 316. Bech JN, Nielsen CB, Ivarsen P, et al. Dietary sodium affects systemic and
25(6):901–910. renal hemodynamic response to NO inhibition in healthy humans. Am J Phyisol.
290. Badzynska B, Sadowski J. Opposed effects of prostaglandin E2 on per- 1998;274(5 Pt 2):F914–F923.
fusion of rat renal cortex and medulla: interactions with the renin-angiotensin 317. Sanders PW. Vascular consequences of dietary salt intake. Am J Physiol Re-
system. Exp Physiol. 2008;93(12):1292–1302. nal Physiol. 2009;297(2):F237–F243.
291. Carey RM. Update on the role of the AT2 receptor. Curr Opin Nephrol 318. Zhao X, Pollock DM, Inscho EW, et al. Decreased renal cytochrome P450
Hypertens. 2005;14(1):67–71. 2C enzymes and impaired vasodilation are associated with angiotensin salt-
292. Zhang Z, Rhinehart K, Kwon W, et al. ANG II signaling in vasa recta peri- sensitive hypertension. Hypertension. 2003;41(3 Pt 2):709–714.
cytes by PKC and reactive oxygen species. Am J Physiol Heart Circ Physiol. 2004; 319. Fujihara CK, De Nucci G, Zatz R. Physiologic role of ETA receptors in the
287(2):H773–H781. regulation of renal hemodynamics in normal and salt-depleted rats. J Cardiovasc
293. Rajapakse NW, Mattson DL. Role of L-arginine in nitric oxide production Pharmacol. 1995;26 Suppl 3:S466–S469.
in health and hypertension. Clin Exp Pharmacol Physiol. 2009;36(3):249–255. 320. Vanni S, Polidori G, Cecioni I, et al. ET(B) receptor in renal medulla is en-
294. Mori T, O’Connor PM, Abe M, et al. Enhanced superoxide production in re- hanced by local sodium during low salt intake. Hypertension. 2002;40(2):179–185.
nal outer medulla of Dahl salt-sensitive rats reduces nitric oxide tubular-vascular 321. Fattal I, Abassi Z, Ovcharenko E, et al. Effect of dietary sodium intake on
cross-talk. Hypertension. 2007;49(6):1336–1341. the expression of endothelin-converting enzyme in the renal medulla. Nephron
295. Abe M, O’Connor P, Kaldunski M, et al. Effect of sodium delivery on super- Physiol. 2004;98(4):89–96.
oxide and nitric oxide in the medullary thick ascending limb. Am J Physiol Renal 322. Taylor TA, Gariepy CE, Pollock DM, et al. Gender differences in ET and
Physiol. 2006;291(2):F350–F357. NOS systems in ETB receptor-de cient rats: effect of a high salt diet. Hyperten-
296. Schnackenberg CG. Physiological and pathophysiological roles of oxygen sion. 2003;41(3 Pt 2):657–662.
radicals in the renal microvasculature. Am J Physiol Regul Integr Comp Physiol. 323. Lai EY, Onozato ML, Solis G, et al. Myogenic responses of mouse isolated
2002;282(2):R335–R342. perfused renal afferent arterioles: effects of salt intake and reduced renal mass.
297. Vera T, Stec DE. Moderate hyperbilirubinemia improves renal hemodynam- Hypertension. 2010;55(4):983–989.
ics in ANG II-dependent hypertension. Am J Physiol Regul Integr Comp Physiol. 324. Lau C, Sudbury I, Thomson M, et al. Salt-resistant blood pressure and salt-
2010;299(4):R1044–R1049. sensitive renal autoregulation in chronic streptozotocin diabetes. Am J Physiol
298. Oliver JJ, Eppel GA, Rajapakse NW, et al. Lipoxygenase and cyclo- Regul Integr Comp Physiol. 2009;296(6):R1761–R1770.
oxygenase products in the control of regional kidney blood ow in rabbits. Clin 325. Saeed A, Dibona GF, Marcussen N, et al. High NaCl intake impairs dy-
Exp Pharmacol Physiol. 2003;30(11):812–819. namic autoregulation of renal blood ow in angiotensin II-infused rats. Am J
299. Kuczeriszka M, Badzynska B, Kompanowska-Jezierska E. Cytochrome Physiol Regul Integr Comp Physiol. 2010;299(5):R1142–R1149.
P-450 monooxygenases in control of renal haemodynamics and arterial pressure 326. Hoagland KM, Flasch AK, Roman RJ. Inhibitors of 20-HETE formation
in anaesthetized rats. J Physiol Pharmacol. 2006;57 Suppl 11:179–185. promote salt-sensitive hypertension in rats. Hypertension. 2003;42(4):669–673.
300. Badzynska B, Sadowski J. Differential action of bradykinin on intrarenal 327. Dobrowolski L, Kompanowska-Jezierska E, Walkowska A, et al. Sodium
regional perfusion in the rat: waning effect in the cortex and major impact in the intake determines the role of adenosine A2 receptors in control of renal medul-
medulla. J Physiol. 2009;587(Pt 15):3943–3953. lary perfusion in the rat. Nephrol Dial Transplant. 2007;22(10):2805–2809.
301. Cheng MK, Doumad AB, Jiang H, et al. Epoxyeicosatrienoic acids mediate 328. Walkowska A, Dobrowolski L, Kompanowska-Jezierska E, et al. Role of
adenosine-induced vasodilation in rat preglomerular microvessels (PGMV) via NO and COX pathways in mediation of adenosine A1 receptor-induced renal
A2A receptors. Br J Pharmacol. 2004;141(3):441–448. vasoconstriction. Exp Biol Med (Maywood). 2007;232(5):690–694.
C H A P T E R

4 Regulation of Water Balance:


Urine Concentration and Dilution
Yumi Noda • Sei Sakaki

W
ater is by far the largest component of the body depending on the requirements for water balance. When the
and accounts for approximately 50% to 65% kidney’s capacity to conserve water is maximized to the limit
of body weight. Maintenance of body uid due to dehydration, a sensation of thirst is activated, causing
homeostasis, including uid volume and solute concentra- oral water intake to be increased.
tion, is essential for cell function and whole-organism sur- Because the solute concentrations in water in the body
vival. The osmolality of body uid, a concentration of all must be kept nearly constant, water loss must be regulated
of the solute in water, is kept within a remarkably narrow by a mechanism that decouples water and the solutes. The
range (280 to 295 mOsm per kilogram of water), in spite kidney can excrete the appropriate amount of water with-
of large uctuations of solute and water intakes and losses. out marked perturbations in solute excretion. When water
Although this constancy is made by a variety of regulatory intake is too large and dilutes blood plasma, urine diluted
mechanisms in the body, the most critical regulatory ca- more than plasma is excreted to concentrate the plasma.
pacities are provided by the kidney’s urine concentration When water intake is too small to concentrate plasma,
and dilution mechanisms. urine concentrated more than plasma is excreted to dilute
Body water homeostasis is maintained by the balance the plasma. In both cases, the solute excretion varies little.
between the input and output of water. Each side has regu- Renal solute-free water excretion is mainly regulated by the
lated and unregulated components. The regulated compo- antidiuretic hormone vasopressin. Vasopressin is secreted
nent of water input is oral intake of uids in response to from the posterior pituitary gland into systemic circulation
a perceived sensation of thirst. The unregulated compo- in response to increases in the tonicity, which is an effective
nents of water input are oral intake of liquids and water osmotic pressure in the plasma, to decreases in the effective
in foods, and metabolic water of oxidation. Oral intake of circulating volume or pressure, or to several other stimuli.
water usually varies enormously in excess of homeostatic In response to changing levels of vasopressin in the plasma,
need because of social, cultural, or psychological in uences. the kidney is capable of wide variations in free water excre-
Solute-free water excretion by the kidney is the only route tion. The molecular entity of a major effector of vasopressin
of regulated water output. The unregulated component of in the kidney is aquaporin-2 (AQP2), which is a member
water excretion occurs via various kinds of insensible water of the aquaporin (AQP) water channel family. Molecular
losses including sweat, evaporative loss, gastrointestinal loss, identi cation of the AQP family has revolutionized the un-
and the obligate amount of water that is required to excrete derstanding of water transport in the body, including urine
the solutes in the urine. Sweat volume is determined by the concentration and dilution.1,2 AQP2 is abundant in the col-
requirements of temperature regulation. Evaporative loss is lecting duct, which is the chief site of regulation of water
determined by body temperature and surface area, ventila- reabsorption.3 Acute stimulation of vasopressin promotes
tion, and environmental temperature and humidity. Gastro- AQP2 translocation from an intracellular reservoir to the lu-
intestinal water loss is affected by disturbance of its function. minal cell surface, and its chronic stimulation increases the
Both the input and output of water have very substantial cellular abundance of AQP2, both of which elevate the wa-
unregulated components, and these can vary tremendously ter permeability of the collecting duct cells, resulting in the
as a result of factors that are unrelated to the maintenance promotion of water reabsorption from the urinary tubule.
of body water homeostasis. Therefore, the regulated com- Its impairments result in various water balance disorders
ponents, which are urine excretion and water intake caused including nephrogenic diabetes insipidus (NDI). In addi-
by thirst, must compensate for whatever perturbations re- tion to AQP2, six other AQP isoforms are expressed in the
sult from the unregulated water gains and losses. The daily kidney and play key roles in water transport activities spe-
urine excretion range is as low as 0.5 L to as high as 25 L ci c for their localization in the nephron segments without

13 2
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1133
33

affecting solute transport. In addition to AQPs, the localiza- are de ned by the length of their Henle loop. Short-looped
tions of urea transporters and ion transporters are highly nephrons originate from super cial glomeruli and have a
speci c through the renal tubule segments. Together with Henle loop, which turns near the inner-outer medullary bor-
a three-dimensional con guration of the nephron, the spe- der and consists of a proximal straight tubule, a thin descend-
ci c transport activities of water, urea, and ion all through ing limb, and a thick ascending limb. Long-looped nephrons
the renal tubule segments enable the mechanism of urine originate from the juxtamedullary glomeruli, extend into the
concentration and dilution. inner medulla, bend at various levels of the inner medulla, and
contain a thin ascending limb in addition to segments found
in short-looped nephrons. Thin ascending limbs are found
ANATOMIC CONSIDERATIONS only in the inner medulla and their transition to thick ascend-
FOR URINE CONCENTRATION AND ing limbs de nes the inner-outer medullary border. Thus, only
DILUTION MECHANISMS thick ascending limbs are found in the outer medulla.
General Features of the Nephron Structure
The kidney consists of two populations of nephrons: short- The Descending Part of the Henle Loop
looped nephrons and long-looped nephrons (Fig. 4.1). Both The descending part of the Henle loop consists of the S2
types of loops have a hairpin or a U-shape con guration, and proximal straight tubule in the medullary ray, the S3 proximal

FIGURE 4.1 The kidney structure. The con gurations of both a long-looped and a short-looped nephron are shown. The vasa recta is
shown in the left. The major portions of the nephron are glomeruli (circles), proximal tubules (hatched), the thin limbs of the Henle loop
(single lines), thick ascending limbs of the Henle loop (solid), distal convoluted tubules (stippled), and the collecting duct system (open).
(Modi ed from Knepper MA, Stephenson JL. Urinary concentrating and diluting processes. In: Andreoli TE, Fanestil DD, Hoffman JF,
Schultz SG, eds. Physiology of Membrane Disorders, 2nd ed. New York: Plenum, 1986:713, with permission.)
134
13 4 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

straight tubule in the outer stripe of the outer medulla, and and computer-assisted reconstruction in rats’ inner medullas
the thin descending limbs in the inner stripe of the outer and show that the thin ascending limb marked by ClC-K1 ex-
inner medulla. AQP1 is abundant in both the apical mem- pression occurs about 160 m before the loop bend.9
brane and the basolateral plasma membrane in the S2 and The thick ascending limb has essentially no water per-
S3 proximal tubules and the descending thin limbs.4–6 AQP1 meability and does not express AQPs. On the other hand,
is constitutively active and is not regulated by vasopressin. Na-K-2Cl cotransporter (NKCC2) is present in the apical
AQP1 is present in suf cient abundance to account for the membrane,17 and Na-K-ATPase, a Na pump, is present in the
observed water permeability of these tubule segments.7 The basolateral membrane in the thick ascending limb. NaCl is
osmotic water permeability of the thin descending limb is ex- actively absorbed by these transporters, resulting in the dilu-
tremely high due to the presence of the AQP1 water channel.6 tion of the luminal uid in this segment, which works as the
However, there is an axial heterogeneity in the expression single effect in the countercurrent multiplication (described
levels of AQP1 in the thin descending limb; AQP1 is present in the following sections). Urea permeability in the cortical
in entirety through the thin descending limb of the short- thick ascending limb is higher than in the medullary thick
looped nephron, whereas it is located in the upper half of the ascending limb.18 This may also contribute to the dilution of
thin descending limb of long-looped nephron.8–10 A com- the luminal uid as it ows up to the cortex by passive urea
parison of water permeability and the abundance of AQP1 absorption.
in these tubule segments indicates that AQP1 is the main
route for water movement across the tubules.8 The high wa- The Distal Convoluted Tubule and
ter permeability of these segments is critical for the counter- Connecting Tubule
current multiplication system that is described in the section
After exiting the Henle loop, the luminal uid enters the
Countercurrent Multiplication in the Outer Medulla and
distal convoluted tubule that expresses the thiazide-sensitive
Other Mechanisms in the Inner Medulla, which follows. As
Na-Cl cotransporter NCC.17,19 NCC is important for NaCl
expected, a severely impaired urine concentrating ability is
reabsorption that reduces uid delivery to the collecting
observed in AQP1 knockout mice.11
duct, leading to increases in urinary concentrating ability.
In addition to AQP1, AQP7 is expressed in the api-
Water permeability in the distal convoluted tubule is low
cal membrane of the S3 proximal tubule. AQP7 transports
and the expression of AQP2 or vasopressin V2 receptor is
glycerol as well as water, thus AQP7 is an aquaglyceropo-
not observed.
rin.12 Studies in AQP7 knockout mice suggest that water
Several distal tubules merge to form a connecting tubule
absorption mediated by AQP7 is small compared to that of
arcade. The connecting tubule cells express both AQP2 and
by AQP1 and minimally contributes to urine concentration;
vasopressin V2 receptor, suggesting that, like the collecting
whereas glycerol absorption by AQP7 may be important in
ducts, the connecting tubules are sites of regulated water
glycerol metabolism in the body.13
absorption. Moreover, vasopressin-regulated water absorp-
A urea transporter UT-A2 is present in the thin limb of
tion in the connecting tubule can result in a considerable
the long-looped nephron in the inner medulla and the thin
amount of free-water absorption during antidiuresis because
descending limb of the short-looped nephron in the outer
the large aggregate epithelial surface area of the connecting
medulla. Thus, these segments have a high urea permeabi-
arcades is estimated to be roughly equivalent to that of the
lity14 and play an important role for urea-recycling pathways
cortical collecting ducts.20
that serve to maintain high urea concentrations within the
An AQP2 knockin mice model of nephrogenic diabe-
inner medullary interstitium, which is critical for the urinary
tes insipidus, a rare disease manifested by an inability to re-
concentration that is described in the section Urea Accumu-
spond to vasopressin, shows a severe urinary concentrating
lation in the Inner Medulla, which follows.
defect that results in neonatal death.21 Meanwhile, AQP2
conditional selective knockout in the collecting duct but
The Ascending Part of the Henle Loop not in the connecting tubule rescues mice from the lethal
phenotype that is observed in mice lacking AQP2 globally.22
The ascending part of the Henle loop consists of the thin
Although all types of mice show a severe urinary concentrat-
ascending limb in the inner medulla, the medullary thick
ing defect, these ndings con rm an important role of the
ascending limb in the inner stripe of the outer medulla, and
connecting duct for urinary concentration.
the cortical thick ascending limb in the medullary rays. The
thin ascending limb is located exclusively in the inner me-
dulla, is present only in the long-looped nephron, and be- The Collecting Duct
comes a thick ascending limb at the inner-outer medullary The collecting duct is the nal structure in the nephron. The
border. The thin ascending limb has extremely low water luminal uid from the connecting tubules then enters the col-
permeability and AQPs are not detected. By contrast, the lecting tubules in the super cial cortex. The collecting duct
ClC-K1 chloride channel is expressed in the apical and ba- spans all the regions of the kidney. From the super cial cor-
solateral membrane of the entire length of the thin ascend- tex, the collecting duct descends through the cortex and the
ing limb.9,15,16 Studies with immunohistochemical labeling outer medulla. In the inner medulla, these collecting ducts
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1135
35

continually merge, nally forming the ducts of Bellini that The urea permeability is extremely high only in the
open into the renal pelvis at the papillary tip. The renal pelvis terminal IMCD and low in other portions of the collecting
is continuous with the ureter. The collecting ducts are arrayed duct.31 UT-A1 and UT-A3 urea transporters are present in
parallel to the Henle loop. The collecting ducts have several the terminal IMCD cells.32 Urea reabsorption in this segment
morphologically discrete tubule segments: the cortical col- is important for the preservation of high urea concentrations
lecting duct, the outer stripe portion of the outer medullary in the inner medullary interstitium, which are essential for
collecting duct, the inner stripe portion of the outer medul- its urine concentrating ability and are also important for
lary collecting duct, the initial part of the inner medullary minimizing the urinary loss of urea that is a major part of
collecting duct (IMCD), and the terminal part of the IMCD. the urea recycling pathway. This process is described in de-
The collecting duct, under the control of vasopressin tail in the section Urea Accumulation in the Inner Medulla,
and other factors, is the nephron segment responsible for which follows.
the nal control of water excretion. The collecting duct ex-
presses the vasopressin V2 receptor and AQP2, which are The Vasculature
the primary targets for vasopressin regulation. In the absence The blood vessels named the vasa recta carry blood into
of vasopressin, AQP2 resides in the vesicles below the api- and out of the renal medulla. Like the Henle loop, there are
cal cell surface and the entire collecting duct is very water the descending and ascending parts in the vasa recta, both
impermeable. When the body is dehydrated, plasma os- of which are arranged in parallel and in mutual proximity.
molality is increased, which is sensed by the hypothalamic Blood from efferent arterioles of juxtamedullary nephrons
osmoreceptors and results in vasopressin secretion by the enters into the descending vasa recta in the medulla, passes
posterior pituitary. Vasopressin binds to V2 receptors in the through the capillary plexus located at various depths within
basolateral membrane of the principal cells of the collect- the medulla, then merges to form the ascending vasa recta.
ing duct, stimulates adenylate cyclase to produce cAMP, ac- Different from the Henle loop, the descending and ascending
tivates protein kinase A, phosphorylates AQP2, and inserts vasa recta are separated by the capillary plexus (see Fig. 4.1).
this water channel into the apical cell surface, which results The AQP1 water channel and the UT-B urea transporter
in a signi cant increase in the collecting duct water perme- are present in the descending vasa recta and enhance the
ability and water reabsorption.23–25 This process is described countercurrent exchange of water and urea between the de-
in detail in the section Aquaporin-2, which follows. Not scending and ascending vasa recta in the medulla.6,33 This
only are AQP2 water channels present, but also are AQP3 countercurrent exchange reduces the effective blood ow,
and AQP4 water channels in the collecting duct principal contributing to the conservation of osmolality gradients in
cells. There is axial heterogeneity in the abundance of AQP3 the medullary interstitium.
and AQP4; AQP3 is abundant in the connecting tubule, the
cortical collecting duct, and the outer medullary collecting The Medullary Interstitium
duct; whereas AQP4 is most abundant in the inner medul-
lary collecting duct.26 AQP3 and AQP4 are located in the The interstitium of the outer medulla and the outer portion
basolateral membrane of these cells and represent a potential of the inner medulla is a narrow space that is important for
exit pathway from the cell to the interstitium for water enter- limiting solute diffusion upward along the medullary axis.34
ing via AQP2. To the contrary, the interstitium of the inner half of the inner
Water reabsorption mainly occurs in the cortex and the medulla is much larger. In this region, the interstitial cells
outer medulla. The inner medulla has the highest osmola- are interspersed in a gelatinous matrix of highly polyme-
lity and is important for the reabsorption of the remaining rized hyaluronic acid,35 which is largely devoid of capillary
water when maximal water reabsorption is required. Rather, plexuses or lymphatics. This structure slows the diffusion of
water reabsorption by the terminal part of IMCD is greater solute and water in the inner medulla.
during diuresis than during antidiuresis27 because of its large
transepithelial osmolality difference and relatively high basal The Pelvis
water permeability compared to other portions of the col- The collecting duct nally opens at the papillary surface and
lecting duct. urine enters the pelvic space. In the kidney with only one
The connecting tubule and the cortical collecting duct papilla, such as in hamsters and rats, the renal pelvis is an
express the amiloride-sensitive sodium channel ENaC.28,29 intrarenal urinary space surrounding the papilla. In the kid-
Sodium is actively absorbed via ENaC in the apical membrane ney with many papillae, such as in humans, each papilla is
of the principal cells and exits the cells via Na-K-ATPase in surrounded by a funnel-shaped calyx. In the human kidney,
the basolateral membrane. Sodium reabsorption via ENaC is it is the compartment between the calyces and the ureter
increased by vasopressin or aldosterone and plays an impor- that is called the pelvis.36 This compartment is not present in
tant role for reducing uid delivery to the medullary collect- kidneys with one papilla, where the pelvis is a direct exten-
ing duct, thus leading to increases in urinary concentrating sion of the ureter. The renal pelvic (calyceal) wall contains
ability.30 On the other hand, the sodium permeability is low smooth muscle layers. Contractions of these muscles in-
in the medullary collecting duct. duce regular peristaltic contractions of the wall that strongly
136
13 6 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

compress the medullary tissue, resulting in intermittent ow uid then enters the thin descending limb of Henle and is
in the collecting ducts and the Henle loop.37 concentrated as it ows down to the bend of the Henle loop
because of water reabsorption. As the uid ows up the as-
cending limb of the Henle loop, the luminal uid becomes
URINE CONCENTRATION diluted because this segment is impermeable to water, and
MECHANISMS the thick ascending limb actively absorbs NaCl. This diluted
General Features of Urine Concentration uid nally enters the collecting duct system. In diuresis
(low AVP), the uid remains hypotonic. On the other hand,
In an adult human, glomerular ltrate is about 180 L per
in antidiuresis (high AVP), the uid is concentrated to a
day, most of which is reabsorbed by the high water perme-
level far greater than plasma by water reabsorption as it
able proximal tubule and descending limb of the Henle
ows down the collecting duct. This is due to a vasopressin-
loop. However, regulation of water excretion mainly occurs
induced signi cant increase in water permeability of this
after the luminal uid reaches the distal tubule. The osmo-
segment and the existence of an axial osmolality gradient
lalities of luminal uid along the rat nephron are shown in
in the medulla. This osmolality gradient with the highest
Figure 4.2. The luminal uid in the proximal tubule is isos-
degree of hyperosmolality at the papillary tip is maintained
motic to plasma, regardless of antidiuresis or diuresis. This
by several mechanisms, including countercurrent multipli-
cation, countercurrent exchange, and urea accumulation in
the inner medulla, as described subsequently.

Countercurrent Multiplication in the


Outer Medulla and Other Mechanisms in
the Inner Medulla
Countercurrent multiplication is an essential process for
generating a medullary osmotic gradient along the cortico-
medullary axis and occurs in the Henle loop.38 The coun-
ter ow arrangement of this loop makes the osmolality
difference between the ascending and descending limbs to
be multiplied, resulting in an enormous increase in osmola-
lity toward the bend of this loop. Figure 4.3 illustrates the
basic components of this mechanism in the short Henle
loop. In the thick ascending limb of Henle, which cor-
responds to the right limb in Figure 4.3, NaCl is actively
reabsorbed at any level of this ascending limb. Because this
segment is impermeable to water, the luminal uid is diluted
and NaCl concentrations of the surrounding interstitium be-
come higher than the luminal uid. On the other hand, the
descending limb of the Henle loop, which corresponds to
the left channel in Figure 4.3, is highly permeable to water
FIGURE 4.2 The typical tubular uid osmolalities (in mil- due to the presence of an AQP1 water channel, but is im-
liosmoles per kilogram of water) found along the nephron permeable to NaCl. Water is passively reabsorbed from the
segments of the rat kidney. Fluid osmolality in the proximal descending limb lumen to the interstitium, which has a high
tubule is isotonic, and it increases to 600 to about 1,200 mOsm osmolality due to NaCl accumulation. The luminal uid in
per kilogram of water at the bend of the loop. Fluid emerg- the descending limb is concentrated by water reabsorption
ing from the thick ascending limb is hypotonic, and nal urine and is continually concentrated downward to the bend of
osmolalities are determined by urine concentration in the col- the loop, and then enters to the ascending limb. High NaCl
lecting duct depending on circulating vasopressin levels (65 to concentrations of this uid further promote NaCl reabsorp-
about 1,150 mOsm per kilogram of water). AVP, arginine vaso- tion in the ascending limb. This uid is then diluted by this
pressin. (Based on micropuncture studies by Wirz H. Der osmo- NaCl absorption while owing toward the top of the loop.
tische Druck in den corticalin Tubuli der Ratten niere. Helv Physiol Because the transverse concentration difference between the
Pharmacol Acta. 1956;14:353; Gottschalk CW, Mylle M. Micro- two limbs is always maintained by active NaCl absorption
puncture study of the mammalian urinary concentrating mecha- by the ascending limb (this is called the single effect) and
nism: evidence for the countercurrent hypothesis. Am J Physiol. water permeability of the descending limb at each level of
1959;196:927; Jamison RL, Buerkert J, Lacy F. A micropuncture the longitudinal axis is high, the bend of the loop achieves a
study of collecting tubule function in rats with hereditary diabe- progressively higher osmolality. This results in an enormous
tes insipidus. J Clin Invest. 1971;50:2444, with permission.) increase in osmolality toward the bend of the Henle loop.
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1137
37

is mentioned previously, results in high urea concentrations


in the inner medullary interstitium. This causes osmotic
withdrawal of water from the thin descending limb and
increases NaCl concentrations of the luminal uid. This
highly concentrated NaCl then exits passively from the thin
ascending limb (works as single effect), and the luminal uid
is progressively diluted as it ows up. This passive transport
process may produce an axial NaCl concentration gradient
in the inner medulla. This model requires that the thin de-
scending limb is highly permeable to water and but not to
NaCl or urea, whereas the thin ascending limb is permeable
to NaCl but not to water or urea. A microperfusion study
of the thin ascending limb shows that the permeability of
chloride and sodium are higher than urea, and that lumi-
nal dilution takes place when tubule segments are perfused
in a condition simulating an in vivo situation.41 Consistent
with these physiologic studies, the ClC-K1 chloride channel
is exclusively expressed in the thin ascending limb. ClC-K1
knockout mice show a large urinary concentrating defect
with impaired urea as well as NaCl accumulation in the in-
ner medulla.42,43 This nding con rms the importance of a
rapid chloride exit in the thin ascending limb and supports
the passive mechanism.
Simulations based on many mathematical models in-
corporating physiologic parameters have been examined to
show the validity of the passive model, but they usually failed
to show satisfactory results. However, this does not neces-
sary neglect the passive model because solute permeability
and water permeability are sometimes different in species, in
nephrons (long looped versus short looped), and within the
segment (axial heterogeneity). Moreover, based on extensive
FIGURE 4.3 A countercurrent multiplication in the short Henle
studies of the rat inner medulla by immunohistochemical
loop. In the thick ascending Henle limb, NaCl is actively reabsorbed.
labeling and computer-assisted reconstruction, Pannabecker
Because this segment is impermeable to water (indicated by bold
et al.10 show that three-dimensional tubular and vascular
line), the luminal uid is diluted and the interstitium around this
relationships are more complex than thought before and a
segment has high NaCl concentrations. On the other hand, the de-
more comprehensive understanding of three-dimensional
scending limb of the Henle loop is highly permeable to water, and
functional architecture is critically important for modeling
water reabsorption occurs from the descending limb lumen to the
urine concentration mechanisms of the inner medulla.
interstitium with high NaCl concentrations.This results in an enor-
mous increase in osmolality toward the bend of the Henle loop.
Countercurrent Exchange
Although the hypertonic medulla is essential for the ability to
A countercurrent mechanism is accepted for generat- concentrate urine, blood ow may decrease the high solute
ing the axial osmolality gradient in the outer medulla, where concentrations in the medulla. The osmolality of blood en-
active NaCl reabsorption occurs in the thick ascending tering to the medulla from the general circulation is far
limb. However, in the inner medulla, there is little activity lower than the medullary interstitium and may decrease the
of NaCl transport despite the fact that osmolality continues osmolality of the interstitium as they come to equilibrium.
to increase toward the tip of the papilla. The thin ascend- Meanwhile, blood going out the medulla to the general cir-
ing limb of the Henle loop in the inner medulla cannot ac- culation can carry out the solutes from the interstitium. To
tively reabsorb NaCl. The high inner medullary interstitial minimize this dissipation of the high solute concentrations in
osmolality provides a critical driving force for osmotic water the medulla, there is a process called countercurrent exchange.
ow across the collecting ducts, where the water permeabil- Blood supply to the medulla is done by the vasa recta with
ity is regulated by vasopressin and AQP2 water channel, as the descending and ascending limbs arranged in a counter-
described later. For explaining this concentrating effect in the ow con guration. The vasa recta are permeable to water,
inner medulla, a passive mechanism is widely accepted.39,40 sodium, and urea. Therefore, in the descending vasa recta,
In this model, urea ef ux from the terminal IMCD, which blood loses water and gains solutes as it ows down because
138
13 8 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

of the increasing osmolality in the medullary interstitium. Af- limbs is then carried upward to the cortex through the thick
ter entering the ascending vasa recta, blood gains water and ascending limb, the distal convoluted tubule, the connecting
loses solutes as it ows up because of the decreasing osmo- tubule, and the cortical collecting duct, and again enters into
lality of the surrounding interstitium. This exchange of wa- the medulla through the outer and inner medullary collect-
ter and solute between the descending and ascending vasa ing ducts. Because these segments before the terminal IMCD
recta is called countercurrent exchange. This mechanism have low urea permeability, urea can be returned to this re-
minimizes the solute washout from the inner medulla to the gion with little loss. Urea is again reabsorbed by the termi-
systemic circulation. Thus, this exchange at each level in the nal IMCD and recycled to the inner medullary interstitium
medulla preserves the axial solute concentration gradients in (Fig. 4.4).
the medullary interstitium. If the blood ow is decreased, for
instance in the conditions of volume depletion in the body,
the ef ciency of countercurrent exchange is further increased
by getting enough time for achieving osmotic equilibration,
leading to an increase in urinary concentrating ability.
The AQP1 water channel and the UT-B urea transporter
are located in the descending vasa recta, which helps the
rapid countercurrent exchange of water and urea.6,33 Indeed,
an impaired urine concentration ability is observed in AQP1
and UT-B knockout mice.11,44

Urea Accumulation in the Inner Medulla


The major solute responsible for the inner medullary osmo-
lality gradient is urea and NaCl; although, in the outer me-
dulla, it is NaCl.45 High osmolality in the inner medulla is
maintained by urea accumulation in this region in addition
to NaCl. There are several mechanisms to maintain the high
urea accumulation in the medulla that are called urea recy-
cling (Fig. 4.4).46,47
Among the collecting duct, only the terminal IMCD has
a high urea permeability due to the presence of UT-A1 and
UT-A3 urea transporters in this segment.31,32 During anti-
diuresis, water is absorbed in the collecting duct segments in
the cortex and the outer medulla, which are impermeable to
urea. Thus, the urea concentrations of the luminal uid are
progressively increased as it ows down through the con-
necting tubule, the cortical collecting duct, and the outer
medullary collecting duct. When the luminal uid reaches
the terminal IMCD, which is highly permeable to urea, urea
is rapidly absorbed from the lumen to the surrounding in-
terstitium. During antidiuresis, the urea permeability of the
inner collecting duct is increased by vasopressin, and this
accounts for further rapid reabsorption of urea.48,49 FIGURE 4.4 The urea recycling pathways within the kidney.
Once reabsorbed, urea is not rapidly washed out to the The terminal portion of the inner medullary collecting duct
general circulation because of countercurrent exchange and (IMCD) ef ciently reabsorbs urea through urea transporters
the low effective blood ow of the vasa recta, resulting in UT-A1 and UT-A3, and accumulates urea in the inner medullary
the high urea accumulation in the inner medullary inter- interstitium. Some of the urea is recycled to the IMCD by be-
stitium. In addition, during antidiuresis, urea reabsorption ing reintroduced to the thin descending limb where UT-A2 is
by the terminal IMCD has another advantage for decreasing located. A high urea concentration in the medulla is maintained
the luminal osmolality and preventing the osmotic diuresis by countercurrent exchange (recycling) of urea between the
when luminal uid is very concentrated by enhanced water descending vasa recta (DVR) and the ascending vasa recta (AVR),
reabsorption in the upper portions of the collecting duct. which is helped by UT-B in DVR. Some of the urea in the AVR is
After being reabsorbed by the terminal IMCD, some reintroduced to the thin descending limb and fed again to the
urea in the inner medullary interstitium enters into the thin IMCD. (Redrawn from Yang B and Bankir L. Urea and concentrat-
limbs of the long Henle loop where urea permeability is high ing ability: new insights from studies in mice. Am J Physiol Renal
due to the presence of UT-A2.50 Urea entered in the thin Physiol. 2005;288:F881, with permission.)
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1139
39

In addition to the recycling pathway via the long loop in signi cant increase in the collecting duct water permeability
the inner medulla, there is another recycling pathway using and water reabsorption. In addition, vasopressin stimula-
the short loop in the outer medulla. Some parts of the urea tion increases AQP2 protein abundance in the cell (Fig. 4.5).
reabsorbed by the terminal IMCD exit the inner medulla via These processes are described in detail in the section Aqua-
the vasa recta. The vasa recta and the short Henle loop are porin-2, which follows. On the basolateral cell surface, AQP3
arranged in parallel and in mutual proximity, and the de- and AQP4 are located and represent a potential exit pathway
scending limb of the short loop expresses the UT-A2 urea from the cell to the interstitium for water entering via AQP2.
transporter and has a high urea permeability. Therefore, urea Water reabsorption mainly occurs in the connecting tu-
carried by the vasa recta is able to enter the short loop, then is bule, the cortical collecting duct, and the outer medullary
convected through the distal convoluted tubule, the connect- collecting duct. In the cortex and the outer medulla, blood
ing tubule, and the collecting duct. When the luminal uid ow is suf ciently high so that absorbed water can be carried
nally reaches the terminal IMCD, urea is again reabsorbed out to the general circulation without diluting the intersti-
to the surrounding inner medullary interstitium (Fig. 4.4). tium. On the other hand, the inner medulla has the highest
The reabsorption sites among the collecting duct system osmolality and is important for reabsorption of the remain-
are different between water and urea. Water is reabsorbed ing water when maximal water reabsorption is required.
mainly in the cortex and the outer medulla, where blood
ow is so high that water can be rapidly supplied to the sys-
temic circulation. Furthermore, this reabsorption does not
URINE DILUTION MECHANISMS
dilute the inner medulla. On the other hand, urea is reab- Approximately 50 mOsm per kilogram of water is the limit
sorbed in the inner medulla. Owing to low blood ow in this of the urinary diluting ability of humans. The mechanisms
region and urea recycling pathways, urea can be trapped in responsible for urinary dilution nearly overlap with that for
the inner medulla, which is required for the passive mecha- urinary concentration. These two conditions between diure-
nism, as described previously. sis and antidiuresis are switched mainly by the collecting
duct water permeability, which is regulated by vasopressin
stimulation and the AQP2 water channel.
Regulated Water Reabsorption in the
In the thick ascending limb, the luminal uid is diluted
Collecting Duct by active NaCl absorption through NKCC256 and Na-H ex-
A countercurrent multiplication in the Henle loop generates changer NHE3,57 regardless of antidiuresis or diuresis. In
a hypertonic medulla. A countercurrent exchange in the vasa addition, water impermeability of this segment preserves
recta minimizes the dissipation of this osmotic gradient. How- the luminal low osmolality by preventing water uxes. The
ever, either of these processes has the ability to regulate wa- distal convoluted tubule also has the ability to actively ab-
ter reabsorption in response to the body water balance. This sorb NaCl due to the presence of Na-Cl cotransporter NCC19
function is performed by the collecting duct. The collecting and is impermeable to water, resulting in the dilution of the
duct is responsible for the nal control of urine concentra- luminal uid to an osmolality of about 100 mOsm per kilo-
tion, and its water permeability is regulated by vasopressin gram of water. In diuresis, this diluted uid passes through
and other factors. In the absence of vasopressin, the collect- the collecting duct because its water permeability is very low.
ing duct has an extremely low water permeability. Because Active Na reabsorption through Na channel ENaC in the col-
the luminal uid exiting the Henle loop is diluted, the uid lecting duct29 can further dilute the luminal uid. 58
remains diluted after passing through the collecting duct, In addition to diuresis, there is another mechanism that
yielding a large volume of hypotonic urine. In the presence further promotes urinary dilution. The terminal IMCD has
of vasopressin, the water permeability of the collecting duct a higher basal water permeability than other portions of the
is dramatically increased. Based on the hypertonic medullary collecting duct and is surrounded by a high inner medul-
interstitium generated by countercurrent multiplication and lary interstitium. Because the osmolality of the luminal
other processes, as described previously, an increase in water uid reaching the terminal IMCD in diuresis is lower than
permeability of the collecting duct results in signi cant water that in antidiuresis, the transepithelial osmolality gradient
reabsorption by the osmotic gradient between the lumen and in diuresis is larger than that in antidiuresis, resulting in a
the surrounding interstitium (Fig. 4.2). The molecular entity higher water reabsorption in this segment.27 This reduces
of this regulation of water permeability of the collecting duct the inner medullary interstitial osmolality, leading to a fur-
is vasopressin V2 receptors and the AQP2 water channel ex- ther decrease in urinary concentrating ability.
pressed in the principal cells of the collecting duct.26,51–55 V2
receptors and AQP2 are present in the connecting tubule and
all segments of the collecting duct. When vasopressin binds VASOPRESSIN
to V2 receptors in the basolateral membrane of these cells, The primary determinant of solute-free water excretion is
it stimulates adenylate cyclase to produce cAMP, activates the regulation of urinary water excretion by circulating levels
protein kinase A, phosphorylates AQP2, and inserts this wa- of vasopressin in plasma. This section describes the regula-
ter channel into the apical cell surface, which results in a tion of vasopressin secretion from the neurohypophysis.
140
14 0 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Structure and Synthesis withdrawal and shrinkage of the osmoreceptor neurons.


Arginine vasopressin is the antidiuretic hormone of most Owing to the presence of mechanosensitive cation channels
mammals, although members of the pig family have lysine in these cells, cell shrinkage induces a positive charge in ux
vasopressin, in which a lysine replaces the arginine in po- and depolarizes the cell membrane, resulting in the trigger-
sition 8 of arginine vasopressin. Vasopressin is produced ing of neuronal action potentials. Such neurons send axonal
by the hypothalamic neurohypophyseal tract. This tract is projections to the SON, where they release the excitatory
composed of magnocellular neurons that arise bilaterally in transmitter glutamate to synaptically excite the magnocellu-
the supraoptic (SON) and paraventricular (PVN) nuclei of lar neurons that release vasopressin in their distal axons.60,61
the hypothalamus and project medially to merge in the pi- Recent studies show that the transient receptor potential
tuitary stalk and form the posterior pituitary gland in the vanilloid (TRPV) channels are likely to be the major compo-
sella tunica. Vasopressin is synthesized as part of a protein nent of the osmoreceptor. The N-terminal truncated TRPV1
precursor of approximately 21,000 Da of molecular mass channel is selectively expressed in osmosensory neurons in
that incorporates a signal peptide at its amino terminus and OVLT. The responses of these neurons to hypertonic stimuli,
vasopressin, neurophysin, and copeptin at its carboxyl ter- including reactive increases in cation channel conductance,
minus. In the endoplasmic reticulum, the signal peptide is membrane depolarization, and increased frequency of neu-
removed. The prohormone then moves through the Golgi ronal action potentials, were greatly inhibited in TRPV1
apparatus and into the neurosecretory granules that travel knockout mice. Furthermore, TRPV1 knockout mice showed
down the axon. In the neurosecretory granules, the prohor- a higher serum osmolality than wild-type mice, indicating
mone is processed to yield amidated vasopressin, neurophy- a decreased sensitivity of osmosensation.62 However, these
sin, and copeptin.59 Vasopressin and neurophysin form an results were not reproduced by another study.63 In addition
insoluble complex in the nerve terminal and dissociate from to TRPV1, TRPV2 and TRPV4 are present in osmorecep-
each other after its release into the general circulation. tor neurons and functional characteristics of these channels
show responsiveness to the tonicity. Further studies, such as
examining a possible hetero-oligomerization of these TRPV
Regulation of Vasopressin Secretion by the channels, are necessary for a molecular understanding of
Tonicity of Body Fluid osomosensation.60,61
Under physiologic conditions, the most important determi- The functional properties of osmoregulatory mecha-
nant of vasopressin secretion is the tonicity of body uid. nisms have a discrete threshold for vasopressin secretion,
Tonicity de nes the forces that determine the net ux of water above which a linear relationship between plasma osmolality
between two solutions separated by a membrane permeable and vasopressin levels occurs.64 When the plasma osmola-
to water but impermeable to certain solutes. On the other lity is below a threshold level, vasopressin secretion is sup-
hand, osmolality refers to the forces generated by solutes that pressed to low or undetectable levels (0.5 pg per milliliter).
reduce the random movement of water molecules. The osmo- Above this threshold, vasopressin secretion increases linearly
lality is a concentration of all of the solute in water, whereas in direct proportion to plasma osmolality. Both the threshold
the tonicity is an effective osmotic pressure and a concen- level and the slope of the regression line relating vasopressin
tration of all of the osmotically effective solutes. There are secretion to plasma osmolality vary between persons due to
two types of solutes: osmotically effective and noneffective. unknown genetic factors and between the different condi-
Osmotically effective solutes, such as Na and Cl, are charac- tions in the same individual. In general, the threshold is ap-
terized by their inability to move across the cell membrane, proximately 280 mOsm per kilogram of water, and above
resulting in a difference in their concentrations between the this threshold, a rise in plasma osmolality of 1% increases
intracellular and extracellular compartments. On the other plasma vasopressin by approximately 0.4 to 1.0 pg per mil-
hand, osmotically noneffective solutes, such as urea and etha- liliter. The renal response to circulating vasopressin is also
nol, are characterized by their ability to diffuse freely across linear, with urinary concentration that is proportional to va-
the cell membrane and their concentrations between these sopressin levels from 0.5 to 5 pg per milliliter, above which
two compartments are similar. Glucose is con ned in the urinary osmolality is maximal and cannot increase further.
extracellular compartment and is osmotically effective in the Therefore, changes of as little as 1% in plasma osmolality
absence of insulin; whereas in the presence of insulin, glu- are suf cient to cause a signi cant increase in urinary con-
cose is able to enter the cell by insulin-activated transporters, centration, and maximal antidiuresis is achieved by an in-
thereby becoming osmotically noneffective. Vasopressin se- crease in plasma osmolality of only about 10 to 15 mOsm
cretion is nely regulated by the tonicity of body uid. per kilogram of water above the threshold. Furthermore,
The changes in the tonicity of body uid are sensed by vasopressin secretion occurs within minutes in response
a group of cells called osmoreceptor neurons. These neurons to changes in plasma osmolality. After release into the sys-
are located in several brain areas including the organum vas- temic circulation, vasopressin distributes quickly because
culosum laminae terminalis (OVLT), the SON, and the PVN of its small size, and the equilibration between the vascular
nuclei of the hypothalamus.60 It is postulated that an increase and extravascular compartments is almost complete within
in the tonicity of the extracellular compartment causes water 10 minutes. The half-life of vasopressin in plasma is within
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1141
41

30 minutes. Therefore, the changes in plasma osmolality are The pathways responsible for this effect are located in the che-
rapidly transferred to changes in urinary concentration. moreceptor zone in the postrema area of the brainstem. Wa-
There are several factors that can alter the threshold and ter loading blunts, but does not abolish, the effect of nausea
the sensitivity, which is the slope of the regression line re- on vasopressin secretion, suggesting that the mechanism of
lating vasopressin secretion to plasma osmolality. The most emetic effect has a possible common pathway with that of an
important factor is hemodynamic changes, including blood osmotic effect.
pressure and effective arterial blood volume, which are The renin-angiotensin system also in uences the os-
described in the next section. Aging enhances the sensitivity motic regulation of vasopressin secretion.73 Neurons in the
of vasopressin secretion.65 In pregnancy, relaxin, an ovarian subfornical organ (SFO) contain angiotensin II and project
hormone produced by the corpora, reduces the threshold axons onto hypothalamic magnocellular neurosecretory cells
and increases vasopressin secretion, contributing partly to (MNCs) located in the SON and PVN nuclei. It is suggested
the increase in blood volume.66 that the release of angiotensin II may contribute to the po-
tentiation of osmotically evoked action potential ring and
Hemodynamic Regulation of vasopressin release.
Vasopressin Secretion Acute hypoglycemia induces a modest increase in plas-
ma vasopressin,65 although the pathways responsible for this
Vasopressin secretion is also affected by changes in blood effect are unknown.
volume and pressure.67 A decrease in blood volume, such The effects of pregnancy on the osmotic regulation of
as with bleeding, sequestration or redistribution of blood, vasopressin secretion are complex. The systemic hemody-
or uid loss by sweating, diarrhea, or vomiting, can increase namic pro le of pregnancy is characterized by a decrease in
vasopressin secretion. The vasopressin release mechanism is mean arterial pressure, a rise in cardiac output and plasma
much less sensitive to small changes in blood volume than volume, and a decrease in body tonicity. This is due to a
to comparable changes in osmolality. Small reductions under decrease in the osmotic threshold for vasopressin secretion
8% in blood volume usually have little effect on plasma vaso- because of arterial under lling secondary to systemic arte-
pressin concentration. On the other hand, further acute re- rial vasodilatation.74,75 In addition, relaxin, an ovarian hor-
duction in blood volume signi cantly stimulates vasopressin mone produced by the corpora, reduces the threshold and
secretion. Usually, 20% to 30% reductions in blood volume increases vasopressin secretion, which contributes partly
increase vasopressin secretion to the levels of 20 to 30 times to the increase in blood volume.66 On the other hand, the
normal. The stimulus-response relationship follows an ex- metabolic clearance of vasopressin is increased, owing to cir-
ponential pattern. The vasopressin response to an acute re- culating cystine aminopeptidase (vasopressinase) produced
duction in blood pressure is similar to the response to blood by the placenta. This mechanism rarely causes transient dia-
volume. Reductions in blood pressure under 10% have little betes insipidus in late pregnancy76 because of the increased
effect on vasopressin secretion, whereas blood pressure de- release of vasopressin.
creases of 20% to 30% result in signi cant increase in plasma
vasopressin. The effects of reductions in blood volume and
pressure are exerted through shifting the threshold and sen- MOLECULAR MECHANISMS OF
sitivity of vasopressin secretion to osmotic stimuli.68
These hemodynamic effects on vasopressin secretion are VASOPRESSIN-REGULATED WATER
mediated at least in part by neural pathways that originate in PERMEABILITY OF THE
stretch-sensitive receptors called baroreceptors in the cardiac COLLECTING DUCT
atria, the aorta, and the carotid sinus. From these receptors, Vasopressin-regulated water permeability of the collecting
afferent nerve bers ascend in the vagus and glossopharyn- duct is responsible for the nal control of urine concentra-
geal nerves to the nuclei of the tractus solitarus (NTS) in tion. Upon vasopressin stimulation, the water permeability
the brainstem.69 A variety of postsynaptic pathways from the of the collecting duct is dramatically increased. Based on the
NTS then project, both directly and indirectly, to the SON hypertonic medullary interstitium, the increased water per-
and the PVN nuclei of the hypothalamus. meability results in signi cant water reabsorption and urine
Vasopressin secretion through the baroreceptor mecha- concentration. The vasopressin V2 receptor and AQP2 in
nism is promoted not only by a blood volume decrease but the collecting duct are the primary targets for vasopressin.
also by the reduction in effective arterial circulating blood Vasopressin-regulated AQP2 translocation and abundance
volume. For example, upright posture, congestive heart change the water permeability of the collecting duct prin-
failure, and cirrhosis stimulate vasopressin secretion.70,71 cipal cells. The essential roles of the vasopressin V2 recep-
tor and AQP2 in urine concentrations are demonstrated
Other In uences for Vasopressin Secretion by mouse models. Male mice that were introduced with a
The sensation of nausea, with or without vomiting, is a pow- nonsense mutation into the AVPR2 gene, which codes the
erful stimulus to vasopressin secretion.72 Nausea increases vasopressin V2 receptor and is located in the X chromo-
plasma vasopressin in excess of 200 to 400 pg per milliliter. some, died within 7 days after birth due to hypernatremic
142
14 2 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 4.5 A schematic representation of the AQP2 protein dynamics within the cell. Vasopressin binds to the V2 receptor and this stim-
ulates adenylate cyclase via G protein, resulting in the production of cAMP. cAMP stimulates the synthesis of the AQP2 protein through the
interaction of the cAMP responsive element (CRE) of the AQP2 gene and the CRE binding protein (CREBP). Synthesized AQP2 are stored in
subapical vesicles. Protein kinase A (PKA) directly phosphorylates AQP2, and this alters the binding dynamics of AQP2 and its binding pro-
teins (AQP2 binding protein complex), which induces a reorganization of actin and stimulates the exocytosis of AQP2 containing vesicles
to the apical membrane. Upon withdrawal of vasopressin stimuli, AQP2 is internalized by endocytosis and degraded. Some of the internal-
ized AQP2 is transferred to the multivesicular body (MVB) and is excreted to the urine as an exosome.Thus, water transport—crossing the
apical membrane through AQP2 and exiting the basolateral membrane through AQP3 and AQP4—is regulated by vasopressin.

dehydration.77 Mice models with AQP2 protein deletion in and this phosphorylation event is required to increase the
the collecting duct also showed a severe defect in urinary water permeability and water reabsorption of renal principal
concentration.22,78 This section describes the molecular cells, which is described in the next section.
mechanisms for the regulation of water permeability of the
collecting duct principal cells. Figure 4.5 illustrates several Aquaporin-2
major mechanisms. AQP2 is a 271 amino acid protein with 6 membrane-spanning
domains and 2 conserved, membrane-embedded aspara-
Vasopressin V2 Receptor gine–proline–alanine (NPA) motifs, and selectively permeates
When circulating vasopressin reaches the kidney, vasopressin water.3 AQP2 is predominantly expressed in the collecting
binds to the vasopressin V2 receptor expressed on the basolat- duct principal cells (Fig. 4.6).81,82 In response to vasopressin,
eral plasma membrane of the collecting duct principal cells and AQP2 recycles between the luminal cell surface membrane
initiates the signal transduction in the cell. The V2 receptor is a (also called the apical membrane) and the intracellular sub-
371 amino acid protein with 7 membrane-spanning domains apical storage vesicles of the collecting duct principal cells.
and couples to heterotrimeric G-proteins.79,80 The binding of In the absence of vasopressin, AQP2 predominantly resides
the V2 receptor to vasopressin promotes the disassembly of in the subapical vesicles. Because water molecules slowly dif-
the bound heterotrimeric G-protein (Gs) into G and G fuse through the lipid bilayer, the water permeability of the
subunits. GDP-GTP exchange occurs in the G subunit and cell membrane without AQPs is low. Therefore, basal water
this activated Gs then stimulates adenylate cyclase, resulting permeability of the principal cells is also low. In the presence
in an increase in intracellular cAMP levels (Fig. 4.5). Increased of vasopressin, AQP2-containing vesicles fuse with the apical
cAMP activates protein kinase A, which phosphorylates AQP2, membrane via exocytosis, thus inducing an exceedingly high
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1143
43

FIGURE 4.6 The immunoperoxidase labeling of AQP2 in the


cortical (COR), the outer medullary (OM), and the inner medul-
lary (IM) collecting duct. AQP2 is very abundant in the apical
plasma membrane domains, as well as in the subapical domains
(arrows), whereas intercalated cells are unlabeled (arrowheads).
In the inner medullary collecting duct, AQP2 is also present
in the basolateral part of the cell. (Magni cation 1,100.)
(Reprinted from Nielsen S, Knepper MA, Kwon TH, Frøkiaer J.
Urinary concentration and dilution. In: Schrier RW, ed. Diseases
of the Kidney &Urinary Tract, 8th ed. Philadelphia: Lippincott
Williams &Wilkins 2005:98, with permission.)

water permeability of this apical membrane (Fig. 4.7).23–25


Vasopressin increases the water permeability by a factor of
10 to 100 in the cortical collecting duct,49 20 to 30 in the
outer medullary collecting duct,83 and 10 to 30 in the initial
IMCD.49 The terminal IMCD has a higher basal water perme-
ability and vasopressin increases the water permeability by a
factor of 10 in the terminal IMCD.49
The water permeability of the basolateral membrane of
the principal cells is always high because of the presence of A
AQP3 and AQP4, and is not rate limiting for water trans-
port. Thus, water that enters into the cell from the lumen via
AQP2 can exit to the hypertonic medullary interstitium by
the osmotic gradient. Upon the binding of vasopressin to its
receptors, AQP2 is phosphorylated and this phosphoryla-
tion event is a requisite for AQP2 translocation to the apical
membrane (Fig. 4.5). B
FIGURE 4.7 AQP2 immunogold labeling in the inner medul-
Aquaporin-2 Phosphorylation and lary collecting duct cells of control (A) and vasopressin-treated
Traf cking (B) Brattleboro rats. Immunogold labeling is obvious in the
AQP2 forms homotetramers,84 and at least three of four subapical cytoplasm in close association with cytoplasmic
monomers in AQP2 tetramers must be phosphorylated for vesicles (arrows) or in the multivesicular body (*) but is sparse
successful apical membrane localization.85 Phosphorylation on the apical membrane (arrowheads). In contrast, heavy AQP2
of serine 256 is required for the traf cking of AQP2 to the labeling is observed on the apical membrane after vasopres-
cell surface in cultured cells.86,87 Protein kinase A and its sub- sin treatment (arrowheads). N, nucleus. (Magni cation
strates are present throughout the cell; therefore, localization 64,000.) (Reprinted from Yamamoto T, Sasaki S, Fushimi K, et al.
of protein kinase A in speci c sites is necessary for PKA to Vasopressin increases AQP-CD water channel in apical mem-
effectively phosphorylate its target. The phosphorylation brane of collecting duct cells in Brattleboro rats. Am J Physiol.
process is assisted by protein kinase A anchoring proteins 1995;268:C1546, with permission.)
144
14 4 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

(AKAPs). The tethering of PKA to AKAPs is required for AQP2 less, Lorenz et al.102 demonstrated that cyclic AMP alone is
shuttling to the cell surface.88 A splice variant of AKAP-18, suf cient to induce AQP2 translocation without the need for
AKAP-18 , is speci cally involved in AQP2 shuttling89 and an increase in cytosolic Ca2 levels in the inner medullary
the involvement of AKAP-220 has also been reported.90 collecting duct cells.
AQP2 phosphorylation by kinases other than PKA Involvement of extracellular Ca2 in AQP2 regulation
might also be involved in the regulation of AQP2 traf ck- has also been indicated by several ndings.103–106 Urinary
ing. Serine 256 in AQP2 is also a substrate for Golgi casein AQP2 excretion correlates with the severity of enuresis, a
kinase. AQP2 transition through the Golgi apparatus is as- disease characterized by nocturnal polyuria and hypercal-
sociated with a PKA-independent increase in AQP2 phos- ciuria.103 Clinical amelioration demonstrated by a low cal-
phorylation at serine 256, suggesting that phosphorylation cium diet is accompanied by the regulation of urine output
by Golgi casein kinase may be required for Golgi transition.91 through the remodulation of AQP2 expression/traf cking.104
van Balkom et al.92 showed that the activation of protein ki- Drug-induced hypercalcemia/hypercalciuria causes polyuria
nase C mediates AQP2 endocytosis, which is independent of and reduces AQP2 expression in rats.105 AQP2 translocation
the phosphorylation state of serine 256. In addition, a cyclic- to the apical membrane prompted by forskolin-induced in-
GMP-dependent pathway is shown to be involved in AQP2 creases in cyclic AMP levels is inhibited by increased levels
exocytosis,93 and an inhibitor of cyclic GMP phosphodiester- of extracellular Ca2 .106 This process is probably mediated
ase is able to induce AQP2 translocation to the cell surface.94 by the endogenous calcium-sensing receptor and is associ-
In addition to serine 256, there are three additional phos- ated with an increase in F-actin levels.
phorylation sites near the AQP2 C-terminus. These modi -
able residues are serine 261, serine 264, and serine 269. Other In uences for Aquaporin-2 Traf cking
Vasopressin induces the phosphorylation of AQP2 also at
Several other factors have recently been reported to affect
serine 264, and serine 264-phosphorylated AQP2 is translo-
AQP2 traf cking. Nejsum et al.107 used Madin–Darby canine
cated to the plasma membrane similarly to serine 256-phos-
kidney epithelial cells transfected with AQP2 and showed
phorylated AQP2.95 On the other hand, vasopressin decreases
that prostaglandin E2 and dopamine induce the internaliza-
the phosphorylation levels at serine 261, and the localization
tion of AQP2, regardless of AQP2 dephosphorylation. de
of serine-261-phosphorylated AQP2 is different from that of
Seigneux et al.108 reported that aldosterone induces a baso-
serine 256-phosphorylated AQP2, which suggests distinct
lateral expression of AQP2, suggesting a role for aldosterone
roles for these residues in AQP2 traf cking.96 Lu et al.97 re-
in water metabolism in conditions of increased sodium reab-
ported that the phosphorylation state of serine 261 does not
sorption in the collecting ducts.
affect AQP2 traf cking. Serine 269 is shown to be involved
in the plasma membrane retention of AQP2.98 Moeller et al.99
showed that the phosphorylation of serine 264 and serine 269 Role of the Cytoskeleton in Aquaporin-2
depends on the prior phosphorylation of serine 256, and that Traf cking
the phosphorylation of serine 261 partially depends on the The actin cytoskeleton is reported to function as a barrier for
phosphorylation of serine 264 and serine 269. In contrast, AQP2 exocytosis. Actin depolymerization is necessary for
serine 256 phosphorylation is not dependent on the state of the cAMP-dependent translocation of AQP2. 109 In fact, the
any of the other phosphorylation sites, suggesting that serine stimulation of prostaglandin E3 receptors has been shown
256 is the most important phosphorylation site of AQP2. to inhibit vasopressin-induced inactivation of Rho GTPase,
vasopressin-induced F-actin depolymerization, and AQP2
Role of Calcium in Aquaporin-2 Regulation translocation induced by vasopressin, cAMP, or forskolin.110
Intracellular Ca2 mobilization is also involved in va- Rho GTPase activation by bradykinin stabilizes cortical
sopressin-mediated AQP2 traf cking,100 although its precise F-actin and inhibits AQP2 traf cking.111
role remains unclear. In addition to increasing cAMP lev- GTPase-activating protein Spa-1 (SPA-1) binds to the
els in the cytoplasm of the principal cells of the collecting C-terminus of AQP2, and this binding is required for AQP2
duct, vasopressin binding to V2 receptor triggers a rapid in- traf cking.112 SPA-1 may inhibit Rap1 GTPase activating pro-
crease of intracellular Ca2 , which is followed by sustained tein, which triggers F-actin disassembly and may maintain
temporal oscillations of the level of this ion. This process the basal mobility of AQP2.55,113 SPA-1-de cient mice show
seems to be involved in AQP2 exocytosis. Balasubramanian impaired AQP2 traf cking and hydronephrosis.112 In hu-
et al.100 suggest several plausible candidates as downstream mans, mutations in the C-terminus of AQP2, which is the
effectors of this signaling cascade, such as calmodulin and binding region of SPA-1, causes nephrogenic diabetes insipi-
MLCK. MLCK is a calmodulin-dependent kinase that reg- dus (NDI), a disease characterized by a massive loss of wa-
ulates actin lament organization by phosphorylating the ter through the kidney.114,115 Furthermore, AQP2 binds to a
regulatory light chain of myosin II, and thus also activates multiprotein complex that includes the actin cytoskeleton,
myosin motor activity. Myosin II and its regulatory light and a pattern of competing interactions between AQP2 and
chain are found in the AQP2-binding protein complex,101 G-actin or tropomyosin directs AQP2 traf cking to the api-
supporting their involvement in AQP2 traf cking. Neverthe- cal membrane (Fig. 4.5).101,116,117
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1145
45

F-actin assembly might have both inhibitory and facili- accumulate the phosphorylation-de cient mutant in the
tatory effects on an AQP2 transport to the cell surface.118 cell surface despite the fact that AQP2-S256A accumulation
Drug-induced actin depolymerization inhibits AQP2 trans- in the cell surface is not induced by vasopressin. Methyl- -
location from the early endosomes that express early endo- cyclodextrin depletes membranes of cholesterol and results
some antigen 1 to the subapical storage vesicles that express in a rapid inhibition of endocytosis.126 These data also sup-
Rab-11.119 Myosin II and its regulatory light chain are found port the constitutive recycling of AQP2 and the presence of
in an AQP2-binding protein complex,101 and vasopressin in- the processes that are not dependent on phosphorylation of
duces myosin light chain phosphorylation, which enhances AQP2 at serine 256.
the myosin–actin lament interaction and the formation of A heat shock protein hsc70, which is important for un-
actin bers.120 Myosin has also been shown to be critical for coating clathrin-coated vesicles, binds to the C-terminus of
AQP2 recycling.121 In addition to acting as a barrier to pre- nonphosphorylated AQP2 and is required for AQP2 endo-
vent AQP2 traf cking, actin bers may function as “cables” cytosis.127) Kamsteeg et al.128 reported that the myelin and
that promote and direct AQP2 transport. Dynamic actin re- lymphocyte protein (also known as MAL), which is involved
organization may be responsible for the transformation of in the organization of the glycosphingolipid-enriched mem-
the actin barrier into actin cables. Further studies are neces- brane, interacts with AQP2 and enhances accumulation of
sary for understanding these sequential events. AQP2 in the apical membrane by decreasing the level of
internalization of the protein. Ubiquitination at lysine 270
Fusion of Aquaporin-2 Vesicles with the of AQP2 is important for AQP2 endocytosis and degrada-
tion.129 Furthermore, LIP5, which is involved in multivesic-
Apical Membrane
ular body formation, interacts with AQP2 and facilitates its
The docking and fusion of AQP2-containing vesicles with the lysosomal degradation.130
apical membrane involves the action of N-ethylmaleimide-
sensitive factor activating protein receptor (SNARE) proteins, Regulation of Water Transport Activity of
including VAMP-2, SNAP-23, syntaxin-3, and syntaxin-4.52 Individual Aquaporin-2s
Syntaxin-binding protein 2 (also called Munc18b) is reported
As described previously, AQP2 phosphorylation induces its
to function as a negative regulator of SNARE complex forma-
apical membrane insertion, rendering the collecting duct
tion and AQP2-vesicle fusion to the apical membrane.122
cells water permeable. However, whether this phosphory-
lation regulates the water permeability of individual AQP2
Aquaporin-2 Recycling and Endocytosis water channels remained unclear until recently. Several
AQP2 is a recycling membrane protein. Upon vasopressin groups had examined the role of phosphorylation on osmot-
stimulation, AQP2 is transported to the apical membrane, ic water permeability (Pf) of individual AQP2s. In the Xeno-
rendering the cell water permeable, as described previously. pus oocytes expression system, one study showed that PKA
After vasopressin stimulation is terminated, AQP2 is shuttled phosphorylated AQP2 at serine 256, which increased the Pf
back to the cell cytoplasm, a process that restores the water- of oocytes without a signi cant increase in the amount of
impermeability of the cell (Fig. 4.5). This internalization AQP2 on the oocyte surface.131 However, another study us-
process consists of AQP2 retrieval into early endosomes that ing a similar method showed that the Pf values that corrected
express early endosome antigen 1, and subsequent trans- for the plasma membrane abundance of AQP proteins were
feral of this water channel to storage vesicles that express not different between WT-AQP2 and the nonphosphoryla-
Rab-11.123 From Rab-11-positive vesicles, AQP2 is able to tion-mimicking mutant S256A-AQP2 expressing oocytes,
go again to the apical membrane. Actually, this recycling indicating a lack of an effect of phosphorylation at S256 on
process occurs constitutively, and many signaling pathways the Pf of individual AQP2 proteins.99 Lande et al.132 puri ed
are involved for the regulation of each part of this recycling endosomes derived from the apical membrane of rat IMCD
itinerary. Vasopressin signaling is the most potent and most cells that were highly enriched for AQP2. Then, these endo-
important factor that enhances the exocytotic process among somes were phosphorylated by an exogenous PKA catalytic
the recycling itinerary. subunit or were dephosphorylated by an exogenous alkaline
During the endocytotic process of the AQP2 recycling phosphatase. There was no signi cant difference in Pf be-
pathway, AQP2 accumulates in clathrin-coated pits and is tween these two samples, suggesting that the Pf of AQP2 is
internalized via a clathrin-mediated process.124,125 Dynamin not changed by its phosphorylation. However, involvement
is a GTPase that is involved in the formation and pinching of other proteins, such as endogenous phosphatases, cannot
off of clathrin-coated pits to form clathrin-coated vesicles, be neglected.132
and its dominant-negative mutant K44A renders the pro- To clarify whether the Pf of AQP2 is regulated by its
tein GTPase de cient and arrests clathrin-mediated endo- phosphorylation event alone, the experimental system that
cytosis. This GTPase-de cient dynamin mutant K44A is does not contain any other regulatory proteins is required.
shown to accumulate AQP2 in the plasma membrane even Eto et al.133 performed a large-scale expression of full-length
without vasopressin stimulation.125 Furthermore, this dy- recombinant human AQP2, puri cation and reconstitution
namin mutant K44A or methyl- -cyclodextrin is able to in proteoliposomes, and examined the protein function.
146
14 6 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

The results showed that the Pf of proteoliposomes was en-


hanced approximately twofold by phosphorylation at serine CHANNELS AND TRANSPORTERS
256. This observation indicates that AQP2 water channel CONTRIBUTING TO URINE
activity is directly regulated by its phosphorylation, and the CONCENTRATION AND DILUTION
mechanism involved may be the channel gating that is well Urine concentration and dilution are enabled by the special-
studied in plant AQPs by X-ray crystal structure analysis.134 ized organization of the renal tubule and vasculature, and the
Thus, in addition to AQP2 translocation to the luminal presence of channels and transporters with their speci c lo-
membrane, the water transport activity of individual AQP2 calizations. This section describes water channel aquaporin
proteins may be involved in the regulation of water reab- family members other than AQP2 and other channels and
sorption in kidney collecting ducts. transporters involved in urine concentration and dilution.
Actually, vasopressin increases the water permeability Table 4.1 summarizes the basic characteristics of AQP mem-
of the collecting ducts more than tenfold.49,83 Thus, vaso- bers in the kidney, and tissue localization and phenotypes of
pressin-induced short-term regulation of Pf of the collecting mice and humans caused by gene knockout or mutations.
ducts still seems to be mainly due to AQP2 translocation, Figure 4.8 shows the localization of these channels and trans-
and the altered water transport activity of individual AQP2s porters along the nephron. Figure 4.9 shows the maximal
may have a role of doubling the effect. urine osmolalities of the knockout mice in which channels and
transporters involved in urinary concentration are deleted.
Long-Term Regulation of Water Permeability
of the Collecting Duct by Altered Aquaporin-1
Aquaporin-2 Abundance AQP1 is localized on the apical and basolateral membrane of
In addition to short-term regulation of collecting duct water the proximal tubules, the descending thin Henle limbs, and
permeability described previously, long-term regulation also the outer medullary descending vasa recta.
plays an important role in body water balance. Long-term AQP1 is constitutively active as a water-selective pore in
regulation of collecting duct water permeability is seen when these segments.1,26
water intake is restricted for 24 hr or more, resulting in an AQP1-null mice show marked polyuria. Urinary os-
increased maximum concentrating ability. This response is molality in AQP1-null mice is low and unresponsive to
mainly induced by an increase in AQP2 abundance due to vasopressin secretion or water deprivation.11,145 As shown
increased transcription of the AQP2 gene.135,136 An increased in Figure 4.9, maximal urine osmolalities of AQP1 knock-
AQP2 expression level during water restriction is a down- out mice is 23% of that of wild-type mice under the same
stream of vasopressin signaling.137,138 It is known that the stimulated condition (i.e., 36 hr water deprivation). This
cAMP responsive element (CRE) is present in the 5 - anking phenotype was explained by two distinct mechanisms:
region of the AQP2 gene and regulates the transcription of impaired near iso-osmolar water reabsorption by the proxi-
the gene.139,140 Hasler et al.141 examined the AQP2 gene mal tubule and reduced medullary hypertonicity resulting
transcription in a cultured cell line, which endogenously ex- from impaired countercurrent exchange.146 Analysis of
presses AQP2 and showed that the vasopressin-stimulated AQP1-null mice clari ed that AQP1 is the principal water
transcription of AQP2 was mediated through CRE. channel in the proximal tubules and the descending thin
Medullary collecting ducts are surrounded by intersti- Henle limbs and provides the major route for transepithe-
tial hypertonicity and the hypertonicity may be another de- lial water permeability in these segments.146 In 2001, two
terminant of AQP2 transcription.142,143 Hypertonicity affects unrelated individuals with a de ciency in AQP1 expression
transcription of many genes through the interaction between were reported.147 One of these individuals was homozygous
the tonicity-responsive enhancer (TonE) and its transcription for a deletion of exon 1 of AQP1. The second individual was
factor TonEBP. TonE is observed in the AQP2 gene, suggest- homozygous for a frame-shift mutation in exon 1 of AQP1.
ing that TonE/TonEBP contributes to AQP2 transcription. Although both patients seemed asymptomatic under normal
This possibility was examined in TonEBP gene knockout conditions, they showed an impaired urinary concentrating
mice, which had mostly embryonic lethality or died within a ability following 24 hr of water restriction as compared with
few weeks after birth. However, the surviving mice showed healthy controls (450 versus 1,000 mOsm per kilogram of
atrophy of the kidney medulla, and the protein expression water). Such a defect can become clinically meaningful in
of AQP2 but not of AQP3 was clearly downregulated in the circumstances that require maximal urinary concentration;
knockout mice, con rming the role of TonE/ToneEBP in for instance, during vomiting and diarrhea.148
AQP2 transcription.144 Taken together, AQP2 protein abun-
dance in long-term regulation is likely to be mediated by Aquaporin-3
stimulated transcription of the AQP2 gene through CRE and The AQP3 water channel is present in the principal cells
TonE motifs. In addition, the presence of AP1 and GATA ele- of the connecting tubule and the collecting duct, and en-
ments are known in the promoter region of the AQP2 gene, ables water entry into the interstitium in these segments.149
but their roles remain speculative (Fig. 4.5). AQP3 is constitutively localized in the basolateral plasma
T
T A
A B
B L
L E
E
A q u a p o r
i n s i n t h e K i
d n e y
4 . 1
A
K
S
y
e
n
d
D
r
a
l
u
l
l
e
c
b
L
n
o
i
t
u
b
i
r
t
s
A
h
t
i
w
e
c
i
M
f
o
e
p
y
t
o
n
e
h
P
l
a
n
e
r
a
r
t
x
E
n
o
i
t
a
z
i
l
a
c
P
s
n
o
i
t
a
t
u
M
P
w
s
n
a
m
u
H
f
o
e
p
y
t
o
n
e
A
s
n
o
i
t
a
t
u
M
P
Q
A
h
t
H
of
s
b
m
i
l
n
i
h
t
g
n
i
d
n
e
c
s
e
D
s
e
l
u
b
u
t
l
a
m
i
ox
r
P
1
P
O
e
l
n
d
y
r
a
l
l
u
d
e
m
r
e
t
n
e
c
s
p
l
a
r
e
t
a
ol
s
a
b
d
n
a
l
a
c
i
p
A
a
t
c
e
r
a
s
a
v
g
n
n
a
r
b
m
e
m
a
m
s
a
,
s
e
t
y
c
o
r
h
t
y
r
E
y
r
a
i
l
i
d
n
y
r
a
n
o
m
l
u
p
,
s
u
x
e
l
p
d
i
o
r
o
h
c
,
m
u
i
l
e
h
t
i
p
e
s
n
e
e
r
a
l
u
c
s
a
e
h
t
o
d
M
on
i
t
a
s
n
e
s
n
i
a
p
d
e
r
i
a
p
m
I
t
c
e
f
e
d
g
n
i
t
a
r
t
n
e
c
on
c
y
r
a
n
i
r
U
m
u
i
A
t
c
e
f
e
d
g
n
i
t
a
r
t
n
e
c
on
c
y
r
a
n
i
r
u
d
l
c
l
a
p
i
c
n
i
r
p
t
c
u
d
g
n
i
t
c
e
l
ol
C
2
P
A
s
l
m
a
m
s
a
l
p
l
a
c
i
m
r
s
d
n
a
e
n
a
b
c
i
e
v
l
c
e
A
s
u
d
i
p
i
s
n
i
s
e
t
e
b
a
i
D
s
u
d
i
p
i
s
n
i
s
e
t
e
b
a
i
D
a
e
l
h
oc
c
,
s
i
m
y
d
i
d
i
p
E
c
l
a
p
i
c
n
i
r
p
t
c
u
d
g
n
i
t
c
e
l
ol
C
3
P
m
a
m
s
a
l
p
l
a
r
e
t
a
ol
s
a
B
s
l
m
r
A
l
a
m
or
n
y
l
t
n
e
r
a
p
p
A
s
or
m
u
t
n
i
k
s
of
on
i
t
a
m
or
f
e
h
t
o
t
e
c
n
a
t
s
i
s
e
R
g
n
i
l
a
e
h
d
n
ou
w
d
e
r
i
a
p
m
i
,
on
i
t
a
r
d
y
h
n
i
k
s
d
e
c
u
d
e
R
t
c
e
f
e
d
g
n
i
t
a
r
t
n
e
c
on
c
y
r
a
n
i
r
U
s
e
t
y
oc
r
h
t
y
r
e
,
s
e
t
y
oc
n
i
t
a
r
e
k
,
a
i
l
e
h
t
i
p
e
c
i
on
ol
c
,
a
i
l
e
h
t
i
p
e
y
a
w
r
i
a
y
r
a
on
m
l
u
p
,
a
v
i
t
c
n
u
j
on
C
e
n
c
l
a
p
i
c
n
i
r
p
t
c
u
d
g
n
i
t
c
e
l
ol
C
4
P
m
a
m
s
a
l
p
l
a
r
e
t
a
ol
s
a
B
s
l
l
l
a
n
i
t
e
r
,
a
m
y
d
n
e
p
e
,
a
i
l
og
r
t
s
A
e
n
a
r
b
m a
l
v
a
a
u
n
l
e
p
a
s
l
s
e
e
o
c e
d
l
e
u
h
i
p
Q
Q
Q
Q
Q
Q
i
e
u
i
e
b
i
e
b
e
i l
l
i P Lo c a l i
z a t i o n i
n g
k
a
p
d
g
n
i
t
a
r
t
n
e
c
on
c
y
r
a
n
i
r
u
d
l
i
M
A
t
or
p
e
r
o
N
on
i
t
c
a
f
l
o
d
n
a
,
g
n
i
r
a
e
h
,
on
i
s
i
v
d
e
r
i
a
p
m
I
t
c
e
f
i
t
c
u
d
g
n
i
t
c
e
l
ol
C
6
P
l
a
c
r
e
t
l
e
c
d
e
t
A
t
or
p
e
r
o
N
t
or
p
e
r
o
N
s
e
l
c
i
s
e
v
c
i
t
p
a
n
y
s
,
m
u
l
l
e
b
e
r
e
C
s
e
l
c
i
s
e
v
r
a
l
u
l
l
e
c
a
r
t
n
I
s
t
l
a
m
i
ox
r
p
of
t
n
e
m
g
e
s
3
S
7
P
A
s
e
l
u
b
m
a
m
s
a
l
p
l
a
c
i
m
l
a
t
e
l
e
k
s
,
s
i
t
s
e
t
,
e
u
s
s
i
t
e
os
p
i
d
A
e
n
a
r
b
m
i
,
n
i
a
r
b
,
t
r
a
e
h
,
e
l
c
s
i
t
s
e
t
o
,
m
s
i
ol
b
a
t
e
m
ol
r
e
c
y
l
g
e
v
i
t
c
e
f
e
D
a
i
r
u
ol
r
e
c
y
l
G
e
A
m
s
i
ol
b
a
t
e
m
ol
r
e
c
y
l
g
e
v
i
t
c
e
f
e
D
s
l
l
e
c
t
e
l
s
i
r
e
l
l
a
m
s
,
y
t
i
s
e
(
m
u
l
u
c
i
t
e
r
c
i
m
s
a
l
op
d
n
E
s
e
l
u
b
u
t
l
a
m
i
ox
r
P
1
1
P
t
or
p
e
r
o
N
s
y
e
n
d
i
k
c
i
t
s
y
c
y
ol
P
r
e
v
i
l
,
e
n
i
t
s
e
t
n
i
,
s
u
m
y
h
t
,
s
i
t
s
e
T
)
R n
a
l
n
n
u
E
i
l
b
e a
e
p
Q
Q
Q
r
i
e
u
r
r a
w
a
i
, e
t
a
m
t i
y
n a
u
l oc
e
c
sp c
e
yi
l l
tt
e
l he
s se
, lb
i p
ae ,
u
r s
l
c
m t om
e
l
on l
s a
,
a c
r
h y
CHAPTER 4
REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION

F
N
N
v
e
R
t
.
on
i
s
s
i
m
r
e
p
h
t
i
w
,
8
6
1
:
6
;
0
1
0
2
.
l
o
r
h
p r
a
e om N od a Y
, S
oh a r a E , O h t a E , e
t a
l . A
q u a
p or i
n s i
n k i
d n e
y p a
t h op h y
s i
ol og
y .
47
1147
148
14 8 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 4.8 Transporters and channels involved


in urine concentration along the nephron. Water
is reabsorbed in the proximal tubule and the thin
descending limb by AQP1. Chloride with sodium
is passively reabsorbed in the thin ascending limb
by the ClC-K1 chloride channel. NaCl is actively re-
absorbed across the thick ascending limb by the
apical plasma membrane Na-K-2Cl cotransporter
(NKCC2). Potassium is recycled through an apical
plasma membrane potassium channel, ROMK,
and chloride is transported across the basolateral
membrane by ClC-K2. Water is reabsorbed across
the apical membrane of the collecting duct by
AQP2 water channels. Water is reabsorbed across
the basolateral membrane by AQP3 in the cortical
and outer medullary collecting ducts and by both
AQP3 and AQP4 in the inner medullary collect-
ing duct. Urea is reabsorbed in the terminal inner
medullary collecting duct by the urea transport-
ers UT-A1 and UT-A3.

FIGURE 4.9 Maximal urine osmolalities of knockout mice in


which transporters or channels involved in urine concentration
are deleted. Data are expressed as a percentage of the maxi-
mal urine osmolalities of the wild-type mice in the same study.
A maximally stimulated urine concentration was induced by
water deprivation for 6 to 36 hr or by vasopressin administra-
tion. Data are taken from References 11, 13, 32, 42, 44, 78, 152,
158, 172, 180, and 186.
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1149
49

membrane. AQP3, together with AQP4, represents poten- The case of a man with a point mutation in AQP7 has
tial exit pathways from these cells for water entering the cell been reported. However, no renal symptoms were mentioned,
via AQP2. The AQP3 expression is shown to be increased although he did have a defective glycerol metabolism.167
by thirst and by vasopressin or aldosterone secretion.150,151
AQP3 knockout mice exhibit an NDI-like phenotype, in- Aquaporin-11
dicating that AQP3 has an important role in urine con- AQP family members have two widely conserved, membrane-
centration (Fig. 4.9).152,153 AQP3 de ciency was reported embedded NPA motifs that are essential for forming the
in humans.154 This defect is caused by homozygous muta- water-permeable pore structure. However, AQP11, together
tion affecting the 5 donor splice site of intron 5 of AQP3. with AQP12, have atypical NPA boxes, suggesting that their
This mutation causes the skipping of exon 5 and generates structure and function are unique.168 In the kidney, AQP11
a frameshift change and premature stop codon. However, is present in the proximal tubular cells. Within the cells,
phenotypes associated with this defect were not reported, AQP11 is localized in the endoplasmic reticulum (ER) mem-
possibly because the patients seemed normal. brane, but not localized in the cell surface plasma mem-
brane. AQP11 knockout mice are born healthy but grow
Aquaporin-4 poorly and die before weaning because of uremia caused by
AQP4 is present in the principal cells of the collecting enlarged multiple cysts in the kidney.169 The cysts originate
duct.155 AQP4 is more abundant in the inner medullary col- from kidney proximal tubule cells, inside which swollen ER
lecting duct cells than is AQP3. The organization of AQP4 are observed. The role of AQP11 is still unclear, although it
into orthogonal arrays of particles might enhance AQP4 wa- seems to have an important role in ER membrane function in
ter permeability,156 and this process might be regulated by proximal tubular cells. Kidney cyst cells in AQP11 knockout
vasopressin.157 Similarly to AQP3, AQP4 is constitutively mice showed an abnormal gene expression pattern, which is
localized in the basolateral plasma membrane. AQP4 knock- similar to that observed in polycystic kidney disease (PKD)
out mice have a very little urinary concentrating defect (Fig. model animals, suggesting that AQP11 knockout mice and
4.9).153,158 In contrast to AQP3, AQP4 expression is not PKD animals share common pathogenic mechanisms for cyst
regulated by thirst or by vasopressin secretion. AQP4 water formation.170 Yakata et al.171 measured the Pf of the vesicles
permeability is decreased by high levels of protein kinase C from the membrane fraction of Sf9 cells expressing AQP11
and dopamine. This effect is mediated by phosphorylation at and showed that AQP11 has slow but constant water trans-
serine 180 of AQP4.159 port activities. Further studies are required for clarifying the
role of AQP11.
Aquaporin-6
Urea Transporters
The water channel family member AQP6 is localized in
In the kidney, there are four urea transporters: UT-A1, UT-A2,
the intracellular vesicles in intercalated cells in the collect-
UT-A3, and UT-B (Figs. 4.4 and 4.8). UT-A1 and UT-A3 are
ing duct.160–162 AQP6 has low water permeability and acts
expressed in the terminal IMCD,32 and UT-A2 is present in
primarily as an anion transporter.163 AQP6 is expressed in
the thin descending limb of the long-looped nephron in the
acid-secreting type-A intercalated cells and colocalizes with
inner medulla and the thin descending limb of the short-
V-type H -ATPase; therefore, AQP6 is suggested to function
looped nephron in the outer medulla.14 Their localization is
to promote urinary acid secretion.
essential for the urea recycling pathway as described in the
section Urea Accumulation in the Inner Medulla, discussed
Aquaporin-7 previously. Vasopressin stimulation promotes the phosphor-
AQP7 is localized in the brush border of proximal straight ylation of UT-A1 and UT-A3 and the accumulation of UT-A1
tubules (S3 segment)12,164,165 and is able to transport glyc- and UT-A3 in the plasma membrane of the IMCD cells. This
erol as well as water.12 AQP7 knockout mice showed marked is responsible for the increased urea permeability of the ter-
glyceroluria,13 indicating that glycerol can be reabsorbed minal IMCD that results in greater urea reabsorption during
through AQP7 in the proximal straight tubule, and that antidiuresis.50 Vasopressin stimulation also increases UT-A2
there might be no other glycerol reabsorbing system to com- protein abundance.14
pensate for this defect in the distal nephron segments. UT-A1/UT-A3 knockout mice show a large urinary
In AQP7 knockout mice, water permeability of the brush concentrating defect (Fig. 4.9). This concentrating defect is
border membrane vesicles was slightly but signi cantly low- ameliorated by a low protein diet, indicating that this de-
er than that in wild-type mice,13 which suggests that AQP7 fect is caused by a urea-dependent osmotic diuresis.32 An-
makes a small contribution to the water permeability of the other possibility may be that a low protein diet reduces urea
proximal straight tubules. This contribution has been esti- concentrations in the medulla in both UT-A1/UT-A3 knock-
mated to be one-eighth that of AQP1.166 As expected from out mice and wild-type mice, and the difference of urine
the small decrease in water permeability, AQP7 knockout concentrating abilities of these two groups becomes small.
mice did not show a urine concentrating defect (Fig. 4.9).13 Nevertheless, UT-A1/UT-A3 greatly contributes to urine
150
15 0 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

concentration by accumulating urea in the medulla when NKCC2 and ROMK


protein intake is normal. NKCC2 is expressed in the thick ascending limb and con-
On the other hand, in UT-A2 knockout mice, a small tributes to NaCl reabsorption in this segment.56 The gene
urinary concentrating defect and a reduction in medullary encoding NKCC2 is another gene, the mutations of which
urea content are observed only on a low protein diet, but cause Bartter syndrome.179 Different from NHE3, NKCC2 is
not on a normal diet (Fig. 4.9; a value on a normal diet also present in the macula densa.56 NKCC2 knockout mice
is shown).172 This nding indicates a relative role of recy- cannot survive to weaning because of severe dehydration
cling urea through the “tubular route”; urea leaves from and polyuria. This may be because the compensatory de-
IMCD to the interstitium, diffuses around, then enters into crease in glomerular ltration does not occur in response to
the tubular lumen at the thin descending limb (mediated increased distal uid delivery.180 This nding indicates that
by UT-A2), and again comes back the IMCD (Fig. 4.4), is NKCC2 plays an important role in the macula densa in the
small, and becomes signi cant when urea delivery to the mediation of tubuloglomerular feedback and is critical for
IMCD is low. distal uid delivery. Treatment with indomethacin rescues
UT-B is localized in the descending vasa recta endo- the knockout mice, and the surviving mice show a large
thelial cells.46 UT-B knockout mice show a decrease in urea vasopressin-resistant defect in urine concentrating ability
accumulation in the medulla and a urinary concentrating (Fig. 4.9).180 Thus, in the same mechanism as in ClC-K2,
defect, indicating that the recycling of urea by a counter- NKCC2 contributes to urine concentration.
current exchange via the vasa recta is important for urine Another Na-K-Cl cotransporter isoform, NKCC1, is ex-
concentration abilities (Fig. 4.9).44 Humans genetically lack- pressed in the collecting duct and contributes to NaCl secre-
ing UT-B (Kidd blood group antigen) are identi ed and their tion.181,182 NKCC1 knockout mice show a reduced capacity
maximal urinary concentrating ability is reduced (UOSM, to excrete free water.183 NKCC1 is present also in the juxta-
max 819 mOsm per kilogram of water), which is consis- glomerular afferent arteriole and the glomerular and extra-
tent with the results of the knockout mice.173 glomerular mesangium, where it is thought to participate in
the process of tubuloglomerular feedback and the regulation
ClC-K1 and ClC-K2 of blood pressure.
The chloride channel ClC-K1 is mainly expressed in the The ATP-sensitive, inwardly rectifying potassium chan-
thin ascending Henle limb and its expression is upregulat- nel, ROMK, is expressed in the apical membrane of the thick
ed by water deprivation.10,15,16 ClC-K2 is present from the ascending limb and throughout the distal nephron segments
thick ascending limb through to the collecting duct.174,175 and is involved in NaCl reabsorption in the thick ascending
ClC-K1 knockout mice show a signi cantly reduced os- limb.176,184 Mutations of the gene encoding ROMK are also
molality of the papilla and a severe defect in urinary con- found in patients with Bartter syndrome.185 Knockout mice
centrating ability (Fig. 4.9).42 This nding indicates that with ROMK have hydronephrosis, are severely dehydrated,
the rapid chloride exit to the medullary interstitium is im- and 95% die before 3 weeks of age. The urine concentrating
portant for the maintenance of the high inner medullary ability of the surviving mice is severely impaired, as expected
interstitial osmolality and supports the passive mechanism (Fig. 4.9).186
as described in the section Countercurrent Multiplication A Na-H exchanger isoform NHE3 is expressed in the
in the Outer Medulla and Other Mechanisms in the Inner thin descending Henle limb and the thick ascending Henle
Medulla, discussed previously. Severities of impaired urine limb and contributes to sodium reabsorption in these seg-
concentrating abilities are comparable in ClC-K1 and ments.57 NHE3 knockout mice show a marked reduction in
UT-A1/UT-A3 knockout mice (Fig. 4.9), implying a func- proximal tubule uid absorption. Nevertheless, these mice
tional linkage between the chloride transport by ClC-K1 maintain a relatively normal distal delivery, and its urinary
and the urea transport by UT-A1/UT-A3, which is again concentrating defect is mild.187,188 This is due to a compen-
consistent with the operation of the passive model in the satory decrease in the glomerular ltration rate owing to
inner medulla. an intact tubulo-glomerular feedback.188 Furthermore, the
The human orthologue of the ClC-K2 gene is one of the concentrating defect of NHE3 knockout mice is explained
genes that, when mutated, causes Bartter syndrome in hu- by the decreased expression of NKCC2, a Na-K-Cl cotrans-
mans, which is a disease manifested by salt wastage, hypo- porter isoform.189
kalemia, metabolic alkalosis, and hyperaldosteronism.176,177
Polyuria and polydipsia are commonly observed in Bartter NCC and ENaC
syndrome and these symptoms are not relieved by vaso- The Na-Cl cotransporter NCC is present in the distal con-
pressin, thus showing the presence of NDI.178 Thus, NaCl voluted tubule,19 and the Na channel ENaC is present in
reabsorption in the thick ascending limb mediated by a ba- the connecting tubule and the cortical collecting tubule
solateral chloride channel, ClC-K2, importantly contributes (Fig. 4.8).28,29 Both the proteins mediate sodium reabsorp-
to urine concentration through the operation of the counter- tion beyond the macula densa and contribute to urine
current multiplication system. concentration by reducing uid delivery to the medullary
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1151
51

collecting duct. Long-term vasopressin stimulation increases their plasma osmolality. There are congenital and acquired
the abundance of NCC and ENaC.190 Short-term vasopres- forms of NDI.
sin stimulation promotes ENaC accumulation in the apical
membrane, resulting in increased sodium reabsorption.191 Congenital Nephrogenic Diabetes Insipidus
In more than 90% of cases of congenital NDI, the condi-
URINE CONCENTRATING DEFECTS tion results from a loss of function mutations in the AVPR2
Central Diabetes Insipidus gene encoding the vasopressin V2 receptor, which makes the
collecting duct cells insensitive to vasopressin. The AVPR2
Central diabetes insipidus is caused by inadequate secretion
gene is located in chromosome region Xq28, and its muta-
of vasopressin from the posterior pituitary in response to
tions cause the X-linked inheritable form of NDI. To date,
osmotic stimulation. In most cases, this is due to the de-
over 200 mutations have been reported in the AVPR2 gene,
struction of the neurohypophysis by a variety of lesions,
which are categorized into ve classes according to the cel-
including granulomas (e.g., histiocytosis, sarcoidosis), neo-
lular fate.197–199 Class I mutations result in premature stop
plasms (e.g., craniopharyngioma, germinoma, lymphoma,
codons or unstable mRNA, and include promoter alterations,
leukemia, meningioma, pituitary tumor, metastasis), infec-
exon skipping, aberrant splicing, frameshift, and non-sense
tions (e.g., meningitis, tuberculosis, encephalitis), trauma,
mutations that result in truncated proteins. On the other
and cerebrovascular diseases (e.g., cerebral hemorrhage,
hand, mutations of class II, III, IV, and V result in fully trans-
infarction). Several cases with inherited central diabetes
lated proteins. Class II comprises the missense or insertion/
insipidus are also reported, although it is a rare disor-
deletion mutations that lead to the misfolding of this pro-
der.192–194 Most of these cases show an autosomal dominant
tein. Misfolded proteins are retained in the ER and, subse-
mode of inheritance and are caused by mutations in the
quently, are mostly targeted for proteasome degradation.
gene coding for the vasopressin-neurophysin precursor. It
Class II is most prevalent, comprising approximately 45% of
is postulated that these mutations cause the production of
all mutations in the AVPR2 gene. Class III and IV mutations
an abnormal precursor protein that accumulates and dam-
do not interrupt the translocation to the plasma membrane.
ages the neurons.
Class III mutations interfere with binding to the stimulatory
Gs protein, leading to the reduced activation of adenylate
Osmoreceptor Dysfunction cyclase, whereas Class IV mutations interfere with binding
The osmoreceptors that control vasopressin secretion and to vasopressin. Class V mutations result in traf cking defects
thirst are located in the anterior hypothalamus. Osmoreceptor beyond the early secretory pathway and misrouting to dif-
dysfunction is caused by their damage by a variety of brain ferent organelles in the cell. Although most female carriers
lesions including granulomas, neoplasms, and cerebrovas- of the X-linked V2 receptor defect have no clinical disease,
cular diseases. In contrast to central diabetes insipidus, os- some females were reported with symptomatic NDI.200 Rare
moreceptor dysfunction-causing lesions usually occur more female patients manifest severe NDI due to the inactivation of
rostrally in the hypothalamus because of the location of the the normal X chromosome.201,202 Furthermore, a signi cant
osmoreceptors. One lesion unique to this disorder is an an- number of de novo mutations have also been reported.203
terior communicating cerebral artery aneurysm. In this disor- Congenital NDI can also result from mutations of the
der, osmoregulation of both vasopressin secretion and thirst is autosomal gene that codes for AQP2. AQP2, the gene that
impaired.195 Patients with osmoreceptor dysfunction typically encodes AQP2, is located in 12q13 and is comprised of
have osmolalities in the range of 300 to 340 mOsm per kilo- four exons.139,204,205 To date, over 43 mutations in the AQP2
gram of water, whereas patients with central diabetes insipi- gene have been reported (Fig. 4.10).51,206,207 Two inheritance
dus maintain their plasma osmolality within the normal range types are possible for the disease: Autosomal-recessive NDI
by polydipsia. This underscores the importance of a normal is associated with 35 mutations, and autosomal-dominant
thirst mechanism. On the other hand, in patients with osmo- NDI is associated with 8 mutations. Almost all of the muta-
receptor dysfunction, the baroreceptor mediated pathway196 tions in recessive NDI are located in the core region of the
and responses to other nonosmotic stimuli such as nausea are protein, and they lead to misfolded proteins that become
intact. These patients have normal vasopressin and renal con- trapped in the ER and then are targeted for rapid degrada-
centrating responses to hypovolemia and hypotension. tion by the proteasome. On the other hand, AQP2 homo-
tetramers composed only of wild-type proteins are properly
Nephrogenic Diabetes Insipidus translocated to the apical membrane. This effect explains the
NDI is caused by the inability of the kidney to respond to healthy phenotype of patients’ parents.206
vasopressin stimulation. Thus, plasma vasopressin levels in All mutations in autosomal-dominant NDI locate in
NDI patients are increased or normal depending on their the cytosolic C-terminus of AQP2 (Fig. 4.10). This region is
plasma osmolality. Hyperosmolar patients with NDI have el- important for AQP2 traf cking and these mutations impair
evated vasopressin levels, whereas those with central NDI traf cking to the apical membrane, although the water chan-
have an absence of blunted vasopressin responses relative to nel function of these mutants is preserved. Arg254Leu and
152
15 2 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 4.10 The AQP2 membrane topology and mutations causing nephrogenic diabetes insipidus. Amino acids that, when mutated,
cause the autosomal-recessive form of nephrogenic diabetes insipidus are shown in grey. Also, grey arrows show AQP2 deletions and
the location of the rst amino acid of the protein affected by these nucleotide deletions, which causes the autosomal-recessive form of
nephrogenic diabetes insipidus. Amino acids, when mutated, that cause the autosomal-dominant form of nephrogenic diabetes insipidus
are shown in black. Similarly, in black are also reported AQP2 deletions or insertions, and the location of the rst amino acid of the protein
affected by these nucleotide changes, which cause the autosomal-dominant form of nephrogenic diabetes insipidus. bp, base pair.

Arg254Gly mutations destroy the site for PKA phosphory- apical sorting of wild-type AQP2. The urine concentrating
lation, so that forskolin-induced traf cking to the plasma ability of these gene knockin mice is severely reduced.
membrane is impaired.207,208 The Glu258Lys mutant of AQP2 Several other mouse models of NDI caused by AQP2
is missorted to multivesicular bodies and/or lysosomes.209 mutations have also been generated. Yang et al.21 created
AQP2 mutants resulting from three gene deletions, 721delG, mice with a T126M knockin mutation in the AQP2 gene.
763–772del, and 812–818del, have similar extended C- These homozygous mutant mice died within 6 days after
terminal tails, which contain the basolateral-membrane- birth, suggesting that the mice may be highly sensitive or-
sorting dileucine (LeuLeu) motif, so these mutated proteins ganisms with regard to water homeostasis, and are unable to
are wrongly translocated to the basolateral membrane.114,210 survive with polyuria. Lloyd et al.212 generated mice with a
In contrast to the AQP2 mutants associated with the reces- F204V mutation in the AQP2 gene that survived beyond the
sive form of the disease, AQP2 mutants associated with the neonatal period and had a much milder form of NDI.
dominant form of the disease are not misfolded, so they are
able to form heterotetramers with wild-type AQP2. Because Acquired Nephrogenic Diabetes Insipidus
of the dominance of the missorting motif in the mutant pro- Acquired NDI is more common than congenital NDI and
teins, tetramers composed of the mutant and wild-type forms is caused by various conditions, including drug treatments,
are missorted, which leads to severely decreased amounts of electrolyte disturbances, and urinary tract obstruction.
AQP2 on the apical membrane. This effect explains the dom- Dysregulation of AQP2 plays a fundamental role in many
inant mode of NDI inheritance in patients with these muta- cases of acquired NDI.
tions. Sohara et al.211 generated gene knockin mice with the Lithium is widely used for treating bipolar disorder
heterozygous mutant AQP2 resulting from a gene deletion and 20% to 30% of patients treated with lithium develop
(763–772del) that produces a mouse model of dominant NDI.213 In lithium-induced NDI, both AQP2 expression and
NDI. The mutant AQP2 is wrongly translocated to the baso- its traf cking to the apical membrane are inhibited. Lithium
lateral membrane; it forms a heterotetramer with wild-type enters cells expressing AQP2 via the epithelial sodium
AQP2 and shows a dominant-negative effect on the normal channel (ENaC) in the apical membrane and accumulates
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1153
53

intracellularly. This accumulation leads to the inhibition of rat models of cardiac failure.226,227 Furthermore, water re-
signaling pathways that involve glycogen synthase kinase- tention and hyponatremia in these rats are reversed by a V2
3 (GSK3 ). Although the mechanism by which AQP2 is receptor antagonist.227 These ndings indicate that hypona-
dysregulated in this context is not established, the involve- tremia is caused by nonosmotic stimulation of vasopressin,
ment of GSK3 is speculated. Inhibition of GSK3 by lith- which promotes the expression and traf cking of AQP2. In
ium increases the expression of cyclo-oxygenase 2 and the patients with heart failure, V2 receptor antagonists promote
local excretion of prostaglandin E2.214 Prostaglandin E2 is electrolyte-free water excretion and elevate serum sodium
suggested to counteract vasopressin activity by causing an concentration.228–230 In these patients, vasopressin antago-
endocytic retrieval of AQP2 from the plasma membrane, nists also have been shown to improve several symptoms of
thus impairing the urinary concentrating ability of the cell. heart failure, such as dyspnea.231
Furthermore, lithium increases the intracellular accumula- Water retention and hyponatremia are observed in pa-
tion of -catenin,215 which can serve as an activator of T-cell tients with hepatic cirrhosis. In these patients, the nonosmotic
factor-dependent transcription. AQP2 downregulation may secretion of vasopressin occurs secondary to splanchnic arte-
be achieved via this transcription mechanism. In addition, rial vasodilatation and relative arterial under lling.225 In cir-
AQP3 expression is also decreased. Moreover, lithium treat- rhotic rats, AQP2 expression was increased and correlated
ment caused a marked reduction in the fraction of the prin- with the volume of ascites.232 In patients with hyponatremic
cipal cells in the collecting duct with a parallel increase in the cirrhosis, V2 receptor antagonists are effective at inducing
population of intercalated cells.216 This restructuring of the free water diuresis and raising plasma sodium levels.229,233
collecting duct, together with the downregulation of collect- During pregnancy, arterial under lling secondary to
ing duct AQPs, may be important in lithium-induced NDI. systemic arterial vasodilatation with nonosmotic vasopressin
Hypokalemia and hypercalcemia cause downregulation secretion and upregulation of AQP2 is observed.75,234
of AQP2 expression, which results in a vasopressin-resistant The administration of a V2 receptor antagonist increases
urinary concentrating defect. With regard to hypercalce- electrolyte-free water excretion in pregnant rats.234
mia, in addition to AQP2, the expression levels of AQP1 The syndrome of inappropriate antidiuretic hormone
and AQP3 are also decreased.217 In addition, hypercalcemia secretion (SIADH) is a condition in which plasma vasopres-
reduces the expression of NKCC2 and ROMK,218 resulting sin levels are not appropriately suppressed despite hypo-
in sodium absorption defects in the thick ascending limb, osmolality. SIADH is the predominant cause of euvolemic
which would affect the countercurrent multiplication. hyponatremia and is a commonly encountered disorder.235
Ureteral obstruction decreases AQP2 expression and SIADH occurs frequently in association with vascular, in-
impairs urine concentrating capacity.219 In addition to fectious, or neoplastic abnormalities in the lung or central
AQP2, AQP1, AQP3, and AQP4 are also decreased by ure- nervous system. In patients with SIADH, a V2 receptor an-
teral obstruction.219 tagonist is shown to be effective in increasing urine volume
A urinary concentrating defect is also observed in pa- and plasma sodium levels.236 However, the long-term effects
tients with nephrotic syndrome.220,221 AQP2 expression of its administration are limited in rats with SIADH.237 Al-
is decreased in nephrotic rats.220,222 However, changes in though the AQP2 protein expression is reduced shortly after
AQP2 expression levels have not yet been con rmed in pa- the administration of the V2 receptor antagonist to rats with
tients with nephrotic syndrome. SIADH, it increases again in parallel with the decline of the
Mouri et al.223 reported that AQP2 translocation to the therapeutic effects.
apical membrane is inhibited by metabolic acidosis, a mech- The nephrogenic syndrome of inappropriate antidiuresis
anism that might be responsible for the diuresis in patients (NSIAD) is a rare hereditary disease caused by gain of func-
with chronic renal failure. tion mutations of the V2 receptor gene. Two causative muta-
tions at the arginine residue at the position 137, Arg137Cys
Water Retention by Urine Diluting Defects and Arg137Leu, have been reported.238 In patients with this
AQP2 has a critical role in the pathophysiology of many disease, endogenous vasopressin is completely suppressed,
diseases associated with water balance disorders. The best whereas antidiuresis persists due to the constitutive activa-
known example is congestive heart failure (CHF). Water re- tion of the receptor, showing the same clinical pictures as
tention and hyponatremia are common and clinically im- SIADH.238 The clinical presentation of the disease starts from
portant complications of CHF. Plasma vasopressin levels childhood, but the same mutations seem to explain some
are suppressed by hyponatremia in healthy individuals; sporadic episodes of euvolemic hyponatremia in adults. To
however, these levels are not suppressed in patients with differentiate NSIAD from SIADH, it may be helpful to observe
hyponatremia who have CHF.224,225 In CHF, effective blood the responses to V2 receptor antagonists; the former does not
volume and arterial lling are decreased, which is sensed by respond to the antagonist, whereas the latter does.239
aortic and carotid baroreceptors, thus resulting in stimula- The urinary AQP2 excretion level is associated with vaso-
tion of vasopressin secretion. Upregulation of AQP2 expres- pressin activity in the kidney and is, therefore, a clinically use-
sion and increased AQP2 traf cking to the apical membrane ful biomarker.240 AQP2 is excreted into the urine through the
of principal cells of the collecting duct have been shown in secretion of exosomes originating from the internal vesicles of
154
15 4 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

multivesicular bodies (Fig. 4.5).241 During this process, the 18. Y


21. Knepper MA, et A,
ang B, Gillespie al.Carlson
Urea transport
EJ, et al.inNeonatal
isolatedmortality
thick ascending limbs and
in an aquaporin-2
collectingmouse
knock-in ducts modelrom rats. Am J Physiol.
of recessive 1983;245:F634.
nephrogenic diabetes insipidus. J Biol Chem.
outer membrane of multivesicular bodies fuses with the api- 19. Kim GH, Masilamani S, Turner R, et al. The thiazide-sensitive Na-
2001;276:2775.
cal plasma membrane. Urinary AQP2 excretion is increased Cl22.cotransporter
Rojek A, Füchtbauer is an aldosterone-induced
EM, Kwon TH, etprotein. al. Severe Proc Natl Acad
urinary Sci USA.
concentrating
by dehydration or vasopressin and is decreased by hydration. 1998;95:14552.
defect in renal collecting duct-selective AQP2 conditional-knockout mice. Proc
http://www.ncbi.nlm.nih.gov/pubmed/9826738
Natl Acad Sci USA. 2006;103:6037.
Urinary AQP2 excretion is also increased in patients with 20. Y
23. Kishore
amamoto BK,T,Mandon
Sasaki S,B, Fushimi
Oza NB, K, et al.
et Rat renal arcadeincreases
al. Vasopressin segment expresses
AQP-CD
CHF and hepatic cirrhosis and in pregnant women.242–244 vasopressin-regulated
water channel in apicalwater membranechannel and vasopressin
of collecting duct cells V2 inreceptor.
BrattleboroJ Clin Invest.
rats. Am
In patients with CHF, the administration of a V2 receptor J1996;97:2763.
Physiol. 1995;268:C1546.
21. Sabolić
24. Yang B,I,Gillespie
Katsura A, T, Carlson
Verbavatz EJ,JM,
et al. Neonatal
et al. The AQP2 mortality
waterinchannel:
an aquaporin-2
effect of
antagonist produced a signi cant increase in urine ow and knock-in mouse
vasopressin modelmicrotubule
treatment, o recessive disruption,
nephrogenic anddiabetes insipidus.
distribution J Biol Chem.
in neonatal rats.
solute-free water excretion, and was accompanied with a dra- J2001;276:2775.
Membr Biol. 1995;143:165.
matic decrease in urinary AQP2 excretion.242 An increased 22. Nielsen
25. Rojek A,S,Füchtbauer
Chou CL,EM, Kwon D,
Marples TH,etet al.al. Vasopressin
Severe urinary concentrating
increases water
de ect in renal
permeability of collecting
kidney collectingduct-selective
duct by AQP2 conditional-knockout
inducing mice. Proc
translocation of aquaporin-
urinary excretion of AQP2 is also found in SIADH and NSI- Natlwater
CD Acad channels
Sci USA. 2006;103:6037.
to plasma membrane. Proc Natl Acad Sci USA. 1995;92:1013.
AD.245,246 Thus, the urinary excretion of AQP2 is a useful 23. Nielsen
26. Yamamoto T, SasakiJ,S,Marples
S, Frøkiaer FushimiD,K,etetal.al.Aquaporins
Vasopressininincreases
the kidney: AQP-CDfrom
marker of the antidiuretic activity of the collecting duct. water channel
molecules in apical membrane
to medicine. Physiol Rev. o2002;82:205.
collecting duct cells in Brattleboro rats. Am
J 27.
Physiol. 1995;268:C1546.
Jamison RL, Buerkert J, Lacy F. A micropuncture study of collect-
ing24.tubule
Sabolić I, Katsura
function in T, Verbavatz
rats JM, et al. The
with hereditary AQP2insipidus.
diabetes water channel:J Cline Invest.
ect o
vasopressin treatment, microtubule disruption, and distribution in neonatal rats.
1971;50:2444.
REFERENCES J 28.
Membr Biol. 1995;143:165.
Canessa CM, Horisberger JD, Schild L, et al. Expression cloning of the
25. Nielsen
epithelial sodium S, Chou
channel. CL,Kidney
Marples Int. D, et al. Vasopressin increases water per-
1995;48:950.
1. Preston
Preston GM,GM, Carroll
CarrollTP TP,, Guggino
Guggino WB, WB, et et al.
al. Appearance
Appearance of o water
water channels
channels meability
29. Lof ongkidney collecting D,
J, Lof ng-Cueni duct by inducing
Macher A, et al. translocation
Localization of o epithelial
aquaporin-CD sodi-
in Xenopus oocytes expressing red cell CHIP28 protein. Science. 1992;256:385. water channels
um channel andtoaquaporin-2
plasma membrane. in rabbitProckidneyNatlcortex.
Acad Sci Am USA. 1995;92:1013.
J Physiol Renal Physiol.
2. Agre P, Sasaki S, Chrispeels MJ. Aquaporins: a family of water channel
http://www.ncbi.nlm.nih.gov/pubmed/1373524 26. Nielsen S, Frøkiaer J, Marples D, et al. Aquaporins in the kidney: rom
2000;278:F530.
proteins.
2. Agre AmP,J Sasaki
Physiol.S,1993;265:F461.
Chrispeels MJ. Aquaporins: a amily o water channel molecules to medicine. Physiol Rev. 2002;82:205.
30. Stockand JD. Vasopressin regulation of renal sodium excretion. Kidney Int.
3. Fushimi
proteins. K, Uchida
Am J Physiol. S, Hara Y, et al. Cloning and expression of apical
1993;265:F461. http://www.ncbi.nlm.nih.gov/pubmed/11773613
2010;78:849.
membrane
3. Fushimi water K,channel
Uchida S,of Hara
rat kidney
Y, et al. collecting
Cloningtubule. Nature. 1993;361:549.
and expression o apical mem- 27. Sands
JamisonJM, RL, Buerkert
31. Knepper J, MA. Lacy FUrea
. A micropuncture
permeabilitystudy o collecting tubule
of mammalian inner
4. Sabolic
brane I, Valenti
water channel o G,
ratVerbavatz JM, et al.tubule.
kidney collecting Localization
Nature.of1993;361:549.
the CHIP28 water unction incollecting
rats with hereditary
medullary duct systemdiabetes insipidus.
and papillary J Clinepithelium.
surface Invest. 1971;50:2444.
J Clin Invest.
channel in rat kidney. Am J Physiol. 1992;263:C1225.
http://www.ncbi.nlm.nih.gov/pubmed/8429910 http://www.ncbi.nlm.nih.gov/pubmed/5096527
1987;79:138.
5.
4. Nielsen
Sabolic I, S, Valenti
Smith G,BL,Verbavatz
Christensen JM, et EI,al.etLocalization
al. CHIP28 owater channelswater
the CHIP28 are 28. Fenton
CanessaRA, CM,Chou Horisberger JD, Schild L, et al. Expression cloning o the
32. CL, Stewart GS, et al. Urinary concentrating defect in
localized
channel ininrat constitutively
kidney. Am water-permeable
J Physiol. 1992;263:C1225. segments of the nephron. J Cell Biol. epithelial
mice withsodium
selectivechannel.
deletionKidney Int. 1995;48:950.
of phloretin-sensitive urea transporters in the renal
1993;120:371.
5. Nielsen S, Smith BL, Christensen EI, et al. CHIP28 water channels are http://www.ncbi.nlm.nih.gov/pubmed/8569104
collecting duct. Proc Natl Acad Sci USA. 2004;101:7469.
6. Nielsen
localized S, Pallone T, water-permeable
in constitutively Smith BL, et al. Aquaporin-1
segments o water channelsJ Cell
the nephron. in short
Biol. 29. Pallone
Lo ng J, LoCharacterization
ng-Cueni D, Macher
33. TL. of theA,urea et al.transporter
Localization in oouter
epithelial sodi-
medullary
and long loop descending thin limbs and in descending vasa recta in rat kidney.
1993;120:371. um channelvasa and recta.
aquaporin-2 in rabbit kidney cortex. Am J Physiol Renal Physiol.
descending Am J Physiol. 1994;267:R260.
Am J Physiol. 1995;268:F1023.
http://www.ncbi.nlm.nih.gov/pubmed/7678419 2000;278:F530.
34. Knepper MA, Danielson RA, Saidel GM, et al. Quantitative analysis of renal
7.
6. Maeda
NielsenY,S,Smith BL,T,Agre
Pallone SmithP, etBL,
al.etQuanti cation of Aquaporin-CHIP
al. Aquaporin-1 water channels inwater short 30. Stockand JD. in Vasopressin regulation o renal sodium excretion. Kidney Int.
medullary anatomy rats and rabbits. Kidney Int. 1977;12:313.
channel
and longprotein in microdissected
loop descending thin limbs renaland tubules by uorescence-based
in descending vasa recta in rat ELISA.
kidney. 2010;78:849.
35. Knepper MA, Saidel GM, Hascall VC, et al. Concentration of solutes in the
JAmClinJ Physiol.
Invest. 1995;95:422.
1995;268:F1023. http://www.ncbi.nlm.nih.gov/pubmed/20736986
renal inner medulla: interstitial hyaluronan as a mechano-osmotic transducer.
8.
7. Chou
MaedaCL, Nielsen
Y, Smith BL,S,AgreKnepper
P, et al.MA.Quanti Structural-functional
cation o Aquaporin-CHIP correlation in
water 31.J Physiol
Sands RenalJM, Knepper MA. Urea permeability o mammalian inner med-
Am Physiol. 2003;284:F433.
chinchilla
channel proteinlong loop of Henle thinrenal
in microdissected limbs: a novel
tubules by papillary subsegment.
f uorescence-based Am JJ
ELISA. ullary collecting
36. Dwyer TM, duct system and B.papillary
Schmidt-Nielsen The renal surpelvis:
ace epithelium.
machinery Jthat Clinconcen-
Invest.
Physiol. 1993;265:F863.
Clin Invest. 1995;95:422. 1987;79:138.
trates urine in the papilla. News Physiol Sci. 2003;18:1.
9. Pannabecker TL, Abbott DE, Dantzler WH. Three-dimensional functional
http://www.ncbi.nlm.nih.gov/pubmed/7529263 http://www.ncbi.nlm.nih.gov/pubmed/3793921
37. Reinking LN, Schmidt-Nielsen B. Peristaltic ow of urine in the renal pap-
reconstruction
8. Chou CL, of inner
Nielsenmedullary
S, Knepper thin MA.limbsStructural-
of Henle’s unctional
loop. Am Jcorrelation
Physiol Renal in 32. collecting
Fenton RA, Chou CL, Stewart GS, Int.
et al. Urinary concentrating de ect in
illary ducts of hamsters. Kidney 1981;20:55.
Physiol.
chinchilla 2004;286:F38.
long loop o Henle thin limbs: a novel papillary subsegment. Am J mice
38. with
Kuhnselective
W, Ryffel deletion o phloretin-sensitive
K. Herstellung konzentrierter urea transporters
Losungen in the renal
aus verdunnten
10. Pannabecker
Physiol. 1993;265:F863. TL, Dantzler WH, Layton HE, et al. Role of three-dimen- collecting duct.Membranwirkung.
Proc Natl Acad Sci Ein USA.Modellversuch
2004;101:7469.
durch blosse zur Funktion der Niere.
sional architecture inTL,
9. Pannabecker theAbbott
urine DE,concentrating
Dantzler WH. mechanism of the rat renal
Three-dimensional inner
unctional 33. Pallone
Hoppe Seylers ZTL. Characterization
Physiol Chemie. 1942;276:145.o the urea transporter in outer medullary
medulla. Am J Physiol
reconstruction o inner Renal Physiol.thin
medullary 2008;295:F1271.
limbs o Henle’s loop. Am J Physiol Renal descending
39. Kokkovasa JP, recta.
RectorAm FCJ Physiol. 1994;267:R260.
Jr. Countercurrent multiplication system without
11. Ma2004;286:F38.
Physiol. T, Yang B, Gillespie A, Carlson EJ, et al. Severely impaired urinary 34. transport
Knepper MA, Danielson RA,Kidney
Saidel Int.
GM,1972;2:214.
et al. Quantitative analysis o renal
active in inner medulla.
concentrating
10. Pannabecker abilityTL,
in transgenic
Dantzler WH, miceLaytonlackingHE, aquaporin-1
et al. Rolewater channels. J
o three-dimen- medullary anatomy
40. Stephenson JL.inConcentration
rats and rabbits. KidneyinInt.
of urine 1977;12:313.
a central core model of the renal
Biol
sionalChem. 1998;273:4296.
architecture in the urine concentrating mechanism o the rat renal inner http://www.ncbi.nlm.nih.gov/pubmed/604620
counter ow system. Kidney Int. 1972;2:85.
12. Ishibashi
medulla. K, Kuwahara
Am J Physiol M, Gu 2008;295:F1271.
Renal Physiol. Y, Kageyama Y, et al. Cloning and functional 35. Imai
Knepper MA, Saidel GM, Hascall VC,urea,
et al.and
Concentration o solutes
41. M, Kokko JP. Sodium chloride, water transport in theinthinthe
expression
11. Ma T,of Y aang
newB,water channel
Gillespie A, abundantly
Carlson EJ, expressed in theimpaired
et al. Severely testis permeable
urinary renal innerlimb medulla: interstitial hyaluronan as gradients
a mechano-osmotic transducer.
ascending of Henle. Generation of osmotic by passive diffusion of
to water, glycerol,
concentrating andinurea.
ability J Biol Chem.
transgenic mice1997;272:20782.
lacking aquaporin-1 water channels. J Am J Physiol
solutes. J ClinRenal
Invest. Physiol. 2003;284:F433.
1974;53:393.
13.Chem.
Biol Sohara E, Rai T, Miyazaki J, et al. Defective water and glycerol transport
1998;273:4296. 36. Matsumura
Dwyer TM, YSchmidt-Nielsen B.YThe renal pelvis: machinery that concen-
42. , Uchida S, Kondo , et al. Overt nephrogenic diabetes insipi-
in12.
the Ishibashi
proximalK,tubules
Kuwahara of AQP7
M, Guknockout
Y, Kageyama mice.Y, etAm
al. JCloning
Physiol and
Renalunctional
Physiol. trates
dus inurine
mice in the papilla.
lacking the CLC-K1News Physiol
chloride Sci.channel.
2003;18:1. Nat Genet. 1999;21:95.
2005;289:F1195.
expression o a new water channel abundantly expressed in the testis permeable 37. Akizuki
ReinkingN,LN, Schmidt-Nielsen
43. Uchida S, Sasaki S, etB.al.Peristaltic
Impairedf soluteow o urine in the renal
accumulation pap-
in inner
to14. Wade
water, JB, Leeand
glycerol, AJ,urea.
Liu JJ,Biol
et al.
Chem.UT-A2: a 55-kDa urea transporter in thin
1997;272:20782. illary collecting ducts mice o hamsters. Kidney Int. 1981;20:55.
medulla of Clcnk1-/- kidney. Am J Physiol Renal Physiol. 2001;280:F79.
descending
13. Sohara limb whose
E, Rai abundance
T, Miyazaki is regulated
J, et al. De ective by vasopressin.
water and glycerolAm J Physiol Re-
transport http://www.ncbi.nlm.nih.gov/pubmed/7300113
44. Yang B, Bankir L, Gillespie A, et al. Urea-selective concentrating defect in
nal Physiol.
in the 2000;278:F52.
proximal tubules o AQP7 knockout mice. Am J Physiol Renal Physiol. 38. Kuhnmice W, lacking
Ry el K.urea Herstellung konzentrierter Losungen aus verdunnten
transgenic transporter UT-B. J Biol Chem. 2002;277:10633.
15. Uchida S, Sasaki S, Furukawa T, et al. Molecular cloning of a chloride
2005;289:F1195. durch blosse
45. Hai MA, Membranwirkung.
Thomas S. The time-course Ein Modellversuch
of changes inzur Funktion
renal der Niere.
tissue composition
channel
14. Wade that JB,
is regulated
Lee AJ, Liuby dehydration
J, et al. UT-A2: anda expressed
55-kDa urea predominantly
transporter ininthin
kidney
de- Hoppe Seylers Z Physiol Chemie. 1942;276:145.
during lysine vasopressin infusion in the rat. P ugers Arch. 1969;310:297.
medulla.
scending Jlimb Biol Chem.
whose1994;269:19192.
abundance is regulated by vasopressin. Am J Physiol Renal 39. Y Kokko
46. ang B, JP , Rector
Bankir L. FC
Urea Jr.and
Countercurrent multiplication
urine concentrating ability:system without
new insights ac-
from
16. Uchida
Physiol. S, Sasaki S, Nitta K, et al. Localization and functional characteriza-
2000;278:F52. tive transport in Aminner medulla. Kidney Int. 2005;288:F881.
1972;2:214.
studies in mice. J Physiol Renal Physiol.
tion of rat kidney-speci c chloride channel, ClC-K1. J Clin Invest. 1995;95:104.
http://www.ncbi.nlm.nih.gov/pubmed/10644655 http://www.ncbi.nlm.nih.gov/pubmed/4657922
47. Sands JM, Layton HE. The physiology of urinary concentration: an update.
17.
15. Gamba
Uchida G. Molecular
S, Sasaki physiologyT, and
S, Furukawa et al.pathophysiology
Molecular cloning of electroneutral
o a chloride 40. Nephrol.
Stephenson JL. Concentration o urine in a central core model o the renal
Semin 2009;29:178.
cation-chloride cotransporters.
channel that is regulated Physiol Rev.
by dehydration and2005;85:423.
expressed predominantly in kidney counterf ow system.
48. Kondo Y, ImaiKidney Int. 1972;2:85.
M. Effects of glutaraldehyde xation on renal tubular
18. Knepper
medulla. MA, et1994;269:19192.
J Biol Chem. al. Urea transport in isolated thick ascending limbs and http://www.ncbi.nlm.nih.gov/pubmed/4671532
function. I. Preservation of vasopressin-stimulated water and urea pathways in
collecting ducts from rats. Am J Physiol. 1983;245:F634.
http://www.ncbi.nlm.nih.gov/pubmed/8034678
rat41. Imai M,
papillary Kokko JP
collecting . Sodium
duct. P ugerschloride, urea, and water transport in the thin
Arch. 1987;408:479.
19.
16. KimUchida GH, Masilamani
S, Sasaki S, NittaS, K,Turner R, et al. The
et al. Localization andthiazide-sensitive Na-Cl
unctional characteriza- ascending limb
49. Sands JM,o Nonoguchi
Henle. Generation H, Knepper o osmotic gradients byeffects
MA. Vasopressin passiveondiurea usion o
and
cotransporter is an aldosterone-induced
tion o rat kidney-speci c chloride channel, protein. ClC-K1. JProcClin Natl
Invest.Acad Sci USA.
1995;95:104. solutes. J Clin Invest. 1974;53:393.
H2O transport in inner medullary collecting duct subsegments. Am J Physiol.
1998;95:14552.
http://www.ncbi.nlm.nih.gov/pubmed/7814604 http://www.ncbi.nlm.nih.gov/pubmed/11344552
1987;253:F823.
20.
17. Kishore
Gamba BK, Mandon B,physiology
G. Molecular Oza NB, etand al. Rat renal arcade segment
pathophysiology expresses
o electroneutral 42. Fenton
Matsumura Y, Uchidarole S, Kondo Y, et al. Overt nephrogenic diabetes insipi-
50. RA. Essential of vasopressin-regulated urea transport processes
vasopressin-regulated water channel
cation-chloride cotransporters. Physioland Rev.vasopressin
2005;85:423. V2 receptor. J Clin Invest. dusthein mammalian
mice lackingkidney. the CLC-K1 chloride channel. Nat Genet. 1999;21:95.
in P ugers Arch. 2009;458:169.
1996;97:2763.
http://www.ncbi.nlm.nih.gov/pubmed/15788703 http://www.ncbi.nlm.nih.gov/pubmed/9916798
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1155
55

43. Noda
51. Akizuki Y, N, Uchida
Sohara E, S,
OhtaSasaki
E, etS, al.
et al. Impaired solute
Aquaporins in kidneyaccumulation in inner
pathophysiology. 69. Stricker EM,M,
80. Birnbaumer Sved AF. Controls
Seibold A, Gilbert o vasopressin secretion
S, et al. Molecular and thirst:
cloning similari-
of the receptor
medulla
Nat o Clcnk1-/-
Rev Nephrol. mice kidney. Am J Physiol Renal Physiol. 2001;280:F79.
2010;6:168. ties and dissimilarities
for human antidiureticin signals. Physiol
hormone. Nature.Behav. 2002;77:731.
1992;357:333.
44. Brown
52. Yang B,D,Bankir
BretonL,S,Gillespie
Ausiello A, DAetetal.
al.Urea-selective
Sensing, signaling concentrating
and sorting deevents
ect in http://www.ncbi.nlm.nih.gov/pubmed/12527027
81. Nielsen S, DiGiovanni SR, Christensen EI, et al. Cellular and subcellular
transgenic
in mice lacking
kidney epithelial urea transporter
cell physiology. Traf c.UT-B. J Biol Chem. 2002;277:10633.
2009;10:275. 70. Schrier RW. Pathogenesis
immunolocalization o sodium and
of vasopressin-regulated water
water retention
channel in kidney.
in rat high-output
Proc
45. Hoffert
53. Hai MA,JD,ThomasChou CL, S. The time-course
Knepper o changes in
MA. Aquaporin-2 in the
renal tissue era.
“-omics” composi-
J Biol and
Natl low-output
Acad Sci USA. cardiac ailure, nephrotic syndrome, cirrhosis, and pregnancy
1993;90:11663.
tion during
Chem. lysine vasopressin in usion in the rat. P ugers Arch. 1969;310:297.
2009;284:14683. (1).
82.NFushimi
Engl J Med. K, 1988;319:1065.
Sasaki S, Yamamoto T, et al. Functional characterization and
46. Boone
54. Yang B,M,BankirDeen L.PM. Urea and urineand
Physiology concentrating
pathophysiologyability:ofnew the insights rom
vasopressin- http://www.ncbi.nlm.nih.gov/pubmed/3050518
cell immunolocalization of AQP-CD water channel in kidney collecting duct.
studies in renal
regulated mice.water
Am J Physiol Renal Physiol.
reabsorption. P ugers2005;288:F881.
Arch. 2008;456:1005. Am71.J Physiol.
Schrier1994;267:F573.
RW. Pathogenesis o sodium and water retention in high-output
47. Noda
55. Sands Y JM, LaytonS.HE.
, Sasaki The physiology
Regulation o urinary
of aquaporin-2 concentration:
traf cking and its an binding
update. and83.low-output
Horster MFcardiac
, Zink H.ailure, nephrotic
Functional syndrome,ofcirrhosis,
differentiation and pregnancy
the medullary collecting
Semin Nephrol.
protein complex. 2009;29:178.
Biochim Biophys Acta. 2006;1758:1117–1125. (2). N Engl
tubule: J Med. of
in uence 1988;319:1127.
vasopressin. Kidney Int. 1982;22:360.
http://www.ncbi.nlm.nih.gov/pubmed/19523568
56. Nielsen S, Maunsbach AB, Ecelbarger CA, et al. Ultrastructual localiza- 72. Rowe JW
84. Schenk AD,Shelton
WertenRL, PJ,Helderman
Scheuring JH, S, etetal.al.The
Inf uence o the emetic
4.5 A structure ref ex
of human
48.ofKondo
tion Na-K-2Cl Y, Imai M. E ects in
cotransporter o thick
glutaraldehyde
ascending limb xationandonmacula
renal tubular
densa ofunc-rat on vasopressin
AQP2. J Mol Biol.release in man. Kidney Int. 1979;16:729.
2005;350:278.
tion. I. Am
kidney. Preservation o vasopressin-stimulated water and urea pathways in rat
J Physiol. 1998;275:F885. http://www.ncbi.nlm.nih.gov/pubmed/548611
85. Kamsteeg EJ, Heijnen I, van Os CH, et al. The subcellular localization of
papillary collecting D,
57. Biemesderfer duct. P ugers Arch.
Rutherford 1987;408:479.
PA, Nagy T, et al. Monoclonal antibodies for an73. Prager-Khoutorsky
aquaporin-2 tetramer dependsM, Bourque on the CW.stoichiometry
Osmosensation in vasopressin neu-
of phosphorylated and
http://www.ncbi.nlm.nih.gov/pubmed/3110736
high-resolution localization of NHE3 in adult and neonatal rat kidney. Am J rons: changing actin monomers.
nonphosphorylated density to optimize
J Cell Biol.unction. Trends Neurosci. 2010;33:76.
2000;151:919.
49. Sands
Physiol. JM, Nonoguchi H, Knepper MA. Vasopressin e ects on urea and
1997;273:F289. http://www.ncbi.nlm.nih.gov/pubmed/19963290
86. Fushimi K, Sasaki S, Marumo F. Phosphorylation of serine 256 is required
H2O transport RL,
58. Jamison in inner
Lacy medullary
FB. Evidence collecting duct subsegments.
for urinary dilution by the Amcollecting
J Physiol. 74.cAMP-dependent
for Lindheimer MD, Davisonexocytosis
regulatory JM. Osmoregulation,
of the aquaporin-2 the secretion o ar-
water channel.
1987;253:F823.
tubule. Am J Physiol. 1972;223:898. ginine vasopressin
J Biol Chem. and its metabolism during pregnancy. Eur J Endocrinol.
1997;272:14800.
50. Brownstein
59. Fenton RA. MJ, Essential
RussellroleJT,
o Gainer
vasopressin-regulated urea transport
H. Synthesis, transport processes
and release of 1995;132:133.
87. Katsura T, Gustafson CE, Ausiello DA, et al. Protein kinase A phosphoryla-
in the mammalian
posterior kidney. P ugers
pituitary hormones. Arch.
Science. 2009;458:169.
1980;207:373. http://www.ncbi.nlm.nih.gov/pubmed/7858729
tion is involved in regulated exocytosis of aquaporin-2 in transfected LLC-PK1
http://www.ncbi.nlm.nih.gov/pubmed/19011892
60. Sharif-Naeini R, Ciura S, Zhang Z, et al. Contribution of TRPV channels 75. Am
cells. Chapman
J Physiol.AB, Abraham WT, Zamudio S, et al. Temporal relationships be-
1997;272:F817.
to51.osmosensory
Noda Y, Sohara E, Ohta E,thirst,
transduction, et al. Aquaporins
and vasopressin in kidney pathophysiology.
release. Kidney Int. tween hormonal and
88. Klussmann hemodynamic
E, Maric K, Wiesner changesB, etinal. early human
Protein pregnancy.
kinase Kidney
A anchoring
Nat Rev Nephrol. 2010;6:168.
2008;73:811. Int. 1998;54:2056.
proteins are required for vasopressin-mediated translocation of aquaporin-2 into
http://www.ncbi.nlm.nih.gov/pubmed/20101255
61. Bourque CW. Central mechanisms of osmosensation and systemic 76.membranes
cell Kalelioglu ofI, renal
Kubatprincipal
Uzum A, Yildirim
cells. A, et al.
J Biol Chem. Transient gestational dia-
1999;274:4934.
52. Brown D, Breton
osmoregulation. Nat RevS,Neurosci.
Ausiello 2008;9:519.
DA et al. Sensing, signaling and sorting events betes
89. insipidus
Henn V, diagnosed
Edemir B, inStefan successive
E, et pregnancies:
al. Identi cation reviewofo apathophysiology,
novel A-kinase
in62.
kidney
Sharif epithelial
Naeini R, cellWitty
physiology. Tra f c.P, 2009;10:275.
MF, Séguéla Bourque CW. An N-terminal variant diagnosis,
anchoring treatment,
protein 18and management
isoform and evidence o delivery.
for itsPituitary.
role in 2007;10:87.
the vasopressin-in-
http://www.ncbi.nlm.nih.gov/pubmed/19170982
of Trpv1 channel is required for osmosensory transduction. Nat Neurosci. http://www.ncbi.nlm.nih.gov/pubmed/17308961
duced aquaporin-2 shuttle in renal principal cells. J Biol Chem. 2004;279:26654.
53. Ho ert JD, Chou CL, Knepper MA. Aquaporin-2 in the “-omics” era. J Biol
2006;9:93. 77. Yun J, Schöneberg
90. Okutsu R, Rai T, Kikuchi T, Liu A,J, etet al.
al. Generation and phenotype
AKAP220 colocalizes o miceinhar-
with AQP2 the
Chem. 2009;284:14683.
63. Taylor AC, McCarthy JJ, Stocker SD. Mice lacking the transient receptor boring a nonsense
inner medullary mutation
collecting in the
ducts. V2 vasopressin
Kidney receptor gene. J Clin Invest.
Int. 2008;74:1429.
http://www.ncbi.nlm.nih.gov/pubmed/19193633
vanilloid potential 1 channel display normal thirst responses and cen- 2000;106:1361.
91. Procino G. Carmosino M, Marin O, et al. Ser-256 phosphorylation
54.Fos
tral Boone M, Deen
activation to PM. Physiology and
hypernatremia. Am Jpathophysiology
Physiol Regul Integr o theComp vasopressin-
Physiol. http://www.ncbi.nlm.nih.gov/pubmed/11104789
dynamics of aquaporin 2 during maturation from the endoplasmic reticulum to
regulated renal water reabsorption. Pf ugers Arch. 2008;456:1005.
2008;294:R1285. 78.vesicular
the Yang B,compartment
Zhao D, Qian L, et cells.
in renal al. MouseFASEBmodel o inducible nephrogenic
J. 2003;17:1886.
55. Robertson
64. Noda Y, Sasaki GL. TheS. Regulation
regulationo of aquaporin-2
vasopressintrafunction
cking and its binding
in health and diabetes
92. vaninsipidus
Balkom produced
BW, Savelkoul by f oxed aquaporin-2
PJ, Markovich D, gene
et al.deletion.
The roleAm J Physiol
of putative
protein complex.
disease. Rec Prog Horm BiochimRes.Biophys Acta. 2006;1758:1117–1125.
1976;33:333. Renal Physiol. 2006;291:F465.
phosphorylation sites in the targeting and shuttling of the aquaporin-2 water
56. Baylis
65. Nielsen PH.S, Osmoregulation
Maunsbach AB, Ecelbarger
and controlCA, et al. Ultrastructual
of vasopressin secretionlocaliza tion
in healthy 79. Lolait
channel. SJ,Chem.
J Biol O’Carroll AM, McBride OW, et al. Cloning and characterization
2002;277:41473.
o Na-K-2Cl
humans. Am cotransporter in thick ascending limb and macula densa o rat kid-
J Physiol. 1987;253:R671. o 93.
a vasopressin
Bouley R, V2 receptor
Breton S, SunandT,possible link tooxide
et al. Nitric nephrogenic diabetes
and atrial insipidus.
natriuretic fac-
ney.
66.Am J Physiol.
Brunton PJ,1998;275:F885.
Arunachalam S, Russel JA. Control of neurohypophysi- Nature. 1992;357:336.
tor stimulate cGMP-dependent membrane insertion of aquaporin 2 in renal
al57. Biemesder
hormone er D, Ruther
secretion, blood ord PA, Nagy
osmolality andT, volume
et al. Monoclonal
in pregnancy. antibodies
J Physiol or http://www.ncbi.nlm.nih.gov/pubmed/1534150
epithelial cells. J Clin Invest. 2000;106:1115.
high-resolution
Pharmacol. localization8):27.
2008;59(Suppl o NHE3 in adult and neonatal rat kidney. Am J Phys- 80. Birnbaumer
94. Bouley M, Seibold A,N,
R, Pastor-Soler Gilbert
Cohen S, etO, al. Molecular cloning o the
et al. Stimulation of recep-
AQP2
iol.
67.1997;273:F289.
Sladek CD, Song Z. Regulation of vasopressin release by co-released tor or human
membrane antidiuretic
insertion in renal hormone.
epithelialNature.
cells 1992;357:333.
in vitro and in vivo by the cGMP
58. Jamison RL, Lacy
neurotransmitters: FB. Evidence
mechanisms or urinary
of purinergic anddilution
adrenergicby the collectingProg
synergism. tu- http://www.ncbi.nlm.nih.gov/pubmed/1534149
phosphodiesterase inhibitor sildena l citrate (Viagra). Am J Physiol Renal Physiol.
bule. Am
Brain Res.J2008;170:93.
Physiol. 1972;223:898. 81. Nielsen S, DiGiovanni SR, Christensen EI, et al. Cellular and subcellular
2005:288:F1103.
http://www.ncbi.nlm.nih.gov/pubmed/5075165
68. Quillen EW Jr, Skelton MM, Rubin J, et al. Osmotic control of vasopressin immunolocalization
95. Fenton RA, Moeller o vasopressin-regulated
HB, Hoffert JD, et al.water Acutechannel in rat
regulation kidney. Proc
of aquaporin-2
59. chronically
with Brownstein MJ, Russell
altered JT, Gainer
volume statesH.inSynthesis,
anephrictransport
dogs. Am and Jrelease
Physiol.o Natl Acad Sci USA. at
phosphorylation 1993;90:11663.
Ser-264 by vasopressin. Proc Natl Acad Sci USA.
posterior pituitary hormones. Science. 1980;207:373.
1984;247:E355. 82. Fushimi K, Sasaki S, Yamamoto T, et al. Functional characterization and
2008;105:3134.
http://www.ncbi.nlm.nih.gov/pubmed/6153132
69. Stricker EM, Sved AF. Controls of vasopressin secretion and thirst: cell
96.immunolocalization
Hoffert JD, Nielseno J,AQP-CD Yu MJ, water channel in of
et al. Dynamics kidney collecting
aquaporin-2 duct. Am
serine-261
60. Shariand
similarities -Naeini R, Ciura S,inZhang
dissimilarities Z, Physiol
signals. et al. Contribution o TRPV channels to
Behav. 2002;77:731. Jphosphorylation
Physiol. 1994;267:F573.
in response to short-term vasopressin treatment in collecting
osmosensory
70. Schrier transduction,
RW. Pathogenesis thirst, of
andsodium
vasopressin release.retention
and water Kidney Int. in2008;73:811.
high-output 83. Am
duct. Horster MF, Renal
J Physiol Zink H. Functional
Physiol. di erentiation o the medullary collecting
2007;292:F691.
http://www.ncbi.nlm.nih.gov/pubmed/18200003
and low-output cardiac failure, nephrotic syndrome, cirrhosis, and pregnancy tubule:
97. LuinfHJ, uence o vasopressin.
Matsuzaki T, Bouley Kidney
R, et Int. 1982;22:360.
al. The phosphorylation state of serine
61.N Bourque
(1). Engl J Med. CW. Central mechanisms o osmosensation and systemic os-
1988;319:1065. http://www.ncbi.nlm.nih.gov/pubmed/7176334
256 is dominant over that of serine 261 in the regulation of AQP2 traf cking in
moregulation.
71. Schrier RW. Nat Pathogenesis
Rev Neurosci. 2008;9:519.
of sodium and water retention in high-output 84. epithelial
renal Schenk AD, Werten
cells. PJ, Scheuring
Am J Physiol S, et al.
Renal Physiol. The 4.5 A structure o human
2008;295:F290.
http://www.ncbi.nlm.nih.gov/pubmed/18509340
and low-output cardiac failure, nephrotic syndrome, cirrhosis, and pregnancy AQP2. J Mol Biol.
98. Moeller HB, 2005;350:278.
Knepper MA, Fenton RA. Serine 269 phosphorylated
62.N Shari
(2). Naeini
Engl J Med. R, Witty MF, Séguéla P, Bourque CW An N-terminal vari-
1988;319:1127. http://www.ncbi.nlm.nih.gov/pubmed/15922355
aquaporin-2 is targeted to the apical membrane of collecting duct principal cells.
ant
72.o RoweTrpv1JW, channel
Sheltonis RL,
required
Helderman or osmosensory
JH, et al. In transduction.
uence of the Nat emeticNeurosci.
re ex 85. Kamsteeg
Kidney EJ, Heijnen I, van Os CH, et al. The subcellular localization o
Int. 2009;75:295.
2006;9:93.
on vasopressin release in man. Kidney Int. 1979;16:729. an99.aquaporin-2
Moeller HB, tetramer
MacAulay depends on theMA,
N, Knepper stoichiometry
et al. Role ofomultiple
phosphorylated
phosphoryla-and
http://www.ncbi.nlm.nih.gov/pubmed/16327782
73. Prager-Khoutorsky M, Bourque CW. Osmosensation in vasopressin neurons: nonphosphorylated monomers. Jtail
tion sites in the COOH-terminal CellofBiol. 2000;151:919.
aquaporin-2 for water transport: evidence
63. Taylor
changing actinAC, McCarthy
density JJ, Stocker
to optimize SD. Trends
function. Mice lacking
Neurosci. the2010;33:76.
transient receptor http://www.ncbi.nlm.nih.gov/pubmed/11076974
against channel gating. Am J Physiol Renal Physiol. 2009;295:F649.
vanilloid potential MD,
74. Lindheimer 1 channel
Davisondisplay
JM. normal thirst responses
Osmoregulation, and cen tral
the secretion of Fos acti-
arginine 86. Balasubramanian
100. Fushimi K, SasakiL,S, Sham Marumo JS, FY. ip
Phosphorylation
KP. Calcium signalingo serine in256vasopressin-
is required
vation to hypernatremia.
vasopressin and its metabolism Am J Physiol
duringRegul Integr Comp
pregnancy. Physiol. 2008;294:R1285.
Eur J Endocrinol. 1995;132:133. or cAMP-dependent
induced aquaporin-2 regulatory
traf cking. exocytosis
P ugers Arch. o the aquaporin-2 water channel. J
2008;456:747–754.
64. Chapman
75. Robertson AB, GL. Abraham
The regulation o vasopressin
WT, Zamudio S, et al.unction
Temporalin health and dis-
relationships Biol
101.Chem.
Noda1997;272:14800.
Y, Horikawa S, Katayama Y, et al. Identi cation of a multiprotein
ease. Rechormonal
between Prog Hormand Res.hemodynamic
1976;33:333.changes in early human pregnancy. Kidney http://www.ncbi.nlm.nih.gov/pubmed/9169447
“motor” complex binding to water channel aquaporin-2. Biochem Biophys Res
http://www.ncbi.nlm.nih.gov/pubmed/801194
Int. 1998;54:2056. 87. Katsura
Commun. T, Gusta son CE, Ausiello DA, et al. Protein kinase A phosphoryla-
2005;330:1041–1047.
65. Kalelioglu
76. Baylis PH.I,Osmoregulation
Kubat Uzum A,and control
Yildirim o al.
A, et vasopressin secretion indiabetes
Transient gestational healthy tion
102.isLorenz
involved D, in regulated
Krylov A, Hahmexocytosis
D, et al.o Cyclic
aquaporin-2
AMP isin suftrans ected
cient LLC-PK1
for triggering
humans. Am
insipidus J Physiol. in
diagnosed 1987;253:R671.
successive pregnancies: review of pathophysiology, cells. Am J Physiol.
the exocytic 1997;272:F817.
recruitment of aquaporin-2 in renal epithelial cells. EMBO Rep.
66. Brunton
diagnosis, PJ, Arunachalam
treatment, and management S, Russel JA. Control
of delivery. o neurohypophysi-al
Pituitary. 2007;10:87. hor- 88. Klussmann E, Maric K, Wiesner B, et al. Protein kinase A anchoring pro-
2003;4:88.
mone
77. Y secretion, blood osmolality
un J, Schöneberg T, Liu J,and et volume in pregnancy.
al. Generation J Physiol Pharmacol.
and phenotype of mice teins
103. are required
Valenti or vasopressin-mediated
G, Laera A, Pace G, et al. translocation
Urinary aquaporin o aquaporin-2 into cell
2 and calciuria
2008;59(Suppl
harboring 8):27. mutation in the V2 vasopressin receptor gene. J Clin Invest.
a nonsense membranes
correlate with o renalthe principal
severity cells. J Biol Chem.
of enuresis in 1999;274:4934.
children. J Am Soc Nephrol.
http://www.ncbi.nlm.nih.gov/pubmed/19258663
2000;106:1361. http://www.ncbi.nlm.nih.gov/pubmed/9988736
2000;11:1873.
67. Y
78. Sladek
ang B,CD, Zhao Song
D, Z. Regulation
Qian L, et al. oMouse
vasopressin
modelrelease by co-released
of inducible nephrogenicneu- 89. Valenti
104. Henn VG, , Edemir
Laera A, B, Gouraud
Ste an E, S, et etal. al.
Identi cation o diet
Low-calcium a novel A-kinase an-
in hypercalciuric
rotransmitters:
diabetes insipidusmechanisms
produced obypurinergic and adrenergic
oxed aquaporin-2 synergism.
gene deletion. AmProg Brain
J Physiol choring
enuretic protein
children18restores
iso ormAQP2 and evidence
excretion or anditsimproves
role in theclinical
vasopressin-in-duced
symptoms. Am
Res. 2008;170:93.
Renal Physiol. 2006;291:F465. aquaporin-2 shuttle in renal principal cells. J Biol Chem. 2004;279:26654.
J Physiol. 2002;283:F895.
http://www.ncbi.nlm.nih.gov/pubmed/18655875
79. Lolait SJ, O’Carroll AM, McBride OW, et al. Cloning and characterization 90. Sands
105. OkutsuJM, R, Rai T, Kikuchi
Flores F, KatoA,A,etetal.al. AKAP220 colocalizes with
Vasopressin-elicited AQP2
water andinurea
the
of68. Quillen EW
a vasopressin Jr, Skelton
V2 receptor andMM,
possibleRubinlinkJ,toetnephrogenic
al. Osmoticdiabetescontrolinsipidus.
o vaso- inner medullaryare
permeabilities collecting
altered in ducts.
IMCD Kidney Int. 2008;74:1429.
in hypercalcemic rats. Am J Physiol. 1998;
pressin 1992;357:336.
Nature. with chronically altered volume states in anephric dogs. Am J Physiol. http://www.ncbi.nlm.nih.gov/pubmed/19008911
274:F978.
1984;247:E355.
156
15 6 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

91. Procino
106. Procino G. G,Carmosino
Carmosino M, M,Marin O, et al.G,Ser-256
Tamma et al.phosphorylation
Extracellular calciumdynam- 134. Harazaki M, Kawai
113. Törnroth-Horse eldYS,
, Su L, etY,al.Hedfalk
Wang Specif cK,recruitment o SPA-1
et al. Structural to the im-
mechanism of
ics o aquaporin
antagonizes 2 during maturation
forskolin-induced aquaporinrom the cking
2 traf endoplasmic reticulum
in collecting duct tocells.the munological
plant aquaporin synapse:
gating.involvement o actin-bundling protein actinin. Immunol
Nature. 2006;439:688.
vesicular
Kidney Int.compartment
2004;66:2245. in renal cells. FASEB J. 2003;17:1886. Lett. 2004;92:221.
135. Yamamoto T, Sasaki S, Fushimi K, et al. Localization and expression of
http://www.ncbi.nlm.nih.gov/pubmed/12897058
107. Nejsum LN, Zelenina M, Aperia A, et al. Bidirectional regulation of AQP2 ahttp://www.ncbi.nlm.nih.gov/pubmed/15081616
collecting duct water channel, aquaporin, in hydrated and dehydrated rats.
92.cking
traf van Balkom
and recycling:BW, Savelkoul
involvement PJ, Markovich
of AQP2-S256 D, et al. The role o putative
phosphorylation. Am J 114.Nephrol.
Exp Kuwahara M, Iwai K, Ooeda T, et al. Three amilies with autosomal domi-
1995;3:193.
phosphorylation
Physiol Renal Physiol. sites2005;288:F930.
in the targeting and shuttling o the aquaporin-2 water nant nephrogenic
136. Terris J, Ecelbarger diabetes CA,insipidus
Nielsen S, caused by aquaporin-2
et al. Long-term mutations
regulation of fourinrenal
the
channel.
108. J Biol Chem.
de Seigneux 2002;277:41473.
S, Nielsen J, Olesen ET, et al. Long-term aldosterone treatment C-terminus.inAm
aquaporins J Hum
rats. Am JGenet.
Physiol.2001;69:738.
1996;271:F414.
http://www.ncbi.nlm.nih.gov/pubmed/12194985
induces decreased apical but increased basolateral expression of AQP2 in CCD of http://www.ncbi.nlm.nih.gov/pubmed/11536078
137. DiGiovanni SR, Nielsen S, Christensen EI, et al. Regulation of collecting
rat93. Bouley
kidney. AmR,J Physiol
Breton Renal
S, Sun T, et al.
Physiol. Nitric oxide and atrial natriuretic actor
2007;293:F87. 115. water
duct Kuwahara
channel M, expression
Asai T, Terada Y, et al. The
by vasopressin in C-terminal
Brattleboro tail rat. oProcaquaporin-2
Natl Acad
stimulate
109. Tamma cGMP-dependent
G, Klussmannmembrane E, Maric insertion
K, et al.o Rho aquaporin
inhibits2 in renal epithe-
cAMP-induced determines
Sci apical tra f cking. Kidney Int. 2005;68:1999.
USA. 1994;91:8984.
lial cells. J Clin
translocation ofInvest. 2000;106:1115.
aquaporin-2 into the apical membrane of renal cells. Am J Physiol http://www.ncbi.nlm.nih.gov/pubmed/16221200
138. Hayashi M, Sasaki S, Tsuganezawa H, et al. Expression and distribution
94. Physiol.
Renal Bouley 2001;281:F1092.
R, Pastor-Soler N, Cohen O, et al. Stimulation o AQP2 mem- 116.
of Noda Yof
aquaporin , Horikawa
collectingS,duct Katayama Y, et al.
are regulated byWater channel
vasopressin V2aquaporin-2
receptor in rat di-
brane Tamma
110. insertionG,inWiesnerrenal epithelial
B, Furkert cellsJ,inetvitro
al. Theand prostaglandin
in vivo by the E2 cGMP phos-
analogue rectly binds
kidney. J ClintoInvest.
actin.1994;94:1778.
Biochem Biophys Res Commun. 2004;322:740.
phodiesterase
sulprostone inhibitor vasopressin-induced
antagonizes sildenaf l citrate (Viagra). Am J through
antidiuresis Physiol Renal Physiol.
activation of http://www.ncbi.nlm.nih.gov/pubmed/15336526
139. Uchida S, Sasaki S, Fushimi K, et al. Isolation of human aquaporin-CD
2005:288:F1103.
Rho. J Cell Sci. 2003;116:3285. 117. JNoda
gene. Y, Horikawa
Biol Chem. S, Kanda E, et al. Reciprocal interaction with G-actin and
1994;269:23451.
95. Tamma
111. Fenton G, RA,Carmosino
Moeller HB, Ho ertM,JD,
M, Svelto et al.
et al. Acute regulation
Bradykinin signaling ocounter- aqua- tropomyosin
140. Sasaki S,is Noda
essential or aquaporin-2
Y. Aquaporin-2 tra fdynamics
protein cking. J Cell Biol.the
within 2008;182:587.
cell. Curr Opin
porin-2
acts phosphorylation
cAMP-elicited aquaporin at Ser-264 by vasopressin.
2 translocation in renal Proc
cells.Natl
J AmAcad Soc Sci USA.
Nephrol. http://www.ncbi.nlm.nih.gov/pubmed/18678705
Nephrol Hypertens. 2007;16:348.
2008;105:3134.
2005;16:2881. 118. Hasler
141. Noda Y,U, Sasaki
NielsenS. TheS,role o actinE,remodeling
Féraille in the tra f cking control
et al. Posttranscriptional o intracel-of
http://www.ncbi.nlm.nih.gov/pubmed/18287043
112. Noda Y, Horikawa S, Furukawa T, et al. Aquaporin-2 traf cking is lular vesicles,abundance
aquaporin-2 transporters, byand channels:inocusing
vasopressin on aquaporin-2.
renal collecting Pf ugers cells.
duct principal Arch.
96. Ho by
regulated ertPDZ-domain
JD, Nielsen containing
J, Yu MJ, etprotein
al. Dynamics
SPA-1. FEBS o aquaporin-2 serine-261
Lett. 2004;568:139. 2008;456:737.
Am J Physiol Renal Physiol. 2006;290:F177.
phosphorylation
113. Harazaki M,inKawai response Y, Suto L,short-term
et al. Speci vasopressin treatment
c recruitment in collecting
of SPA-1 to the http://www.ncbi.nlm.nih.gov/pubmed/18066585
142. Kasono K, Saito T, Saito T, et al. Hypertonicity regulates the aquapo-
duct. Am J Physiol
immunological Renal Physiol.
synapse: involvement 2007;292:F691.
of actin-bundling protein actinin. Immunol 119. promoter
rin-2 Tajika Y, Matsuzaki
independently T, Suzuki T, et al.vasopressin.
of arginine Di erential regulation
Nephrol Dial o Transplant.
AQP2 tra -
97.2004;92:221.
Lett. Lu HJ, Matsuzaki T, Bouley R, et al. The phosphorylation state o serine f cking in endosomes by microtubules and actin f laments. Histochem Cell Biol.
2005;20:509.
256 isKuwahara
114. dominant M, overIwai
that K,o serine
Ooeda261 T, inet the
al. regulation
Three familieso AQP2 withtraautosomal
f cking in 2005;124:1.
143. Hasler U, Jeon US, Kim JA, Tonicity-responsive enhancer binding protein
renal epithelial
dominant cells. Amdiabetes
nephrogenic J Physiolinsipidus
Renal Physiol.
caused 2008;295:F290.
by aquaporin-2 mutations in http://www.ncbi.nlm.nih.gov/pubmed/17153517
is an essential regulator of aquaporin-2 expression in renal collecting duct prin-
the98.C-terminus.
Moeller HB, AmKnepper
J Hum Genet. MA, 2001;69:738.
Fenton RA. Serine 269 phosphorylated aqua- 120. cells.
cipal ChouJ Am CL,Soc Christensen BM, Frische S, et al. Non-muscle myosin II and
Nephrol. 2006;17:1521.
porin-2
115. is targeted
Kuwahara M, toAsai theT, apical
Teradamembrane
Y, et al. The o C-terminal
collecting ducttail of principal
aquaporin-2 cells. myosin
144. light chain kinase
López-Rodríguez C,are downstream
Antos CL, Shelton targets
JM, et oral.
vasopressin
Loss of NFAT5 signaling in the
results in
Kidney Int. 2009;75:295.
determines apical traf cking. Kidney Int. 2005;68:1999. collecting
renal atrophy andduct.lackJ of
Bioltonicity-responsive
Chem. 2004;279:49026. gene expression. Proc Natl Acad Sci
http://www.ncbi.nlm.nih.gov/pubmed/18843259
116. Noda Y, Horikawa S, Katayama Y, et al. Water channel aquaporin-2 directly 121. 2004;101:2392.
USA. Nedvetsky PI, Ste an E, Frische S, et al. A role o myosin Vb and Rab11-
99. to
binds Moeller
actin. HB, MacAulay
Biochem BiophysN,Res Knepper
Commun. MA,2004;322:740.
et al. Role o multiple phosphory- FIP2 in
145. the aquaporin-2
Verkman AS. Dissecting shuttle.theTra f c. 2007;8:110.
roles of aquaporins in renal pathophysiology
la-tionNoda
117. sites inY, the COOH-terminal
Horikawa S, Kandatail E, oet aquaporin-2
al. Reciprocal or interaction
water transport: with evi- G- http://www.ncbi.nlm.nih.gov/pubmed/17156409
using transgenic mice. Semin Nephrol. 2008;28:217.
dence and
actin against channel gating.
tropomyosin Am J Physiol
is essential Renal Physiol. traf
for aquaporin-2 2009;295:F649.
cking. J Cell Biol. 122. Pallone
146. Procino TL, G, Barbieri
EdwardsC,A,Tamma Ma T, G, et al.
et al. AQP2 exocytosis
Requirement in the renal
of aquaporin-1 for
100. Balasubramanian L, Sham JS, Yip KP. Calcium signaling in vasopressin-
2008;182:587. collecting duct
NaCl-driven water - involvement
transport across o SNARE iso orms
descending vasaand theJ regulatory
recta. Clin Invest. role
2000;o
induced
118. Noda aquaporin-2
Y, Sasakitra S. f The
cking.rolePf ugers Arch.remodeling
of actin 2008;456:747–754.
in the traf cking of Munc18b. J Cell Sci. 2008;121:2097.
105:215.
http://www.ncbi.nlm.nih.gov/pubmed/17957381
intracellular vesicles, transporters, and channels: focusing on aquaporin-2. http://www.ncbi.nlm.nih.gov/pubmed/18505797
147. King LS, Choi M, Fernandez PC, et al. Defective urinary-concentrating
P101.
ugersNodaArch.Y2008;456:737.
, Horikawa S, Katayama Y, et al. Identif cation o a multiprotein 123. Takata
ability due to K, Matsuzaki
a complete deT,ciency
Tajikaof Y,aquaporin-1.
et al. LocalizationN Engl and tra f2001;345:175.
J Med. cking o aqua-
“motor”
119. complex
Tajika binding T,
Y, Matsuzaki to Suzuki
water channel
T, et al.aquaporin-2. Biochem Biophys
Differential regulation of AQP2 Res porin Schrier
148. 2 in the RW, kidney. Histochem Cell Biol.
Cadnapaphornchai 2008;130:197.
MA. Renal aquaporin water channels:
Commun.
traf cking2005;330:1041–1047.
in endosomes by microtubules and actin laments. Histochem Cell http://www.ncbi.nlm.nih.gov/pubmed/18566824
from molecules to human disease. Prog Biophys Mol Biol. 2003;81:117.
http://www.ncbi.nlm.nih.gov/pubmed/15823548
Biol. 2005;124:1. 124. Ishibashi
149. Brown D,K,Orci SasakiL. Vasopressin
S, Fushimi K, stimulates ormation
et al. Molecular o coated
cloning pits in rat
and expression
102. Chou
120. LorenzCL, D, Christensen
Krylov A, Hahm D, et al.S,Cyclic
BM, Frische et al. AMP is su f cient
Non-muscle myosinor trigger-
II and kidney
of collecting
a member of the ducts. Nature.family
aquaporin 1983;302:253.
with permeability to glycerol and urea in
ing the light
myosin exocytic
chain recruitment
kinase are odownstream
aquaporin-2 in renal
targets epithelial cells.
for vasopressin EMBO
signaling in Rep.
the 125. SuntoTX,
addition water Vanexpressed
Hoek A, at Huang Y, et al. Aquaporin-2
the basolateral membrane localization in clath-
of kidney collecting
2003;4:88.
renal collecting duct. J Biol Chem. 2004;279:49026. rin-coated
duct pits:Natl
cells. Proc inhibition
Acad Sci o USA.
endocytosis by dominant-negative dynamin. Am J
1994;91:6269.
http://www.ncbi.nlm.nih.gov/pubmed/12524527
121. Nedvetsky PI, Stefan E, Frische S, et al. A role of myosin Vb and Rab11- Physiol.
150. 2002;282:F998.
Ishibashi K, Sasaki S, Fushimi K, et al. Immunolocalization and effect of
103. inValenti
FIP2 G, Laera A,shuttle.
the aquaporin-2 Pace G, et al.
Traf Urinary aquaporin 2 and calciuria cor-
c. 2007;8:110. 126. Lu H, on
dehydration Sun AQP3,TX, aBouley R, etwater
basolateral al. Inhibition
channel ofokidney endocytosis
collecting causes phos-
ducts. Am
relate Procino
122. with the G, severity
Barbierio enuresis
C, Tamma in children.
G, et al.J Am AQP2Soc Nephrol.
exocytosis 2000;11:1873.
in the renal Jphorylation (S256)-independent plasma membrane accumulation o AQP2. Am
Physiol. 1997;272:F235.
http://www.ncbi.nlm.nih.gov/pubmed/11004218
collecting duct – involvement of SNARE isoforms and the regulatory role of J Physiol.
151. Kwon 2004;286:F233.
TH, Nielsen J, Masilamani S, et al. Regulation of collecting duct
104. Valenti
Munc18b. J CellG,Sci.Laera A, Gouraud S, et al. Low-calcium diet in hypercalciuric
2008;121:2097. 127. Lu
AQP3 HA, Sun TX,
expression: Matsuzaki
response T, et al. Heat shock
to mineralocorticoid. Amprotein
J Physiol 70 Renal
interacts with
Physiol.
enuretic
123. children
Takata restores AQP2
K, Matsuzaki excretion
T, Tajika Y, etand al.improves clinical
Localization andsymptoms.
traf ckingAmofJ aquaporin-2 (AQP2) and regulates its tra f cking. J Biol Chem. 2007;282:28721.
2002;283:F1403.
Physiol. 2002;283:F895.
aquaporin 2 in the kidney. Histochem Cell Biol. 2008;130:197. 128. Ma
152. Kamsteeg
T, Song EJ,YDu f eldB,AS,
, Yang Konings IB, diabetes
Nephrogenic et al. MALinsipidus
decreasesinthe internaliza-
mice lacking
105. Brown
124. Sands JM, FloresL.F, Vasopressin
D, Orci Kato A, et al.stimulates
Vasopressin-elicited
formationwater and urea
of coated pitsperme-
in rat tion o the aquaporin-2
aquaporin-3 water channels. waterProc
channel. Proc Natl
Natl Acad Acad2000;97:4386.
Sci USA. Sci USA. 2007;104:16696.
abilitiescollecting
kidney are alteredducts.in IMCD in hypercalcemic
Nature. 1983;302:253.rats. Am J Physiol. 1998; 274:F978. 129. V
153. Klumperman
erkman AS. Aquaporins:J, Deen PM. translating
Short-chainbench ubiquitination
research tomediates
human the reg-
disease.
106. Sun
125. ProcinoTX, G,Van Carmosino
Hoek A,M,Huang TammaY,G,etet al. al. Aquaporin-2
Extracellular calcium antago-
localization in Julated endocytosis
Exp Biol. 2009;212:1707. o the aquaporin-2 water channel. Proc Natl Acad Sci USA.
nizes orskolin-induced
clathrin-coated aquaporin
pits: inhibition 2 tra f ckingbyindominant-negative
of endocytosis collecting duct cells. Kidney
dynamin. 2006;103:18344.
154. Roudier N, Ripoche P, Gane P, et al. AQP3 de ciency in humans and the
Int. J2004;66:2245.
Am Physiol. 2002;282:F998. 130. van basis
molecular Balkom of a BW,novelBoone
bloodM, Hendriks
group system,G, et al.
GIL. LIP5
J Biol Chem.Interacts with aqua-
2002;277:45854.
http://www.ncbi.nlm.nih.gov/pubmed/15569313
126. Lu H, Sun TX, Bouley R, et al. Inhibition of endocytosis causes porin Hasegawa
155. 2 and acilitates H, Maits T,lysosomal
Skach W, degradation.
Molecular cloning J Am Soc of aNephrol.
mercurial- 2009;20:990.
insensitive
107. Nejsum LN,
phosphorylation Zelenina M, Aperiaplasma
(S256)-independent A, et al. Bidirectional
membrane regulationofo AQP2.
accumulation AQP2 http://www.ncbi.nlm.nih.gov/pubmed/19357255
water channel expressed in selected water-transporting tissues. J Biol Chem.
tra fJ cking
Am Physiol.and recycling: involvement o AQP2-S256 phosphorylation. Am J
2004;286:F233. 131. Kuwahara M, Fushimi K, Terada Y, et al. cAMP-dependent phosphoryla-
1994;269:5497.
PhysiolLuRenal
127. HA,Physiol.
Sun TX,2005;288:F930.
Matsuzaki T, et al. Heat shock protein 70 interacts with tion stimulates
156. Yang B, van water Hoek permeability
AN, Verkman o aquaporin-collecting
AS. Very high single duct channel water channel
water per-
108. de Seigneux
aquaporin-2 (AQP2)S,and Nielsen J, Olesen
regulates its trafET, et al.J Biol
cking. Long-term aldosterone treat-
Chem. 2007;282:28721. protein expressed
meability of aquaporin-4in Xenopus in oocytes. J Biol Chem. 1995;270:10384.
baculovirus-infected insect cells and liposomes
ment Kamsteeg
128. induces decreasedEJ, Duf eld apical
AS, but increased
Konings IB, etbasolateral expression
al. MAL decreases o AQP2 in
the internaliza- http://www.ncbi.nlm.nih.gov/pubmed/7537730
reconstituted with puri ed aquaporin-4. Biochemistry. 1997;36:7625.
CCDofothe
tion rataquaporin-2
kidney. Am Jwater Physiol Renal Physiol.
channel. Proc Natl 2007;293:F87.
Acad Sci USA. 2007;104:16696. 132. V
157. Lande
an Hoek MB, AN, Jo I, Zeidel
BouleyML, R, et
Lual.Y,Phosphorylation o aquaporin-2
et al. Vasopressin-induced does not
differential
109. Klumperman
129. Tamma G, Klussmann J, Deen PM. E, Maric K, et al. Rho
Short-chain inhibits cAMP-induced
ubiquitination mediates thetrans- reg- alter the membrane
stimulation of AQP4 water permeability
splice o rat papillary
variants regulates water channel-containing
the in-membrane assembly of
locationendocytosis
ulated o aquaporin-2 of theinto the apicalwater
aquaporin-2 membrane
channel. o Proc
renalNatlcells.Acad
Am SciJ Physiol
USA. vesicles. J Biol
orthogonal Chem.
arrays. Am1996;271:5552.
J Physiol Renal Physiol. 2009;296:F1396.
Renal Physiol. 2001;281:F1092.
2006;103:18344. 133. Ma
158. Eto T,
K, Y Noda
ang B,Y, Gillespie
HorikawaA,S, Generation
et al. Phosphorylation
and phenotype o aquaporin-2
of a transgenicregu-
110. van
130. TammaBalkom G, BW,
Wiesner
BooneB,M, Furkert
Hendriks J, etG,al. The
et al. prostaglandin
LIP5 Interacts with E2aquaporin
analogue lates its water
knockout mouse permeability.
lacking the J Biol Chem. 2010;285:40777.
mercurial-insensitive water channel aquaporin-4.
2sulprostone
and facilitates antagonizes
its lysosomalvasopressin-induced
degradation. J Amantidiuresis
Soc Nephrol.through activation o
2009;20:990. Jhttp://www.ncbi.nlm.nih.gov/pubmed/20971851
Clin Invest. 1997;100:957.
Rho. JKuwahara
131. Cell Sci. 2003;116:3285.
M, Fushimi K, Terada Y, et al. cAMP-dependent phosphoryla- 134. Zelenina
159. Törnroth-Horsef
M, Zelenin eldS,S,Bondar
Wang Y , Hed
AA, alkWater
et al. K, et permeability
al. Structuralofmechanism
aquaporin-4 o
http://www.ncbi.nlm.nih.gov/pubmed/12829746
tion stimulates water permeability of aquaporin-collecting duct water channel plant
is aquaporin
decreased by gating.
proteinNature.
kinase2006;439:688.
C and dopamine. Am J Physiol Renal Physiol.
111. Tamma
protein expressed G, Carmosino M, Svelto
in Xenopus oocytes. M, Chem.
J Biol et al. Bradykinin
1995;270:10384. signaling counter- http://www.ncbi.nlm.nih.gov/pubmed/16340961
2002;283:F309.
acts cAMP-elicited
132. Lande MB, Jo aquaporinI, Zeidel ML, 2 ettranslocation in renal of
al. Phosphorylation cells. J Am Socdoes
aquaporin-2 Nephrol.not 135. Y
160. Yasui
amamoto
M, Hazama T, Sasaki S, Fushimi
A, Kwon TH, etK,al.etRapid
al. Localization
gating and and anionexpression
permeabilityo a
2005;16:2881.
alter the membrane water permeability of rat papillary water channel-containing collecting
of duct water
an intracellular channel,Nature.
aquaporin. aquaporin, in hydrated and dehydrated rats. Exp
1999;402:184.
http://www.ncbi.nlm.nih.gov/pubmed/16093449
vesicles. J Biol Chem. 1996;271:5552. Nephrol.
161. 1995;3:193.
Yasui M, Kwon TH, Knepper MA, et al. Aquaporin-6: an intracellular
112. Eto
133. Noda Y, Horikawa
K, Noda Y, Horikawa S, Furukawa T, et al. Aquaporin-2
S, et al. Phosphorylation tra f ckingregulates
of aquaporin-2 is regu- http://www.ncbi.nlm.nih.gov/pubmed/7542539
vesicle water channel protein in renal epithelia. Proc Natl Acad Sci USA. 1999;
lated
its by PDZ-domain
water permeability. containing
J Biol Chem.protein SPA-1. FEBS Lett. 2004;568:139.
2010;285:40777. 136. Terris J, Ecelbarger CA, Nielsen S, et al. Long-term regulation o our renal
96:5808.
http://www.ncbi.nlm.nih.gov/pubmed/15196935 aquaporins in rats. Am J Physiol. 1996;271:F414.
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1157
57

137. Ohshiro
162. DiGiovanni K, YSR,aoitaNielsen
E, Yoshida S, Christensen EI, et al. and
Y, et al. Expression Regulation o collecting
immunolocalization 161. Yasui M,LK,Kwon
192. Hansen RittigTH, Knepper GL.
S, Robertson MA,Genetic
et al. basis
Aquaporin-6:
of familialan intracellular
neurohypoph-
duct
of water
AQP6 in channel
intercalated expression
cells ofby thevasopressin in Brattleboro
rat kidney collecting duct.rat.ArchProc NatlCytol.
Histol Acad vesicle water channel
yseal diabetes insipidus. protein
TrendsinEndocrinol
renal epithelia. Proc Natl Acad Sci USA. 1999;
Metab. 1997;8:363.
Sci USA. 1994;91:8984.
2001;64:329. 96:5808.
193. Repaske DR, Phillips JA III, Kirby LT, et al. Molecular analysis of autosomal
138. Ikeda
163. Hayashi M,M, BeitzSasaki S, Tsuganezawa
E, Kozono D, et al.H,Characterization
et al. Expressionof and distribution
aquaporin-6 as 162.
dominant Ohshiro K, Yaoita E, Yoshida
neurohypophyseal Y, et al.
diabetes Expression
insipidus. J and
Clin immunolocalization
Endocrinol Metab.
ao nitrate
aquaporin o collecting
channel duct are cells.
in mammalian regulated by vasopressin
Requirement V2 receptorresidue
of pore-lining in rat o1990;70:752.
AQP6 in intercalated cells o the rat kidney collecting duct. Arch Histol Cytol.
kidney. J Clin
threonine 63. Invest. 1994;94:1778.
J Biol Chem. 2002;277:39873. 2001;64:329.
194. Rittig S, Robertson GL, Siggaard C, et al. Identi cation of 13 new mutations
139. Nejsum
164. Uchida S, LN,Sasaki
ElkjaerS, M,Fushimi
Hager K, H,etet al.
al. Isolation
Localization o human aquaporin-CD
of aquaporin-7 in rat http://www.ncbi.nlm.nih.gov/pubmed/11575429
in the vasopressin-neurophysin II gene in 17 kindreds with familial autosomal
gene.mouse
and J Biol Chem.
kidney1994;269:23451.
using RT-PCR, immunoblotting, and immunocytochemistry. 163.
dominant Ikeda M, Beitz E, Kozono
neurohypophyseal D, et al.
diabetes Characterization
insipidus. Am J Hum o Genet.
aquaporin-6 as a ni-
1996;58:107.
http://www.ncbi.nlm.nih.gov/pubmed/7522228
Biochem Biophys Res Commun. 2000;277:164. trate
195. channel
Baylis PH, in mammalian
Thompson cells. Requirement o pore-lining
CJ. Osmoregulation of vasopressin residue threonine
secretion and
140. Rojek
165. SasakiA, S, Praetorius
Noda Y. Aquaporin-2
J, Frøkiaer protein
J, et al. dynamics
A currentwithinview of thethe
cell. Curr Opin
mammalian 63. J Biol
thirst Chem.and
in health 2002;277:39873.
disease. Clin Endocrinol (Oxf). 1988;29:549.
Nephrol Hypertens. 2007;16:348.
aquaglyceroporins. Annu Rev Physiol. 2008;70:301. http://www.ncbi.nlm.nih.gov/pubmed/12177001
196. Smith D, McKenna K, Moore K, et al. Baroregulation of vasopressin release
http://www.ncbi.nlm.nih.gov/pubmed/17565277
166. Sohara E, Rai T, Sasaki S, et al. Physiological roles of AQP7 in the kidney: 164. Nejsum
in adipsic LN, insipidus.
diabetes Elkjaer M,JHager H, et al. Localization
Clin Endocrinol o aquaporin-7 in rat
Metab. 2002;87:4564.
141. Hasler
lessons from AQP7U, Nielsen S, Feraille
knockout mice. E, et al. Biophys
Biochim Posttranscriptional control o aqua-
Acta. 2006;1758:1106. and
197.mouse
Spanakis kidney using RT-PCR,
E, Milord E, Gragnoliimmunoblotting,
C. AVPR2 variants and immunocytochemistry.
and mutations in
porin-2
167. abundance
Kondo by vasopressin
H, Shimomura in renal
I, Kishida K,collecting
et al. Human duct principal
aquaporin cells. Am J
adipose Biochem
nephrogenic Biophys Res Commun.
diabetes insipidus:2000;277:164.
review and missense mutation signi cance.
Physiol Renal
(AQPap) gene.Physiol.
Genomic 2006;290:F177.
structure, promoter analysis and functional mutation. http://www.ncbi.nlm.nih.gov/pubmed/11027658
J Cell Physiol. 2008;217:605.
142.J Biochem.
Eur Kasono K, Saito T, Saito T, et al. Hypertonicity regulates the aquapo-rin-2 pro-
2002;269:1814. 165. Rojek A,JH,
198. Robben Praetorius
Knoers NV J, ,Frøkiaer
Deen PM. J, Cell
et al.biological
A currentaspects
view oofthethe mammalian
vasopressin
moter Ishibashi
168. independently o arginine vasopressin.
K. Aquaporin subfamily with Nephrol Dial Transplant.
unusual NPA boxes. 2005;20:509.
Biochim aquaglyceroporins.
type-2 receptor and Annu Rev Physiol.
aquaporin 2 2008;70:301.
water channel in nephrogenic diabetes
http://www.ncbi.nlm.nih.gov/pubmed/15671070
Biophys Acta. 2006;1758:989. http://www.ncbi.nlm.nih.gov/pubmed/17961083
insipidus. Am J Physiol Renal Physiol. 2006;291:F257.
143. Morishita
169. Hasler U, Jeon US, Kim JA,T,Tonicity-responsive
Y, Matsuzaki Hara-chikuma M,enhancer et al. binding
Disruption proteinof 166.
199. Los SoharaEL,E,DeenRai T,PM,
Sasaki S, et al.JH.
Robben Physiological
Potential of roles o AQP7 in(ant)agonists
nonpeptide the kidney:
is an essential regulator
aquaporin-11 produceso polycystic
aquaporin-2 expression
kidneys in renalvacuolization
following collecting ductofprin- the lessons
to rescue romvasopressin
AQP7 knockout mice. Biochim
V2 receptor mutants Biophys
for Acta. 2006;1758:1106.
the treatment of X-linked
cipal cells.tubule.
proximal J Am Soc MolNephrol.
Cell Biol. 2006;17:1521.
2005;25:7770. http://www.ncbi.nlm.nih.gov/pubmed/16860289
nephrogenic diabetes insipidus. J Neuroendocrinol. 2010;22:393.
144. Okada
170. López-Rodríguez
S, Misaka C, T, Antos
TanakaCL, Y, Shelton JM, et al. Lossknockout
et al. Aquaporin-11 o NFAT5 mice resultsandin 167.
200. van Kondo H, Shimomura
Lieburg AF, Verdijk I, Kishida
MA, SchouteK, et al. FHuman
, et al. aquaporin adipose (AQ-
Clinical phenotype of
renal atrophy
polycystic and disease
kidney lack o animals
tonicity-responsive
share a common gene mechanism
expression. of Proc Natl
cyst Acad Sci
formation. Pap) gene. Genomic
nephrogenic diabetesstructure,
insipiduspromoter
in femalesanalysis and unctional
heterozygous mutation.type
for a vasopressin Eur2J
USA. 2004;101:2392.
FASEB J. 2008;22:3672. Biochem. 2002;269:1814.
receptor mutation. Hum Genet. 1995;96:70.
http://www.ncbi.nlm.nih.gov/pubmed/14983020
171. Yakata K, Tani K, Fujiyoshi Y. Water permeability and characterization of http://www.ncbi.nlm.nih.gov/pubmed/11952783
201. Faerch M, Corydon TJ, Rittig S, et al. Skewed X-chromosome inactivation
145. VerkmanJ AS.
aquaporin-11. Struct Dissecting the roles
Biol. 2011;174: 315.o aquaporins in renal pathophysiology 168.
causing Ishibashi
diagnosticK. Aquaporin sub amily in
misinterpretation withcongenital
unusual NPA boxes. Biochim
nephrogenic Bio-
diabetes
using Uchida
172. transgenic S, mice.
Sohara Semin
E, Rai Nephrol.
T, et2008;28:217.
al. Impaired urea accumulation in the phys Acta. 2006;1758:989.
insipidus. Scand J Urol Nephrol. 2010;44:324.
http://www.ncbi.nlm.nih.gov/pubmed/18519083
inner medulla of mice lacking the urea transporter UT-A2. Mol Cell Biol. http://www.ncbi.nlm.nih.gov/pubmed/16579962
202. Chan Seem CP, Dossetor JF, Penney MD. Nephrogenic diabetes insipidus
146. Pallone TL, Edwards A, Ma T, et al. Requirement o aquaporin-1 or NaCl-
2005;25:7357. 169.
due toMorishita
a new mutationY, Matsuzaki
of the T,arginine
Hara-chikumavasopressin M, V2 et al. Disruption
receptor gene oin aqua-
a girl
drivenSands
173. water JM,transport
Gargus across descending
JJ, Fröhlich O, et vasaal.recta. J Clin
Urinary Invest. 2000; ability
concentrating 105:215. in porin-11
presentingproduces polycystic kidneys
with non-accidental injury. Ann ollowing vacuolization
Clin Biochem. o the proximal
1999;36:779.
http://www.ncbi.nlm.nih.gov/pubmed/10642600
patients with Jk(a-b-) blood type who lack carrier-mediated urea transport. J Am tubule. Mol Cell Biol.
203. Birnbaumer M.2005;25:7770.
Vasopressin receptor mutations and nephrogenic diabetes
147.Nephrol.
Soc King LS, Choi M, Fernandez PC, et al. De ective urinary-concentrating
1992;2:1689. 170. Okada
insipidus. ArchS,Med Misaka T, Tanaka Y, et al. Aquaporin-11 knockout mice and
Res. 1999;30:465.
abilityYdue
174. to a complete
oshikawa M, Uchida def ciency o aquaporin-1.
S, Yamauchi A, et al. N Engl J Med. of
Localization 2001;345:175.
rat CLC-K2 polycystic
204. Sasaki kidney disease
S, Fushimi animals
K, Saito H, etshare a common
al. Cloning, mechanism oandcyst
characterization, ormation.
chromosomal
http://www.ncbi.nlm.nih.gov/pubmed/11463012
chloride channel mRNA in the kidney. Am J Physiol. 1999;276:F552. FASEB
mapping J. 2008;22:3672.
of human aquaporin of collecting duct. J Clin Invest. 1994;93:1250.
148. Kobayashi
175. Schrier RW, K. Cadnapaphornchai
Uchida S, Mizutani S, MA. Renal
et al. aquaporin
Intrarenal water channels:
and cellular localiza- 171.
205. DeenYakataPM, K, Verdijk
Tani K, MA,
Fujiyoshi
Knoers Y.NVWater permeability
, et al. Requirement and characterization
of human renal watero
romofmolecules
tion to human
ClC-K2 protein disease.
in the mouse Prog Biophys
kidney. J Am MolSocBiol. 2003;81:117.
Nephrol. 2001;12:1327. aquaporin-11.
channel aquaporin-2 J Structfor
Biol. 2011;174: 315.
vasopressin-dependent concentration of urine. Science.
http://www.ncbi.nlm.nih.gov/pubmed/12565698
176. Hebert SC. Bartter syndrome. Curr Opin Nephrol Hypertens. 2003;12:527. http://www.ncbi.nlm.nih.gov/pubmed/21251984
1994;264:92.
149. Simon
177. Ishibashi DB,K,Bindra
SasakiRS, S, Mans
Fushimi eldK,TA, et et
al.al.
Molecular
Mutations cloning
in the and expression
chloride chan- 172. Uchida S,AJ,Sohara
206. Loonen Knoers E, Rai
NV, T,vanet al.OsImpaired
CH, et al. ureaAquaporin
accumulation in the inner
2 mutations in
o agene,
nel member o the cause
CLCNKB, aquaporinBartter’ amily with permeability
s syndrome type III. NattoGenet.glycerol and urea in
1997;17:171. medulla
nephrogenic o mice lacking
diabetes the urea Semin
insipidus. transporter
Nephrol.UT-A2. Mol Cell Biol. 2005;25:7357.
2008;28:252.
addition
178. to water
Bartter expressed
FC, Pronove at the
P, Gill basolateral
JR Jr, membrane
et al. Hyperplasia of othekidney collecting
juxtaglomerular http://www.ncbi.nlm.nih.gov/pubmed/16055743
207. Savelkoul PJ, De Mattia F, Li Y, et al. p.R254Q mutation in the aquaporin-2
duct cells.with
complex Prochyperaldosteronism
Natl Acad Sci USA. 1994;91:6269.
and hypokalemic alkalosis. A new syndrome. 173. Sands JM,
water channel Gargus
causing JJ, Fröhlich
dominant O, et al.diabetes
nephrogenic Urinaryinsipidus
concentrating
is dueability in
to a lack
150.J Med.
Am Ishibashi K, Sasaki S, Fushimi K, et al. Immunolocalization and e ect o
1962;33:811. patients
of arginine with Jk(a-b-) blood typephosphorylation.
vasopressin-induced who lack carrier-mediated Hum Mutat. urea transport. J Am
2009;30:E891.
dehydration
179. Simon on DB,AQP3,
Karet aFE, basolateral
Hamdanwater JM, etchannel o kidney
al. Bartter’ collecting
s syndrome, ducts. Am
hypokalaemic Soc
208.Nephrol.
de Mattia 1992;2:1689.
F, Savelkoul PJ, Kamsteeg EJ, et al. Lack of arginine vasopressin-
J Physiol. 1997;272:F235.
alkalosis with hypercalciuria, is caused by mutations in the Na-K-2Cl http://www.ncbi.nlm.nih.gov/pubmed/1498276
induced phosphorylation of aquaporin-2 mutant AQP2-R254L explains
151. Kwon TH,
cotransporter NKCC2.Nielsen NatJ,Genet.
Masilamani S, et al. Regulation o collecting duct
1996;13:183. 174.
dominant Yoshikawa
nephrogenicM, Uchida
diabetes S, insipidus.
Yamauchi JA,Am etSocal. Nephrol.
Localization o rat CLC-K2
2005;16:2872.
AQP3Takahashi
180. expression: response to mineralocorticoid.
N, Chernavvsky DR, Gomez RA, et al. AmUncompensated
J Physiol Renalpolyuria Physiol. chloride
209. KamsteegchannelEJ, mRNA in thePJ,kidney.
Savelkoul Hendriks Am G, J Physiol. 1999;276:F552.
et al. Missorting of the aquaporin-2
2002;283:F1403.
in a mouse model of Bartter’s syndrome. Proc Natl Acad Sci USA. 2000;97:5434. 175.
mutantKobayashi
E258K to K. Uchida S, Mizutani
multivesicular S, et al. Intrarenal
bodies/lysosomes in dominant and NDI
cellular localiza-
is associated
152. Kaplan
181. Ma T, Song MR, YPlotkin
, Yang B, MD,Nephrogenic
Brown D, diabetes insipidus inofmice
et al. Expression the lacking
mouse tion
with oitsClC-K2 protein in theand
monoubiquitination mouse kidney.
increased J Am Soc Nephrol.
phosphorylation by 2001;12:1327.
PKC but is due to
aquaporin-3
Na-K-2Cl water channels.
cotransporter, mBSC2,Proc Natl Acadterminal
in the Sci USA. inner
2000;97:4386.
medullary collecting http://www.ncbi.nlm.nih.gov/pubmed/11423561
the loss of E258. P ugers Arch. 2008;455:1041.
http://www.ncbi.nlm.nih.gov/pubmed/10737773
duct, the glomerular and extraglomerular mesangium and the glomerular 176.
210. AsaiHebert T, SC. Bartter syndrome.
Kuwahara M, Kurihara Curr OpinH, etNephrol Hypertens. 2003;12:527.
al., Pathogenesis of nephro-
153. Verkman
afferent arteriole.AS. Aquaporins:
J Clin translating bench research to human disease. J
Invest. 1996;98:723. 177.
genic Simon
diabetes DB,insipidus
Bindra RS, byMansf eld TA, etC-terminus
aquaporin-2 al. Mutations in the chloride
mutations. Kidneychan-
Int.
Exp Biol.
182. Ginns2009;212:1707.
SM, Knepper MA, Ecelbarger CA, et al. Immunolocalization of the nel gene, CLCNKB, cause Bartter’s syndrome type III. Nat Genet. 1997;17:171.
2003;64:2.
http://www.ncbi.nlm.nih.gov/pubmed/19448080
secretory isoform of the Na-K-Cl cotransporter in rat renal intercalated cells. http://www.ncbi.nlm.nih.gov/pubmed/9326936
211. Sohara E, Rai T, Yang SS, et al., Pathogenesis and treatment of autosomal-
J154. Roudier
Am Soc Nephrol. N, 1996;7:2533.
Ripoche P, Gane P, et al. AQP3 def ciency in humans and the 178.
dominant Bartter FC, Pronove
nephrogenic P, Gillinsipidus
diabetes JR Jr, et al. Hyperplasia
caused o the juxtaglomerular
by an aquaporin 2 mutation.
molecular
183. WallbasisSM, oKnepper
a novel blood group system,
MA, Hassell KA, et GIL. J Biol Chem. 2002;277:45854.
al. Hypotension in NKCC1 null complex
Proc Natl withAcadhyperaldosteronism
Sci USA. 2006;103:14217. and hypokalemic alkalosis. A new syndrome.
http://www.ncbi.nlm.nih.gov/pubmed/12239222
mice: role of the kidneys. Am J Physiol Renal Physiol. 2006;290:F409. Am
212.J Med.
Lloyd1962;33:811.
DJ, Hall FW, Tarantino LM, et al. Diabetes insipidus in mice with a
155. Xu
184. Hasegawa
JZ, HallH, AE,MaPeterson
T, Skach LN, W,etMolecular cloning
al. Localization of othea ROMK
mercurial-insensi-
protein on http://www.ncbi.nlm.nih.gov/pubmed/13969763
mutation in aquaporin-2. PLoS Genet. 2005;1(2):e20.
tive water
apical channelof
membranes expressed
rat kidney innephron
selected segments.
water-transporting
Am J Physiol. tissues. J Biol Chem.
1997;273:F739. 179. Simon DB,
213. Grünfeld JP,Karet FE,BC.
Rossier Hamdan
Lithium JM, et al. Bartter’srevisited.
nephrotoxicity syndrome, Nathypokalaemic
Rev Nephrol.
1994;269:5497.
185. Simon DB, Karet FE, Rodriguez-Soriano J, et al. Genetic heterogeneity of Bartter’s alkalosis
2009;5:270. with hypercalciuria, is caused by mutations in the Na-K-2Cl cotrans-
156. Yang
syndrome B, vanby
revealed Hoek AN, Verkman
mutations in the K AS. Very ROMK.
channel, high single channel
Nat Genet. water per-
1996;14:152. porter
214. Rao NKCC2.
R, ZhangNat Genet.
MZ, Zhao 1996;13:183.
M, et al. Lithium treatment inhibits renal GSK-3
meability
186. Lorenz o aquaporin-4
JN, Baird NR, in baculovirus-in
Judd LM, et al. ected insect cells
Impaired renaland NaCl liposomes
absorption re- http://www.ncbi.nlm.nih.gov/pubmed/8640224
activity and promotes cyclooxygenase 2-dependent polyuria. Am J Physiol Renal
constituted
in mice lackingwith purif
the ROMKed aquaporin-4.
potassiumBiochemistry.
channel, a 1997;36:7625.
model for type II Bartter’s 180.
Physiol.Takahashi
2005;288:F642.N, Chernavvsky DR, Gomez RA, et al. Uncompensated polyuria
157. Van Hoek
syndrome. AN, Bouley
J Biol Chem. R, Lu Y, et al. Vasopressin-induced di erential stim-
2002;277:37871. in a mouse
215. Nielsen model o Bartter’
J, Hoffert JD, sKnepper
syndrome. MA, Proc
et Natl Acad Sci USA.
al. Proteomic 2000;97:5434.
analysis of lithium-
ulationSchultheis
187. o AQP4 splicePJ, Clarkevariants regulates the
LL, Meneton P, etin-membrane
al. Renal and assembly
intestinaloabsorptive
orthogo- http://www.ncbi.nlm.nih.gov/pubmed/10779555
induced nephrogenic diabetes insipidus: mechanisms for aquaporin 2 down-
nal arrays.
defects Am Jlacking
in mice Physiol theRenal Physiol.
NHE3 Na 2009;296:F1396.
/H exchanger. Nat Genet. 1998;19:282. 181. Kaplan
regulation andMR, Plotkin
cellular MD, Brown
proliferation. Proc D,Natl
et al.Acad
Expression
Sci USA.o2008;105:3634.
the mouse Na-K-
158. Lorenz
188. Ma T, JN, Yang B, Gillespie
Schultheis A, Generation
PJ, Traynor and phenotypeanalysis
T, et al. Micropuncture o a transgenic
of single- 2Cl
216.cotransporter,
Christensen BM, mBSC2, in the
Marples D,terminal
Kim YH,inner et al.medullary
Changes incollecting duct, the
cellular composi-
knockoutfunction
nephron mouse lacking
in NHE3-de the mercurial-insensitive
cient mice. Am J Physiol. water1999;277:F447.
channel aquaporin-4. J glomerular
tion of kidney andcollecting
extraglomerular
duct cellsmesangium
in rats withand the glomerular
lithium-induced NDI. a erent arteri-
Am J Physiol
Clin Invest.
189. Amlal1997;100:957.
H, Ledoussal C, Sheriff S, et al. Downregulation of renal AQP2 water ole.
Cell JPhysiol.
Clin Invest. 1996;98:723.
2004;286:C952.
http://www.ncbi.nlm.nih.gov/pubmed/9276712
channel and NKCC2 in mice lacking the apical Na -H exchanger NHE3. http://www.ncbi.nlm.nih.gov/pubmed/8698864
217. Wang W, Li C, Kwon TH, et al. AQP3, p-AQP2, and AQP2 expression
J159. Zelenina
Physiol. M, Zelenin S, Bondar AA, et al. Water permeability o aquapor-
2003;553:511. 182. Ginns in
is reduced SM,polyuric
Knepperrats MA,withEcelbarger CA, et al. prevention
hypercalcemia: Immunolocalization
by cAMP-PDE o the
in-4 isEcelbarger
190. decreased CA, by protein
Kim GH, kinase
TerrisCJ,and dopamine.
et al. Am J Physiol Renal
Vasopressin-mediated Physiol.
regulation of secretory
inhibitors.iso Amorm o the Renal
J Physiol Na-K-Cl cotransporter
Physiol. in rat renal intercalated cells. J Am
2002;283:F1313.
2002;283:F309.
ENaC abundance in rat kidney. Am J Physiol Renal Physiol. 2000;279:F46. Soc
218.Nephrol.
Wang 1996;7:2533.
W, Kwon TH, Li C, et al. Reduced expression of Na-K-2Cl
160. Snyder
191. Yasui M,PM. Hazama A, Kwon
Minireview: TH, et al.ofRapid
regulation gating
epithelial Naandchannel
anion permeability
traf cking. http://www.ncbi.nlm.nih.gov/pubmed/8989731
cotransporter in medullary TAL in vitamin D-induced hypercalcemia in rats. Am
o an intracellular
Endocrinology. aquaporin. Nature. 1999;402:184.
2005;146:5079. 183.
J PhysiolWall SM,Physiol.
Renal Knepper MA, Hassell KA, et al. Hypotension in NKCC1 null
2002;282:F34.
http://www.ncbi.nlm.nih.gov/pubmed/10647010 mice: role o the kidneys. Am J Physiol Renal Physiol. 2006;290:F409.
158
15 8 SECTION 1 STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

184. Li
219. XuC,JZ,Wang
Hall AE,W, Peterson
Knepper LN, MA,etetal.al.Localization o the ROMK
Downregulation of renalprotein on
aquapo- 206. Gerbes
233. LoonenAL, AJ,Gülberg
Knoers V,NV , vanP, Os
Ginès et al.CH, et al. ofAquaporin
Therapy hyponatremia2 mutations in
in cirrhosis
apical
rins inmembranes
response too unilateral
rat kidneyureteral
nephronobstruction.
segments. Am AmJ Physiol. 1997;273:F739.
J Physiol Renal Physiol. nephrogenic
with diabetes
a vasopressin insipidus.
receptor Semin Nephrol.
antagonist: 2008;28:252.
a randomized double-blind multicenter
185. Simon DB, Karet FE, Rodriguez-Soriano J, et al. Genetic heterogeneity o
2003;284:F1066. http://www.ncbi.nlm.nih.gov/pubmed/18519086
trial. Gastroenterology. 2003;124:933.
Bartter’
220. s syndrome
Apostol revealed byCA,
E, Ecelbarger mutations
Terris inJ, the
et K+al. channel,
ReducedROMK. Nat Genet.
renal medullary 207. Ohara
234. Savelkoul M, PJ, De Mattia
Martin PY, Xu F, Li
DL,Y, et
et al.
al.p.R254Q mutation
Upregulation in the aquaporin-2
of aquaporin 2 water
1996;14:152.
water channel expression in puromycin aminonucleoside–induced nephrotic water channel
channel expressioncausing dominantrats.
in pregnant nephrogenic
J Clin Invest.diabetes insipidus is due to a lack
1998;101:1076.
http://www.ncbi.nlm.nih.gov/pubmed/8841184
syndrome. J Am Soc Nephrol. 1997;8:15. o arginine
235. Ishikawa vasopressin-induced
SE, Saito T, Saito phosphorylation. Hum Mutat.
T, et al. Pathophysiological 2009;30:E891.
role of aquaporin-2
186. Bohlin
221. Lorenz AB,JN, Berg
Baird U.NR,Renal
Juddwater
LM, ethandling
al. Impaired renal NaCl
in minimal absorption
change nephrotic in 208.
in de Mattia
impaired water F, excretion.
Savelkoul PJ,ProgKamsteeg
Brain Res.EJ,2008;170:581.
et al. Lack o arginine vasopressin-
mice lacking
syndrome. Int the ROMK
J Pediat potassium
Nephrol. channel, a model or type II Bartter’s syn-
1984;5:93–98. induced
236. Saitophosphorylation
T, Ishikawa S,o Abe aquaporin-2
K, et al. mutant AQP2-R254L
Acute aquaresis by theexplains domi-
nonpeptide
drome.
222. J Biol Chem. 2002;277:37871.
Fernández-Llama P, Andrews P, Ecelbarger CA, et al. Concentrating defect nant nephrogenic
arginine vasopressin diabetes
(AVP)insipidus.
antagonist J AmOPC-31260
Soc Nephrol. 2005;16:2872.
improves hyponatremia
http://www.ncbi.nlm.nih.gov/pubmed/12122007
in experimental nephrotic syndrome: altered expression of aquaporins and thick http://www.ncbi.nlm.nih.gov/pubmed/16120822
in patients with syndrome of inappropriate secretion of antidiuretic hormone
187. Schultheis
ascending limb NaPJ, Clarke LL, Meneton
transporters. Kidney PInt.
, et 1998;54:170.
al. Renal and intestinal absorptive 209. Kamsteeg
(SIADH). EJ, Savelkoul
J Clin Endocrinol Metab.PJ, 1997;82:1054.
Hendriks G, et al. Missorting o the aquapor-
de ectsMouri
223. in mice T, lacking
Inoue T, theNonoguchi
NHE3 Na+/H+ H, et exchanger.
al. AcuteNat andGenet. 1998;19:282.
chronic metabolic in-2 mutant
237. KazamaE258K to multivesicular
I, Hatano R, Michimatabodies/lysosomes in dominant
M, et al. BSC1 inhibition NDI is as-
complements
http://www.ncbi.nlm.nih.gov/pubmed/9662405
acidosis interferes with aquaporin-2 translocation in the rat kidney collecting sociatedofwith
effects its monoubiquitination
vasopressin and increased
V2 receptor antagonist phosphorylation
on hyponatremia by PKCrats.
in SIADH but
188. Lorenz
ducts. Hypertens JN,Res.Schultheis PJ, Traynor T, et al. Micropuncture analysis o sin-
2009;32:358. is due to
Kidney Int.the loss o E258. Pf ugers Arch. 2008;455:1041.
2005;67:1855.
gle-nephron
224. SzatalowiczunctionVL, inArnold
NHE3-def cient mice. Am
PE, Chaimovitz C, JetPhysiol. 1999;277:F447. of
al. Radioimmunoassay 210. Feldman
238. Asai T, Kuwahara M, Kurihara
BJ, Rosenthal SM, VargasH, et GA,al., Pathogenesis o nephro-genic
et al. Nephrogenic syndromedia- of
189. Amlal
plasma H, Ledoussal
arginine vasopressin C, Sheri S, et al. Downregulation
in hyponatremic patients with o renal AQP2 heart
congestive water betes insipidusantidiuresis.
inappropriate by aquaporin-2N Engl C-terminus mutations. Kidney Int. 2003;64:2.
J Med. 2005;352:1884.
channelNand
failure. EnglNKCC2 in mice lacking the apical Na + -H+ exchanger NHE3. J
J Med. 1981;305:263. http://www.ncbi.nlm.nih.gov/pubmed/12787389
239. Decaux G, Vandergheynst F, Bouko Y, et al. Nephrogenic syndrome of
Physiol.
225. 2003;553:511.
Schrier RW. Vasopressin and aquaporin 2 (AQP2) in clinical disorders of 211. Sohara E,antidiuresis
inappropriate Rai T, YanginSS,adults:
et al., Pathogenesis
high phenotypic and treatment
variabilityoinautosomal-
men and
http://www.ncbi.nlm.nih.gov/pubmed/14500765
water homeostasis. Semin Nephrol. 2008;28:289. dominant
women from nephrogenic diabetes
a large pedigree. J Aminsipidus caused
Soc Nephrol. by an aquaporin 2 mutation.
2007;18:606.
190. Nielsen
226. Ecelbarger CA, Kim
S, Terris GH, TerrisD,J, et al. Vasopressin-mediated
J, Andersen Congestive heart failure regulation o
in rats is Proc Natl
240. Kanno AcadK,SciSasaki
USA. 2006;103:14217.
S, Hirata Y, et al. Urinary excretion of aquaporin-2 in
ENaC abundance
associated in rat kidney.
with increased Am Jand
expression Physiol Renal Physiol.
targeting 2000;279:F46.
of aquaporin-2 water channel 212. Lloyd
patients withDJ, Hall FW,
diabetes Tarantino
insipidus. LM,J et
N Engl Med.al. 1995;332:1540.
Diabetes insipidus in mice with a
191.
in Snyderduct.
collecting PM. ProcMinireview:
Natl Acadregulation o epithelial Na+ channel tra f cking.
Sci USA. 1997;94:5450. mutation
241. in aquaporin-2.
Pisitkun T, Shen PLoS Genet. 2005;1(2):e20.
RF, Knepper MA. Identi cation and proteomic
Endocrinology.
227. Xu DL, 2005;146:5079.
Martin PY, Ohara M, et al. Upregulation of aquaporin-2 water 213. ling
pro Grünofeld JP, RossierinBC.human
exosomes Lithiumurine.
nephrotoxicity
Proc Natlrevisited.
Acad SciNatUSA.
Rev Nephrol.
2004;
192. Hansen
channel LK, Rittig
expression S, Robertson
in chronic GL. Genetic
heart failure. basis o1997;99:1500.
J Clin Invest. amilial neurohypoph- 2009;5:270.
101:13368.
yseal diabetes
228. Gheorghiade insipidus. TrendsWA,
M, Gattis Endocrinol
O’Connor Metab. CM,1997;8:363.
et al. Effects of tolvaptan, a http://www.ncbi.nlm.nih.gov/pubmed/19384328
242. Martin PY, Abraham WT, Lieming X, et al. Selective V2-receptor
http://www.ncbi.nlm.nih.gov/pubmed/18406826
vasopressin antagonist, in patients hospitalized with worsening heart failure: 214. Rao R,antagonism
vasopressin Zhang MZ,decreases
Zhao M,urinary
et al. Lithium treatment
aquaporin-2 inhibits
excretion renal GSK-3
in patients with
193.
A Repaskecontrolled
randomized DR, Phillips JAJAMA.
trial. III, Kirby LT, et al. Molecular analysis o auto-
2004;291:1963. activity and
chronic heartpromotes
failure. J cyclooxygenase
Am Soc Nephrol. 2-dependent
1999;10:2165. polyuria. Am J Physiol Renal
somalSchrier
229. dominant RW,neurohypophyseal
Gross P, Gheorghiade diabetes
M, et al.insipidus.
Tolvaptan,J Clin Endocrinol
a selective oralMetab.
vaso- Physiol.
243. 2005;288:F642.
Ivarsen P, Frøkiaer J, Aagaard NK, et al. Increased urinary excretion of
1990;70:752.
pressin V2-receptor antagonist, for hyponatremia. N Engl J Med. 2006;355:2099. 215. Nielsen
aquaporin 2 inJ,patients
Ho ertwithJD, Knepper MA, et
liver cirrhosis. Gut.al. 2003;52:1194.
Proteomic analysis o lithium-
http://www.ncbi.nlm.nih.gov/pubmed/1968469
230. Abraham WT, Shamshirsaz AA, McFann K, et al. Aquaretic effect of induced
244. Bueminephrogenic
M, D’Annadiabetes insipidus:
R, Di Pasquale G, etmechanisms or aquaporin
al. Urinary excretion 2 down-
of aquaporin-2
194. RittiganS,oral
lixivaptan, Robertson GL, Siggaard
non-peptide, selectiveC,V2etreceptor
al. Identif cation o antagonist,
vasopressin 13 new mu- in regulation
water channel and during
cellularpregnancy.
proli eration.CellProc
PhysiolNatlBiochem.
Acad Sci2001;11:203.
USA. 2008;105:3634.
tations
the NewinYork the Heart
vasopressin-neurophysin
Association functional II class
gene IIinand17 III
kindreds
chronic with amilial
heart failure http://www.ncbi.nlm.nih.gov/pubmed/18296634
245. Ishikawa SE, Saito T, Fukagawa A, et al. Close association of urinary
autosomal
patients. J Amdominant neurohypophyseal
Coll Cardiol. 2006;47:1615. diabetes insipidus. Am J Hum Genet. 216. Christensen
excretion BM, Marples
of aquaporin-2 D, Kim YH,
with appropriate andet inappropriate
al. Changes inarginine
cellular vasopres-
composi-
1996;58:107.
231. Gheorghiade M, Konstam MA, Burnett JC Jr, et al. Short-term clinical tion o kidney collecting
sin-dependent antidiuresis duct
in cells in rats withinlithium-induced
hyponatremia elderly subjects.NDI.
J ClinAm J Physiol
Endocrinol
http://www.ncbi.nlm.nih.gov/pubmed/8554046
effects of tolvaptan, an oral vasopressin antagonist, in patients hospitalized for Cell Physiol.
Metab. 2004;286:C952.
2001;86:1665.
195. failure:
heart Baylis PH, Thompson
the EVEREST CJ. Osmoregulation
Clinical Status Trials. JAMA. o vasopressin secretion and
2007;297:1332. 217. Ranchin
246. Wang W,B,Li Boury-Jamot
C, Kwon TH, M, et al. AQP3, p-AQP2,
Blanchard G, et al.and AQP2 expression
Familial nephrogenic is
thirst Asahina
232. in healthYand disease.
, Izumi Clin Endocrinol
N, Enomoto N, et al.(Ox ). 1988;29:549.
Increased gene expression of water reduced inofpolyuric
syndrome rats with
inappropriate hypercalcemia:
antidiuresis: prevention
dissociation by cAMP-PDE
between aquaporin-2 inhibi-
and
http://www.ncbi.nlm.nih.gov/pubmed/3075528
channel in cirrhotic rat kidneys. Hepatology. 1995;21:169. tors. Am J Physiol
vasopressin RenalJPhysiol.
excretion. 2002;283:F1313.
Clin Endocrinol Metab. 2010;95:E37.
196. Smith D, McKenna K, Moore K, et al. Baroregulation o vasopressin release 218. Wang W, Kwon TH, Li C, et al. Reduced expression o Na-K-2Cl cotrans-
in adipsic diabetes insipidus. J Clin Endocrinol Metab. 2002;87:4564. porter in medullary TAL in vitamin D-induced hypercalcemia in rats. Am J Physiol
http://www.ncbi.nlm.nih.gov/pubmed/12364435 Renal Physiol. 2002;282:F34.
197. Spanakis E, Milord E, Gragnoli C. AVPR2 variants and mutations in 219. Li C, Wang W, Knepper MA, et al. Downregulation o renal aquapo-
nephrogenic diabetes insipidus: review and missense mutation signif cance. J Cell rins in response to unilateral ureteral obstruction. Am J Physiol Renal Physiol.
Physiol. 2008;217:605. 2003;284:F1066.
http://www.ncbi.nlm.nih.gov/pubmed/18726898 220. Apostol E, Ecelbarger CA, Terris J, et al. Reduced renal medullary wa-
198. Robben JH, Knoers NV, Deen PM. Cell biological aspects o the vasopressin ter channel expression in puromycin aminonucleoside-induced nephrotic syn-
type-2 receptor and aquaporin 2 water channel in nephrogenic diabetes insipi- drome. J Am Soc Nephrol. 1997;8:15.
dus. Am J Physiol Renal Physiol. 2006;291:F257. http://www.ncbi.nlm.nih.gov/pubmed/9013444
199. Los EL, Deen PM, Robben JH. Potential o nonpeptide (ant)agonists to res- 221. Bohlin AB, Berg U. Renal water handling in minimal change nephrotic
cue vasopressin V2 receptor mutants or the treatment o X-linked nephrogenic syndrome. Int J Pediat Nephrol. 1984;5:93–98.
diabetes insipidus. J Neuroendocrinol. 2010;22:393. http://www.ncbi.nlm.nih.gov/pubmed/6490323
http://www.ncbi.nlm.nih.gov/pubmed/20163515 222. Fernandez-Llama P, Andrews P, Ecelbarger CA, et al. Concentrating de ect
200. van Lieburg AF, Verdijk MA, Schoute F, et al. Clinical phenotype o nephro- in experimental nephrotic syndrome: altered expression o aquaporins and thick
genic diabetes insipidus in emales heterozygous or a vasopressin type 2 receptor ascending limb Na+ transporters. Kidney Int. 1998;54:170.
mutation. Hum Genet. 1995;96:70. http://www.ncbi.nlm.nih.gov/pubmed/9648076
http://www.ncbi.nlm.nih.gov/pubmed/7607658 223. Mouri T, Inoue T, Nonoguchi H, et al. Acute and chronic metabolic acido-
201. Faerch M, Corydon TJ, Rittig S, et al. Skewed X-chromosome inactivation sis inter eres with aquaporin-2 translocation in the rat kidney collecting ducts.
causing diagnostic misinterpretation in congenital nephrogenic diabetes insipi- Hypertens Res. 2009;32:358.
dus. Scand J Urol Nephrol. 2010;44:324. http://www.ncbi.nlm.nih.gov/pubmed/19300448
202. Chan Seem CP, Dossetor JF, Penney MD. Nephrogenic diabetes insipidus 224. Szatalowicz VL, Arnold PE, Chaimovitz C, et al. Radioimmunoassay o
due to a new mutation o the arginine vasopressin V2 receptor gene in a girl plasma arginine vasopressin in hyponatremic patients with congestive heart ail-
presenting with non-accidental injury. Ann Clin Biochem. 1999;36:779. ure. N Engl J Med. 1981;305:263.
http://www.ncbi.nlm.nih.gov/pubmed/10586320 225. Schrier RW. Vasopressin and aquaporin 2 (AQP2) in clinical disorders o
203. Birnbaumer M. Vasopressin receptor mutations and nephrogenic diabetes water homeostasis. Semin Nephrol. 2008;28:289.
insipidus. Arch Med Res. 1999;30:465. http://www.ncbi.nlm.nih.gov/pubmed/18519089
http://www.ncbi.nlm.nih.gov/pubmed/10714359 226. Nielsen S, Terris J, Andersen D, et al. Congestive heart ailure in rats is
204. Sasaki S, Fushimi K, Saito H, et al. Cloning, characterization, and associated with increased expression and targeting o aquaporin-2 water channel
chromosomal mapping o human aquaporin o collecting duct. J Clin Invest. in collecting duct. Proc Natl Acad Sci USA. 1997;94:5450.
1994;93:1250. http://www.ncbi.nlm.nih.gov/pubmed/9144258
http://www.ncbi.nlm.nih.gov/pubmed/7510718 227. Xu DL, Martin PY, Ohara M, et al. Upregulation o aquaporin-2 water
205. Deen PM, Verdijk MA, Knoers NV, et al. Requirement o human renal water channel expression in chronic heart ailure. J Clin Invest. 1997;99:1500.
channel aquaporin-2 or vasopressin-dependent concentration o urine. Science. 228. Gheorghiade M, Gattis WA, O’Connor CM, et al. E ects o tolvaptan, a
1994;264:92. vasopressin antagonist, in patients hospitalized with worsening heart ailure: A
http://www.ncbi.nlm.nih.gov/pubmed/8140421 randomized controlled trial. JAMA. 2004;291:1963.
(contd.)
CHAPTER 4 REGULATION OF WATER BALANCE: URINE CONCENTRATION AND DILUTION 1159
158
57

229. Ohshiro
162. Schrier RW, K, YGross P, Gheorghiade
aoita E, Yoshida Y, et M, al. et al. Tolvaptan,
Expression and aimmunolocalization
selective oral vaso- 238. FeldmanLK,
192. Hansen BJ,Rittig
Rosenthal SM, Vargas
S, Robertson GA, et basis
GL. Genetic al. Nephrogenic syndrome o
of familial neurohypoph-
pressin
of AQP6V2-receptor
in intercalatedantagonist, or hyponatremia.
cells of the rat kidney collecting N Engl duct.
J Med.Arch
2006;355:2099.
Histol Cytol. inappropriate
yseal diabetes antidiuresis.
insipidus. TrendsN Engl J Med. 2005;352:1884.
Endocrinol Metab. 1997;8:363.
http://www.ncbi.nlm.nih.gov/pubmed/17105757
2001;64:329. http://www.ncbi.nlm.nih.gov/pubmed/15872203
193. Repaske DR, Phillips JA III, Kirby LT, et al. Molecular analysis of autosomal
230. Ikeda
163. Abraham WT, Shamshirsaz
M, Beitz E, Kozono D, AA,etMcFann K, et al. Aquaretic
al. Characterization e ect o lix-
of aquaporin-6 as 239.
dominantDecaux G, Vandergheynstdiabetes
neurohypophyseal F, Bouko insipidus.
Y, et al. Nephrogenic syndromeMetab.
J Clin Endocrinol o in-
aivaptan,
nitrateanchannel
oral non-peptide,
in mammalian selective
cells.V2Requirement
receptor vasopressin antagonist,
of pore-lining residuein appropriate
1990;70:752. antidiuresis in adults: high phenotypic variability in men and wom-
the New Y63.
threonine ork JHeart Association
Biol Chem. unctional class II and III chronic heart ailure
2002;277:39873. en
194.rom a large
Rittig pedigree. GL,
S, Robertson J AmSiggaard
Soc Nephrol. 2007;18:606.
C, et al. Identi cation of 13 new mutations
patients.
164. J Am Coll
Nejsum LN, Cardiol.
Elkjaer M,2006;47:1615.
Hager H, et al. Localization of aquaporin-7 in rat http://www.ncbi.nlm.nih.gov/pubmed/17229917
in the vasopressin-neurophysin II gene in 17 kindreds with familial autosomal
http://www.ncbi.nlm.nih.gov/pubmed/16630999
and mouse kidney using RT-PCR, immunoblotting, and immunocytochemistry. 240.
dominantKanno K, Sasaki S, Hirata
neurohypophyseal Y, etinsipidus.
diabetes al. Urinary Amexcretion o aquaporin-2
J Hum Genet. 1996;58:107. in
231. Gheorghiade
Biochem Biophys Res M, Konstam
Commun. MA, Burnett JC Jr, et al. Short-term clinical e -
2000;277:164. patients
195. BayliswithPH,diabetes insipidus.
Thompson CJ. NOsmoregulation
Engl J Med. 1995;332:1540.
of vasopressin secretion and
ects oRojek
165. tolvaptan, an oralJ, vasopressin
A, Praetorius Frøkiaer J, et antagonist,
al. A current in patients hospitalized
view of the mammalian or http://www.ncbi.nlm.nih.gov/pubmed/7537863
thirst in health and disease. Clin Endocrinol (Oxf). 1988;29:549.
heart ailure: the EVEREST
aquaglyceroporins. Annu Rev Clinical
Physiol.Status Trials. JAMA. 2007;297:1332.
2008;70:301. 241. Pisitkun
196. Smith D, T, Shen RF,K,
McKenna Knepper
Moore K, MA.et Identif cation and proteomic
al. Baroregulation profrelease
of vasopressin ling o
http://www.ncbi.nlm.nih.gov/pubmed/17384438
166. Sohara E, Rai T, Sasaki S, et al. Physiological roles of AQP7 in the kidney: exosomes
in adipsic in humaninsipidus.
diabetes urine. Proc NatlEndocrinol
J Clin Acad Sci USA.
Metab. 2004; 101:13368.
2002;87:4564.
232. Asahina
lessons from AQP7 Y, Izumi N, Enomoto
knockout N, et al.Biophys
mice. Biochim Increased
Acta.gene expression o water
2006;1758:1106. http://www.ncbi.nlm.nih.gov/pubmed/15326289
197. Spanakis E, Milord E, Gragnoli C. AVPR2 variants and mutations in
channel
167. in cirrhotic
Kondo rat kidneys.
H, Shimomura I, Hepatology.
Kishida K,1995;21:169.
et al. Human aquaporin adipose 242. Martin PY
nephrogenic , Abraham
diabetes WT, Lieming
insipidus: reviewX,and et al.missense
Selectivemutation
V2-receptor vasopres-
signi cance.
http://www.ncbi.nlm.nih.gov/pubmed/7528708
(AQPap) gene. Genomic structure, promoter analysis and functional mutation. sin antagonism
J Cell decreases urinary aquaporin-2 excretion in patients with chronic
Physiol. 2008;217:605.
233.J Biochem.
Eur Gerbes AL, Gülberg V Ginès P, et al. Therapy o hyponatremia in cirrhosis
2002;269:1814. heart ailure. JJH,
198. Robben AmKnoers
Soc Nephrol.
NV, Deen1999;10:2165.
PM. Cell biological aspects of the vasopressin
with aIshibashi
168. vasopressin K. receptor
Aquaporin antagonist:
subfamily a randomized
with unusual double-blind
NPA boxes. multicenter
Biochim http://www.ncbi.nlm.nih.gov/pubmed/10505693
type-2 receptor and aquaporin 2 water channel in nephrogenic diabetes
trial. Gastroenterology.
Biophys 2003;124:933.
Acta. 2006;1758:989. 243. Ivarsen
insipidus. Am JP,Physiol
Frøkiaer
RenalJ, Aagaard NK, et al. Increased urinary excretion o
Physiol. 2006;291:F257.
http://www.ncbi.nlm.nih.gov/pubmed/12671890
169. Morishita Y, Matsuzaki T, Hara-chikuma M, et al. Disruption of aquaporin
199. Los 2EL, in patients
Deen PM, withRobben
liver cirrhosis. Gut. 2003;52:1194.
JH. Potential of nonpeptide (ant)agonists
234. Ohara M,produces
aquaporin-11 Martin PY , Xu DL, et
polycystic al. Upregulation
kidneys following ovacuolization
aquaporin 2ofwater the 244. Buemi
to rescue M, D’Anna V2
vasopressin R, Direceptor
Pasqualemutants
G, et al. Urinary
for the excretion
treatmento aquaporin-2
of X-linked
channel expression
proximal tubule. Mol inCell
pregnant rats. J Clin Invest. 1998;101:1076.
Biol. 2005;25:7770. water channeldiabetes
nephrogenic during insipidus.
pregnancy.JCell Physiol Biochem.
Neuroendocrinol. 2001;11:203.
2010;22:393.
http://www.ncbi.nlm.nih.gov/pubmed/9486978
170. Okada S, Misaka T, Tanaka Y, et al. Aquaporin-11 knockout mice and 245.
200. vanIshikawa
LieburgSE, AF
Saito T, Fukagawa
, Verdijk A, et al. FClose
MA, Schoute association
, et al. o urinary ex-
Clinical phenotype of
235. Ishikawa
polycystic kidney SE,disease
Saito T, Saito T,
animals et al.a common
share Pathophysiological
mechanismrole o aquaporin-2
of cyst formation. cretion o aquaporin-2
nephrogenic with appropriate
diabetes insipidus in females and inappropriate
heterozygous arginine vasopres-
for a vasopressin type 2
in impaired
FASEB water excretion. Prog Brain Res. 2008;170:581.
J. 2008;22:3672. sin-dependent
receptor mutation. antidiuresis
Hum Genet. in hyponatremia
1995;96:70. in elderly subjects. J Clin Endocrinol
http://www.ncbi.nlm.nih.gov/pubmed/18655911
171. Yakata K, Tani K, Fujiyoshi Y. Water permeability and characterization of Metab. 2001;86:1665.
201. Faerch M, Corydon TJ, Rittig S, et al. Skewed X-chromosome inactivation
236. Saito T, Ishikawa
aquaporin-11. S, 2011;174:
J Struct Biol. Abe K, et al. 315. Acute aquaresis by the nonpeptide ar- 246.
causing Ranchin B, Boury-Jamot
diagnostic M, Blanchard
misinterpretation G, et al. Familial
in congenital nephrogenic
nephrogenic syn-
diabetes
ginineUchida
172. vasopressin (AVP)E,antagonist
S, Sohara Rai T, et OPC-31260
al. Impaired improves hyponatremia
urea accumulation in thein drome o inappropriate
insipidus. antidiuresis:
Scand J Urol Nephrol. dissociation between aquaporin-2 and vaso-
2010;44:324.
patients
inner with syndrome
medulla of mice olacking
inappropriate
the urea secretion o antidiuretic
transporter UT-A2. Mol hormone (SI-
Cell Biol. pressin
202. Chan excretion.
Seem JCP Clin Endocrinol
, Dossetor JF, Metab.
Penney2010;95:E37.
MD. Nephrogenic diabetes insipidus
ADH). J Clin Endocrinol Metab. 1997;82:1054.
2005;25:7357. due to a new mutation of the arginine vasopressin V2 receptor gene in a girl
237. Sands
173. Kazama JM,I, Gargus
Hatano JJ,R, Fröhlich
Michimata O,M, et et
al.al.Urinary
BSC1 inhibition complements
concentrating ability in presenting with non-accidental injury. Ann Clin Biochem. 1999;36:779.
e ects owith
patients vasopressin V2 receptor
Jk(a-b-) blood type who antagonist on hyponatremia
lack carrier-mediated in SIADH Jrats.
urea transport. Am 203. Birnbaumer M. Vasopressin receptor mutations and nephrogenic diabetes
Kidney
Soc Int. 2005;67:1855.
Nephrol. 1992;2:1689. insipidus. Arch Med Res. 1999;30:465.
http://www.ncbi.nlm.nih.gov/pubmed/15840033
174. Yoshikawa M, Uchida S, Yamauchi A, et al. Localization of rat CLC-K2 204. Sasaki S, Fushimi K, Saito H, et al. Cloning, characterization, and chromosomal
chloride channel mRNA in the kidney. Am J Physiol. 1999;276:F552. mapping of human aquaporin of collecting duct. J Clin Invest. 1994;93:1250.
175. Kobayashi K. Uchida S, Mizutani S, et al. Intrarenal and cellular localiza- 205. Deen PM, Verdijk MA, Knoers NV, et al. Requirement of human renal water
tion of ClC-K2 protein in the mouse kidney. J Am Soc Nephrol. 2001;12:1327. channel aquaporin-2 for vasopressin-dependent concentration of urine. Science.
176. Hebert SC. Bartter syndrome. Curr Opin Nephrol Hypertens. 2003;12:527. 1994;264:92.
177. Simon DB, Bindra RS, Mans eld TA, et al. Mutations in the chloride chan- 206. Loonen AJ, Knoers NV, van Os CH, et al. Aquaporin 2 mutations in
nel gene, CLCNKB, cause Bartter’s syndrome type III. Nat Genet. 1997;17:171. nephrogenic diabetes insipidus. Semin Nephrol. 2008;28:252.
178. Bartter FC, Pronove P, Gill JR Jr, et al. Hyperplasia of the juxtaglomerular 207. Savelkoul PJ, De Mattia F, Li Y, et al. p.R254Q mutation in the aquaporin-2
complex with hyperaldosteronism and hypokalemic alkalosis. A new syndrome. water channel causing dominant nephrogenic diabetes insipidus is due to a lack
Am J Med. 1962;33:811. of arginine vasopressin-induced phosphorylation. Hum Mutat. 2009;30:E891.
179. Simon DB, Karet FE, Hamdan JM, et al. Bartter’s syndrome, hypokalaemic 208. de Mattia F, Savelkoul PJ, Kamsteeg EJ, et al. Lack of arginine vasopressin-
alkalosis with hypercalciuria, is caused by mutations in the Na-K-2Cl induced phosphorylation of aquaporin-2 mutant AQP2-R254L explains
cotransporter NKCC2. Nat Genet. 1996;13:183. dominant nephrogenic diabetes insipidus. J Am Soc Nephrol. 2005;16:2872.
180. Takahashi N, Chernavvsky DR, Gomez RA, et al. Uncompensated polyuria 209. Kamsteeg EJ, Savelkoul PJ, Hendriks G, et al. Missorting of the aquaporin-2
in a mouse model of Bartter’s syndrome. Proc Natl Acad Sci USA. 2000;97:5434. mutant E258K to multivesicular bodies/lysosomes in dominant NDI is associated
181. Kaplan MR, Plotkin MD, Brown D, et al. Expression of the mouse with its monoubiquitination and increased phosphorylation by PKC but is due to
Na-K-2Cl cotransporter, mBSC2, in the terminal inner medullary collecting the loss of E258. P ugers Arch. 2008;455:1041.
duct, the glomerular and extraglomerular mesangium and the glomerular 210. Asai T, Kuwahara M, Kurihara H, et al., Pathogenesis of nephro-
afferent arteriole. J Clin Invest. 1996;98:723. genic diabetes insipidus by aquaporin-2 C-terminus mutations. Kidney Int.
182. Ginns SM, Knepper MA, Ecelbarger CA, et al. Immunolocalization of the 2003;64:2.
secretory isoform of the Na-K-Cl cotransporter in rat renal intercalated cells. 211. Sohara E, Rai T, Yang SS, et al., Pathogenesis and treatment of autosomal-
J Am Soc Nephrol. 1996;7:2533. dominant nephrogenic diabetes insipidus caused by an aquaporin 2 mutation.
183. Wall SM, Knepper MA, Hassell KA, et al. Hypotension in NKCC1 null Proc Natl Acad Sci USA. 2006;103:14217.
mice: role of the kidneys. Am J Physiol Renal Physiol. 2006;290:F409. 212. Lloyd DJ, Hall FW, Tarantino LM, et al. Diabetes insipidus in mice with a
184. Xu JZ, Hall AE, Peterson LN, et al. Localization of the ROMK protein on mutation in aquaporin-2. PLoS Genet. 2005;1(2):e20.
apical membranes of rat kidney nephron segments. Am J Physiol. 1997;273:F739. 213. Grünfeld JP, Rossier BC. Lithium nephrotoxicity revisited. Nat Rev Nephrol.
185. Simon DB, Karet FE, Rodriguez-Soriano J, et al. Genetic heterogeneity of Bartter’s 2009;5:270.
syndrome revealed by mutations in the K channel, ROMK. Nat Genet. 1996;14:152. 214. Rao R, Zhang MZ, Zhao M, et al. Lithium treatment inhibits renal GSK-3
186. Lorenz JN, Baird NR, Judd LM, et al. Impaired renal NaCl absorption activity and promotes cyclooxygenase 2-dependent polyuria. Am J Physiol Renal
in mice lacking the ROMK potassium channel, a model for type II Bartter’s Physiol. 2005;288:F642.
syndrome. J Biol Chem. 2002;277:37871. 215. Nielsen J, Hoffert JD, Knepper MA, et al. Proteomic analysis of lithium-
187. Schultheis PJ, Clarke LL, Meneton P, et al. Renal and intestinal absorptive induced nephrogenic diabetes insipidus: mechanisms for aquaporin 2 down-
defects in mice lacking the NHE3 Na /H exchanger. Nat Genet. 1998;19:282. regulation and cellular proliferation. Proc Natl Acad Sci USA. 2008;105:3634.
188. Lorenz JN, Schultheis PJ, Traynor T, et al. Micropuncture analysis of single- 216. Christensen BM, Marples D, Kim YH, et al. Changes in cellular composi-
nephron function in NHE3-de cient mice. Am J Physiol. 1999;277:F447. tion of kidney collecting duct cells in rats with lithium-induced NDI. Am J Physiol
189. Amlal H, Ledoussal C, Sheriff S, et al. Downregulation of renal AQP2 water Cell Physiol. 2004;286:C952.
channel and NKCC2 in mice lacking the apical Na -H exchanger NHE3. 217. Wang W, Li C, Kwon TH, et al. AQP3, p-AQP2, and AQP2 expression
J Physiol. 2003;553:511. is reduced in polyuric rats with hypercalcemia: prevention by cAMP-PDE
190. Ecelbarger CA, Kim GH, Terris J, et al. Vasopressin-mediated regulation of inhibitors. Am J Physiol Renal Physiol. 2002;283:F1313.
ENaC abundance in rat kidney. Am J Physiol Renal Physiol. 2000;279:F46. 218. Wang W, Kwon TH, Li C, et al. Reduced expression of Na-K-2Cl
191. Snyder PM. Minireview: regulation of epithelial Na channel traf cking. cotransporter in medullary TAL in vitamin D-induced hypercalcemia in rats. Am
Endocrinology. 2005;146:5079. J Physiol Renal Physiol. 2002;282:F34.
C H A P T E R

5 Tubular Sodium Transport


Arohan R. Subramanya • Núria M. Pastor-Solar • W. Brian Reeves •
Kenneth R. Hallows

N
a , the primary extracellular cation, is of critical im- high rates of Na transport (proximal segments) and highly
portance to the maintenance of extracellular uid regulated Na transport (distal segments).
volume. The kidneys play the dominant role in reg- Substantial progress has been made recently in identify-
ulating Na excretion. Each day, the glomeruli lter roughly ing the proteins that mediate Na transport in each nephron
25,000 mEq of Na . From this quantity, almost 10 times the segment and in de ning their interactions and regulation
total exchangeable Na in the body, the kidneys typically ab- within each segment. Many Na transport proteins have been
sorb over 99%. A remarkable feature of the Na absorptive linked to speci c genetic disorders (Table 5.1). Given the pri-
process is the precision with which it is regulated. An indi- macy of renal Na transport to the control of extracellular uid
vidual consuming a typical diet containing 6 g of Na will volume, it is not surprising that the majority of these genetic
excrete 260 mEq of Na per day. The same individual placed disorders are characterized by either hypotension or hyperten-
on a 2-g Na -restricted diet will promptly reduce Na excre- sion. An updated and curated database of these genetic disor-
tion to 87 mEq per day. Although the fraction of ltered Na ders is maintained at the Online Mendelian Inheritance in Man
absorbed by the kidney changes from 99.0% on a standard website (www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=OMIM).
diet to 99.6% on a Na -restricted diet, this small change This chapter considers the transepithelial transport of
represents the addition or removal of over 1 L per day to Na by the various nephron segments. The discussion of
extracellular uid volume. Thus, the kidneys absorb large each nephron segment begins with a description of the gen-
amounts of ltered Na with remarkably precise control. eral features of Na transport in that segment along with
The exquisitely sensitive regulation of Na absorption pertinent structure–function relations. The mechanism of
by the kidneys relies on sequential actions of the various Na transport is then considered on a cellular or subcellular
nephron segments, each with highly specialized transport level, with emphasis on recent electrophysiologic, biochemi-
capabilities. Figure 5.1 provides an overview of Na trans- cal, and molecular ndings. Finally, each section includes a
port along the nephron. In general, the absolute rates of Na consideration of the factors that regulate Na transport in
reabsorption are greatest in the proximal tubule and fall as the individual segments.
the tubular uid proceeds from proximal to distal segments.
Conversely, the ability to transport Na against steep tubu-
lar uid to blood gradients and its physiologic control in- PRINCIPLES OF MEMBRANE
crease along the nephron. For example, the proximal tubule TRANSPORT
reabsorbs the bulk (60% to 70%) of the ltered Na load,
This section describes physical principles that underlie the
but as will be detailed later, does so against at most small
movement of ions across individual membranes and epithelia.
electrochemical gradients. Moreover, the ability to alter Na
However, it is not intended to be an extended treatment of
transport in the proximal tubule, in relative terms, is rather
the thermodynamic aspects of membrane transport processes.
limited, usually varying by less than 25%. The collecting
duct, in contrast, reabsorbs only a minor fraction ( 2% to
4%) of the ltered Na load. However, the collecting duct Diffusion Processes
can transport Na against a large electrochemical gradient Solute transport across membranes may occur by diffu-
to produce urine, which is almost Na free ( 10 mEq/L). In sion or convection, or by a mediated process. Diffusion is
addition, the rate of Na transport in the collecting duct can the random Brownian motion of a molecule with respect
vary over a wide range (tenfold) in response to physiologic to adjacent molecules and occurs as the consequence of
stimuli. The different nephron segments thus permit both thermal energy.1 Because the diffusional movement of an

159
160
16 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Convective Processes
Convection is the vectorial movement of an ensemble of
molecules and is driven by an externally imposed force
(e.g., hydrostatic pressure). Examples of convective transport
include glomerular ltration and solvent drag, a process in
which solute movement is coupled to water movement.
Bulk water ow may be driven by hydrostatic pressure
and/or osmotic pressure. The familiar Starling equation:

Jv K( P ) (3)

describes net volume ow (Jv) in response to hydrostatic


( P) and osmotic ( ) pressure differences. The equiv-
alence of osmotic and hydrostatic pressure is explicit
in the Starling equation. The degree to which a solute
exerts an osmotic pressure depends on the degree to
which it permeates membranes. The ratio of the observed
osmotic pressure to that predicted if a solute were
excluded absolutely from a membrane is termed the re-
ection coef cient, :

FIGURE 5.1 The contribution of various nephron segments obs/ theoretical (4)
to Na transport. PCT, proximal convoluted tubule; DCT, distal
convoluted tubule; CCD, cortical collecting duct; TAL, thick For impermeant solutes, 1; for highly permeable solutes,
ascending limb; IMCD, inner medullary collecting duct. approaches 0.
For solutes with 1, transmembrane solute ux
will be accelerated in the direction of volume ow. This
individual molecule is random, a concentration gradient is
acceleration is known as solvent drag.4 Thus, the net pas-
required for any net transfer of molecules to occur across
sive ux of a permeable solute across a membrane may be
a membrane. Thus, the concentration gradient represents
driven by both diffusion and entrainment with solvent ow
the driving force for net transport.
(i.e., solvent drag).
For charged solutes, the driving force for transport is the
sum of the chemical and electrical potential gradients. The
Nernst equation describes the equilibrium condition for a Facilitated Diffusion
membrane permeable only to a single ionic species: Biologic membranes are composed primarily of lipid bi-
layers. Because the permeability of many hydrophilic sol-
Vm V2 V1 (RT/ZF) ln (C2/C1) (1) utes through lipid membranes is low, membranes contain
proteins that facilitate the transport of certain solutes.
where R is the gas constant, T is the absolute temperature, Transport proteins, often termed carriers or transporters,
Z is the valence of the solute, F is the Faraday constant, and have a high degree of speci city for the transported solute.
C and V are concentration and electrical potential terms, Flux through the limited number of transporters saturates
respectively. At equilibrium, then, the voltage (Vm) across an as the solute concentration is increased. An example of
ideally selective membrane is de ned by the concentrations carrier-mediated facilitated diffusion is the entry of glucose
of the permeant ion on both sides of the membrane, C2 and into renal tubular cells mediated by the hexose transporter,
C1, respectively. For systems containing more than one per- GLUT-1.5 The movement of ions through ion channels rep-
meant ion, the equilibrium voltage can be described by the resents another form of facilitated diffusion. In this case,
Goldman–Hodgkin–Katz (GHK) equation 2,3: integral membrane proteins containing several membrane-
spanning domains form pores in cell membranes through
Vm RT/F ln [(PNaC2Na PKC2K PClC1Cl)/ which ions permeate. Ion channels generally have a high
(PNaC1Na PK C1K PCl C2Cl)] (2) degree of speci city for the ions being transported and very
high transport rates. Facilitated diffusion mechanisms, like
where Px is the permeability of the respective solutes, in this enzymes, serve only to accelerate the rate of transport,
case, Na , K , and Cl . Thus, in a system containing mul- but do not affect the equilibrium distribution of solutes.
tiple charged solutes, the transmembrane voltage is a func- In other words, facilitated diffusion, like simple diffusion
tion of the relative concentrations and permeabilities of each and convection, is a passive process that tends to dissipate
solute on the two sides of the membrane. transmembrane gradients.
T A B L E
I
n h e r
i t e d D
i s o r
d e r
s o f R
e n a l S
o d i
u m T
r a n s p o r
t
5 . 1
D
A
G
d
e
t
c
e
f
N
#
M
I
M
O
t
c
u
d
o
r
P
e
n
D
n
o
r
h
p
B
s
r
e
d
r
o
s
i
D
e
v
i
s
n
e
t
o
yp
H
s
e
r
u
t
a
e
F
l
a
c
i
n
i
l
C
s
e
c
n
e
u
q
e
s
n
o
C
l
a
n
o
i
t
c
n
u
F
n
o
i
t
u
b
i
r
t
s
N
)
l
a
t
a
n
e
t
n
a
(
1
e
p
y
t
,
r
e
t
t
r
(
2
C
C
6
)
1
A
2
1
C
L
D
L
A
T
a
N
d
e
s
a
e
r
c S
0
a
f
e
e
e
i
i
K s
0 o
8 r
3
d 9
e r
- K - 2 C l B
s
i
os
l
a
k
l
a
c
i
ol
b
a
t
e
m
,
a
i
m
e
l
a
ok
p
y
h
,
on
i
s
n
e
ot
p
y
H
t
or
p
s
n
a
r
ot
c
R
)
l
a
t
a
n
e
t
n
a
(
2
e
p
y
t
,
r
e
t
t
r
D
D
C
C
,
L
A
T
9
5
3
0
0
6
)
1
J
N
C
K
(
K
M
K
l
a
c
i
p
a
d
e
s
a
e
r
c a
O e
B
s
ou
i
v
e
r
p
s
a
e
m
a
S
l
C
l
a
r
e
t
a
ol
s
a
b
d
e
s
a
e
r
c
e
D
L
A
T
3
2
0
2
0
6
b
K
-
C
l
C
)
e
l
i
t
n
a
f
n
i
(
3
e
p
y
t
,
r
e
t
t
r r a
e c
y c l i
n g
B
a
i
m
e
n
i
n
e
r
r
e
p
y
h
,
s
i
os
l
a
k
l
a
,
a
i
m
e
l
a
ok
p
y
H
l
C
l
a
r
e
t
a
l
o
s
ba
d
e
s
a
e
r
c
e
D
r
a
e
r
e
n
n
i
,
L
A
T
,
L
A
t
2
1
4
6
0
6
n
i
t
t
r
a
b
4
e
p
y
t
,
r
e
t
t
r a u x
a
C
of
on
i
t
a
t
u
m
g
n
i
t
a
v
i
t
c
A
n
i
a
r
b
,
d
oi
r
y
h
t
,
d
oi
r
y
h
t
a
r
a
p
,
L
A
T
,
T
P
9
9
1
1
0
6
R
S
a
C
5
e
p
y
t
,
r
e
t
t
r
a
B
s
s
e
n
f
a
e
d
l
a
r
u
e
n
i
or
s
n
e
s
h
t
i
w
,
s
ou
i
v
e
r
p
s
A u x
2
K
d
e
s
a
e
r
c
e
d
h
t
i
w
or
t
p
e
c
e
r
g
n
i
s
n
e
s
-
E
a
i
m
e
s
e
n
g
a
om
p
y
h
,
a
i
r
u
i
c
l
a
oc
p
y
h
,
s
i
os
l
a
k
l
a
c
i
ol
b
a
t
e
m
,
a
i
m
e
l
a
ok
p
y
H
t
or
p
s
n
a
r
t
o
c
l
C
a
N
d
e
s
a
e
r
c
e
D
T
C
D
8
6
9
0
0
6
)
3
A
2
1
L
C
S
(
T
C
C
N
n
a
m
l
e
t
i
G
a
i
m
e
c
l
a
oc
p
y
h
,
a
i
m
e
n
i
n
e
r
r
e
p
y
h
,
s
i
os
l
a
k
l
a
,
a
i
m
e
l
a
ok
p
y
H
on
i
t
p
or
s
b
a
e
r
l
C
a
N
K
l
a
r
e
t
a
l
o
s
ba
d
e
s
a
e
r
c
e
D
T
C
D
0
8
7
2
1
6
)
0
1
J
N
C
K
(
1
.
4
r
i
K
E
M
A
S
e
S
/
T
S r
A e
c y
c l i
n g a n d
CHAPTER 5

d
a
N
l
a
r
e
t
a
l
o
s
a
b
d
e
s
a
e
r
c r e
e c
y c l i
n g
;
K
,
-
d
l
a
r
u
e
n
i
or
s
n
e
s
,
a
i
x
a
t
a
,
y
s
p
e
l
i
p
e
,
a
i
m
e
s
e
n
g
a
om
p
y
h
,
a
i
r
u
i
c
l
a
oc
p
y
h
,
s
i
os
l
a
k
l
a
c
i
ol
b
a
t
e
m
,
a
i
m
e
l
a
ok
p
y
H
n
f
a
P
s
s
B
,
1A
N
N
C
S
(
s
t
i
n
bu
u
s
C
a
N
E
)
e
v
i
s
s
e
c
e
r
l
a
m
o
s
o
t
u
a
(
1
e
p
y
t
m
s
i
n
o
r
e
t
s
o
d
l
a
o
p
y
h
o
d
u
e
N
e
v
i
t
i
s
n
e
s
-
e
d
i
or
l
i
m
a
d
e
s
a
e
r
c
e
D
t
c
u
d
g
n
i
t
c
e
l
ol
C
0
5
3
4
6
2
)
G A
s
,
e
e
a T P a s
e a ct i v i t
y
H
P
s
i
os
d
i
c
a
c
i
ol
b
a
t
e
m
,
a
i
m
e
l
a
k
r
e
p
y
h
,
on
i
s
n
e
ot
p
y
M
)
t
n
a
n
i
m
o
d
l
a
m
o
s
o
t
u
a
(
1
e
p
y
t
m
s
i
n
o
r
e
t
s
o
d
l
a
o
p
y
h
o
d
u
e
r
d
i
o
c
i
t
r
o
c
o
l
a
r
e
n
(
r
o
t
p
e
c
(
a
i
m
e
l
a
k
r
e
p
y
h
,
on
i
s
n
e
ot
p
y
H
s
d
oi
c
i
t
or
oc
l
a
r
e
n
i
m
o
t
e
s
on
p
s
e
r
d
e
s
a
e
r
c
e
D
t
c
u
d
g
n
i
t
c
e
l
ol
C
5
3
7
7
7
1
)
2
C
3
R
(
)
e
v
i
s
s
e
c
e
r
n
a
h
t
e
r
e
v
e
s
s
s
e
)
d
e
u
n
i
t
n
o t
N
c
e
l
s r
i a
n s p or t
TUBULAR SODIUM TRANSPORT
61
1161
162
16 2

T A B L E
SECTION I

I
(
)
d
e
u
n
i
t
n
o n
c h e r i
t e d D i
s o r d e r
s o f R e n a l S o d i
u m T
r a n s p o r t
5 . 1
D
A
G
d
e
t
c
e
f
N
#
M
I
M
O
t
c
u
d
o
r
P
e
n
D
n
o
r
h
p
L
s
r
e
d
r
o
s
i
D
e
v
i
s
n
e
t
r
e
yp
H
s
e
r
u
t
a
e
F
l
a
c
i
n
i
l
C
s
e
c
n
e
u
q
e
s
n
o
C
l
a
n
o
i
t
c
n
u
F
n
o
i
t
u
b
i
r
t
s
e
l
d
d i
f
e
e
i
i s o r
d e r or (
C
a
N
S
,
B
1
N
N
C
P
e
d
i
or
l
i
m
a
o
t
e
v
i
s
on
p
s
e
r
,
s
i
os
l
a
k
l
a
c
i
ol
b
a
t
e
m
,
a
i
m
e
l
a
ok
p
y
h
,
on
i
s
n
e
t
r
e
p
y
H
s
l
e
n
n
a
h
c
C
a
N
E
of
y
t
i
v
i
t
c
a
d
n
a
on
i
s
s
e
r
p
x
e
e
c
a
f
r
u
s
l
l
e
c
d
e
s
a
e
r
c
n
I
t
c
u
d
g
n
i
t
c
e
l
ol
C
0
0
2
7
7
1
)
G
I
N
N
C
r
a
l
u
l
l
e
c
a
r
a
p
d
e
s
a
e
r
c
n
i
,
t
or
p
s
n
a
r
ot
c
l
C
a
N
d
e
s
a
e
r
c
n
I
t
c
u
d
g
n
i
t
c
e
l
ol
c
,
T
C
D
0
6
2
5
4
1
4
K
N
W
or
1
K
N
W
)
n
o
d
r
o
G
(
2
e
p
y
t
m
s
i
n
o
r
e
t
s
o
d
l
a
o
p
y
h
o
d
u
e E
S
C
s l
-
H
or
t
p
e
c
e
r
d
oi
c
i
t
or
oc
l
a
r
e
n
i
m
of
on
i
t
a
v
i
t
c
a
g
n
i
s
u
a
c
s
d
oi
c
i
t
or
oc
c
u
l
g
of
on
i
t
a
d
i
ox
d
e
s
a
e
r
c
e
D
t
c
u
d
g
n
i
t
c
e
l
ol
c
,
T
C
D
0
3
0
8
1
2
2
D
S
t
e
v
i
s
n
o
p
s
e
r
,
s
i
s
o
l
ka
l
a
c
i
l
bo
a
t
e
m
,
a
i
m
e
l
a
k
o
p
y
h
,
n
o
i
s
n
e
t
r
e
p
m
a
x
e
d
t
O
e
n
o
s
a
D
e
l
u
b
u
t
l
a
m
i
ox
r
P
0
0
1
3
3
2
)
2
A
5
C
L
S
(
2
T
L
G
S
a
i
r
u
os
c
u
l
g
l
a
n
e
R
s
r
e
d
r
o
s
i
D
r
e
h
a
N
d
e
s
a
e
r
c o
h
H
e
y
te A
s
c
i
t
e
r
u
i
d
e
d
i
z
a
i
h
t
o
t
e
v
i
s
on
p
s
e
r
,
s
i
os
d
i
c
a
c
i
ol
b
a
t
e
m
,
a
i
m
e
l
a
k
r
e
p
y
h
,
on
i
s
n
e
t
r
e
p
y
H
e
d
oi
c
i
t
or
oc
l
a
r
e
n
i
m
t
n
e
r
a
p
c
1 p
x
e
1
p e
s
s
r m e
a b i
l i
t y
-
D
e
l
u
b
u
t
l
a
m
i
ox
r
P
5
4
3
3
0
6
)
4
A
4
C
L
S
(
1
C
B
N
A
T
R
l
a
m
i
ox
r
P
a
i
r
u
os
c
u
l
g
or
f
d
ol
h
s
e
r
h
t
d
e
s
a
e
r
c
e
D
on
i
t
p
or
s
b
a
a
N
d
e
s
a
e
r
c c
e
ou p l
e d g
l u c
os e
- c
ou p l
e d H C O m
l
a
r
e
t
a
ol
s
a
b
s
os
r
c
a
t
or
p
s
n
a
r
t
m
r
on
i
t
a
d
r
a
t
e
r
l
a
t
n
e
m
d
n
a
h
t
ow
r
g
,
s
e
i
t
i
l
a
m
or
n
b
a
l
a
t
n
e
d
d
n
a
r
a
l
u
oc
,
s
i
os
d
i
c
a
c
i
ol
b
a
t
e
M
e
n a e
b
3
STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

O
.
M
I
M
O
=
b
d
?
i
g
c
f
.
y
r
e
u
q
/
z
e
r
t
n
e
/
ov
g
.
h
i
n
.
m
l
n
.
i
b
c
n
.
w
w
w
/
/
:
p
t
t
h
t
a
e
l
b
a
l
i
a
v
a
s
e
c
n
e
r
e
f
e
r
d
e
t
a
d
p
U
.
s
i
os
d
i
c
a
r
a
l
u
b
u
t
l
a
n
e
r
,
A
T
R
;
e
c
n
a
l
a
b
m
i
e
t
y
ol
r
t
c
e
l
e
d
n
a
,
on
i
t
a
d
r
a
t
e
r
l
a
t
n
e
m
,
a
i
x
a
t
a
,
s
s
e
n
f
a
e
d
l
a
r
u
e
n
i
or
s
n
e
s
,
s
e
r
u
z
i
e
s
,
E
M
A
S
e
S
;
y
h
t
a
op
l
u
b
u
t
d
n
a
,
s
s
e
n
f
a
e
d
,
l
a
r
u
e
n
i
or
s
n
e
s
,
a
i
x
a
t
a
,
y
s
p
e
l
i
p
e
,
T
S
A
E
;
e
l
u
b
u
t
d
e
t
u
ol
v
on
c
l
a
t
s
i
d
,
T
C
D
;
e
l
u
b
u
t
l
a
m
i
ox
r
p
,
T
P
;
b
m
i
l
g
n
i
d
n
e
c
s
a
n
i
h
t
,
L
A
t
;
b
K
l
e
n
n
a
h
c
e
d
i
or
l
h
c
,
b
K
-
C
l
C
;
l
e
n
n
a
h
c
K
y
r
a
l
l
u
d
e
m
r
e
t
ou
l
a
n
e
r
,
K
M
O
R
;
e
l
u
b
u
t
d
e
t
u
ol
v
on
c
l
a
t
s
i
d
, M I M # ; a
c c e s
s i
on n u m b e
r f or O n l
i n e Me n d e l
i a n I
n h e
r i
t a n c
e i
n M a
n ; T
A L
, t h i
c k a
s c
e n d i
n g l
i m b ; C
C D
, c
or t i
c a
l c
ol le ct i
n g d u c
t ; D
C T
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1163
63

Active Transport Processes ltrate.14 Second, the absorption of sodium in the proximal
Active transport is a special case of facilitated transport in tubule provides, by way of coupled processes, the driving
which chemical bond energy is supplied to the transport force for the absorption of other solutes, such as bicarbon-
process so that the nal distribution of the solute is remote ate, glucose, phosphate, and amino acids.
from equilibrium. The coupling of solute transport to the en- Under most circumstances, uid at any given point
ergy source can take two forms. In primary active transport, along the proximal tubule has virtually the same Na con-
solute transport is coupled directly to an energy-yielding centration and osmolality as plasma.13 The isosmotic na-
reaction. ture of proximal tubule uid absorption derives from the
The most widely recognized example of primary active high water permeability of this segment,15 which effectively
transport is the transport of Na and K by the Na ,K - clamps the osmolality of the tubular uid at that of plasma.
ATPase. This enzyme, often referred to as the sodium pump, Although Na transport in the proximal tubule occurs in
couples the extrusion of cellular Na to cellular K uptake.6 the absence of large electrical or chemical gradients, the
In renal tubules, this enzyme is localized to the basolateral bulk of Na absorption in the proximal tubule involves ac-
membrane. In general, segments with high rates of active tive transport. For example, Na can be reabsorbed against
Na transport have high Na ,K –ATPase activity.7 The hy- both concentration 13,16 and electrical17 gradients. In ad-
drolysis of each ATP molecule ordinarily pumps three so- dition, uid absorption and sodium transport cease when
dium ions out of the cell coupled to two potassium ions Na ,K –ATPase activity is inhibited or when cell metabo-
moving inward.8 Therefore, the pump is electrogenic. The lism is slowed.18,19
Na ,K –ATPase is responsible for maintaining the cell Na A signi cant amount of proximal Na transport also
activity at a low level, which provides the energy for the occurs passively.20 For example, in the late convoluted and
Na -coupled transport of many other solutes. Thus, the straight tubules (S2 and S3 segments), Na diffuses passively
inhibition of Na ,K –ATPase (e.g., by peritubular ouabain out of the tubule driven by the lumen-positive electrical
addition) causes a signi cant rise in the cell Na activity.9 potential difference in those segments. This potential differ-
The af nity constant (Km) of the pump for intracellular so- ence derives from a Cl concentration gradient across the
dium, about 15 to 30 mM,10 is similar to the intracellular tubule wall. Even in this case, however, it is the active trans-
sodium activity measured in proximal tubule cells.9 There- port of Na in upstream portions of the proximal nephron
fore, the pump is unsaturated with respect to sodium, and that ultimately accounts for these gradients and potentials.
pump activity is very sensitive to changes in the intracellular
sodium concentration activity. Nephron Heterogeneity
In secondary active transport, solute movement against Analyses of proximal tubular Na transport are complicated
its electrochemical gradient is energized by the movement by two factors: a nonhomogeneous nephron population and
of another solute down its own gradient.11 Na , because of axial changes in uid composition. There is considerable het-
its steep inward electrochemical gradient maintained by the erogeneity of both morphologic and functional characteris-
sodium pump, often participates in the transport of other tics along the proximal tubule. The S1 segment cells have
solutes, either in the same (cotransport or symport) or op- extensive basal interdigitations, numerous mitochondria, and
posite (exchange or antiport) direction. Thus, by coupling a well-developed luminal brush border.21 S3 segment cells, in
solute transport with sodium movement into cells, cellular contrast, are atter and have fewer mitochondria, lower brush
metabolic energy generated by the Na ,K –ATPase is stored borders, and much less extensive basolateral membranes
in the form of a Na concentration gradient, analogous to a than S1 cells.21 The Na ,K –ATPase activity of the S3 seg-
battery, and then dissipated in the transport of a variety of ment is only 25% of that for the S1 segments.22 As might be
different solutes. Some examples of symport processes in- expected on the basis of these observations, the net rates of
clude Na -glucose and Na -amino acid cotransport; Na - the Na and uid transport in the S3 straight segment are, in
proton and Na -calcium exchange are two examples of an- general, lower than in the S1 convoluted tubule.23
tiport processes. Juxtamedullary proximal convoluted tubules have high-
er rates of volume and bicarbonate absorption than their
PROXIMAL TUBULE super cial counterparts,23,24 although this disparity has not
been noted between juxtamedullary and super cial straight
General Features segments.25
The proximal tubule is the major site for Na absorption Glomerular ultra ltrate in the early proximal tubule
within the kidney and serves two major purposes. First, undergoes axial composition changes. Figure 5.2 illus-
the proximal tubule protects the extracellular uid volume trates that the chloride concentration rises as a conse-
by reclaiming the bulk, approximately 60% to 80%, of the quence of the preferential absorption of NaHCO3 over
glomerular ltrate.12,13 The proximal tubule with its well- NaCl in this segment.26 Glucose, amino acids, and other
developed brush border membrane is optimally designed organic compounds are also absorbed avidly in associa-
to perform the reabsorption of such a large fraction of the tion with Na so that their luminal concentrations in this
164
16 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Electrical Resistance
The electrical resistance of the mammalian proximal tubule
is remarkably low, making this tubule a classic example of
a leaky epithelium, with resistances of 5 to 10 -cm2.36,37
In the proximal tubule, the total cellular resistance (i.e., the
sum of the apical and basolateral resistance) is 20- to 70-fold
greater than the transepithelial resistance. This indicates that
the paracellular resistance is low and that the predominant
route for passive ion ows in the proximal tubule involves
the paracellular pathway.

Ionic Selectivity
The initial convolution of the super cial proximal tubule is Na
selective; thereafter, the super cial convoluted and straight
tubules are Cl– selective.29,38 In contrast, juxtamedullary
proximal tubules are Na selective throughout their course.38
Because the Cl– concentration rises as uid ows along the
convoluted tubule (Fig. 5.2), oppositely directed gradients for
Cl– and HCO3– in late convoluted and straight segments give
rise to a lumen-positive transepithelial potential difference.26
The latter indicates a higher permeability for Cl– than HCO3–,
FIGURE 5.2 The pro le of transepithelial voltage and solute both in super cial and juxtamedullary proximal tubules.23,39
concentrations along the mammalian proximal tubule.
TF/P, tubular uid/plasma concentration ratio; PD, potential
difference. (From: Rector FC Jr. Sodium, bicarbonate, and MECHANISMS OF SODIUM
chloride absorption by the proximal tubule. Am J Physiol. REABSORPTION
1983;244:F461, with permission.)
Apical Membrane Sodium Entry
26,27
In the proximal tubule, Na entry into the cell may be
segment approach zero. The omission of glucose and coupled to the movement of other solutes, such as glu-
amino acids from luminal uids reduces both the potential cose, chloride, or protons; or Na may enter independently.
difference and the volume absorptive rate.24,28 In the S3 In either case, the driving force for Na entry is the steep
segment, on the other hand, the omission of glucose and electrochemical gradient favoring Na in ux.
alanine has no effect either on the potential difference or The intracellular Na activity ranges from 15 to 35 mM.10,40
on the uid absorptive rate,25,29 although the deletion of all The entry of Na into cells appears to be rate limiting for trans-
organic solutes does reduce volume absorption by 50%.30 epithelial Na transport. Amphotericin B, a polyene antifungal,
The rates of transport of glucose, amino acids, phosphate, increases the permeability of the luminal membrane to Na and
and Na in the early proximal convoluted tubule exceed causes a large rise in net sodium absorption.41
those in the proximal straight tubule.31–33 These rates cor-
relate well with the relative basolateral membrane areas of Na /H Exchange
the respective segments.34
Directly coupled Na /H exchange in the proximal tubular
Electrophysiology of the Proximal Tubule brush border is responsible for most proton secretion and
for a large fraction of Na reabsorption in the proximal tu-
Transepithelial Potential Difference bule.42 The mechanism whereby Na /H exchange effects
The electrogenic nature of the transport of Na coupled to Na reabsorption is presented in Figure 5.3. Brie y, entry of
glucose and amino acids creates a lumen-negative transepi- Na is coupled to extrusion of a proton into the lumen. The
thelial electrical potential difference in the early proximal proton titrates a ltered HCO3 molecule to form carbonic
convoluted tubule. The deletion of glucose and alanine from acid. Carbonic acid subsequently is dehydrated to CO2 in
the luminal uid reduces the potential difference from about a reaction catalyzed by carbonic anhydrase IV in the brush
5.0 mV, lumen-negative, nearly to zero.24,28 This transepi- border membrane.43 Within the cell, the reverse process oc-
thelial potential difference becomes lumen-positive ( 2 to curs: carbonic acid formed by the hydration of CO2 dissoci-
4 mV) when the tubular uid to plasma chloride concen- ates into H and HCO3 . The H is extruded into the lumen
tration ratio is approximately 1.3.35 This lumen-positive by Na /H exchange or by the vacuolar H –ATPase to
voltage is probably a diffusion potential arising from the Cl- repeat another cycle while the HCO3 is transported into the
concentration gradient. blood via a 1Na :3HCO3 cotransport process (vide infra).
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1165
65

over, the transporter in the early cortical proximal tubule has


a 1:1 Na to glucose stoichiometry,61 whereas the transporter
in the straight medullary segment has a 2:1 stoichiometry.62
By coupling the energy from two Na ions moving down
their electrochemical gradient to the transport of each glu-
cose molecule, the medullary transporter is able to establish
a much greater cellular to extracellular glucose concentra-
tion ratio than a 1:1 Na :glucose transporter.27 The 2 Na :1
glucose transporter is, therefore, well suited to the straight
segment, where tubular uid glucose concentrations have
already been reduced by glucose absorption in the more
proximal segments.
FIGURE 5.3 The scheme of NaHCO3 transport mediated by
A Na -glucose cotransporter (SGLT-1), which belongs to
Na /H exchange. See text for explanation.
the SLC5 gene family,63 mediates high af nity Na -glucose
cotransport with a sodium-to-glucose coupling ratio of 2:1,
whereas SGLT-264 shares 59% homology to SGLT-1 and
The mammalian Na /H exchanger (NHE) is electro-
mediates low-af nity Na -glucose cotransport with a sodi-
neutral with a stoichiometry of one proton for one sodium.44
um-to-glucose coupling ratio of 1:1.65 In situ hybridization
The exchanger is reversibly inhibited by high concentrations
revealed high levels of a SGLT-2 message in the S1 segment
of amiloride.45 Intracellular protons, via an internal activator
of the proximal tubule.66 Recently, better antibodies have
site,46 increase Na /H exchange in response to intracellular
con rmed that, in rat kidney, SGLT-1 immunolocalizes to the
acidosis.47
brush border membrane of all three segments of the proximal
The apical and basolateral membranes of kidney cells
tubule.5 By immunohistochemistry, SGLT-2 was detected at
contain different forms of NHEs with different af nities for
the brush border of the early proximal tubule in mice, which
amiloride.48 The apical Na /H exchanger is involved in uri-
was absent in SGLT-2 knockout animals.67 Thus, it appears
nary acidi cation and has a low amiloride af nity, whereas
that SGLT-2 may represent the low-af nity, high-capacity
the basolateral exchanger has a high af nity for amiloride.
sodium–glucose cotransporter in the early proximal tubule,
The sensitivity of NHE1 to amiloride49 and its basolateral
whereas SGLT-1 may represent the high-af nity, low-capacity
localization 50 suggest that it represents the “housekeeping”
transporter of the proximal straight tubule.65
NHE. In contrast, NHE3 is in the brush border membrane
Mutations in SGLT-2 form the basis for renal glycosuria
of proximal tubule cells.51 NHE3 knockout mice have sig-
(Table 5.1), an inherited condition characterized by a low-
ni cantly reduced rates of Na and HCO3– transport in
ered threshold for tubular reabsorption of glucose.68 In con-
proximal tubules.52 In addition, pharmacologic inhibitors of
trast, the dominant clinical manifestations of inactivating
NHE3 reduce proximal tubule Na reabsorption by about
mutations of SGLT-169 relate to the failure to absorb sugars
one third.53 However, a signi cant rate of amiloride-sensitive
in the intestinal tract (glucose–galactose malabsorption).
HCO3 transport still persists in the proximal tubules of
These ndings suggest that SGLT-2 plays a much more sig-
NHE3 knockout mice,54 indicating that the NHE3 is respon-
ni cant role, quantitatively, than SGLT-1 in proximal tubule
sible for much, but not all, proximal tubular Na -coupled
glucose reabsorption. SGLT-2 inhibitors are currently under
luminal acidi cation (vide infra). NHE8 is also expressed in
investigation as potential therapeutic agents for the treat-
the apical membranes of cortical tubules and may contribute
ment of diabetes.70
to these processes.55

Sodium–Glucose Cotransport Sodium–Amino Acid Cotransport


Electrophysiologic studies in kidney proximal tubules show The proximal tubule reabsorbs amino acids from the tubular
that apical membranes depolarize with addition of glucose uid via an active transport step at the luminal membrane.31
to luminal uids.56 The depolarization occurs because the Samarzija and Frömter,71 using double-perfusion micro-
Na -glucose transporter is electrogenic. The Na -glucose puncture techniques, observed a depolarization of the lumi-
transporter is speci c for the D-stereoisomers of glucose, nal membrane during amino acid transport, and they were
galactose, and -methyl-D-glucoside.57 The Na -glucose able to identify ve classes of amino acid transporters in the
cotransporter has little af nity for cations other than Na .58 luminal membrane. Over the last decade, many of the trans-
Phlorizin inhibits Na -glucose cotransport by competing port proteins that mediate the different amino acid transport
with glucose for its binding site.59 systems have been identi ed in kidney and intestine. This
The rate of glucose transport by the early proximal topic has been recently reviewed.72
tubule is greater than in late proximal segments.60 In the Both Na -dependent and Na -independent amino acid
proximal straight tubule, the Km for D-glucose is 5 to 20 uptake pathways have been characterized in the kidney.72
times lower than in the proximal convoluted tubule. More- Neutral amino acid transport appears to involve at least
166
16 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

three separate transport systems,73 one that transports all


neutral amino acids, one speci c for imino acids, and one
for the -amino acids. Glycine may also have a speci c
transporter.74 In the kidney, neutral amino acid transport is
driven by a Na gradient, as supported by experiments in
slices, perfused tubules, and brush border membranes. The
neutral amino acid transporter B0AT1 (SLC6A19) cotrans-
ports one Na per amino acid.75 The Km of the substrate
decreases with an increasing cosubstrate concentration and
vice versa. The initial step for transport involves the bind-
ing of the amino acid to B0AT1, and this binding af nity
increases under hyperpolarizing conditions.76
The acidic and basic amino acid groups each have their
own transport systems.77,78 At least one amino acid trans- FIGURE 5.4 The scheme of neutral NaCl transport mediated
porter, a Na -independent transporter for neutral and diba- by the parallel action of Na H exchange and formate/Cl
sic amino acids, has been cloned from the kidney.79,80 exchange. Formate (HCO2 ) combines with H in the tubular
lumen to form formic acid (H2CO2), which reenters the cell by non-
NaCl Transport ionic diffusion. A similar scheme applies for oxalate–Cl exchange.
Two basic mechanisms account for NaCl reabsorption in the
proximal tubule. In simple electrogenic Na entry, sodium in Figure 5.4 uses formate/Cl exchange as the anionic com-
is transported actively through the cell, thereby creating a ponent of electroneutral NaCl transport.
lumen-negative potential difference. Cl reabsorption then As indicated previously, there is abundant evidence
proceeds through the paracellular pathway driven by the for a Na /H exchanger in proximal tubule brush border
lumen-negative potential difference. In electrically neutral membranes. With respect to NaCl transport, the inhibition
NaCl transport, both Na and Cl move through the cell at of Na /H exchange by high concentrations of amiloride86
equal rates, such that no transepithelial potential and, hence, or more speci c inhibitors of NHE353 results in a dramatic
no driving force for paracellular Cl movement is generated. fall in transcellular NaCl transport. Likewise, knockout
of NHE3 also reduces NaCl and uid reabsorption in the
Neutral NaCl Transport proximal tubule.52 Several Cl /base exchangers have been
Several lines of evidence indicate that a sizable fraction of implicated in NaCl transport. Recent interest has focused on
proximal NaCl transport is transcellular and electroneu- the role of Cl /formate (HCO2 ) and Cl /oxalate (C2O42–)
tral.81 First, by virtue of the coupling of Cl entry to apical exchange in NaCl transport.
Na entry, the intracellular Cl activity of proximal tubule A Cl /formate exchanger is present in brush bor-
cells is greater than predicted from an equilibrium distribu- der membrane vesicles.87 A role for Cl /formate exchange
tion.82 Second, Cl absorption persists even when the driv- in neutral NaCl transport is suggested by the nding that
ing force for passive, paracellular movement is abolished.83 the addition of formate to the luminal perfusate increases
Conversely, Cl reabsorption is inhibited by cyanide in the the rate of NaCl reabsorption in rabbit proximal tubules.88
absence of any change in the passive driving forces for Cl As depicted in Figure 5.4, formate is presumed to leave the
movement.83 Finally, the luminal application of SITS,84 an cell in exchange for Cl . The secreted formate then com-
anion-exchange inhibitor, or removal of chloride from the bines with a proton, which was transported by the Na /H
tubule perfusate,85 reduces net Na reabsorption. exchanger to form formic acid. The formic acid then reen-
In principle, electroneutral NaCl transport across the ters the cell by nonionic diffusion and dissociates to supply
apical membrane of proximal tubule cells could occur as di- substrate for the continuation of both exchange processes.
rectly coupled NaCl cotransport or as parallel Na /H and CFEX (SLC26A6), a homolog of pendrin, is a protein ca-
Cl /base exchangers. There is no good evidence for the for- pable of mediating Cl /formate exchange and is present in
mer process in the mammalian proximal tubule.81 However, apical membranes of the proximal tubule.89
considerable evidence supports the view that electroneutral Cl /oxalate (C2O42–) exchange has also been demonstrated
NaCl transport in the proximal tubule involves parallel ex- in brush border membrane vesicles.90 It has been suggested that
changers. The coupling of Na absorption to Cl absorption Cl /oxalate exchange may mediate neutral NaCl transport in a
in this case occurs because of the relation between cell pH manner analogous to that described for Cl /formate exchange.
and concentration of base within the cell. With reference to It has also been suggested that NaCl absorption proceeds via
Figure 5.4, the extrusion of H in exchange for Na results the operation of three parallel transporters: the Na -sulfate
in the liberation of the base for participation in Cl /base cotransport, the sulfate/oxalate exchange, and the Cl /oxalate
exchange. The uphill entry of Cl , then, is indirectly cou- exchange.91 Indeed, the CFEX protein, in addition to Cl /for-
pled to the downhill entry of Na , because both are coupled mate exchange, is also able to mediate Cl /oxalate, oxalate/for-
to the transport of an acid–base pair. The model illustrated mate, oxalate/oxalate, and oxalate/sulfate exchange.92
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1167
67

Other Na -dependent transport processes have been for Na extrusion. A number of Na -HCO3 cotransport-
described in the apical membrane of the proximal tubule. ers have been cloned, along with different splice variants.98
However, they do not contribute signi cantly to Na reabsorp- One of these, NBC1 (encoded by SLC4A4 and subsequently
tion because of the low concentrations of substrate present. renamed NBCe1-A) is localized to the basolateral membrane
of the S1 and S2 segments of the proximal tubule.99 As il-
Simple Electrogenic Na Entry lustrated in Figure 5.3, the net result of these steps is the
The classic Ussing model for salt reabsorption involves pas- reabsorption of Na and HCO3 , accounting for the bulk
sive entry of Na across apical membranes and extrusion of HCO3 reabsorption and about 20% of Na reabsorption
across the basolateral membrane by Na ,K –ATPase. A in the proximal tubule. It is believed that this cotransport
problem in assessing the contributions of electrogenic pro- process accounts for the basolateral transport of most of the
cesses to Na transport in the proximal tubule is the presence bicarbonate reclaimed from the luminal uid.97 Mutations
of other mechanisms of Na entry. However, when the con- in NBCe1-A cause proximal (type II) renal tubular acidosis
tributions of Na /H exchange and Na cotransport to net and other defects in the eye, teeth, and mental development
Na absorption are minimized by deleting glucose, amino (Table 5.1). The regulation and role of NBC in acid–base
acids, and bicarbonate from the perfusate, a fraction of uid transport in the proximal tubule is an area of active research
absorption in isolated perfused straight segments persists that has been reviewed recently.100
and the transepithelial potential is 1.0 mV.30 These results The pathways for Cl exit across the basolateral membrane
indicate that in the proximal straight tubule simple electro- are less well de ned. Several pathways for Cl exit across the
genic Na transport constitutes a mechanism for Na ab- basolateral membrane have been proposed: conductive Cl
sorption. A conductive Na pathway has been demonstrated channels, KCl cotransport, and Na (HCO3 )2/Cl exchange
in brush border membrane vesicles.93 Unlike the Na chan- (Fig. 5.5). Studies of rat proximal convoluted tubules101 and
nel found in the distal nephron segments, the Na channel rabbit convoluted and straight proximal tubule segments102,103
in the proximal tubule is not blocked by amiloride.93 using intracellular microelectrodes have indicated that the
In the proximal convoluted tubule, however, the deletion proximal tubule cell has a very low Cl conductance. Thus, in
of glucose, bicarbonate, and amino acids completely abolishes normal proximal convoluted tubule and proximal straight tu-
uid absorption.23 Consequently, simple electrogenic proxi- bule segments, conductive Cl ef ux across basolateral mem-
mal Na transport may be limited to straight segments. branes appears to play a minor role in NaCl absorption. Under
hypotonic conditions, however, cell swelling dramatically in-
Passive NaCl Absorption creases the basolateral membrane Cl conductance.104
The rise in tubular uid Cl concentration, and the attendant Because the chemical gradient for K to leave cells ex-
lumen-positive voltage (Fig. 5.2), provides a mechanism for ceeds that for Cl entry, KCl cotransport can mediate ba-
passive NaCl absorption in late regions of the proximal neph- solateral Cl exit from proximal tubule cells. Ion-selective
ron. In the Cl-selective super cial pars recta, approximately microelectrode studies have demonstrated KCl cotransport in
one-third of net NaCl absorption can be accounted for by this basolateral membranes of rabbit proximal tubule cells.103,105
mechanism.20 Stilbene-sensitive, Na -dependent Cl /HCO3 exchange
has been demonstrated in rat106 and rabbit107,108 proximal tu-
Basolateral Membrane bules. In this case, the entry of 1 Na and 2 HCO3 across the
basolateral membrane is coupled to the ef ux of Cl . The Na
The proximal tubule, particularly the S1 segment, possesses
high Na ,K –ATPase activity in the basolateral membrane.
The Na ,K –ATPase pumps Na , which entered cells apical-
ly, across basolateral membranes. In other words, the pump
keeps the cell Na activity low and maintains the electro-
chemical gradient for Na entry across the apical membrane.
Consequently, the inhibition of Na ,K –ATPase activity with
ouabain decreases transepithelial Na reabsorption and in-
creases the intracellular Na activity in the proximal tubule.94
Na also exits across the basolateral membrane in con-
cert with HCO3 . Studies in intact tubules and in membrane
vesicles have demonstrated an electrogenic, stilbene-sensitive
Na -HCO3 cotransporter in the basolateral membrane
of rat and rabbit proximal tubules.95,96 The cotransporter
transfers two net negative charges across the basolateral FIGURE 5.5 The transport pathways for Na and Cl absorption
membrane. The stoichiometry of this process is 1 Na : across the basolateral membrane of proximal tubular cells. Cl
1 HCO3 : 1 CO32 (or SO32 ).97 Thus, this transport moiety can leave the cell via KCl cotransport, Na –2HCO3 /Cl
for Na extrusion, [Na (HCO3 )3]2 is electronegative, with exchange, and Cl channels (minor). Na exits via the
the lumen-negative cell interior providing a major driving Na ,K –ATPase and Na –3(HCO3 ) cotransport.
168
16 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

and HCO3 that enter the cell are thought to be recycled isolated, perfused rabbit nephrons. The key observations are
through the [Na (HCO 3)3]2 cotransporter (see previous). (1) the volume absorptive rate is clearly dependent on the
Indeed, Na (HCO 3)2/Cl exchange may account for much perfusion rate, (2) the ow dependence persists in the ab-
more Cl movement than KCl transport.108 Na -independent sence of active transport when anion gradients are present,
Cl /HCO3 exchange is also present in the basement mem- and (3) the ow dependence is abolished in the absence of
brane of proximal tubules.106–108 However, under physiologic active transport and anion gradients.
conditions, this process mediates net Cl in ux and does not An explanation for these results lies in a consideration
contribute to net NaCl absorption. of axial versus radial changes in uid composition along
the tubule.117 The rate of passive Na absorption in the late
proximal tubule is dependent on the magnitude of the chlo-
CONTROL OF PROXIMAL TUBULAR ride gradient between the lumen and the bath. At low axial
SODIUM REABSORPTION perfusion rates, the radial Cl gradient tends to dissipate as
a function of distance along the tubule, but this dissipation
Glomerulotubular Balance (GTB) is minimized at higher axial perfusion rates. Hence, the in-
The proximal tubule responds to an increase in glomerular l- tegrated driving force for passive NaCl absorption increases
tration with an increase in the absolute rate of uid absorption with the rate of tubule perfusion. In addition, the availability
(APR) to minimize variations in the fractional proximal uid of solutes, such as glucose, amino acids, and bicarbonate, is
absorption. This phenomenon is termed glomerulotubular also partly responsible for the ow dependence of proximal
balance (GTB). The ef ciency of GTB—that is, the extent to uid absorption.118
which APR/GFR remains constant—is subject to physiologic The ow dependence of reabsorption in the proximal
and pathologic control. The prime factor modulating GTB in tubule has also been tested in the mouse proximal tubule.
vivo is the effective circulating volume. Thus, at a constant or In this experimental model, which included the use of the
near constant GFR, volume expansion and volume contrac- NHE3 knockout mouse, the data supported the hypothesis
tion decrease and increase, respectively, the absolute rate of that the “brush border” microvilli act as mechanosensors
proximal Na absorption. In other words, volume expansion that transmit uid dynamic torque to the actin cytoskeleton
and volume contraction reset GTB upward and downward, and thus modulate Na absorption.119
respectively.109,110 This section considers some of the factors
that modulate proximal Na absorption. Catecholamines
Peritubular capillary oncotic pressure is one of the fac-
Renal denervation reduces proximal tubule Na and uid
tors regulating the rate of salt and water absorption from the
absorption.120 Both - and -adrenergic receptors exist in
proximal tubule.111 The oncotic pressure of the peritubular
the proximal tubule.121,122 The rate of salt and water reab-
proteins favors the movement of uid across the basement
sorption in the proximal tubule is stimulated by - and -
membrane, whereas capillary hydrostatic pressure retards
adrenergic agonists.123 -Adrenergic agonists increase api-
this movement. Thus, at a given renal blood ow, an in-
cal Na entry via the stimulation of Na /H exchange122
crease in the glomerular ltration rate and, hence, the ltra-
and also increase basolateral Na ef ux via Na ,K –ATPase
tion fraction, will cause an increase in the oncotic pressure in
activity in rat proximal tubules by a pathway that involves
the postglomerular peritubular capillaries. At constant single
the activation of calcineurin.124 The effects of -adrenergic
nephron glomerular ltration rates (SNGFR), the perfusion
agonists on Na ,K –ATPase activity are less clear, with one
of efferent capillaries with hypo-oncotic uids decreases the
study showing that they increase Na ,K –ATPase activity
absolute rate of proximal uid absorption, whereas perfu-
via protein kinase C (PKC).125 However, others have found
sion of the capillaries with hyperoncotic uids increases
that -adrenergic agonists inhibit Na ,K –ATPase activity
proximal absorption.112 The effects of the peritubular pro-
via PKA.126
tein concentration can also be demonstrated in isolated per-
Dopamine, which is produced by proximal tubular
fused tubules.113
cells,127 inhibits Na reabsorption. Dopamine inhibits
It is not precisely clear how the peritubular protein
Na ,K –ATPase activity via its receptors DA-1 and DA-2.128
concentration modulates proximal uid absorption. The ef-
fect is not simply because of the oncotic pressure exerted
by the proteins, because changes in absorption do not oc- Parathyroid Hormone
cur when active transport is inhibited or from comparable Parathyroid hormone (PTH) causes a 30% to 50% reduction
changes in the transtubular hydrostatic pressure.114 The pre- in proximal tubular Na and phosphate absorption.129 PTH
vailing views are that the peritubular protein may directly stimulates adenylyl cyclase, cAMP production, and PKA,
affect transcellular Na transport115 or the back leak of Na which in turn inhibits Na /H exchange in several proxi-
through the paracellular pathway.113,116 mal tubule systems.130 Weinman et al.131 demonstrated that
Luminal factors also contribute to glomerulotubular bal- phosphorylation by PKA inhibits Na /H exchange activ-
ance. The ow dependence of proximal absorption has been ity via a PDZ domain-dependent interaction with the NHE
investigated in the convoluted 15 and straight segments30 of regulatory factor (NHERF). In the absence of NHERF, cAMP
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1169
69

does not inhibit the exchange activity of NHE3.132 The cur- Nitric Oxide
rent model for this inhibition is that NHE3 associates with Various forms of nitric oxide synthase (NOS) are expressed
PKA indirectly via NHERF and the cytoskeletal protein ezrin. in the proximal tubule.150 Low basal production of nitric ox-
PKA, when active, phosphorylates NHE3 at serines 552 and ide (NO) by the proximal tubule is boosted dramatically by
605, which mediates the inhibition of the exchanger.133 lipopolysaccharide (LPS) and cytokines. Even under basal
NHE3 is directly phosphorylated by other protein kinases, conditions, the proximal tubule may be affected by NO pro-
including calmodulin-dependent protein kinase II, which duced by adjacent cells, such as endothelium or other neph-
inhibits Na /H exchange activity and PKC, which stimu- ron segments. The overall effect of NO on proximal tubule
lates the exchanger.134 Na transport is controversial and may be biphasic, with
acute inhibition and chronic stimulation of Na reabsorp-
Angiotensin II tion as assessed by pharmacologic agents and genetic knock-
The systemic administration of low doses of angiotensin II outs of NOS, respectively.151 In vitro, NO decreased Na /H
(Ang II) inhibits the excretion of Na ,135 whereas inhibitors exchange and Na ,K –ATPase activity in cultured proximal
of Ang II increase Na excretion.136 Systemic Ang II causes tubule cells.152
changes in renal blood ow, aldosterone secretion, ltration
fraction, and catecholamine release from renal sympathetic
nerve endings.137 Low concentrations of Ang II ( 10 9 M) MECHANISM OF ISOTONIC FLUID
cause an increase in proximal tubule uid and bicarbonate ABSORPTION
reabsorption, effects that are mediated by the AT1 subtype
of Ang II receptors present in both the brush border and Proximal tubule water absorption is coupled tightly to sol-
basolateral membranes of the proximal tubule.138 Higher ute absorption, because the measured osmolality of the tu-
concentrations ( 10 8 M) depress uid and bicarbonate ab- bular uid is generally identical to plasma. Three general
sorption, presumably via counterbalancing effects mediated mechanisms of solute–solvent coupling have been suggested
by the lower af nity AT2 receptor.138 Studies have demon- to account for this isotonic absorption: lateral interspace hy-
strated that the stimulatory effect of Ang II on uid and bi- pertonicity, effective osmotic gradients because of different
carbonate reabsorption occurs via enhanced apical Na /H re ection coef cients for solutes in the tubular and peritu-
exchange via NHE3 and basolateral Na -(HCO3 )3 cotrans- bular uids, and luminal uid hypotonicity.
port via NBC-1 in the proximal tubule.139 The physiologic The standing gradient theory argues that active trans-
effects of Ang II may involve the coupling of these receptors port of salt into the lateral intercellular space raises the os-
to both phospholipase A2 and inhibitory G proteins.140,141 molality of the space, thus providing an osmotic gradient
for uid transport from the lumen to the interspace.153 The
tight junctions in this model are presumed to be imperme-
Thyroid Hormone
able to water, so that the osmotic ow of water from the cell
On a clinical level, hypothyroidism is associated with a de- into the hypertonic interspace raises the hydrostatic pressure
creased cardiac output, renal blood ow, and glomerular in this compartment and forces uid across the basement
ltration rate (GFR). Clearance studies in hypothyroid rats membrane.
have documented decreases in GFR, renal Na reabsorp- An alternative explanation 117 proposes that an effective
tion, and renal Na ,K –ATPase activity.142 These changes osmotic driving force for uid absorption can exist between
are reversible after thyroid hormone replacement.143 The solutions of identical osmolalities if the re ection coef cients
thyroid hormone may exert direct effects to stimulate proxi- of the membrane for the solutes in the solutions differ. Spe-
mal tubular salt and uid reabsorption via increased baso- ci cally, the elevated tubular uid-to-plasma Cl concentra-
lateral K permeability144 and/or direct stimulation of Na / tion found in the late proximal convolution and in the pars
H exchange through an increase in NHE3 transcription.145 recta provides an effective osmotic driving force for uid ab-
sorption because HCO3 exceeds Cl . That is, the bicar-
Corticosteroids bonate in the peritubular uid is a more “effective” osmotic
Although mineralocorticoids do not have an effect on proxi- agent than is the chloride in the tubular uid, and, thus,
mal tubular sodium reabsorption,146 there is evidence for net water ows out of the tubule. Although this mechanism
glucocorticoid receptors in the proximal tubule.147 Dexa- may be applicable to the proximal straight tubule, the re ec-
methasone inhibits apical membrane Na -phosphate co- tion coef cients for NaCl and NaHCO3 measured across the
transport in cultured proximal tubular cells via PKC rabbit proximal convoluted tubule are virtually identical,154
activation.148 Dexamethasone also enhances the activity so oppositely directed Cl and HCO3 gradients may make
of apical NHE3 and the mRNA expression and functional only a negligible contribution to uid absorption in convo-
activity of basolateral NBC-1.149 The resulting increase in luted segments.
proximal tubule HCO3 reabsorption could contribute to Finally, because of the high osmotic water permeability
the maintenance of the metabolic alkalosis that is associated of the proximal tubule, only small degrees of absolute lumi-
with increased glucocorticoid production in vivo. nal hypotonicity are needed to provide a suf cient driving
170
17 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

force to account for the observed rates of uid reabsorp- interspecies heterogeneity.165 Although the Na and Cl per-
tion.20 Experimental evidence supports the view that abso- meabilities appear to vary widely among rabbits, hamsters,
lute luminal hypotonicity is a signi cant driving force for and rats, one consistent nding was that the DTL is relatively
uid reabsorption in the proximal tubule. Thus, when prox- less permeable to NaCl than the ATL. Coupled with the DTL’s
imal tubules are perfused and bathed by symmetric NaCl high permeability to water, the relative lack of solute perme-
solutions, the luminal uid becomes slightly hypotonic.155 ability ensures that the osmotic pressure of uid entering
The development of luminal hypotonicity can be ampli ed the renal papilla is greater than that of the uid leaving it.166
by maneuvers that decrease the water permeability of the Thus, the formation of dilute urine by the loop of Henle
proximal tubule. The aquaporin 1 (AQP1) water channel begins in the ATL.
is abundantly expressed in the proximal tubule. In AQP1 The decrease in osmolality in the ATL is due primarily
knockout mice, the osmolality of tubular uid at the end to a fall in the NaCl content of the luminal uid. The elec-
of the proximal tubule is signi cantly lower than in normal troneutral transport of NaCl appears to occur through two
mice.156 As the luminal uid becomes more hypotonic, the key mechanisms. The transepithelial movement of sodium
resorbate becomes more hypertonic, and the degree of re- from the ATL lumen occurs via the paracellular pathway,167
sorbate hypertonicity correlates with the rate of volume re- whereas Cl diffusion occurs through a transcellular route.
absorption by the tubules.157 Yoshitomi et al.168 detected conductive pathways for Cl
in both the apical and basolateral membranes of ATL cells.
THE LOOP OF HENLE The transcellular movement of Cl in the ATL is regulated,
because the basolateral Cl conductance is inhibited at low
The dissociation of salt and water absorption by the loop of
pH168 and by low intracellular Ca2 concentrations.169 Uchi-
Henle is ultimately responsible for the capacity of the kidney,
da et al.170 cloned a Cl channel in 1993 from the rat renal
either to concentrate or to dilute the urine. The active ab-
medulla, ClC-K1 (termed ClC-Ka in humans), which rep-
sorption of NaCl in the water-impermeable thick ascending
resents the major mediator of transcellular Cl movement
limb of Henle (TALH) serves both to dilute the urine and to
in the thin ascending limb. This channel, which belongs
supply the energy for the “single effect” of countercurrent
to the ClC family of Cl channels, is expressed exclusively
multiplication. A functionally similar segment, known as the
within the kidney and has been localized by immunohisto-
diluting segment, is found in amphibians and teleosts.158
chemistry to both the apical and basolateral membranes of
The mammalian loop of Henle contains the descend-
the thin ascending limb of Henle.171 Its activity is depen-
ing thin limb (DTL), the ascending thin limb (ATL), and
dent on the coexpression of barttin, an accessory protein
the thick ascending limb (TAL). The loop of Henle absorbs
that forms a complex with ClC-K1, increases its abundance
about 25% to 40% of the ltered Na load.159 Furthermore,
at plasma membranes, and modi es channel gating.172–175
the uid leaving the loop is dilute, indicating that more NaCl
The expression of ClC-K1 is increased by dehydration.170
is absorbed in the loop than water.
Genetic knockout of the ClC-K1 gene in mice produced a
urinary-concentrating defect, con rming the role of passive
SALT TRANSPORT BY THE THIN NaCl transport in the thin ascending limb in the urinary
DESCENDING AND THIN ASCENDING concentrating mechanism.176
SEGMENTS
As the tubular uid enters the descending thin limb and NaCl ABSORPTION IN THE THICK
ows toward the tip of the renal papilla, it becomes more ASCENDING LIMB
concentrated.160 Passive models for urinary concentration
indicate that this increase in osmotic pressure is attributable General Features
to water extraction rather than solute entry.161 Current ob- The studies of Rocha and Kokko177 and Burg and Green 178
servations indicate that the aquaporin water channel AQP1 were the rst to investigate salt absorption in the thick as-
mediates water movement across the luminal surface of the cending limb, and their work de ned several key features
DTL.162 According to some reports, however, AQP1 expres- of this unique epithelium. First, salt absorption in the med-
sion is signi cantly lower in short loop nephrons than long ullary and cortical TAL generates a lumen-positive transepi-
loop nephrons,163 suggesting that (1) not all DTLs in the thelial voltage, which is sensitive to furosemide. Second, the
kidney extract water to the same degree, and/or (2) water transport of Cl occurs against both electrical and chemical
movement in short loop DTLs is facilitated by alternative gradients and involves an active transport process that is
water channels or via the paracellular route. In contrast to dependent on intact basolateral Na ,K –ATPase activity.179
the DTL, in vitro microperfusion studies demonstrate that A nal important feature of the TAL is that this segment
the ATL is relatively impermeable to water.164 consists of a tight epithelium, which despite its high ionic
The study of NaCl transport by the DTL and ATL has conductance, possesses a very low permeability to water.
been complicated by the fact that the transport character- The apical membrane of the TAL constitutes the major bar-
istics of these two nephron segments exhibit signi cant rier to transcellular and paracellular water ow.180 The high
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1171
71

provides a pathway for net K secretion by the TAL. In


mouse TAL, for example, the rate of K secretion amounts to
about 10% of the rate of net Cl absorption.182 K secretion
in this segment is an active process, ultimately driven by the
Na ,K –ATPase, proceeding in the face of a lumen-positive
transepithelial potential. Third, under open circuit condi-
tions, the transcellular and paracellular pathways form a cur-
rent loop in which the currents traversing the two pathways
are of equal size, but which traverse in the opposite direction.
The potassium current from cell to lumen polarizes the lu-
men and causes an equivalent current to ow from the lumen
to the bath through the paracellular pathway.186 Because the
paracellular pathway is cation selective (PNa/PCl 2 to 6), the
majority of the current through the paracellular pathway is
carried by Na moving from the lumen to the interstitium.
This paracellular absorption of Na increases the ef ciency
of Na transport by the TAL.187 With reference to Figure 5.6,
FIGURE 5.6 A model depicting the major elements of the for each Na transported through the cell and requiring the
mechanism of NaCl absorption by the thick ascending limb. use of ATP, one Na is transported through the paracellular
Dashed lines indicate passive ion movements down electro- pathway without any additional energy expenditure. Finally,
chemical gradients. ROMK, renal outer medullary K channel; the apical K current satis es the continuity requirement im-
ClC-Kb, chloride channel Kb. posed by a high degree of conductive Cl ef ux across baso-
lateral membranes.182
A small component of Na transport by the TAL is
ionic conductance and low water permeability effectively accounted for by NaHCO3 absorption.188 In the rat TAL, the
further dilutes uid entering the TAL from the ascending rate of NaHCO3 absorption is roughly 5% to 10% of that
thin limb. for NaCl absorption. NaHCO3 absorption appears to be me-
Figure 5.6 integrates the results of several electrophysi- diated by an apical membrane amiloride-sensitive Na /H
ologic and biochemical studies to provide a model of salt exchanger and a basolateral membrane electrogenic Na -
reabsorption in the thick ascending limb. According to this 3(HCO3 ) cotransporter.188
model, net Cl absorption by the TAL is a secondary ac- The following sections will describe the individual com-
tive transport process. Luminal Cl entry into the cell is ponents of the mechanism for TAL salt transport (Fig. 5.6)
mediated by an electroneutral Na -K -2Cl cotransport in greater detail.
process driven predominantly by the favorable electrochem-
ical gradient for Na entry.181 Because the Na gradient is
maintained by the continuous operation of the basolateral Apical Na -K -2Cl Cotransport
membrane Na ,K –ATPase pump, the apical entry of Cl Studies of Cl transport across apical membranes of intact TAL
via the cotransporter ultimately depends on the operation of segments189 and in isolated membrane vesicle preparations190
the basolateral Na ,K –ATPase. established that the predominant mode for Cl entry into the
In contrast to the electroneutral entry of Cl across the TAL cell is via a Na -K -2Cl cotransporter. A characteristic
apical membrane, the majority of Cl ef ux across the basolat- feature of this transporter is its sensitivity to inhibition by
eral membrane proceeds through conductive pathways.182,183 furosemide, bumetanide, and other 5-sulfamoylbenzoic acid
A favorable electrochemical gradient for Cl ef ux through derivatives.191 The measurement of isotope ux into TAL
dissipative pathways has been demonstrated by Greger et al.184 cells or membrane vesicles prepared from the inner stripe of
in the rabbit cTAL. Intracellular Cl is maintained at concen- the outer medulla yielded a stoichiometry of 1 Na :1 K :2
trations above electrochemical equilibrium by the continued Cl cotransport.190 K -independent NaCl cotransport has
entry of Cl via the apical Na -K -2Cl cotransporter.185 also been described under certain conditions.192
According to the model in Figure 5.6, K that enters The proteins that mediate the Na -K -2Cl cotransport
TAL cells via the Na -K -2Cl cotransporter recycles, to a have been cloned. An absorptive form of the Na -K -2Cl
large extent, across the apical membrane via a K -conductive cotransporter, referred to as NKCC2 or BSC1, was initially
pathway. This apical K recycling serves several purposes. cloned by Gamba et al.193 based on sequence homology to the
First, it ensures a continued supply of luminal K to sus- thiazide-sensitive Na -Cl– cotransporter (see the following).
tain Na -K -2Cl cotransport. Without recycling, the lumi- A second Na -K -2Cl cotransporter, NKCC1, was cloned
nal K concentration would fall rapidly as a consequence of by Payne et al.194 NKCC2 (BSC1) is the primary mediator of
K entry via Na -K -2Cl cotransport and would limit net apical salt entry in the thick ascending limb. In situ hybrid-
NaCl absorption. Second, the apical membrane K current ization and single-nephron reverse transcriptase polymerase
172
17 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

chain reaction (RT-PCR) studies demonstrated the expres- domain. Each of the A, B, and F isoforms can have either
sion of NKCC2 in the MTAL and CTAL,195 and immuno- a long C-terminus, or a truncated C-terminus; although to
histochemical studies indicate that NKCC2 is localized to date, the short isoforms have only been described in the mu-
the apical membrane of these nephron segments.196 The im- rine TAL.204 In addition, several “tandem” transcripts have
portance of NKCC2 in mediating salt reabsorption in the been described in the human kidney; these contain combina-
TAL is illustrated by the fact that loss-of-function mutations tions of exons 4A, 4B, and 4F spliced alongside one another
of NKCC2 cause Bartter syndrome (Table 5.1),197 a Men- into the NKCC2 pre-mRNA.205 Transcripts containing exons
delian salt-wasting disorder characterized by hypokalemia, 4A/4F, 4B/4A, and 4B/4A/4F have been reported. Because
metabolic alkalosis, hyperaldosteronism, and normal-to-low these tandem transcripts contain redundant sequences en-
blood pressure, results from a defect in salt absorption by coding for the second transmembrane domain, they probably
the thick ascending limb. cause the misfolding of NKCC2, resulting in the formation
The NKCC2 cDNA encodes a glycoprotein contain- of nonfunctional isoforms. Because these isoforms may still
ing 1100 amino acids and having a predicted molecular form oligomers with NKCC2, they likely exert a dominant-
weight of 115 to 120 kDa.193 The full-length protein contains negative effect on NKCC2 function and inhibit its activity.206
12 transmembrane domains containing a sizable extracellu- The A, B, and F isoforms show differential expression
lar loop with N-glycosylation sites positioned between trans- within the thick ascending limb. In the rat nephron, the A
membrane segments 7 and 8, and large intracellular amino isoform was found in both the cortical and medullary TAL,
and carboxy termini ank the transmembrane regions. the B isoform is restricted to the cortical TAL, whereas the F
NKCC2 belongs to the SLC12A family of cation chloride isoform is present in the medullary, but not the cortical, the
cotransporters, which is part of the amino acid polyamine TAL, and to a lesser extent, in the outer medullary collecting
organocation cotransporter (APC) superfamily.198 Based on duct. Although some interspecies discrepancies have been
the homology to other crystallized APC family members, the noted, similar ndings have generally been observed in the
cotransporter structure probably consists of two clustered embryonic mouse and human kidney.205,207,208
groups of ve transmembrane helices that are positioned in When the 4A, 4B, and 4F exons are spliced into tran-
a symmetric, inverted orientation.199 The details regarding scripts containing a long C-terminus, all three products are
how this fold facilitates the three-ion cotransport remain ob- capable of mediating Na -K -2Cl cotransport. However,
scure but surely will provide an initial framework for more the A, B, and F isoforms have different transport properties
detailed structure-function studies in the coming years. that may have physiologic relevance. Isoforms A and B have
The two cytoplasmic domains of NKCC2 mediate spe- higher af nities for Na , K , and Cl than the F isoform.
ci c regulatory functions. The amino terminus is believed The A isoform possesses the highest transport capacity of all
to be unstructured and contains several cytosol-exposed three isoforms. Based on the known distribution of the A, B,
serines and threonines, which are phosphoacceptor sites. and F isoforms in the TAL, it is currently thought that the A
These residues are phosphorylated by at least three pro- isoform accounts for the high transport capacity of the med-
tein kinases that stimulate NKCC2 activity and/or plasma ullary TAL, whereas the presence of the more active A and
membrane expression (discussed in detail below).200 The B isoforms in the cortical TAL allows for the continued reab-
carboxy terminus is large and comprises 40% of the total sorption of salt to take place, even though the tubular uid
NKCC2 sequence. It may also contain phosphorylation sites, in this segment is more dilute than plasma. Supporting this
although to date this has not been explored in detail. It is is experimental evidence demonstrating that the NKCC2 A
clear, however, that the NKCC2 C-terminus serves as a hub and B isoforms can both be strongly activated by Na , K ,
for interactions with proteins that regulate its traf cking, and Cl at concentrations that are much more dilute than
including the glycolytic enzyme aldolase201 and secretory the composition of tubular uid in the cortical TAL.208
membrane carrier protein 2 (SCAMP2).202 It also serves as
the interface for the formation of NKCC2 homodimers,203 Apical K Conductance
which was con rmed recently when the crystal structure of An important feature of the luminal membrane of the TAL is
the C-terminus of the related prokaryotic cation chloride a barium-inhibitable potassium conductance.185 This apical
cotransporter MaCCC was solved.199 membrane K conductance allows K to be recycled from
At least six isoforms of NKCC2 have been identi ed.200 the cell back into the luminal uid to support further NaCl
These isoforms are the result of alternative splicing of two absorption via the NaCl cotransporter. Using measured
regions of the NKCC2 gene: the rst region is a 96 base pair values of intracellular K activity (rabbit cTAL),209 apical
region that encodes part of the second transmembrane do- membrane conductance, and intracellular voltage, it can be
main, whereas the second region encompasses the extreme shown that the measured apical membrane conductance is
C-terminus. Three variants of the 96 base pair region are en- suf cient to provide for the recycling of all of the potassium
coded by different versions of exon 4 of the NKCC2 gene. uptake via the Na -K -2Cl cotransporter.
These exons are differentially spliced into NKCC2 pre-mRNAs Three types of K conductances have been characterized
to generate three distinct isoforms (A, B, and F), which alter in the apical membrane of the thick ascending limb: a high-
the amino acid composition of the second transmembrane conductance (150 pS) calcium-activated K channel, which
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1173
73

does not contribute to net K transport,210 an intermediate- Similar to ClC-K1/ClC-Ka channels in the ATL, ClC-
conductance (70 pS) K channel,211 and a low-conductance K2/ClC-Kb channels require barttin accessory subunits to
(30 to 35 pS) K channel.211,212 The latter two conductances be fully functional. Barttin was originally identi ed by po-
account for the majority of the apical K secretion in the sitional cloning of the BSND gene, which is responsible for
TAL.213 Both the intermediate- and the low-conductance type IV Bartter syndrome (Table 5.1), a severe form of he-
channels are inhibited by intracellular ATP and are increased reditary salt wasting that is accompanied by sensorineural
by high dietary potassium.214 deafness.224 Barttin is located in the basolateral membranes
A major breakthrough in elucidating the molecular of the thin and thick ascending limb, distal nephron, and
mechanism of K secretion in the TAL was made when also in the stria vascularis of the inner ear, where it is be-
Ho et al.215 cloned the ATP-dependent K channel ROMK lieved to play a role in K secretion into the endolymph. The
from rat kidney. This channel is the prototype for a large regulation of ClC-K channels by barttin appears to be mul-
family of inward-rectifying K channels (Kir channels), and tifaceted, because the accessory subunit in uences channel
hence, is also called Kir 1.1. Based on its biophysical prop- protein stability, subcellular localization, and gating.172,173,175
erties and regulation, investigators had long suspected that The mutations of barttin identi ed in Bartter syndrome pa-
ROMK was the sole mediator of the 30-pS K conductance tients generally impair the ability of barttin to produce a Cl
in the TAL. This hypothesis was con rmed when studies of conductance when expressed with ClC-Kb.172,173
the thick ascending limb in the ROMK knockout mouse re- Although less completely studied, there do appear to
vealed the absence of a 30-pS channel.216 Subsequent stud- be additional transport pathways that mediate basolater-
ies of the ROMK knockout mouse demonstrated that these al Cl ux in the TAL. For example, a barium-sensitive
mice also lack a 70pS conductance.214 Thus, it is likely that transcellular K -Cl cotransport mechanism has been
ROMK also comprises at least a portion of the intermediate proposed (Fig. 5.6).225 The expression of two KCl cotrans-
K secretory conductance in this nephron segment. It has porters that belong to the same family of SLC12 electro-
been proposed that the intermediate-conductance channel neutral cotransporters as NKCC2, KCC1,226 and KCC4227
may be a heteromeric protein containing ROMK and other, have been observed. Thus, both of these cotransporters
as yet, unidenti ed subunits. may participate in the extrusion of K and Cl from cells
The notion that ROMK is the predominant channel in the cortical and medullary TAL. Studies in knockout
responsible for apical K recycling in the thick ascending animals, however, indicate that KCC4 is not the primary
limb is also supported by the nding of mutations in the mediator of basolateral Cl ux in the TAL, because these
ROMK gene in families with type II Bartter syndrome.217,218 mice do not develop a salt-wasting phenotype. Rather, the
These mutations have generally been con rmed to result KCC4 knockout mouse develops metabolic acidosis and
in defective K -channel function.219 As noted, Bartter syn- sensorineural deafness, suggesting that KCC4 plays im-
drome results from a defect in thick ascending limb salt portant physiologic functions in the acid-secreting inter-
transport. Thus, the presence of ROMK mutations as a calated cells of the collecting duct and in the cells of the
cause of Bartter syndrome indicates the important role of inner ear.228
ROMK in net salt absorption by the thick ascending limb
(Table 5.1). Synchronous Na /H :Cl /HCO3 Exchange
Friedman and Andreoli229 found that net Cl absorption
Basolateral Membrane Cl Transport and the transepithelial voltage were doubled when CO2 and
Cl exit across the basolateral membrane of TAL cells is HCO3 were added to the external solutions bathing corti-
largely conductive, proceeding down its electrochemical cal TAL segments. Because the (CO2 HCO3 )-stimulated
gradient through Cl -selective channels in the basolateral rate of NaCl absorption did not result in net CO2 transport
membrane.220 The primary mediator of basolateral chloride and could be abolished by the lipophilic carbonic anhydrase
transport in the TAL in humans is thought to be the chloride inhibitor ethoxyzolamide or by the luminal addition of the
channel ClC-Kb, the sequence of which is closely related to anion-exchange inhibitor SITS or DIDS, it was proposed that
the aforementioned human ClC-Ka channel expressed in the apical membrane of the mouse cortical TAL contains par-
the ATL (see previously). The name for the corresponding allel, near synchronous Na /H :Cl /HCO3 exchangers in
rodent ortholog of ClC-Kb is ClC-K2. In contrast to the rel- addition to a Na -K -2Cl cotransporter. The addition of
atively narrow expression of ClC-K1, ClC-K2 is expressed CO2 and HCO3 to the bathing solutions had no effect on
broadly throughout the basolateral membranes of multiple net NaCl transport in either the rabbit cTAL or the mouse
segments in the rodent nephron, including the TAL, the mTAL. Both the rat and mouse medullary TAL do contain
distal convoluted tubule, the connecting tubule, and the Na /H exchangers in their apical membranes. However, in
collecting duct.171,221,222 Inherited mutations of the ClC-Kb these segments, Na /H exchange plays a role in net HCO3
cause type III Bartter syndrome, which can manifest as a transport and cell pH regulation rather than transcellular
mixed Bartter/Gitelman phenotype, possibly owing to the NaCl absorption.230 The NHE3 isoform of the Na /H ex-
expression of ClC-K2/ClC-Kb in the distal convoluted tu- changer is the major isoform expressed in the apical mem-
bule (DCT) and TAL.223 brane of the thick ascending limb.231,232
174
17 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

protein expression yields a mild salt-wasting phenotype that


REGULATION OF SALT ABSORPTION approaches Gitelman syndrome, likely owing to the elimina-
IN THE TAL tion of its activity in the DCT (see the following). However, the
Vasopressin absence of a Bartter-like phenotype in these animals does not
refute the importance of SPAK and OSR1 in the TAL. Rather,
The peptide hormone arginine vasopressin (AVP, also known
the Gitelman-like salt-wasting phenotype in these animals
as antidiuretic hormone, ADH) remains the most extensively
appears to be due to two factors. First, knockout of the SPAK
characterized stimulatory hormone for NaCl reabsorption in
gene not only ablates full-length kinase-active SPAK, but
the TAL. The reabsorption of NaCl in the TAL is crucial for
also eliminates the expression of a truncated kidney-speci c
ef cient urinary concentration, because it is this process that
kinase-defective SPAK isoform (KS-SPAK) that suppresses
plays a key role in maintaining a hypertonic medullary in-
baseline SPAK and OSR1 activity in the TAL.237 Second, in
terstitial solute gradient for water reabsorption in more distal
the SPAK knockout mouse, TAL-expressed OSR1 appears to
portions of the nephron.233 Thus, from a teleologic perspec-
compensate for the absence of SPAK activity. Both of these
tive, vasopressin ought to be an important regulator of this
effects ultimately result in increased rather than decreased
process. The cognate receptor for vasopressin, the V2 recep-
NKCC2 abundance and phosphorylation, which probably
tor (V2R), is expressed in both the cortical and medullary
compensates for the lack of DCT salt reabsorption and gives
TAL, where it participates in signaling cascades that stimu-
rise to a relatively mild salt-wasting phenotype. The domi-
late NKCC2 activity.234
nance of OSR1 over SPAK in the TAL was recently veri ed
Binding of AVP to V2R results in increased intracellu-
in studies of the OSR1 knockout mouse, which, in contrast
lar levels of cAMP in the TAL. The increase in cAMP levels
to the SPAK knockout animal, exhibits a Bartter-like pheno-
ultimately drives the translocation of NKCC2 from intracel-
type.241 On the other hand, knock-in mice bearing a muta-
lular subapical vesicles to the luminal plasma membrane.
tion that ablates a key catalytic activation site in SPAK do
In addition, cAMP serves as a signal that increases the phos-
exhibit decreased NKCC2 phosphorylation; presumably,
phorylation of residues in the NKCC2 amino terminus that
this inactivating mutation exerts a dominant-negative effect
stimulate cotransporter activity in vitro.235,236 Although this
on NKCC2 phosphorylation by preventing the binding of
process may be mediated upstream by the cAMP-dependent
stimulatory kinases such as OSR1 to the NKCC2 N-terminal
protein kinase A (PKA), it is unclear whether PKA directly
docking site.242 Although it is clear that SPAK and OSR1 are
phosphorylates the N-terminal residues that stimulate its ac-
important kinases that are linked to tubular salt reabsorp-
tivity. Rather, it appears that two other kinases in particular
tion in the TAL, it is unclear how they directly connect to
are important downstream mediators of this process. These
the well-established upstream cAMP-dependent signaling
kinases, the Ste20 SPS1-related proline alanine-rich kinase
cascades, which are mediated by vasopressin. Nevertheless,
(SPAK) and oxidative stress responsive kinase 1 (OSR1),
given the fact that vasopressin activates SPAK and OSR1
are structurally homologous, activated by vasopressin, bind
and stimulates NKCC2 phosphorylation at key SPAK/OSR1
to a de ned docking site harbored within the NKCC2 N-
phosphorylation sites, it would appear that these kinases
terminus, and directly phosphorylate these previously iden-
are important downstream intermediaries of the vasopressin
ti ed stimulatory residues (Fig. 5.7).237–239
signaling pathway.
The importance of SPAK and OSR1 in NKCC2 function
In addition to its effects on apical Na -K -2Cl co-
has recently been established in transgenic and knockout
transport, vasopressin increases the transcellular electrical
models.237,240,241 A complete knockout of SPAK mRNA and
conductance of the mouse medullary TAL, and this conduc-
tance increase is a major element in the mechanism for the
hormone-dependent increase in the rate of net salt absorp-
tion.243,244 The available evidence suggests that both the api-
cal and basolateral membrane conductances are increased
by vasopressin. In apical membranes, AVP increases conduc-
tance by increasing the functional number of K channels182;
this increase occurs even when net salt absorption is abol-
ished by furosemide. Ecelbarger at al.245 showed that this
increase in apical K conductance was at least in part due
to a dramatic upregulation in ROMK abundance and api-
cal localization. Thus, the machinery for apical recycling of
K is increased in the TAL, which would aid in augmenting
NKCC2-mediated NaCl reabsorption.
FIGURE 5.7 The current model for SPS1-related proline alanine- The predominant portion of the ADH-induced in-
rich kinase /oxidative stress responsive kinase 1 (SPAK/OSR1) crease in cellular conductance is accounted for by an in-
and kidney-speci c (KS)-SPAK regulation of NKCC2 via phos- crease in the basolateral membrane Cl conductance.182
phorylation in the thick ascending limb of the loop of Henle. Two mechanisms have been suggested for the hormone-
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1175
75

dependent increase in basolateral Cl conductance. Schlat- Hypercalcemia


ter and Greger183 have proposed that the ADH-induced Hypercalcemia often results in an ADH-resistant urinary con-
increase in intracellular cAMP results in a direct increase centrating defect, that is, nephrogenic diabetes insipidus.255
in Cl channel activity. Such a mechanism has been amply At least part of this concentrating defect results from the
demonstrated in Cl -secreting epithelia, such as the tra- inhibition of ADH-stimulated cAMP production in the TAL
chea and intestine. Alternatively, ADH might enhance Cl by calcium.256 Preincubation of tubule segments with per-
conductance indirectly by increasing apical membrane Cl tussis toxin abolishes the effect of hypercalcemia on cAMP
entry.182 According to this proposal, an ADH-dependent generation, indicating that the inhibition of cAMP genera-
activation of apical membrane NKCC2 and ROMK leads tion is mediated through the activation of Gi.257 The effects of
to an increase in intracellular Cl concentration. Because hypercalcemia are mediated by a G protein–coupled calcium-
the activity of basolateral Cl channels is exquisitely sensi- sensing receptor (CaSR) present on the basolateral membrane
tive to changes in intracellular Cl concentration,246 this of TAL cells.258 Activation of this receptor also inhibits the
increase will translate into an increase in the basolateral activity of the 70-pS apical K channel (ROMK, see previous)
membrane Cl conductance. via the production of 20-HETE, a P450 metabolite of arachi-
donic acid.259,260 Even changes in serum calcium within the
Prostaglandins physiologic range can alter NaCl absorption via the CaSR.261
Prostaglandin 1 (PGE2), the major product of prostaglan- This may help to explain the hypotensive effect of high cal-
din synthesis in the renal medulla, participates in a local cium intake in salt-sensitive hypertensive individuals.262
negative-feedback system that modulates the rate of NaCl
absorption by the TAL. PGE2 resulted in a 50% reduction Modulation of NaCl Absorption by Other
in ADH-stimulated net Cl absorption in isolated perfused Peptide Hormones
mammalian mTAL segments, but had no effect in the absence In addition to ADH, a number of other peptide hormones
of ADH.247 It is likely that PGE2 inhibits ADH-stimulated stimulate adenylate cyclase activity in the TAL. In the mouse
generation of cAMP in the mTAL by activating an inhibitory and rat, glucagon stimulates NaCl reabsorption and increases
G protein, Gi.247 Although interstitial cells are a major source the transepithelial potential in isolated microperfused mTAL
of PGE2 production in the renal medulla, thick ascending segments. Calcitonin and PTH stimulate sodium transport
limb cells can metabolize arachidonic acid through at least in the cortical, but not the medullary, portions of the TAL.263
two pathways. Escalante et al.248 demonstrated that puri ed Ang II receptors are present in the thick ascending
medullary thick ascending limb cells produce 20-hydroxy- limb.264 Ang II has been reported to both stimulate and inhibit
eicosatetraenoic acid (20-HETE) via the cytochrome P450 sodium transport in the thick ascending limb.265,266 Chronic
enzyme, -hydroxylase. 20-HETE was subsequently shown infusion of Ang II increased the abundance of NKCC2 in the
to inhibit NaCl transport in the thick ascending limb at rat outer medulla by 87%.267 In a model of heart failure, in
steps, which include the apical K channel249 and the Na - which Ang II levels are elevated, NKCC2 expression was also
K -2Cl cotransporter.248 Thick ascending limb cells, par- increased and this increase could be prevented by treatment
ticularly in the macula densa (MD), also express COX-2.250 with an angiotensin receptor antagonist.268
COX-2 expression in these cells may be coupled to renin
secretion. Thus, COX-2 expression is increased by salt re- Adrenergic Agents
striction, diuretics, and, in Bartter syndrome, all conditions -Agonist-sensitive adenylate cyclase activity is present in the
characterized by hyperreninemia.251 Moreover, COX-2 in- rat, but not the rabbit TAL.269 Likewise, -adrenoceptors have
hibitors reduce renin secretion in these settings.252 been detected along the rat TAL by autoradiographic local-
ization.270 The physiologic effects of adrenergic agents have
Osmolality been tested in micropuncture and in in vitro microperfusion
Peritubular osmolality is a third factor regulating mTAL salt studies. DiBona and Sawin 271 demonstrated an enhancement
absorption. In isolated mouse and rabbit TAL segments, of loop NaCl absorption during low-frequency renal nerve
increases in peritubular osmolality rapidly and reversibly stimulation. Acute renal denervation, on the other hand,
inhibit the ADH-stimulated rate of net Cl absorption.243 depressed NaCl absorption by the loop of Henle.272
Molony and Andreoli253 determined that hypertonicity in- Micromolar concentrations of isoproterenol stimulate
hibits the basolateral membrane Cl conductance. This net Cl absorption by in vitro perfused mouse medullary
inhibition of transcellular salt absorption occurs at a locus and cortical TAL.273 The effects of isoproterenol on NaCl ab-
beyond the generation of cAMP, because supramaximal con- sorption in these segments can be blocked by propranolol.
centrations of either ADH or cAMP are unable to reverse the
hypertonicity-mediated effect.254 Thus, increasing the abso- Nitric Oxide
lute magnitude of interstitial osmolality provides a negative Acute administration of nitric oxide donor or L-arginine, the
feedback signal, which can reduce ADH-dependent salt substrate for NOS, decreases NaCl absorption in isolated
absorption by the medullary TAL. perfused thick ascending limb segments.274 The effect of
176
17 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

L-arginine on NaCl transport can be blocked by L-NAME, DCT, so that the Na concentration averages 50 mM at a
an inhibitor of NOS, indicating that endogenous production point 200 to 300 m from the macula densa.285 From there,
of NO mediates the effect of L-arginine. The inhibitory effect tubular Na concentration decreases along the DCT to a
of NO on net NaCl absorption appears to involve, at least in value of approximately 30 mM at the end.286 Tubular uid
part, the inhibition of NKCC2 activity.275 The thick ascend- to plasma Na ratios as low as 0.10 have been observed dur-
ing limb expresses all three isoforms of NOS.276 Plato et al.277 ing stationary microperfusion.287 This nding, together with
used mice de cient in the various NOS isoforms to deter- the presence of the lumen-negative potential difference (see
mine that the effect of L-arginine is mediated by eNOS rather the following), clearly establishes the active nature of Na
than iNOS or nNOS. In contrast to its effect on NaCl ab- absorption in this segment.
sorption, NO stimulates NaHCO3 absorption in the thick Na absorption by the DCT is load dependent. That is,
ascending limb.266 Finally, although short-term exposure to over a wide range of delivery rates, the proportion of Na
NO inhibits NaCl absorption, chronic exposure increases absorbed by the DCT remains constant at 80%.286 At high
NKCC2 expression,278 which could translate into increased tubular uid ow rates, the fall in luminal Na concentra-
NaCl absorption. tion along the tubule is attenuated; thus, more Na is avail-
able to distal Na absorptive sites at high rather than at low
Sodium Balance ow rates.
Dietary Na restriction in rats results in a transient decrease
in NKCC2 expression,279 whereas high Na intake has no Electrophysiologic Considerations
major effect on NKCC2 expression.280 Chronic treatment Depending on the type of electrodes that were used, mea-
with furosemide in conjunction with a high sodium diet in- surements of the transepithelial voltage in the earliest loops
creases NKCC2 expression.281 The latter phenomenon may of the DCT vary from slightly lumen negative to slightly lu-
account for the development of diuretic resistance and the men positive.288–291 Consistent among all measurements,
interdose rebound in sodium absorption in patients chroni- however, is the observation that the transepithelial voltage
cally treated with loop diuretics. becomes progressively more lumen negative as tubular uid
passes to the end of the DCT and into the CNT and CCD.
The progressively lumen-negative transepithelial elec-
THE DISTAL NEPHRON trical potential is primarily due to a change in Na trans-
Anatomic Considerations port pathways from the early DCT to more downstream
nephron segments. As shown previously by Ellison et al.,292
The distal nephron may be divided into three segments: the
Na reabsorption in the early DCT is largely mediated by
DCT, the connecting tubule (CNT), and the collecting duct.
an electroneutral, thiazide-sensitive NaCl cotransporter,
Perhaps owing to the nature of the original micropuncture
whereas Na absorption in the late DCT also involves an
studies characterizing the distal nephron, the DCT was ini-
amiloride-sensitive electrogenic pathway.282 These and other
tially thought to be a segment consisting of a homogeneous
mechanisms of Na absorption are discussed in detail in the
population of epithelial cells. However, more recent work
following sections.
clearly indicates that the DCT can be further divided into
two functionally distinct subsegments, referred to as the
“early” and “late” DCT (DCT1 and DCT2, respectively).282 The Mechanism of Na Absorption
One of the primary features that distinguishes between the A general model for these mechanism by which Na is reab-
early and late portions of the DCT is the differential sensi- sorbed in the early and late DCT is presented in Figure 5.8.
tivity of these segments to the mineralocorticoid hormone
aldosterone. More speci cally, the late DCT expresses the en- The Apical NaCl Cotransport. The absorption of Na and
zyme 11beta-hydroxysteroid dehydrogenase 2 (11 -HSD2), Cl in the early DCT are coupled.293 The coupling of Cl to
which metabolizes cortisol, thereby rendering mineralocor- Na entry provides a mechanism to maintain the intracellu-
ticoid receptors sensitive to aldosterone.283 For this reason, lar Cl activity above its electrochemical equilibrium.294 The
the late DCT, CNT, and the cortical collecting duct (CCD) early distal tubule is the site of action of thiazide diuretics.295
are collectively termed the aldosterone-sensitive distal neph- Autoradiographic studies296 and immunocytochemical stud-
ron (ASDN). ies297 have demonstrated thiazide-binding sites in the apical
membranes of DCT and connecting tubule cells.
Na Transport in the Distal Convoluted The thiazide-sensitive NaCl cotransporter (NCC, TSC,
Tubule and the Connecting Segment SLC12A3) mediates electroneutral NaCl reabsorption in
the early and late DCT (Fig. 5.8).298 The cotransporter
General Characteristics shares considerable sequence homology to the bumetanide-
The DCT absorbs roughly 5% to 10% of the ltered Na sensitive Na -K -2Cl cotransporter (NKCC2) present in
load.284 Fluid enters the DCT with a Na concentration of the TAL,193 yet exhibits markedly different inhibitor sensi-
25 to 30 mM, but salt is added along the initial 20% of the tivity and ionic requirements. NCC transports NaCl with a
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1177
77

FIGURE 5.8 A model of NaCl absorption by cells of the


early and late distal convoluted tubule (DCT). The early
and late DCT express NCC, a thiazide-sensitive, electro-
neutral NaCl cotransporter. The late DCT also expresses
the amiloride-sensitive Na channel ENaC. Basolateral K
channels include Kir4.1, mutated in EAST/SeSAME (epi-
lepsy, ataxia, sensorineural deafness, and tubulopathy/
seizures, sensorineural deafness, ataxia, mental retardation,
and electrolyte imbalance) syndrome, a hereditary salt-
wasting disorder. ROMK, renal outer medullary Kchannel;
NCC, thiazide-sensitive NaCl cotransporter.

1:1 stoichiometry, is K -independent, and is inhibited by Gordon syndrome), have yielded additional insights into the
thiazide diuretics.299 NCC is expressed in the apical mem- regulation of NCC function. FHHt is the phenotypic opposite
brane of DCT cells and extends, in most species, into the of Gitelman syndrome and is characterized by hypertension,
connecting segment.300 hyperkalemia, and metabolic acidosis (Table 5.1). The disor-
der is largely corrected by the infusion of sodium with poorly
Gitelman Syndrome. Loss-of-function mutations in the NCC reabsorbable anions, or by treatment with thiazide diuretics.304
gene have been linked to the pathogenesis of Gitelman These features suggested that the hypertension in FHHt is
syndrome.218 This syndrome resembles Bartter syndrome chloride dependent, and that an increase in NCC activity may
(hypokalemia, alkalosis, sodium wasting), except that uri- be involved in the pathogenesis of PHA II. Positional cloning
nary calcium excretion is reduced in Gitelman syndrome and demonstrated that FHHt is caused by mutations in either of
is elevated in most cases of Bartter syndrome. Thus, physi- two with-no-lysine (WNK) serine-threonine kinases, WNK1
ologically, Gitelman syndrome mimics the effects of thiazide or WNK4.305 These kinases have been intensely studied since
diuretics. Several studies have evaluated the consequences their linkage to FHHt in 2001, and are well-established regu-
of mutations causing Gitelman syndrome on NCC function, lators of NCC, but the mechanisms by which they affect NCC
and in most cases, these mutations alter the NCC coding se- function are complex. Both WNK1 and WNK4 have been
quence in such a way that they reduce NCC expression and shown to regulate NCC plasma membrane traf cking and ac-
traf cking to the apical plasma membrane of the DCT.301,302 tivity (Fig. 5.9). In the current model it is believed that WNK4
In this regard, most of these disease-causing mutations re- acts as an inhibitor of NCC traf cking at the baseline state
sult in the conformational misfolding of NCC, resulting in because it prevents the cotransporter from traf cking from the
the recognition of mutant NCC by chaperone-dependent biosynthetic pathway to the cell surface.306–308 In contrast to
endoplasmic reticulum (ER) quality control machinery that this inhibitory effect on NCC traf c, it has been hypothesized
targets the cotransporter for proteasomal degradation.301,303 that under certain physiologic states that favor NaCl reabsorp-
tion, WNK4 stimulates NCC phosphorylation. Because NCC
Familial Hyperkalemic Hypertension. Studies of another in- is structurally similar to NKCC2, it is activated by the serine–
herited disorder of distal Na transport, familial hyperkale- threonine kinases SPAK and OSR1 (see previous section).
mic hypertension ([FHHt] type 2 pseudohypoaldosteronism, WNK4 and WNK1 activate SPAK and OSR1, which then can
178
17 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Thus, the WNK1 gene mutations that cause FHHt presum-


ably do so by reversing the inhibitory effect of WNK4 on
NCC traf c and by stimulating NCC phosphorylation.315
As with SPAK gene expression in the TAL (see previous),
long and short WNK1 isoforms are expressed in the DCT.
The long form of WNK1 (L-WNK1) is kinase active and
stimulates NCC activity and traf cking through the mecha-
nisms described previously. A short kidney-speci c kinase-
defective product (KS-WNK1), in contrast, suppresses
L-WNK1 activity (Fig. 5.9).316 The balance of these two iso-
forms has been postulated to act as a “switch” that controls
overall L-WNK1 kinase activity in the DCT, which should
in turn regulate NCC activity.310 Consistent with this idea,
selective knockout of KS-WNK1 expression increases NCC
activity, whereas overexpression of the kidney-speci c
isoform in KS-WNK1 transgenic mice causes salt-wasting
through NCC inhibition.317
FIGURE 5.9 A model of thiazide-sensitive cotransporter (NCC)
regulation by the with-no-lysine SPS1-related proline alanine- The Basolateral Electrogenic Na Pump. In both subseg-
rich kinase/oxidative stress responsive kinase 1 (WNK-SPAK/ ments, the basolateral extrusion of Na from the cytosol into
OSR1) signaling pathway. With-No-Lysine (WNK) kinases regu- the peritubular space (and eventually, the plasma) occurs via
late NCC traf cking and phosphorylation. L-WNK1, full-length the electrogenic Na ,K –ATPase. The activity of this pump
kinase-active (“long”) WNK1; KS-WNK1, short kinase-defective results in the generation of a constant transepithelial voltage
“kidney-speci c”WNK1. across the basolateral membrane of 60 to 90 mV.318,319
A reduction in the luminal sodium concentration causes Vbl
to depolarize, whereas increases in the sodium concentra-
bind to NCC and stimulate its activity by phosphorylating a tion hyperpolarizes Vbl. In addition, Vbl depolarizes after
cluster of serines and threonines in its NH2-terminus.307 In ouabain treatment.320 These observations are consistent with
contrast to the TAL, only the full-length kinase active SPAK is the notion that apical Na entry stimulates the electrogenic
expressed in the DCT (i.e., no inhibitory KS-SPAK isoforms Na ,K –ATPase system in the basolateral membrane.
are expressed).237 Due to the observations that WNK4 inhib-
its NCC under some experimental conditions and stimulates Basolateral K Ef ux. Recent insights from rare Mendelian
it under others, several have proposed that the WNK signal- diseases highlight the importance of basolateral K trans-
ing pathway acts as a “switch” that converts the DCT from port in tubular sodium transport in the distal nephron. In
a salt-wasting to a salt-reabsorptive segment, although more 2008, two groups identi ed that patients with mutations in
work is needed to con rm this hypothesis.309–311 In FHHt, the K channel gene KCNJ10 (Kir4.1) develop hereditary
missense mutations of WNK4 stimulate NCC activity through salt wasting.321,322 Patients with these mutations develop a
two mechanisms. First, mutant WNK4 is incapable of revers- complex constellation of neurologic defects in addition to
ing the inhibitory effect on NCC at the baseline; this results in the renal salt wasting. The disease has been named EAST
increased NCC surface expression.308,312 Second, the FHHt- syndrome (epilepsy, ataxia, sensorineural deafness, and tu-
associated mutant increases NCC phosphorylation, presum- bulopathy) by some investigators and SeSAME syndrome
ably through enhanced SPAK/OSR1 activity. Thus, through (seizures, sensorineural deafness, ataxia, mental retarda-
incompletely de ned mechanisms, mutant WNK4 appears to tion, and electrolyte imbalance) by others (Table 5.1). The
“lock” the DCT into a high Na reabsorptive state by increas- salt-wasting phenotype in these patients is reminiscent of
ing the total number of active NCC molecules at the cell sur- Gitelman syndrome, suggesting that the disorder results in
face, resulting in thiazide-sensitive hypertension.313 an impairment of salt transport in the DCT. Indeed, Kir4.1
In contrast, WNK1 appears to in uence NCC traf c is expressed on the basolateral membrane of DCT cells,
indirectly by suppressing the inhibitory effect of WNK4 and patients with EAST/SeSAME syndrome mutations de-
(Fig. 5.9). This effect requires intact WNK1 kinase activity velop markedly reduced infoldings of the DCT basolateral
and releases NCC from intracellular retention, thereby fa- membrane. This de ciency in total basolateral membrane
cilitating NCC delivery to the cell surface.307,308 Unlike content results in a decrease in the number of surface
WNK4, WNK1 mutations that cause FHHt do not alter Na ,K –ATPase molecules, resulting in decreased sodium
the kinase’s coding sequence. Rather, WNK1 mutations pump activity and impaired salt reabsorptive capacity.323
are large intronic deletions that increase the mRNA abun- Additionally, it is thought that loss-of-function mutations of
dance of WNK1 isoforms.305 Kinase-active WNK1 is capa- Kir4.1 impair K recycling across the basolateral membrane,
ble of phosphorylating and activating SPAK and OSR1.314 which reduces the ef cacy of Na ,K –ATPase (Fig. 5.8).321
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1179
79

Basolateral Cl Transport. As mentioned previously, a signi cant increase in NCC expression, which may have
ClC-Kb channels and barttin are both expressed in the been because of the stimulation of mineralocorticoid secre-
DCT, where they mediate basolateral Cl reabsorption.172 tion.334 In contrast, rabbits fed a high Na diet developed an
Because the apical transport mechanism in the DCT via NCC increased rate of DCT Na reabsorption and an increase in
occurs through the coupled reabsorption of Na and Cl , Na ,K –ATPase activity.335
basolateral Cl transport is essential to the development
of a gradient for Na entry and to reduce the intracellular Steroid Hormones. Currently available evidence suggests
Cl concentration, which stimulates SPAK/OSR1–mediated that adrenal steroid hormones regulate Na transport in the
NCC phosphorylation.324 Mutations in either ClC-Kb or the DCT. The presence of both mineralocorticoid and gluco-
accessory subunit result in impaired distal salt reabsorption. corticoid receptors in the DCT has been demonstrated by
Because barttin is required for adequate basolateral Cl re- immunohistochemistry and by hormone binding.283,336,337
absorption in the loop of Henle and DCT (see previous), In addition, an adrenalectomy resulted in a decrease in
patients with barttin mutations develop a more severe form Na ,K –ATPase activity in the DCT.338 The Na ,K –ATPase
of Bartter syndrome.172 activity could be restored by replacement doses of gluco-
corticoids, but not by mineralocorticoids.338,339 Microper-
Apical Conductive Na Channels. The entry of sodium fusion studies of super cial distal tubules (containing both
into the Amphiuma distal tubular cell325 and late rat DCT DCT and CNT), however, demonstrated an increase in Na
cell326 is inhibited by amiloride, a Na channel blocker. transport in animals receiving aldosterone infusions.340,341
A Na channel in the apical membrane would serve to depo- Both the thiazide-sensitive and the thiazide-insensitive
larize the membrane and create the observed lumen-negative components of Na transport were increased by aldoste-
transepithelial potential. This transepithelial voltage, in turn, rone.341 The former may re ect neutral NaCl cotransport
is a driving force for passive Cl reabsorption. Na chan- in the DCT, whereas the latter re ects electrogenic Na
nel subunits have been found by immunolocalization in the absorption in the late DCT or CNT. Aldosterone infusion
late DCT in mouse and rat kidney,327,328 but not the human also resulted in an increase in thiazide-binding sites in the
kidney.329 renal cortex, as determined by [3H]metolazone binding, an
increase in the natriuretic response to thiazide diuretics,
The Regulation of NaCl transport in the Distal and a large increase in NCC protein.341–343 These ndings
establish NCC as an aldosterone-regulated transporter. By
Convoluted Tubule combining immunohistochemical and in situ hybridization
Na Delivery. NaCl reabsorption in the DCT is dependent techniques, Bostanjoglo et al.283 determined that DCT cells
on the delivered load of NaCl.286 The DCT responds to coexpress NCC, mineralocorticoid receptors, and 11beta-
chronic increases in the delivery of NaCl with an increase hydroxysteroid dehydrogenase type 2, an enzyme typically
in the capacity for NaCl transport,330 as well as marked ul- found in mineralocorticoid target sites. Thus, DCT cells,
trastructural changes in the DCT cell. These morphologic particularly those in the late portions of the DCT, express
changes include an increase in the size of the DCT cell, the key elements required for selective mineralocorticoid
an increase in the basolateral membrane surface area, and actions.
an increase in the size of mitochondria.330 Accompanying In 1998, Kim et al.343 showed that aldosterone in-
the functional and morphologic changes are an increase in creases the total protein abundance of NCC. This effect
Na ,K –ATPase activity and an increase in thiazide-binding did not correlate with changes in NCC mRNA abun-
sites.331 These effects appear to result from an increase in dance, suggesting that aldosterone regulates NCC abun-
Na entry into the DCT cell rather than the increase in dis- dance by posttranslational mechanisms. Recent work in-
tal NaCl delivery or changes in plasma aldosterone or ADH dicates that at least two molecular mechanisms account
levels that occur with chronic furosemide treatment. Inhi- for this effect. Aldosterone rapidly induces and activates
bition of NaCl entry into DCT cells with chronic thiazide the serine–threonine kinase serum and glucocorticoid-
treatment resulted in a loss of cell height, loss of normal po- regulated kinase 1 (SGK1), which directly phosphorylates
larity, and apoptosis of the DCT cells.332 The cellular mech- and inactivates two inhibitors of NCC traf cking, WNK4
anisms whereby NaCl entry affects transport function and (described previously), and neural precursor cell-derived,
morphology are not known. developmentally downregulated 4-2 (Nedd4-2), an E3
ubiquitin ligase.344,345 SGK1-mediated phosphorylation of
Dietary Na . Studies in rats and rabbits have yielded con- two residues located at the C-terminus of WNK4 releases
icting results regarding the effects of increased dietary Na NCC from inhibition. This results in WNK4 inactivation
on DCT morphology and Na transport. In rats, no con- and diverts the cotransporter away from intracellular deg-
sistent effect of a high Na diet on either cell morphology, radation pathways, allowing NCC to traf c directly to the
transport rates, or thiazide-receptor density could be plasma membrane from the biosynthetic pathway.344 By
demonstrated.330,331,333 Rats fed a low Na diet279 or phosphorylating Nedd4-2, SGK1 suppresses the ability of
treated with thiazide diuretics,281 however, demonstrated the E3 ligase to attach ubiquitin molecules to NCC; this
180
18 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

reduces NCC degradation and increases its plasma mem-


brane expression.345 Both of these effects provide an expla- Na TRANSPORT IN THE CORTICAL
nation for the aldosterone-induced increase in NCC total COLLECTING DUCT
protein abundance that was initially seen by Kim et al343 General Considerations
Aldosterone also appears to stimulate NCC transport ac-
The transport processes in the collecting duct mediate nal
tivity by promoting NCC phosphorylation, an effect that
adjustments in urinary composition. The collecting duct is
occurs independently of the effects of mineralocorticoids
a major locus of action of mineralocorticoid hormones and
on NCC traf c.346,347 The mechanism by which this effect
plays a major role in K homeostasis and acid–base balance.
occurs remains unclear but appears to correlate directly
Quantitatively, it is a minor site of Na absorption, reclaim-
with increased SPAK/OSR1 activity, suggesting that ad-
ing only about 2% to 4% of the ltered Na load.354
ditional connections between aldosterone and the WNK/
SPAK/OSR1 signaling pathway may exist.
Electrophysiologic Aspects
As mentioned previously, glucocorticoids increase
Na ,K –ATPase activity following an adrenalectomy in The transepithelial voltage in the CCD varies widely from
the DCT.338,339 This effect was not blocked by spironolac- 10 to 100 mV,295,355,356 which largely results from dif-
tone, a mineralocorticoid receptor antagonist, suggesting ferences in mineralocorticoid levels at the time of measure-
that glucocorticoids were acting via glucocorticoid recep- ment in the animals. The reported values for transepithelial
tors rather than mineralocorticoid receptors.339 In addition, resistance also vary widely depending on apical Na and
dexamethasone infusions increased thiazide-sensitive NaCl K channel activities, as shown by the effects of luminal
transport and [3H]metolazone binding sites in adrenalecto- amiloride and barium, respectively, on transepithelial resis-
mized rats.341,342 Nevertheless, the role of glucocorticoids in tance.356,357 The basolateral membrane is conductive to K
the physiologic regulation of Na transport in the DCT re- and, at least in rabbits, Cl .357
mains unclear.
Gonadal steroid hormones may also in uence NaCl Mechanisms of Salt Absorption in
transport in the DCT. Chen et al.348 reported gender Collecting Ducts
differences in the density of thiazide receptors and in the A proposed model for Na absorption and K secretion
natriuretic response to thiazides in rats. Female rats had in the CCD is presented in Figure 5.10. As indicated previ-
higher levels of thiazide-binding sites in the renal cortex ously, apical membranes of CCD principal cells possess con-
than males. The levels in females fell following ovariectomy, ductive pathways for Na and K .358,359 Na enters principal
whereas levels rose in males following orchiectomy. More- cells through Na channels in the apical membrane down its
over, the increase in urinary Na excretion caused by thia- electrochemical gradient. Na is then pumped across the ba-
zides was greater in females than in males, suggesting that solateral membrane by the Na ,K –ATPase in exchange for
the differences in thiazide-binding sites were re ective of dif- K . The Na current across the apical membrane depolarizes
ferences in thiazide-sensitive salt transport in vivo. Likewise,
using antibodies against NCC, Verlander et al.349 found that
estrogen treatment increased NCC expression in the DCT.
These results are consistent with the view that male sex hor-
mones (e.g., testosterone) may downregulate NCC expres-
sion and salt transport, whereas estrogens increase NCC
expression and salt transport in the DCT. The authors are
not aware of gender differences in the response of humans
to thiazide diuretics.

Vasopressin. Recent work indicates that vasopressin is a


potent activator of the thiazide-sensitive cotransporter.350,351
Two groups have shown that the vasopressin analog
deamino-Cys-1, d-Arg-8 vasopressin (dDAVP) stimulates
increases in NCC abundance, traf cking to the plasma mem-
brane, and activation through SPAK/OSR1-mediated phos-
phorylation of its amino terminus. This effect is probably
dependent on cyclic AMP and protein kinase A (PKA), FIGURE 5.10 A model of salt transport by the principal cell
well-established intermediaries of vasopressin-dependent of the cortical collecting duct. Apical Na entry proceeds via
signaling.352 The effect on NCC abundance may be a Nedd4- amiloride-sensitive Na channels (ENaC). Apical K channels
2-dependent process, because PKA can phosphorylate and (ROMK) mediate K secretion by this segment. Cl absorption is
inactivate Nedd4-2 through mechanisms similar to SGK1353; driven by the lumen-negative voltage through the paracellular
however, this hypothesis is yet to be tested. pathway.
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1181
81

the cell, so that the cellular K is above its equilibrium con- cause excessive ENaC accumulation at the apical plasma
centration, and thus leaves the cell through conductive path- membrane, increasing Na absorption. Pseudohypoaldoste-
ways in the apical or basolateral membrane.360 ronism type 1 (PHA-1), the clinical opposite of Liddle syn-
drome, is caused by homozygous inactivating mutations in
Apical Na Channels the ENaC channel, resulting in a syndrome of Na -wasting,
Apical Na entry depolarizes the apical membrane relative to hypotension, and hyperkalemia.379 The majority of muta-
the basolateral membrane, causing a lumen-negative trans- tions causing PHA-1 are frameshift or nonsense mutations
epithelial voltage, which in turn provides the driving force that result in truncated, nonfunctional ENaC proteins.374
for Cl reabsorption through the paracellular pathway. Patch-
clamp studies of collecting duct cells and urinary bladder Apical Electroneutral Na Transport
cells provided some details regarding the electrophysiologic It was rst observed in rats that a portion of Na entry across
properties and regulation of apical membrane Na chan- the apical membrane in perfused CCD segments was sen-
nels.361 The amiloride-sensitive epithelial Na channel sitive to luminal hydrochlorothiazide, which inhibited Na
(ENaC) has been cloned,362,363 has a single-channel conduc- and Cl absorption without changing the transepithelial volt-
tance to Na of 4 to 5 pS, and is highly selective for Na over age.380 In addition, amiloride inhibited Na transport in this
K (PNa/PK 20). ENaC exhibits slow gating, with openings segment by only 50%, and the effects of amiloride and hydro-
and closings lasting several seconds. Amiloride blocks ENaC chlorothiazide were additive. It was thus concluded that the
at submicromolar concentrations.364 CCD may possess two parallel transport pathways for Na :
ENaC consists of three homologous subunits ( , , an electrogenic pathway involving amiloride-sensitive Na
and ).363 The subunits share a common structure consist- channels and a thiazide-sensitive neutral NaCl cotransport
ing of two transmembrane domains, intracellular N- and C- pathway.380 More recently, a Na -dependent Cl /HCO3
termini, and a large extracellular loop.365,366 Although the exchanger (NDCBE/SLC4A8) was identi ed in intercalated
subunit of ENaC can form Na channels on its own,362 cells and found to mediate the amiloride-resistant, thiazide-
coexpression of all three subunits dramatically increases the sensitive electroneutral Na reabsorption in the CCDs of
membrane Na conductance.363 Based on homology to the mice.381 That NCC knockout mice also exhibited signi cant
related acid-sensing ion channel (ASIC1), the crystal struc- natriuresis following treatment with thiazide diuretics sug-
ture of which was recently solved,367 native ENaC likely gests the importance of this pathway. The mechanism for
exists as an , , and heterotrimer. The single channel net NaCl reabsorption in this setting involves the parallel ac-
properties of the expressed channel closely resemble those tivity of the NDCBE and Na -independent Cl /HCO3 ex-
of the 4 to 5 pS highly selective channel studied in native change via pendrin/SLC26A4 at the apical membrane of type
tissues.363 The large extracellular domains of the and B intercalated cells. The basolateral reabsorption pathways
subunits can get proteolytically cleaved by furin in the bio- for the Na and Cl that enter at the apical membrane in
synthetic pathway.368,369 Other proteases like prostasin and this model are the Na ,K –ATPase and ClC-K Cl channels.
plasmin may cleave the subunit.370 These cleavage events Thus, a novel target of thiazide diuretics distinct from NCC
release small inhibitory fragments of the and subunits, has been identi ed in the collecting duct with the implica-
causing an increase in the open probability of ENaC.370 tion that Cl transport by intercalated cells plays an impor-
ENaC activity is also enhanced by increasing luminal ow tant role in blood pressure regulation.382 Because pendrin is
rates, as the extracellular domain of ENaC subunits respond a key mediator of bicarbonate secretion, and thus acid–base
to laminar shear stress.371,372 Thus, collecting duct Na ab- regulation, these ndings also suggest a strong link between
sorption is enhanced with increased distal uid delivery via acid–base and volume/blood pressure regulation.
ENaC, a mechanosensitive channel.373
The key importance of ENaC for Na reabsorption in Control of Na Absorption in the Cortical
the collecting duct is highlighted by naturally occurring Collecting Duct
mutations in ENaC that are responsible for Liddle syn-
drome, an autosomal dominant form of hypertension, and Aldosterone
pseudohypoaldosteronism type 1, an autosomal recessive Aldosterone is one of the key regulators of Na transport
form of salt wasting (see Table 5.1).374 In Liddle syndrome, in the collecting duct, where it increases the rates of Na
mutations in the ENaC subunits result in an increase in absorption and K secretion.383,384 The major target site for
amiloride-sensitive Na channel activity with a consequent mineralocorticoid effects is the principal cell of the CCD,
increase in sodium reabsorption and volume-mediated hy- although actions in the DCT have also been documented
pertension.375–377 Most Liddle mutations occur in the cy- (see previous). Mineralocorticoid effects are produced by the
toplasmic C-termini of the (SCNN1G) and (SCNN1B) binding of either mineralocorticoids or glucocorticoids385
subunits.375 These mutations affect a conserved PY motif in to mineralocorticoid receptors found predominantly in the
the C-terminus, which is necessary for interaction with the CCD.336,337 In addition, the binding of glucocorticoids to
ubiquitin ligase Nedd4-2 and subsequent internalization glucocorticoid receptors can also produce mineralocorti-
and degradation of ENaC.378 Liddle syndrome mutations coid responses.385 This lack of speci city results from two
182
18 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

factors: rst, mineralocorticoid receptors do not discrimi- As discussed earlier, the binding of Nedd4-2 to ENaC
nate between aldosterone and glucocorticoids,386 so that induces internalization and degradation of the channel.
either class of steroids can bind to and activate the recep- Recently, additional aldosterone-induced proteins have
tor. Second, the DNA binding domains of mineralocorticoid been discovered, including the glucocorticoid-induced leu-
and glucocorticoid receptors are highly conserved such that cine zipper protein (GILZ1), the transcription of which is
both receptors can activate many of the same genes.387 The rapidly induced by aldosterone in collecting duct principal
speci city for mineralocorticoids in vivo is provided by the cells. GILZ1 stimulates ENaC-mediated Na transport and
selective degradation of glucocorticoids, but not mineralo- ENaC surface expression by inhibiting Raf-1 in the extracel-
corticoids, by the enzyme 11 -hydroxysteroid dehydroge- lular signal-regulated kinase (ERK) signaling pathway.404,405
nase.386 11 -Hydroxysteroid dehydrogenase activity is high Soundararajan and colleagues406 have also identi ed another
in CCD segments.388,389 Illustrating the important role of this aldosterone-induced protein, CNK3 (connector enhancer of
enzyme in regulating access of hormones to the mineralocor- kinase suppressor of Ras 3), which also stimulates ENaC and
ticoid receptor, genetic de ciency of this enzyme produces appears to be a scaffolding protein. The emerging story is
a syndrome, apparent mineralocorticoid excess, resembling that a multiprotein complex exists in CCD cells that pro-
hyperaldosteronism (hypertension, hypokalemia, metabolic motes context-speci c aldosterone signal transduction to
alkalosis), except that aldosterone levels are low.390 The induce ENaC activity and apical Na conductance. Speci -
clinical manifestations result from the stimulation of cally, aldosterone stimulates ENaC activity through synergis-
mineralocorticoid receptors by circulating glucocorticoids. tic aldosterone-dependent activation by SGK1 via Nedd4-2
In isolated perfused CCDs from mineralocorticoid- and by GILZ1 via Raf-1 and CNK3.
treated rabbits, there is an increase in both the Na and K Nongenomic effects of aldosterone (rapid effects that are
conductance of the apical membrane and an increase in ba- mineralocorticoid receptor-independent) have also been de-
solateral Na ,K –ATPase activity.356 The functional changes scribed in many tissues, including the kidney.407 Elucidating
after aldosterone treatment are accompanied by morpho- the mechanisms of such effects is an area of active investiga-
logic changes in principal cells. The basolateral membrane tion and appears to include the mitogen-activated protein
length of principal cells falls by 35% after an adrenalectomy, kinase (MAPK) pathway and the methylation of proteins and
and the administration of aldosterone, but not dexametha- lipids. Aldosterone also increases the activity of a number of
sone, restores the membrane length to control levels.391 cellular methyltransferase enzymes.408 Moreover, the sub-
An early effect of aldosterone, occurring within a few unit of ENaC itself is a substrate for methylation, and, when
hours of exposure, is an increase in the sodium permeability methylated, exhibits increased Na transport activity.409 The
of the apical membrane of the CCD principal cell. Results effects of aldosterone on Na channel activity may also in-
from electrophysiologic and immunologic studies support volve small GTP-binding proteins, for example, Ras,410 and
the view that the early aldosterone-induced increase in apical phosphatidylinositol 3-kinase.411 SGK1 is a downstream me-
sodium permeability is because of the activation of quiescent diator of phosphoinositide-3-kinase.412
Na channels rather than the synthesis and/or the insertion Sometime after the increase in apical membrane Na con-
of new Na channels into the membrane.392–394 Several ductance occurs, the basolateral membrane Na ,K –ATPase
pathways have been implicated in the aldosterone-mediated activity and pump current increase.413 This increase is due
increase in Na channel activity. An important downstream initially to the effect of increased cell sodium activity on exist-
mediator of aldosterone is the serum and glucocorticoid- ing pump units.414 Later, aldosterone induces the synthesis
regulated kinase SGK1.395 SGK1 is expressed in the thick of additional Na ,K –ATPase pump subunits.415 Increased
ascending limb, DCT, connecting segment, and cortical apical membrane Na entry may promote the late synthesis
collecting tubules.396 Aldosterone increases the transcrip- of Na ,K –ATPase, because the inhibition of sodium entry
tion of SGK1 in vitro,395 although the effects of aldosterone by amiloride markedly reduced the aldosterone-induced in-
on SGK1 protein expression in vivo appear to be minor.396 crease in Na ,K –ATPase activity.416
Coexpression of SGK1 with ENaC results in markedly greater Aldosterone also exerts an additional, late effect on
sodium currents than seen with ENaC alone.395,397 SGK1 is the amiloride-sensitive Na conductance. Patch-clamp
required for the stimulation of sodium transport by aldoste- studies in rats exposed to high levels of aldosterone for sev-
rone both in vitro397 and in vivo.398 SGK1 increases sodium eral days demonstrated a large increase in the amiloride-
currents by increasing cell surface expression of ENaC399 sensitive whole cell Na conductance,417 which correlated
and also increasing the open probability of individual ENaC with an increase in the number of active Na channels in
channels.400 The effect of SGK1 on cell surface expression of the apical membrane.418 Increases in ENaC mRNA419 and
ENaC is largely mediated by Nedd4-2. Speci cally, Nedd4-2 protein 334 levels in aldosterone-treated tissues suggest that
is a substrate for SGK1. Upon phosphorylation by SGK1, the synthesis of new ENaC channels may contribute to the
the af nity of Nedd4-2 for the PY domains of ENaC is di- late aldosterone-induced increase in Na conductance.
minished,401 whereas Nedd4-2 binding to 14-3-3 scaffold- SGK1, in addition to increasing the cell surface expression
ing proteins is increased.402,403 Thus, 14-3-3 proteins act of ENaC, also regulates the transcription of ENaC subunits,
to sequester Nedd4-2 and prevent it from binding to ENaC. principally .420
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1183
83

Antidiuretic Hormone (ADH) in the apical membrane resistance, re ecting decreased Na


Exposure of rat CCD segments in vitro to ADH results in a entry through Na channels. These changes appear to result
sustained stimulation of Na absorption.421 ADH increases from an inhibition of adenylate cyclase and antagonism of
the transepithelial potential, depolarizes the apical mem- ADH by 2 agonists.435
brane, and increases the conductance of the apical membrane Prostaglandin E2 exerts diuretic and natriuretic effects
of principal cells.422 These changes are entirely reversed by on the kidney. Part of this action is mediated by an inhi-
luminal amiloride, indicating that ADH increases the apical bition of Na absorption in the CCD.436 The application
membrane sodium conductance of the principal cell. These of PGE2 to the basolateral surface of perfused rabbit CCD
effects of ADH are mediated intracellularly by cAMP.423 segments reversibly inhibits the negative transepithelial
Moreover, the effects of ADH on Na transport in the rat voltage and net sodium absorption.436 The effect of PGE2
CCD are enhanced by prior treatment of the animals with on sodium transport is coupled to a rise in intracellu-
mineralocorticoids.421 lar [Ca2 ] and is dependent on the activation of PKC.437
ADH and aldosterone increase apical membrane ENaC Four PGE2 receptor subtypes, designated EP1, EP2, EP3,
activity through both overlapping and distinct mechanisms and EP4, have been characterized. Studies using receptor
of action. Like aldosterone via SGK1, ADH induces enhanced subtype–speci c agonists and antagonists suggest that the
PKA-dependent phosphorylation of Nedd4-2 at phosphory- EP1 receptor mediates PGE2-dependent inhibition of Na
lation sites that overlap those of SGK1.353 As described pre- transport in the CCD.438 This inhibition of Na transport is
viously, these phosphorylation events promote sequestration associated with depolarization of the apical membrane volt-
of Nedd4-2 through enhanced binding to 14-3-3 proteins. In age, consistent with inhibition of the basolateral Na ,K -
contrast to aldosterone, which can activate quiescent chan- ATPase.439 In contrast to the inhibitory effect of basolateral
nels, ADH, via cAMP, promotes the insertion of additional PGE2 on Na transport, luminal PGE2 increases transepi-
Na channels into the apical membrane.424 In addition, thelial voltage and presumably Na absorption via the EP4
PKA-mediated phosphorylation may also directly stimulate receptor.440
the activity of ENaC channels already present in the apical Epidermal growth factor (EGF) reduces Na absorption
membrane, potentially both directly425 and indirectly via in rabbit CCD by about 50% via the inhibition of apical elec-
Nedd4-2 inhibition, causing enhanced channel residency trogenic Na entry via ENaC.441,442 More recent studies per-
time at the membrane and thus proteolytic cleavage.370 formed on cultured mouse CCD cells suggest that the effects
Long-term exposure to ADH may also increase the capacity are mediated by the ErbB2 EGF receptor and that there is
of the collecting duct for Na transport by increasing the actually a biphasic effect of EGF on ENaC activity.443 Acutely
expression of both Na ,K –ATPase and ENaC.426 ( 4 h), EGF treatment increases the ENaC current, an effect
that appears to be mediated via the PI-3-kinase pathway.
Chronically ( 8 h), the ENaC current was inhibited via
Other Agents effects through the MEK/ERK pathway.443
Bradykinin is produced in the connecting duct427 and binds Endothelin-1 (ET-1) is heavily secreted from the basolat-
to speci c receptors in the CCD.428 An intrarenal infu- eral aspect of CD epithelial cells where it binds to basolateral
sion of bradykinin produces a diuresis. Bradykinin has ETB receptors and may thus act in an autocrine fashion to
been reported to reduce Na absorption in rat CCD seg- inhibit both Na and water reabsorption in this segment, as
ments.429 More recently, bradykinin was shown to acutely shown in vitro.444 The physiologic relevance of these nd-
and reversibly decrease the open probability of ENaC in ings has been con rmed in vivo, because mice with CD-
patch-clamp studies performed ex vivo on split open CCDs speci c knockout of ET-1 or ET receptors are hypertensive
isolated from rats. This effect appears to be mediated speci - on a normal Na diet and have exacerbated Na retention
cally through the bradykinin B2 receptor.430 and hypertension when placed on a high-Na diet. ET-1
As will be discussed in the following section, the ma- inhibits ENaC activity through Src- and MAPK-dependent
jor site of action of atrial natriuretic peptide (ANP) is the pathways.444
inner medullary collecting duct. ANP stimulates cGMP Nitric oxide is also a downstream mediator of ET-1,444
production in the CCD,431 and inhibits the hydro-osmotic where it decreases Na transport in rat CCD segments by
actions of ADH in the CCD.432 Biphasic effects of ANP on 40% to 80%.445 The addition of nitric oxide to tubules de-
conscious, sedated rats have been observed with acute water creased the intracellular [Na ], but did not affect the activity
and sodium excretion within minutes, followed by retention of basolateral Na ,K –ATPase, suggesting that the primary
after 90 minutes, which was associated with increased api- effect of nitric oxide is to inhibit apical Na entry via ENaC.
cal membrane expression of - and -ENaC. These delayed The inhibitory mechanism involving NO remains to be
effects may represent a compensatory response to increase determined.444
sodium and water reabsorption and prevent volume deple- Dopamine inhibits ADH-dependent Na transport and
tion in response to prolonged ANP infusion.433 transepithelial voltage in rat and rabbit CCD segments, al-
2-Adrenergic agonists inhibit sodium reabsorption in though the particular basolateral dopamine receptor subtype
the rat CCD.434 This inhibition is associated with an increase may be species speci c.446,447
184
18 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Ang II stimulates sodium channel activity in rabbit and


mouse CCDs.448 Chronic Ang II infusion also increases NA TRANSPORT IN THE INNER
the abundance of the subunit of ENaC, the rate-limit- MEDULLARY COLLECTING DUCT
ing subunit for ENaC assembly. 449 Both the acute effects Mechanism of Na Transport
of Ang II on ENaC activity and the chronic effects on
The analysis of salt transport by the inner medullary collecting
ENaC abundance are mediated by the AT1 receptor.448,449
duct (IMCD) has been confounded by problems of axial tu-
A more recent study performed on rats suggests that the
bule heterogeneity, species variability, and differences in exper-
Ang II-induced ENaC stimulation downstream of the AT1
imental approach. Based on morphologic factors, the IMCD
receptor involves a Ca2 -independent PKC pathway that
has been divided into three subsegments: IMCD1, IMCD2,
induces superoxide generation. The blocking of arachi-
and IMCD3.459 This morphologic heterogeneity is paralleled,
donic acid–induced inhibition of ENaC may also play
to some extent, by functional heterogeneity. For example, the
a role.450
urea permeability and its responsiveness to ADH increases
Bicarbonate (HCO3 ) changes in the CCD lumen, which
from IMCD1 to IMCD3 with increasing medullary depth.460
may re ect changes in total-body acid–base status or local
Further complicating the analysis is the observation that simi-
HCO3 secretion from neighboring intercalated cells via the
lar subsegments from different species exhibit different proper-
apical Na -independent Cl /HCO3 exchanger, pendrin,
ties relating to salt transport.461,462 Finally, for unclear reasons,
have been shown recently to modulate Na reabsorption
studies examining IMCD function in vivo (e.g., by microcathe-
in the CCD.451,452 Speci cally, the bicarbonate-stimulated
terization)463 have yielded markedly different results than have
soluble adenylyl cyclase (sAC) appears to stimulate both
in vitro studies of isolated perfused tubules.461,464
basal and agonist-stimulated Na reabsorption in the kidney
Microelectrode impalement studies of IMCD segments
collecting duct, acting to enhance Na ,K –ATPase catalytic
from rats demonstrated that the apical membrane consti-
activity.451 Another study suggests that pendrin increases
tuted the major cellular resistance, and the luminal appli-
ENaC abundance and activity, at least in part by increasing
cation of amiloride increased the apical membrane voltage
luminal [HCO3 ].452
and resistance and decreased the transepithelial voltage.461
The elucidation of other cellular signaling pathways and
These results, and others,465 are consistent with the presence
kinases that are important in the regulation of ENaC, and
of an amiloride-sensitive sodium conductance in the apical
Na transport in the CCD is a very active area of research and
membrane of IMCD cells. Patch-clamp studies of cultured
involves signaling pathways downstream of diverse stimuli,
rat IMCD cells indicate that Na entry is mediated by a 20 to
including in ammation (IkappaB kinase/NF-kappaB/NF- B
30 pS amiloride-sensitive, nonselective, cGMP-gated cation
pathway)453 and metabolic stress (AMP-activated protein
channel in the apical membrane.465,466 The basolateral mem-
kinase).454 A full treatment of these signaling pathways is
brane of IMCD cells contains the Na ,K –ATPase, a K con-
beyond the scope of this chapter, but the interested reader is
ductance and a HCO3 conductance.461
referred to a recent review.455
The results discussed previously can be combined into
a model for sodium transport in the IMCD (Fig. 5.11). Na
NA TRANSPORT IN THE OUTER
MEDULLARY COLLECTING DUCT
The transport properties of the outer medullary collecting
duct (OMCD) have been studied by in vitro perfusion of
isolated tubule segments. The functional properties of the
OMCD differ depending on the location of the segment with-
in the outer medulla. Segments within the outer stripe of the
outer medulla (OMCDo) exhibit electrophysiologic proper-
ties resembling the cortical collecting duct; that is, a lumen
negative transepithelial voltage and electrogenic apical Na
entry.456 Compared to the CCD, the OMCDo displays a less
negative transepithelial voltage, much lower ionic perme-
abilities, and a lower rate of active reabsorption of Na .457
As the collecting duct descends into the medulla, principal
cells, which mediate Na and K transport in the CCD (see
previous), are replaced by cells the electrical properties of
which are similar to intercalated cells of the CCD (i.e., the
apical membrane lacks a demonstrable Na or K conduc- FIGURE 5.11 A model of Na transport in the inner medullary
tance).458 Within the inner stripe of outer medulla (OMCDi), collecting duct. Apical Na entry proceeds via a nonselective
principal cells are virtually absent and no net Na absorp- amiloride-sensitive cation channel. Basolateral Na -K -2Cl
tion occurs.457 cotransport may be involved in Na secretion.
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1185
85

entry across the apical membrane occurs down its steep elec- REFERENCES
trochemical gradient through amiloride-sensitive Na chan- 1. Einstein
Einstein A. The elementary theory of o the Brownian motion. Z. Elecktro-
nels. Na is extruded across the basolateral membrane by the chem. 1908;14:235.
Na ,K –ATPase. The basolateral membrane K conductance 2. Goldman
Goldman D. D. Potential, impedance, and recti cation in membranes. J Gen
Physiol. 1944;27:37.
serves to recycle the K that enters via the Na ,K –ATPase. http://www.ncbi.nlm.nih.gov/pubmed/19873371
3. Hodgkin AL, Katz B. The effect of sodium ions on the electrical activity of
The K conductance also hyperpolarizes the cell, thereby 3. axon
giant Hodgkinof theAL, KatzJB.
squid. The e 1949;108:37.
Physiol. ect o sodium ions on the electrical activity o
favoring Na entry across the apical membrane. giant
4. axon
Kedem o the squid. J Physiol.
O, Katchalsky 1949;108:37. analysis of the permeability of
A. Thermodynamic
http://www.ncbi.nlm.nih.gov/pubmed/18128147
biological membranes to non-electrolytes. Biochim Biophys Acta. 1958;27:229.
There is some evidence for additional electroneutral 4. KedemK,O,Kasahara
5. Takata Katchalsky A. Thermodynamic
T, Kasahara analysis oofthe
M, et al. Localization Na(permeability
)-dependent o
Na entry pathways in rat IMCD cells, although their im- biological
active typemembranes to non-electrolytes.glucose
and erythrocyte/HepG2-type Biochimtransporters
Biophys Acta.in1958;27:229.
rat kidney: im-
portance in net Na absorption by the IMCD in other spe- http://www.ncbi.nlm.nih.gov/pubmed/13522722
muno uorescence and immunogold study. J Histochem Cytochem. 1991;39:287.
5. TakataL,K,Lewis
6. Reuss Kasahara T, Kasahara
SA, Wills NK, etM, al. etIon
al.transport
Localization o Na(+)-dependent
processes in basolateral
cies remains unclear. Furosemide and thiazide diuretics both active
membranestype and erythrocyte/HepG2-type
of epithelia. Fed Proc. 1984;43:2488.glucose transporters in rat kidney: im-
inhibit a portion of Na absorption by rat IMCD segments in munof uorescence
7. Schmidt and immunogold
U, Dubach UC. Na Kstudy. ATPase J Histochem
in the ratCytochem.
nephron 1991;39:287.
related to so-
vivo.467,468 However, Zeidel and colleagues469 failed to dem- http://www.ncbi.nlm.nih.gov/pubmed/1993828
dium transport; results with quantitative histochemistry. Part II. Curr Probl Clin
6. Reuss
Biochem. L, Lewis SA, Wills NK, et al. Ion transport processes in basolateral
1971;3:320.
onstrate sensitivity of rabbit IMCDs to loop diuretics and membranes
8. Kirk KL, o epithelia.
Halm DR,Fed Proc. 1984;43:2488.
Dawson DC. Active sodium transport by turtle colon
found that conductive Na entry at the apical membrane via7.an Schmidt
electrogenic U, Dubach UC. Napump.
Na-K exchange K ATPase Nature.in the rat nephron related to so-
1980;287:237.
predominated. A Na -K -2Cl cotransporter appears to be dium transport;
9. Wang results with
W, Messner quantitative histochemistry.
G, Oberleithner H, et al. The effect Part of
II. ouabain
Curr Probl onClin
in-
Biochem.
tracellular1971;3:320.
activities of K , Na , Cl-, H and Ca2+ in proximal tubules of frog
present in the basolateral membrane of terminal IMCD seg- http://www.ncbi.nlm.nih.gov/pubmed/4280963
kidneys. P ugers Arch. 1984;401:6.
ments.470 The cloned Na -K -2Cl cotransporter from cul- 8. Soltoff
10. Kirk KL, SP,Halm
Mandel DR,LJ.
Dawson
Active DC.ion Active
transport sodium
in thetransport by turtletubule.
renal proximal colon
tured IMCD cells represents the “secretory” isoform of the via an electrogenic
II. Ionic dependence Na-K exchange
of the Na pump. pump.J Gen Nature.
Physiol. 1980;287:237.
1984;84:623.
http://www.ncbi.nlm.nih.gov/pubmed/6253798
11. Schultz SG, Curran PF. Coupled transport of sodium and organic solutes.
Na -K -2Cl cotransporter (NKCC1) rather than the ab- 9. Wang
Physiol W, Messner G, Oberleithner H, et al. The e ect o ouabain on in-
Rev. 1970;50:637.
sorptive isoform present in the apical membrane of the thick tracellular
12. Lassiter activities o K+, Na+,
WE, Gottschalk CW,Cl-,Mylle
H+ and Ca2+ in proximal
M. Micropuncture studytubules
of netotrans-
rog
ascending limb.471 Finally, the Na -HCO3 cotransporter kidneys. Pf ugers Arch.
tubular movement 1984;401:6.
of water and urea in nondiuretic mammalian kidney. Am J
10. Solto
Physiol. 1961;SP, 200:1139.
Mandel LJ. Active ion transport in the renal proximal tubule. II.
NBCn1 has been found in the basolateral membrane of rat Ionic
13. dependence o the Na pump. JB,Gen
Ullrich KJ, Schmidt-Nielson Physiol.
O’Dell R, 1984;84:623.
et al. Micropuncture study of
IMCDs, which may contribute to cellular defense against http://www.ncbi.nlm.nih.gov/pubmed/6094705
composition of proximal and distal tubular uid in rat kidney. Am J Physiol.
both acidi cation and volume changes in this segment.472 11. Schultz SG, Curran PF. Coupled transport o sodium and organic solutes.
1963;204:527.
Physiol Rev. 1970;50:637.
14. Welling LW, Welling DJ. Surface areas of brush border and lateral cell walls
http://www.ncbi.nlm.nih.gov/pubmed/4919599
in the rabbit proximal nephron. Kidney Int. 1975;8:343.
Regulation of Na Transport 12. Lassiter WE,
15. Andreoli Gottschalk
TE, Schafer CW, MylleSL.
JA, Troutman M.Perfusion
Micropuncture study o net
rate-dependence of trans-
tubular
epithelialmovement
osmosis in o isolated
water and urea in convoluted
proximal nondiuretictubules:
mammalian kidney.ofAm
estimation theJ
The IMCD appears to be the major target site for the potent Physiol.
hydraulic 1961; 200:1139.Kidney Int. 1978;14:263.
conductance.
diuretic hormone ANP.473 This hormone, working through http://www.ncbi.nlm.nih.gov/pubmed/13759324
16. Giebisch G, Klose RM, Malnic G, et al. Sodium movement across single
cGMP,474 inhibits Na entry via apical Na channels. In cell- 13. Ullrich
perfused KJ, Schmidt-Nielson
proximal tubules of rat kidneys. B, O’Dell
J GenR,Physiol.
et al. 1964;47:1175.
Micropuncture study o
composition
17. Barratt LJ, o proximal
Rector FCand distal JP
Jr, Kokko tubular f uid ingoverning
, et al. Factors rat kidney. theAm J Physiol.
transepithe-
attached and excised patches of cultured inner medullary 1963;204:527.
lial potential difference across the proximal tubule of the rat kidney. J Clin Invest.
cells, both ANP and dibutyryl cGMP inhibited the activity of http://www.ncbi.nlm.nih.gov/pubmed/13995307
1974;53:454.
the cation channel.475 14. Welling LW,
18. Andreoli TE,Welling
SchaferDJ. JA,Sur ace areasSL,
Troutman o etbrush border and
al. Solvent draglateral cell walls
component of
in
Cl-theux rabbit proximal
in super nephron.straight
cial proximal Kidney tubules:
Int. 1975;8:343.
evidence for a paracellular com-
In addition to inhibiting Na reabsorption, ANP may http://www.ncbi.nlm.nih.gov/pubmed/1206850
ponent of isotonic uid absorption. Am J Physiol. 1979;237:F455.
stimulate Na secretion in the IMCD.470 In isolated perfused 15. Andreoli
19. Burg MB,TE, SchaJ.erElectrical
Orloff JA, Troutman SL. Per
potential usion rate-dependence
difference across proximaloconvo- trans-
IMCD segments, ANP increased the bath to lumen ux rate epithelial
luted tubules.osmosisAm in isolated1970;219:1714.
J Physiol. proximal convoluted tubules: estimation o the hy-
draulic conductance.
20. Schafer JA, Patlak Kidney Int. 1978;14:263.
CS, Andreoli TE. A component of uid absorption linked
of Na and Cl , an effect that was inhibited by peritubu- http://www.ncbi.nlm.nih.gov/pubmed/723152
to passive ion ows in the super cial pars recta. J Gen Physiol. 1975;66:445.
lar furosemide and by omission of either counterion, thus 16. Giebisch G,AB.
21. Maunsbach Klose RM, MalniconG,the
Observations et segmentation
al. Sodium movement acrosstubule
of the proximal single
suggesting the role of a basolateral membrane Na -K -2Cl per
in theusedratproximal tubules o ratofkidneys.
kidney. Comparison J Genphase
results from Physiol. 1964;47:1175.
contrast, uorescence and
http://www.ncbi.nlm.nih.gov/pubmed/14192552
electron microscopy. J Ultrastruct Res. 1966;16:239.
cotransporter.476 Sands et al.,464 however, found no effect of 17. Barratt LJ,
22. Doucet A, Rector
Katz AI, FC Morel
Jr, Kokko JP, et al. Factorsofgoverning
F. Determination Na-K-ATPase the transepithe-
activity in
ANP on the Na permeability of the rat IMCD. lial potential
single segments di oference across the proximal
the mammalian nephron.tubule o the rat
Am J Physiol. kidney. J Clin Invest.
1979;237:F105.
In one study,477 ADH stimulated amiloride-sensitive 1974;53:454.
23. Jacobson HR. Transport characteristics of in vitro perfused proximal con-
http://www.ncbi.nlm.nih.gov/pubmed/11344559
voluted tubules. Kidney Int. 1982;22:425.
Na absorption by terminal IMCD segments perfused in vi- 18. Andreoli HR.
24. Jacobson TE, Scha er JA, Troutman
Characteristics of volume SL, et al. Solvent in
reabsorption drag component
rabbit super cialo
tro. Other studies, however, failed to nd an effect of ADH Cl-
andfjuxtamedullary
ux in super cialproximalproximalconvoluted
straight tubules:
tubules. evidence or a paracellular
J Clin Invest. 1979;63:410. com-
or cAMP on the amiloride-sensitive cation channel in apical ponent o isotonicS,f uid
25. Kawamura Imaiabsorption.
M, Seldin Am DW,J Physiol. 1979;237:F455.
et al. Characteristics of salt and water
19. BurginMB,
transport OrlocialJ. Electrical
super potential di straight
and juxtamedullary erence across
segmentsproximal convo-
of proximal
membranes of cultured IMCD cells.464,465 luted
tubules.tubules.
J Clin Am J Physiol.
Invest. 1970;219:1714.
1975;55:1269.
Micropuncture studies by Ullrich and Papavassiliou478 20. Scha erFC
26. Rector JA,Jr.
Patlak CS, Andreoli
Sodium, bicarbonate,TE. Aand component
chloride oabsorption
f uid absorption
by the linked
proxi-
demonstrated that mineralocorticoids increased net Na ab- to
malpassive
tubule.ion Am f ows in the1983;244:F461.
J Physiol. super cial pars recta. J Gen Physiol. 1975;66:445.
http://www.ncbi.nlm.nih.gov/pubmed/1181377
27. Schafer JA, Williams JC Jr. Transport of metabolic substrates by the proxi-
sorption in the terminal IMCD, which was attributed to a de- 21.nephron.
mal Maunsbach AnnuAB. RevObservations on the segmentation o the proximal tubule
Physiol. 1985;47:103.
crease in the passive Na permeability of the tubule leading to a in28.
theKokko
rat kidney. Comparison
JP. Proximal tubule o potential
results rom phase contrast,
difference. Dependencef uorescence and
on glucose,
decrease in back leak of NaCl into the lumen. Mineralocorticoids electron
HCO3, and microscopy.
amino acids.J Ultrastruct Res. 1966;16:239.
J Clin Invest. 1973;52:1362.
http://www.ncbi.nlm.nih.gov/pubmed/5333381
29. Schafer JA, Troutman SL, Andreoli TE. Volume reabsorption, transepithe-
may also increase active Na reabsorption in the IMCD. For ex- 22.potential
lial Doucetdifferences,
A, Katz AI,and Morel F. Determination
ionic o Na-K-ATPase
permeability properties activity in
in mammalian sin-
super-
ample, aldosterone produced a three- to seven-fold stimulation gle segments
cial proximalo straight
the mammalian
tubules. Jnephron.
Gen Physiol.Am 1974;64:582.
J Physiol. 1979;237:F105.
of electrogenic Na transport in cultured rat IMCD cells,479 and 23. Jacobson
30. Schafer JA,HR. Transport
Troutman SL,characteristics
Watkins ML, eto al.inVvitro olumeper used proximal
absorption in the con-
pars
voluted
recta. I. tubules.
“Simple”Kidney
active NaInt. 1982;22:425.
transport. Am J Physiol. 1978;234:F332.
chronic mineralocorticoid exposure in vivo increased the activ- http://www.ncbi.nlm.nih.gov/pubmed/6759752
31. Barfuss DW, Schafer JA. Active amino acid absorption by proximal convo-
ity of Na ,K ATPase in the IMCD.480 luted and proximal straight tubules. Am J Physiol. 1979;236:F149.
186
18 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

24. McKeown
32. Jacobson HR. JW, Characteristics
Brazy PC, Dennis o volume reabsorption
VW. Intrarenal in rabbit super
heterogeneity for uid,cial 51. Wright
65. Biemesder EM,erLooD, Pizzonia
DD, HirayamaJ, Abu-Al BA.a A, et al. NHE3:
Biology of human a Na+/H+
sodium exchanger
glucose
and juxtamedullary
phosphate, and glucose proximal convoluted
absorption in the tubules.
rabbit. Am J Clin Invest.1979;237:F312.
J Physiol. 1979;63:410. iso orm o renal
transporters. brush
Physiol Rev.border. Am J Physiol. 1993;265:F736.
2011;91:733.
http://www.ncbi.nlm.nih.gov/pubmed/429562
33. Schafer JA, Barfuss DW. The study of pars recta function by the perfusion http://www.ncbi.nlm.nih.gov/pubmed/8238556
66. Kanai Y, Lee WS, You G, et al. The human kidney low af nity Na /glucose
of25. Kawamura
isolated S, Imai M,Kidney
tubule segments. SeldinInt. DW, et al. Characteristics o salt and water
1982;22:434. 52. Schultheis
cotransporter PJ, Clarke
SGLT2. LL, Meneton
Delineation P, et al.
of the major Renal
renal and intestinal
reabsorptive absorptive
mechanism for
transport
34. Welling in super cial andDJ.juxtamedullary
LW, Welling Relationship between straight structure
segments and o proximal
function tu-in de ects in mice
D-glucose. J Clinlacking
Invest. the NHE3 Na+/H+ exchanger. Nat Genet. 1998;19:282.
1994;93:397.
bules.proximal
renal J Clin Invest.tubule.1975;55:1269.
J Electron Microsc Tech. 1988;9:171. http://www.ncbi.nlm.nih.gov/pubmed/9662405
67. Vallon V, Platt KA, Cunard R, et al. SGLT2 mediates glucose reabsorption
http://www.ncbi.nlm.nih.gov/pubmed/1133172
35. Seely JF, Chirito E. Studies of the electrical potential difference in rat proxi- in53. Vallonproximal
the early V Schwark JR, Richter
tubule. J Am Soc K, Nephrol.
et al. Role o Na(+)/H(+) exchanger NHE3
2010;22:104.
26.tubule.
mal RectorAm FCJ Physiol.
Jr. Sodium, bicarbonate, and chloride absorption by the proxi-
1975;229:72. in68.
nephron
van den unction:
Heuvelmicropuncture
LP, Assink K, studies Willemsen with M,S3226,
et al.anAutosomal
inhibitor orecessive
NHE3.
mal
36.tubule.
Lutz MD, Am JCardinal
Physiol. 1983;244:F461.
J, Burg MB. Electrical resistance of renal proximal Am J Physiol
renal glucosuria Renalattributable
Physiol. 2000;278:F375.
to a mutation in the sodium glucose cotransporter
27. Scha
tubule er JA,inWilliams
perfused vitro. AmJCJ Physiol.
Jr. Transport o metabolic substrates by the proxi-
1973;225:729. 54. Choi
(SGLT2). Hum JY, Genet.
Shah 2002;111:544.
M, Lee MG, et al. Novel amiloride-sensitive sodium-de-
mal
37.nephron.
Seely JF.Annu Rev Physiol.
Variation 1985;47:103.
in electrical resistance along length of rat proximal pendent
69. Martin proton MG, secretion
Turk E, in the mouse
Lostao MP, et proximal
al. Defectsconvoluted
in Na /glucose tubule.cotransporter
J Clin Invest.
http://www.ncbi.nlm.nih.gov/pubmed/2581501
convoluted tubule. Am J Physiol. 1973;225:48. 2000;105:1141.
(SGLT1) traf cking and function cause glucose-galactose malabsorption. Nat
28. Jacobson
38. Kokko JP.HR, Proximal
Kokkotubule potential
JP. Intrinsic di erence.
differences in Dependence
various segments on glucose,
of the 55. Goyal
Genet. S, Vanden Heuvel G, Aronson PS. Renal expression o novel Na+/
1996;12:216.
HCO3, and
proximal amino acids.
convoluted J Clin
tubule. Invest.
J Clin 1973;52:1362.
Invest. 1976;57:818. H+70.exchanger
Patel AK,iso orm NHE8.
Fonseca Am Jglucosuria
V. Turning Physiol Renal into Physiol. 2003;284:F467.
a therapy: Ef cacy and safety
29. Berry
39. Scha er CA, JA,Warnock
TroutmanDG, SL, Rector
Andreoli FCTE. Jr. VIon
olume reabsorption,
selectivity transepithe-
and proximal salt 56. SGLT2
with Biagi B, Kubota T,Curr
inhibitors. Sohtell
DiabM, et al.
Rep. Intracellular potentials in rabbit proxi-
2010;10:101.
lial potential di
reabsorption. Amerences,
J Physiol.and ionic permeability properties in mammalian super-
1978;235:F234. mal
71.tubules
Samarzija per I,used in vitro.
Frömter Am J Physiol. 1981;240:F200.
E. Electrophysiological analysis of rat renal sugar and
cial proximal
40. Gullans SR, straight
Avison tubules.
MJ, Ogino J GenT,Physiol.
et al. NMR 1974;64:582.
measurements of intracellular 57. Sacktor
amino B. Sodium-coupled
acid transport. I. Basic phenomena.hexose transport.
P ugers Arch. Kidney Int. 1989;36:342.
1982;393:179.
http://www.ncbi.nlm.nih.gov/pubmed/4443793
sodium in the rabbit proximal tubule. Am J Physiol. 1985;249:F160. http://www.ncbi.nlm.nih.gov/pubmed/2687560
72. Broer S. Amino acid transport across mammalian intestinal and renal epi-
30. Stroup
41. Scha er RF JA,, Troutman
Weinman SL, E, Watkins
HayslettML, JP, etetal.al.
Volume
Effectabsorption
of luminal in the pars
perme- 58. Aronson
thelia. Physiol Rev. PS,2008;88:249.
Sacktor B. The Na+ gradient-dependent transport o D-glu-
recta. I.on
ability “Simple” active Na+
net transport transport.
across Am J Physiol.
the amphibian 1978;234:F332.
proximal tubule. Am J Physiol. cose
73. inSamarzija
renal brush border E.
I, Fromter membranes. J Biol Chem.analysis
Electrophysiological 1975;250:6032.
of rat renal sugar and
31. Bar uss DW, Scha er JA. Active amino acid absorption by proximal convo-
1974;226:1110. http://www.ncbi.nlm.nih.gov/pubmed/1150669
amino acid transport. III. Neutral amino acids. P ugers Arch. 1982;393:119.
luted
42. and
Warnockproximal DG,straight
Rector tubules.
FC Jr. ProtonAm J Physiol.
secretion1979;236:F149.
by the kidney. Annu Rev 59. Hammerman
74. Aronson PS. Energy-dependence
MR, Sacktor B. Na o-dependent phlorizin binding
transport toofisolated
glycinereinnalrenal
mi-
32. McKeown
Physiol. 1979; 41:197. JW Brazy PC, Dennis VW Intrarenal heterogeneity or f uid, crovillus
brush membranes.
border membrane Evidence
vesicles.concerning
Evidencethe formechanism
a single speci o coupling between
c transport system.the
phosphate,
43. Wistrand and PJ, glucose absorption
Knuuttila KG. Renal in themembrane-bound
rabbit. Am J Physiol. 1979;237:F312.
carbonic anhydrase. electrochemical
Biochim Biophys Na+ Acta.gradient and the sugar transport. J Membr Biol. 1978;42:81.
1982;686:189.
33. cation
Puri Scha er and JA,properties.
Bar uss DW. The Int.
Kidney study o pars recta unction by the per usion
1989;35:851. http://www.ncbi.nlm.nih.gov/pubmed/671529
75. Bohmer C, Broer A, Munzinger M, et al. Characterization of mouse amino
o44.isolated
Kinsellatubule JL, segments.
Aronson PS. Kidney Int. 1982;22:434.
Properties of the Na -H exchanger in renal 60. transporter
acid Bar uss DW, Scha (slc6a19).
B0AT1 er JA. Di erences
BiocheminJ. active and passive glucose transport
2005;389:745.
http://www.ncbi.nlm.nih.gov/pubmed/6759753
microvillus membrane vesicles. Am J Physiol. 1980;238:F461. along the proximal
76. Camargo SM, nephron.
Makrides Am J Physiol.
V, Virkki 1981;241:F322.
LV, et al. Steady-state kinetic characteriza-
34. Aronson
45. Welling LW, Welling DJ. Relationship
PS. Mechanisms of active H between secretionstructure and unction
in the proximal tubule.in 61. ofTurner
tion the mouse RJ, Moran
B(0)AT1 A. sodium-dependent
Stoichiometric studies neutralo the
amino renalacidouter cortical
transporter.
renal
Am proximal
J Physiol. tubule. J Electron Microsc Tech. 1988;9:171.
1983;245:F647. brush
P ugersborder membrane D-glucose transporter. J Membr Biol. 1982;67:73.
Arch. 2005;451:338.
http://www.ncbi.nlm.nih.gov/pubmed/3199236
46. Aronson PS, Nee J, Suhm MA. Modi er role of internal H in activat- http://www.ncbi.nlm.nih.gov/pubmed/7201526
77. Samarzija I, Fromter E. Electrophysiological analysis of rat renal sugar and
35.theSeely
ing Na JF-H , Chirito E. Studies
exchanger in orenalthe electrical
microvillus potential
membranedi erence in ratNature.
vesicles. proxi- 62. Turner
amino RJ, Moran
acid transport. A. Further
IV. Basic amino studies
acids. Po ugersproximal tubular brush border
Arch. 1982;393:210.
mal tubule. Am J Physiol. 1975;229:72.
1982;299:161. membrane
78. Samarzija D-glucose transport
I, Fromter heterogeneity. J Membr
E. Electrophysiological Biol.of1982;70:37.
analysis rat renal sugar and
http://www.ncbi.nlm.nih.gov/pubmed/1147059
47. Boron WF, Boulpaep EL. Intracellular pH regulation in the renal proximal http://www.ncbi.nlm.nih.gov/pubmed/7142902
amino acid transport. V. Acidic amino acids. P ugers Arch. 1982;393:215.
36. Lutz
tubule of theMD, Cardinal Na-H
salamander. J, Burgexchange.
MB. Electrical resistance
J Gen Physiol. o renal proximal tu-
1983;81:29. 63. Tate
79. HedigerSS, YMA,
an N,Coady
UdenfriendMJ, Ikeda TS, et al.cloning
S. Expression Expression
of a Na(cloning and cDNA
)-independent
bule
48. per used inJG,
Haggerty vitro. Am J N,
Agarwal Physiol.
Reilly1973;225:729.
RF, et al. Pharmacologically different Na/H sequencing
neutral amino o the acidNa+/glucose
transporter co-transporter.
from rat kidney. Nature.Proc1987;330:379.
Natl Acad Sci USA.
http://www.ncbi.nlm.nih.gov/pubmed/4726511
antiporters on the apical and basolateral surfaces of cultured porcine kidney cells http://www.ncbi.nlm.nih.gov/pubmed/2446136
1992;89:1.
37. Seely Proc
(LLC-PK1). JF. Variation
Natl Acad in Sci
electrical resistance along length o rat proximal con-
USA. 1988;85:6797. 64. Wells
80. Wells RG,RG, Hediger
Pajor AM, MA. Kanai Y, et of
Cloning al. aCloning
rat kidney o acDNA
human thatkidney cDNAdiba-
stimulates with
voluted
49. Sardettubule. Am J Physiol.
C, Franchi 1973;225:48.
A, Pouyssegur J. Molecular cloning, primary structure, similarity
sic to theamino
and neutral sodium-glucose
acid transport cotransporter.
and has sequenceAm J Physiol.
similarity1992;263:F459.
to glucosidases.
http://www.ncbi.nlm.nih.gov/pubmed/4714410
and expression of the human growth factor-activatable Na /H antiporter. Cell. 65.Natl
Proc Wright
AcadEM, Loo 1992;89:5596.
Sci USA. DD, Hirayama BA. Biology o human sodium glucose
38. Jacobson HR, Kokko JP. Intrinsic di erences in various segments o the
1989;56:271. transporters.
81. Berry CA, Physiol Rev.FC
Rector 2011;91:733.
Jr. Mechanism of proximal NaCl reabsorption in the
proximal convoluted
50. Biemesderfer D,tubule.
Reilly RF J Clin
, ExnerInvest.
M, 1976;57:818.
et al. Immunocytochemical character- http://www.ncbi.nlm.nih.gov/pubmed/21527736
proximal tubule of the mammalian kidney. Semin Nephrol. 1991;11:86.
39. Berry
ization of Na(CA,)-H Warnock DG, Rector
exchanger isoformFC NHE-1Jr. Ioninselectivity
rabbit kidney.and proximal
Am J Physiol.salt 66. Cassola
82. Kanai Y,AC, LeeMollenhauer
WS, You G, etM,al.Fromter The human E. Thekidney low a nity
intracellular Na+/glucose
chloride activity
reabsorption. Am J Physiol. 1978;235:F234.
1992;263:F833. cotransporter
of SGLT2. Delineation
rat kidney proximal tubular cells. o P theugers
major renal
Arch. reabsorptive mechanism or
1983;399:259.
40. Biemesderfer
51. Gullans SR, Avison MJ, Ogino
D, Pizzonia T, et al.
J, Abu-Alfa A,NMR measurements
et al. NHE3: a Na /H o intracellular
exchanger D-glucose.
83. Alpern J Clin Invest. 1994;93:397.
RJ, Howlin KJ, Preisig PA. Active and passive components of chlo-
sodium of
isoform in renal
the rabbitbrushproximal
border. Am tubule. Am J 1993;265:F736.
J Physiol. Physiol. 1985;249:F160. http://www.ncbi.nlm.nih.gov/pubmed/8282810
ride transport in the rat proximal convoluted tubule. J Clin Invest. 1985;76:1360.
41. Schultheis
52. Stroup RF, PJ, Weinman
Clarke E, LL,Hayslett
Meneton JP,Pet
, etal.al.ERenal
ect o and
luminal perme
intestinal ability on
absorptive 67. Lucci
84. VallonMS, V, Platt KA, Cunard
Warnock R, et al.ofSGLT2
DG. Effects mediates glucose
anion-transport inhibitors reabsorption
on NaCl
net transport
defects in mice across
lackingthe the
amphibian
NHE3 Na proximal
/H tubule.
exchanger. Am JNat
Physiol. 1974;226:1110.
Genet. 1998;19:282. in the early proximal
reabsorption in the rattubule.
superJ Am cialSoc Nephrol. convoluted
proximal 2010;22:104. tubule. J Clin Invest.
42. Vallon
53. Warnock DG, Rector
V, Schwark JR,FCRichter
Jr. ProtonK, etsecretion
al. Rolebyofthe kidney.
Na( )/H(Annu Rev Phys-
) exchanger http://www.ncbi.nlm.nih.gov/pubmed/20616166
1979;64:570.
iol. 1979;
NHE3 41:197. function: micropuncture studies with S3226, an inhibitor of
in nephron 68. Green
85. van den R, Heuvel
Bishop LP JH,, Assink
Giebisch K, G.
Willemsen M, et al. Autosomal
Ionic requirements of proximal recessive
tubu-
http://www.ncbi.nlm.nih.gov/pubmed/35088
NHE3. Am J Physiol Renal Physiol. 2000;278:F375. renal
lar glucosuria
sodium attributable
transport. to a mutation
III. Selective luminal in anion
the sodium glucose Am
substitution. cotransporter
J Physiol.
43. Choi
54. Wistrand JY, ShahPJ, Knuuttila
M, Lee KG. MG,Renal et al.membrane-bound carbonic anhydrase.
Novel amiloride-sensitive sodium- (SGLT2). Hum Genet. 2002;111:544.
1979;236:F268.
Puri cationproton
dependent and properties.
secretionKidneyin theInt. 1989;35:851.
mouse proximal convoluted tubule. J Clin http://www.ncbi.nlm.nih.gov/pubmed/12436245
86. Baum M. Evidence that parallel Na -H and Cl(-)-HCO3-(OH-) anti-
http://www.ncbi.nlm.nih.gov/pubmed/2496258
Invest. 2000;105:1141. 69. Martin
porters transport MG,NaClTurkinE,theLostao MP, ettubule.
proximal al. De ects
Am JinPhysiol.
Na+/glucose cotransporter
1987;252:F338.
44. Goyal
55. Kinsella JL, Aronson
S, Vanden Heuvel PS.G,Properties
Aronson PS. o the Na+
Renal -H+ exchanger
expression of novelinNa renal/ (SGLT1)
87. Karniskitra cking and unction
LP, Aronson cause glucose-galactose
PS. Chloride/formate exchange with malabsorption.
formic acid Nat re-
microvillus
H exchanger membrane
isoform vesicles.
NHE8. Am AmJJPhysiol
Physiol.Renal1980;238:F461.
Physiol. 2003;284:F467. Genet. 1996;12:216.
cycling: a mechanism of active chloride transport across epithelial membranes.
45. Biagi
56. Aronson PS. Mechanisms
B, Kubota T, Sohtell M, o etactive H+ secretion
al. Intracellular in the proximal
potentials in rabbit tubule.
proxi- http://www.ncbi.nlm.nih.gov/pubmed/8563765
Proc Natl Acad Sci USA. 1985;82:6362.
Am Jtubules
mal Physiol.perfused
1983;245:F647.
in vitro. Am J Physiol. 1981;240:F200. 70. Schild
88. Patel AK, Fonseca V
L, Giebisch G,Turning
Karniskiglucosuria into a of
LP, et al. Effect therapy:
formateE oncacy and sa
volume ety
reab-
46. Sacktor
57. AronsonB.PS, Nee J, Suhm hexose
Sodium-coupled MA. Modi er roleKidney
transport. o internal H+ in activat-
Int. 1989;36:342. with SGLT2
sorption in theinhibitors. Curr Diab
rabbit proximal Rep. 2010;10:101.
tubule. J Clin Invest. 1987;79:32.
ing
58.the Na+ -H+
Aronson PS, exchanger
Sacktor B.inThe renal Namicrovillus membrane vesicles.
gradient-dependent transport Nature.
of D- http://www.ncbi.nlm.nih.gov/pubmed/20425568
89. Knauf F, Yang CL, Thomson RB, et al. Identi cation of a chloride-formate
1982;299:161.
glucose in renal brush border membranes. J Biol Chem. 1975;250:6032. 71. Samarzija
exchanger expressedI, Frömter
on theE.brushElectrophysiological
border membrane analysis o ratproximal
of renal renal sugar and
tubule
http://www.ncbi.nlm.nih.gov/pubmed/7110335
59. Aronson PS. Energy-dependence of phlorizin binding to isolated re- aminoProc
cells. acidNatl
transport.
Acad SciI. USA.
Basic2001;98:9425.
phenomena. Pf ugers Arch. 1982;393:179.
47.microvillus
nal Boron WFmembranes.
, Boulpaep EL. Intracellular
Evidence pH regulation
concerning in the renal
the mechanism proximal
of coupling http://www.ncbi.nlm.nih.gov/pubmed/7099920
90. Karniski LP, Aronson PS. Anion exchange pathways for Cl- transport in
tubule o the
between theelectrochemical
salamander. Na-H Na exchange.
gradient and J Genthe Physiol.
sugar 1983;81:29.
transport. J Membr Biol. 72. Broer
rabbit S. Amino acid
renal microvillus transport Am
membranes. across mammalian
J Physiol. intestinal and renal epi-
1987;253:F513.
http://www.ncbi.nlm.nih.gov/pubmed/6833996
1978;42:81. thelia. Physiol Rev.
91. Aronson PS. 2008;88:249.
Ion exchangers mediating NaCl transport in the renal proxi-
48. Barfuss
60. Haggerty DW, JG,Schafer
Agarwal JA.N,Differences
Reilly RF, et in al. Pharmacologically
active and passive glucose di erent Na/H
transport http://www.ncbi.nlm.nih.gov/pubmed/18195088
mal tubule. Cell Biochem Biophys. 2002;36:147.
antiporters
along on the apical
the proximal nephron.and basolateral
Am J Physiol. sur1981;241:F322.
aces o cultured porcine kidney cells 73. Jiang
92. Samarzija I, Fromter E. II,
Z, Grichtchenko, Electrophysiological
Boron WF, et al. Speci analysis o of
city ratanion
renal sugar
exchangeand
(LLC-PK1).
61. TurnerProc Natl Acad
RJ, Moran Sci USA. 1988;85:6797.
A. Stoichiometric studies of the renal outer cortical brush amino acidbytransport.
mediated III. Neutral
mouse Slc26a6. J Biolamino
Chem.acids. Pf ugers Arch. 1982;393:119.
2002;277:33963.
http://www.ncbi.nlm.nih.gov/pubmed/2901105
border membrane D-glucose transporter. J Membr Biol. 1982;67:73. http://www.ncbi.nlm.nih.gov/pubmed/7099922
93. Warnock DG, Yee VJ. Sodium uptake mechanisms in brush-border membrane
49. Turner
62. Sardet C, RJ,Franchi
Moran A, A. Pouyssegur
Further studies J. Molecular
of proximal cloning, primary
tubular brush structure,
border 74. Hammerman
vesicles prepared from MR, Sacktor
rabbit renalB.cortex.
Na+ -dependent
Biochim Biophys transport o glycine in renal
Acta. 1982;684:137.
and expression
membrane o thetransport
D-glucose human growth actor-activatable
heterogeneity. J Membr Biol. Na+/H+ antiporter. Cell.
1982;70:37. brush borderSA,
94. Lewis membrane
Wills NK.vesicles. Evidence or
Apical membrane a single speci
permeability andckinetic
transport system.
properties
1989;56:271.
63. Hediger MA, Coady MJ, Ikeda TS, et al. Expression cloning and cDNA Biochim
of Biophyspump
the sodium Acta. in
1982;686:189.
rabbit urinary bladder. J Physiol. 1983;341:169.
http://www.ncbi.nlm.nih.gov/pubmed/2536298
sequencing of the Na /glucose co-transporter. Nature. 1987;330:379. http://www.ncbi.nlm.nih.gov/pubmed/7082661
95. Alpern RJ. Mechanism of basolateral membrane H /OH-/HCO-3 trans-
50. Wells
64. Biemesder
RG, Pajor er D,AM,
Reilly RF, Exner
Kanai Y, et al.M,Cloning
et al. Immunocytochemical
of a human kidney cDNA character-
with 75. inBohmer
port the ratC,proximal
Broer A, Munzinger
convoluted M, et al. Characterization
tubule. A sodium-coupled o mouse amino
electrogenic
ization o toNa(+)-H+
similarity exchanger iso
the sodium-glucose orm NHE-1Am
cotransporter. inJrabbit
Physiol.kidney. Am J Physiol.
1992;263:F459. acid transporter
process. B0AT11985;86:613.
J Gen Physiol. (slc6a19). Biochem J. 2005;389:745.
1992;263:F833. http://www.ncbi.nlm.nih.gov/pubmed/15804236
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1187
87

76. Akiba
96. Camargo SM, Makrides
T, Alpern RJ, EveloffV, Virkki
J, et Lal.
V, et al. Steady-state
Electrogenic kinetic characteriza-
sodium/bicarbonate co- 100. Boron WF
124. Aperia A, . Ibarra
Acid-base transport LB,
F, Svensson by the renal
et al. proximal tubule.
Calcineurin mediates J Am Soc
alpha-
tion o theinmouse
transport rabbit B(0)AT1
renal corticalsodium-dependent
basolateral membrane neutral amino acidJ transporter.
vesicles. Clin Invest. Nephrol.
adrenergic 2006; 17:2368.of Na ,K( )-ATPase activity in renal tubule cells. Proc
stimulation
P ugers Arch. 2005;451:338.
1986;78:1472. http://www.ncbi.nlm.nih.gov/pubmed/16914536
Natl Acad Sci USA. 1992;89:7394.
http://www.ncbi.nlm.nih.gov/pubmed/16133263
97. Aronson PS, Soleimani M, Grassl SM. Properties of the renal Na( )- 101. Burckhardt
125. Singh H, Linas BC,SL. Cassola
Role of AC, Fromter
protein E. Electrophysiological
kinase C in beta 2-adrenoceptor analysis o
func-
77. Samarzija
HCO3- I, Fromter
cotransporter. SeminE.Nephrol.
Electrophysiological
1991;11:28. analysis o rat renal sugar and bicarbonate
tion in cultured permeation
rat proximalacross the peritubular
tubule cell membrane
epithelial cells. Am J Physiol. o 1997;273:F193.
rat kidney prox-
amino acid transport.
98. Aalkjaer C, FrischeIV Basic amino J,acids.
S, Leipziger et al.PSodium
ugers Arch.
coupled1982;393:210.
bicarbonate trans- imal
126. tubule.
Cheng II. XJ,Exclusion
Fisone G,oAizman HCO3(-)-e O, et al.ectsPKA-mediated
on other ion permeabilities
phosphorylation andando
http://www.ncbi.nlm.nih.gov/pubmed/6808460
porters in the kidney, an update. Acta Physiol Scand. 2004;181:505. coupled
inhibitionelectroneutral
of Na( )-K( HCO3(-)-transport.
)-ATPase in response Pf ugers Arch. 1984;401:43.
to beta-adrenergic hormone. Am J
78. Maunsbach
99. Samarzija I, AB, Fromter
Vorum E. Electrophysiological analysis o rat renal
H, Kwon TH, et al. Immunoelectron sugar and
microscopic http://www.ncbi.nlm.nih.gov/pubmed/6089091
Physiol. 1997;273:C893.
amino acid transport.
localization V. Acidic Na/HCO(3)
of the electrogenic amino acids.cotransporter
P ugers Arch.in 1982;393:215.
rat and ambystoma 102. Cardinal
127. Seri I, Kone J, Lapointe
BC, Gullans JY, Laprade
SR, et al.R.Locally
Luminal and peritubular
formed dopamine ionic substitu-
inhibits Na -
http://www.ncbi.nlm.nih.gov/pubmed/6124929
kidney. J Am Soc Nephrol. 2000;11:2179. tions and intracellular
K -ATPase activity in rat potential o the tubule
renal cortical rabbit cells.
proximal Am Jconvoluted tubule. Am J
Physiol. 1988;255:F666.
79. Boron
100. Tate SS, WF Y.anAcid-base
N, Uden transport
riend S. Expression
by the renal cloning
proximalo a tubule.
Na(+)-independ-
J Am Soc Physiol. 1984; 247:F352.
128. Bertorello A, Aperia A. Inhibition of proximal tubule Na( )-K( )-ATPase
ent neutral
Nephrol. 2006;amino acid transporter rom rat kidney. Proc Natl Acad Sci USA.
17:2368. 103.
activityIshibashi
requiresK, Sasaki S, Yoshiyama
simultaneous activationN.ofIntracellular
DA1 and DA2 chloride activity
receptors. Am JoPhysiol.
rabbit
1992;89:1.
101. Burckhardt BC, Cassola AC, Fromter E. Electrophysiological analysis of proximal straight tubule per used in vitro. Am J Physiol. 1988;255:F49.
1990;259:F924.
http://www.ncbi.nlm.nih.gov/pubmed/1729674
bicarbonate permeation across the peritubular cell membrane of rat kidney prox- 104. Welling PA,
129. Baumann K, O’Neil
Chan YL, RG.Bode Cell Fswelling activates
, et al. Effect basolateral membrane
of parathyroid hormone and Cl
80. tubule.
imal Wells RG, Hediger MA.
II. Exclusion Cloning o a rat on
of HCO3(-)-effects kidney
othercDNA that stimulatesand
ion permeabilities diba-
of and
cyclicK adenosine
conductances 3 ,5in-monophosphate
rabbit proximal tubule. on isotonicAm J Physiol. 1990;258:F951.
uid reabsorption: polarity
sic and neutral
coupled amino acid
electroneutral transport and hasP sequence
HCO3(-)-transport. ugers Arch. similarity to glucosidases.
1984;401:43. 105. Sasakitubular
of proximal S, Ishibashi K, Yoshiyama
cells. Kidney N, et al. KCl co-transport across the
Int. 1977;11:77.
Proc Natl
102. Acad Sci
Cardinal USA. 1992;89:5596.
J, Lapointe JY, Laprade R. Luminal and peritubular ionic substitu- basolateral
130. Gesekmembrane FA, Schoolwertho rabbitAC. renal proximalinteractions
Hormonal straight tubules.
with the J Clin Invest.
proximal
81. and
tions Berry CA, Rectorpotential
intracellular FC Jr. Mechanism
of the rabbit o proximal
proximal NaCl reabsorption
convoluted tubule.inAm theJ 1988;81:194.
Na( )-H exchanger. Am J Physiol. 1990;258:F514.
proximal1984;
Physiol. tubule o the mammalian kidney. Semin Nephrol. 1991;11:86.
247:F352. http://www.ncbi.nlm.nih.gov/pubmed/3335635
131. Weinman EJ, Steplock D, Wang Y, et al. Characterization of a protein
http://www.ncbi.nlm.nih.gov/pubmed/2034928
103. Ishibashi K, Sasaki S, Yoshiyama N. Intracellular chloride activity of rabbit 106.
cofactorAlpern
that RJ, Chambers
mediates protein M. Basolateral membraneofCl/HCO3
kinase A regulation the renalexchange in the
brush border
82. Cassola
proximal straightAC,tubule
Mollenhauer
perfusedM,inFromter
vitro. Am E. JThe intracellular
Physiol. chloride activity
1988;255:F49. rat proximalNa(
membrane convoluted tubule. Na-dependent
)-H exchanger. and -independent modes. J Gen
J Clin Invest. 1995;95:2143.
o rat kidney
104. Wellingproximal
PA, O’Neil tubular
RG. Cellcells.swelling
P ugers Arch. 1983;399:259.
activates basolateral membrane Cl Physiol.
132. Yun 1987;89:581.
CH, Oh S, Zizak M, et al. cAMP-mediated inhibition of the epithelial
http://www.ncbi.nlm.nih.gov/pubmed/6664828
and K conductances in rabbit proximal tubule. Am J Physiol. 1990;258:F951. http://www.ncbi.nlm.nih.gov/pubmed/2953859
brush border Na /H exchanger, NHE3, requires an associated regulatory
83. Sasaki
105. Alpern S,RJ,Ishibashi
Howlin KJ, K, Preisig
YoshiyamaPA. Active
N, et and passive
al. KCl components
co-transport o chlo-
across the 107.
protein.Sasaki
Proc S, NatlYoshiyama
Acad Sci USA. N. Interaction
1997;94:3010. o chloride and bicarbonate transport
ride transport
basolateral in the rat of
membrane proximal
rabbit convoluted
renal proximal tubule. J Clintubules.
straight Invest. 1985;76:1360.
J Clin Invest. across the basolateral
133. Alexander RT, membrane
Grinstein S. o rabbit
Tethering,proximal straight
recycling and tubule. Evidence
activation or
of the
84. Lucci MS, Warnock DG. E ects o anion-transport inhibitors on NaCl
1988;81:194. sodium
epithelialcoupled chloride/bicarbonate
sodium-proton exchanger, NHE3. exchange.J ExpJ Clin
Biol. Invest. 1988;81:1004.
2009;212:1630.
reabsorption
106. Alpern RJ, in the rat super
Chambers cial proximal
M. Basolateral convoluted
membrane Cl/HCO3 tubule. J Clin Invest.
exchange in the http://www.ncbi.nlm.nih.gov/pubmed/2450891
134. Weinman EJ, Dubinsky WP, Fisher K, et al. Regulation of reconstituted
1979;64:570.
rat proximal convoluted tubule. Na-dependent and -independent modes. J Gen 108.
renal NaIshibashi K, Rector by
/H exchanger FC calcium-dependent
Jr, Berry CA. Chloride protein transport
kinases.across
J Membr theBiol.
ba-
http://www.ncbi.nlm.nih.gov/pubmed/457869
Physiol. 1987;89:581. solateral
1988;103:237.membrane o rabbit proximal convoluted tubules. Am J Physiol.
85. Sasaki
107. Green S, R, YBishop
oshiyama JH,N.Giebisch
InteractionG. Ionic requirements
of chloride o proximal
and bicarbonate tubu-
transport 1990;258:F1569.
135. Waugh WH. Angiotensin II: local renal effects of physiological increments
lar sodium
across transport.membrane
the basolateral III. Selective luminal
of rabbit anionstraight
proximal substitution.
tubule.Am J Physiol.
Evidence for 109. Daugharty Can
in concentration. TM, J Ueki
PhysiolIF,Pharmacol.
Nicholas 1972;50:711.
DP, et al. Comparative renal e ects
1979;236:F268.
sodium coupled chloride/bicarbonate exchange. J Clin Invest. 1988;81:1004. o136.isoncotic
Hall JE,and colloid-
Guyton AC,reeTrippodo
volume NC, expansion in the rat.control
et al. Intrarenal Am J Physiol. 1972;
of electrolyte
86. Ishibashi
108. Baum M. K, Evidence
Rector FC thatJr,parallel
Berry Na+ -H+ and transport
CA. Chloride Cl(-)-HCO3-(OH-)
across theanti- ba- 222:225.
excretion by angiotensin II. Am J Physiol. 1977;232:F538.
porters transport
solateral membrane NaClofin rabbit
the proximal
proximal tubule. Am J Physiol.
convoluted 1987;252:F338.
tubules. Am J Physiol. http://www.ncbi.nlm.nih.gov/pubmed/5060222
137. Seely JF, Levy M. Control of extracellular uid volume. In: Brenner BM,
87. Karniski LP, Aronson PS. Chloride/ ormate exchange with ormic acid re-
1990;258:F1569. 110.
RectorDaugharty
FC Jr, eds.TM, The Ueki
Kidney. IF, Nicholas
Philadelphia,DP, etPA: al.W.
Renal response to
B. Saunders; chronic
1982: 371.intra-
cycling:
109. a mechanism
Daugharty TM, Uekio active chloride DP
IF, Nicholas transport
, et al. across epithelial
Comparative renalmembranes.
effects of venous salt loading
138. Valles P, Wysocki in the rat. J D.
J, Batlle Clin Invest. 1973;52:21.
Angiotensin II and renal tubular ion transport.
Proc Natl Acad
isoncotic and Sci USA. 1985;82:6362.
colloid-free volume expansion in the rat. Am J Physiol. 1972; http://www.ncbi.nlm.nih.gov/pubmed/4682387
Sci World J. 2005;5:680.
88. Schild L, Giebisch G, Karniski LP, et al. E ect o ormate on volume reab-
222:225. 111. BrennerJ, Giebisch
139. Geibel BM, TroyG,JL,Boron Daugharty TM, et al.IIQuantitative
WF. Angiotensin stimulates both importance
Na( )-H o
sorption in the rabbit proximal tubule. DP J Clin -
110. Daugharty TM, Ueki IF, Nicholas , et Invest.
al. Renal 1987;79:32.
response to chronic intra- changes
exchangeinand postglomerular
Na /HCO3 colloid cotransportosmoticin thepressure
rabbit in mediating
proximal glomerulotu-
tubule. Proc Natl
http://www.ncbi.nlm.nih.gov/pubmed/3793930
venous salt loading in the rat. J Clin Invest. 1973;52:21. bular balance
Acad Sci USA. in the rat. J Clin Invest. 1973;52:190.
1990;87:7917.
89. Brenner
111. Knau F, BM, YangTroyCL, Thomson
JL, Daugharty RB, etTM, al. Identi cation o a chloride-
et al. Quantitative importanceormate of http://www.ncbi.nlm.nih.gov/pubmed/4682382
140. Li L, Wang YP, Capparelli AW, et al. Effect of luminal angiotensin II on
exchanger
changes in expressed
postglomerular on thecolloid
brush osmotic
border membrane
pressure in omediating
renal proximal tubule
glomerulotu- 112.
proximalBrenner
tubule BM,uid Troy JL, Daugharty
transport: role ofTM. apicalOnphospholipase
the mechanismA2. o Aminhibition in
J Physiol.
cells. Proc
bular balanceNatlinAcad Sci USA.
the rat. J Clin2001;98:9425.
Invest. 1973;52:190. f1994;266:F202.
uid reabsorption by the renal proximal tubule o the volume-expanded rat. J
http://www.ncbi.nlm.nih.gov/pubmed/11459928
112. Brenner BM, Troy JL, Daugharty TM. On the mechanism of inhibition in Clin
141.Invest.
Liu FY 1971;50:1596.
, Cogan MG. Angiotensin II stimulates early proximal bicarbonate
90. reabsorption
uid Karniski LP, by Aronson
the renal PS. Anion
proximal exchange
tubule pathways or Cl- transportrat.
of the volume-expanded in http://www.ncbi.nlm.nih.gov/pubmed/5125390
absorption in the rat by decreasing cyclic adenosine monophosphate. J Clin
Jrabbit renal microvillus
Clin Invest. 1971;50:1596. membranes. Am J Physiol. 1987;253:F513. 113.
Invest. Imai M, Kokko JP. E ect o peritubular protein concentration on reabsorp-
1989;84:83.
91. Imai
113. AronsonM, KokkoPS. IonJP.exchangers mediatingprotein
Effect of peritubular NaCl transport
concentrationin theon renal proxi-
reabsorp- tion
142. oKatzsodium and water MD.
AI, Lindheimer in isolated per used and
Renal sodium- proximal tubules. J Clinadeno-
potassium-activated Invest.
mal tubule.
tion of sodium Cell and
Biochem
water Biophys. 2002;36:147.
in isolated perfused proximal tubules. J Clin Invest. 1972;51:314.
sine triphosphatase and sodium reabsorption in the hypothyroid rat. J Clin Invest.
http://www.ncbi.nlm.nih.gov/pubmed/12139400
1972;51:314. http://www.ncbi.nlm.nih.gov/pubmed/5009115
1973;52:796.
92. Grantham
114. Jiang Z, Grichtchenko,
JJ, Qualizza PB, II, Welling
Boron WF LW. , etInal.uence
Speciofcityserum o anion
proteinsexchange
on net 114.
143. De Grantham
Santo NG, JJ, Qualizza
Capasso G, PB,Paduano
Welling L C,WetInfal.uence
Tubular o serum
transportproteins on net
processes in
mediated by mouseofSlc26a6.
uid reabsorption isolated Jproximal
Biol Chem. 2002;277:33963.
tubules. Kidney Int. 1972;2:66. fproximal
uid reabsorption
tubules of o hypothyroid
isolated proximal tubules. Kidney studies
rats. Micropuncture Int. 1972;2:66.
on isotonic uid,
http://www.ncbi.nlm.nih.gov/pubmed/12119287
115. Baum M, Berry CA. Peritubular protein modulates neutral active NaCl http://www.ncbi.nlm.nih.gov/pubmed/4671530
amino acid and buffer reabsorption. P ugers Arch. 1980;384:117.
93. Warnock
absorption in rabbitDG,proximal
Yee VJ.convoluted
Sodium uptake tubule. mechanisms in brush-border
Am J Physiol. 1985;248:F790. 115. Baum M,G,
144. Capasso Berry
Lin CA.
JT, DePeritubular
Santo NG, protein
et al. modulates
Short termneutral active
effect of low NaCl
dosesab-of
membrane
116. Grandchampvesicles A,prepared
Boulpaeprom rabbit renal
EL. Pressure control cortex.
of sodiumBiochim Biophys Acta.
reabsorption and sorption in rabbit proximal
tri-iodothyronine on proximal convoluted tubule. Am Na-K-ATPase
tubular membrane J Physiol. 1985;248:F790.
and potassium
1982;684:137.
intercellular back ux across proximal kidney tubule. J Clin Invest. 1974;54:69. 116. Grandchamp
permeability A, Boulpaep EL.
in thyroidectomized Pressure
rats. P ugers control o sodium reabsorption and
Arch. 1985;403:90.
http://www.ncbi.nlm.nih.gov/pubmed/7055550
117. Schafer JA, Troutman SL, Watkins ML, et al. Flow dependence of uid intercellular
145. Cano A, backfBaum ux M,across
Moeproximal
OW. Thyroid kidneyhormone
tubule. Jstimulates
Clin Invest.the 1974;54:69.
renal Na/H
94. Lewis
transport in SA,
the Wills
isolatedNK.super
Apicalcialmembrane
pars recta: permeability
evidence that and kinetic
osmoticproperties
disequi- http://www.ncbi.nlm.nih.gov/pubmed/4834883
exchanger NHE3 by transcriptional activation. Am J Physiol. 1999;276:C102.
o the sodium
librium between pump in rabbit
external urinary
solutions bladder.
drives J Physiol.
isotonic uid 1983;341:169.
absorption. Kidney Int. 117. Scha erRE,
146. Lynch JA,Schneider
TroutmanEG, SL, Willis
Watkins LR,ML, et al.et Absence
al. Flow of dependence o f uid
mineralocorticoid-
http://www.ncbi.nlm.nih.gov/pubmed/6312027
1981;20:588. transport
dependentinsodium the isolated super incial
reabsorption dogpars recta:tubule.
proximal evidence Am Jthat osmotic
Physiol. disequi-
1972;223:40.
95. Haberle
118. Alpern RJ. DA,Mechanism
Shiigai TT,o Maier basolateral
G, et membrane
al. Dependency H+/OH-/HCO-3
of proximaltrans port
tubular librium
147. Garg between
LC, Narangexternal N, solutions
Wingo CS. drives isotonic f uid
Glucocorticoid absorption.
effects Kidney Int.
on Na-K-ATPase in
inuid
thetransport
rat proximal on theconvoluted tubule. A sodium-coupled
load of glomerular ltrate. Kidney Int. electrogenic
1981;20:18. process. J 1981;20:588.
rabbit nephron segments. Am J Physiol. 1985;248:F487.
Gen Physiol.
119. Du Z, 1985;86:613.
Duan Y, Yan Q, et al. Mechanosensory function of microvilli of the http://www.ncbi.nlm.nih.gov/pubmed/7343709
148. Vrtovsnik F, Jourdain M, Cherqui G, et al. Glucocorticoid inhibition of
http://www.ncbi.nlm.nih.gov/pubmed/2999293
kidney proximal tubule. Proc Natl Acad Sci USA. 2004;101:13068. 118.
Na-Pi Haberle
cotransport DA,inShiigai TT, Maiercells
renal epithelial G, etis al. Dependency
mediated o proximal
by protein kinase C. tubular
J Biol
96. Szenasi
120. Akiba T,G,Alpern Bencsath RJ, PEvelo
, Takacs J, L.
et al. Electrogenic
Proximal tubularsodium/bicarbonate
transport and urinary co- fChem.
uid transport on the load o glomerular ltrate. Kidney Int. 1981;20:18.
1994;269:8872.
transport of
excretion in sodium
rabbit renal after cortical basolateral in
renal denervation membrane vesicles. rats.
sodium depleted J ClinP Invest.
ugers http://www.ncbi.nlm.nih.gov/pubmed/7300109
149. Ali R, Amlal H, Burnham CE, et al. Glucocorticoids enhance the expression
1986;78:1472.
Arch. 1985;403:146. 119.
of the Du Z, DuanNa
basolateral Y, Yan Q, et al.
:HCO3- Mechanosensory
cotransporter in renalunction
proximal o microvilli o the
tubules. Kidney
97. Morgunov
121. Aronson PS, NS,Soleimani
You YD,M, GrasslDJ.
Hirsch SM.Response
Properties ofomouse
the renal Na(+)-HCO3-
proximal straight kidney proximal tubule. Proc Natl Acad Sci USA. 2004;101:13068.
Int. 2000;57:1063.
cotransporter.
tubule Semin Nephrol.
and medullary 1991;11:28.
thick ascending limb to beta-agonist. J Am Soc Nephrol. http://www.ncbi.nlm.nih.gov/pubmed/15319475
150. Liang M, Knox FG. Production and functional roles of nitric oxide in the
http://www.ncbi.nlm.nih.gov/pubmed/1900129
1993;4:1151. 120.
proximalSzenasi
tubule. G, AmBencsath
J Physiol P, Takacs L. Proximal
Regul Integr tubular
Comp Physiol. transport and urinary
2000;278:R1117.
98. Liu
122. Aalkjaer C, Frische
F, Nesbitt S, Leipziger
T, Drezner MK, etJ,al.etProximal
al. Sodium coupledNabicarbonate
nephron /H exchange trans- excretion
151. Garvin o sodium
JL, Herrera a ter M, renal denervation
Ortiz PA. Regulation in sodium of renaldepleted
NaCl rats. Pf ugers
transport by
porters
is in the
regulated bykidney,
alpha 1A- an update.
and alpha Acta1B-adrenergic
Physiol Scand.receptor 2004;181:505.
subtypes. Mol Phar- Arch.
nitric 1985;403:146.
oxide, endothelin, and ATP: clinical implications. Annu Rev Physiol.
http://www.ncbi.nlm.nih.gov/pubmed/15283764
macol. 1997;52:1010. http://www.ncbi.nlm.nih.gov/pubmed/3982965
2011;73:359.
99. Bello-Reuss
123. Maunsbach E. AB,Effect
Vorum of H, Kwon TH, eton
catecholamines al. Immunoelectron
uid reabsorptionmicroscopic
by the iso- 121. MorgunovA,NS,
152. Roczniak BurnsYouKD. YD,Nitric
Hirsch DJ. stimulates
oxide Response guanylate
o mouse proximal
cyclase and straight
regu-
localization
lated proximal o the electrogenic
convoluted tubule.Na/HCO(3)
Am J Physiol. cotransporter
1980;238:F347. in rat and ambystoma tubule and medullary
lates sodium transport in thick
rabbit ascending
proximal limb to beta-agonist.
tubule. Am J Physiol.J1996;270:F106.
Am Soc Nephrol.
kidney. J Am Soc Nephrol. 2000;11:2179. 1993;4:1151.
188
18 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

122. Diamond
153. Liu F, Nesbitt JM, T, Drezner
Bossert WH. MK,Standing-gradient
et al. Proximal nephron osmoticNa+/H+ ow. Aexchange
mecha- 184. Garg LC,
147. Greger R, Narang
Oberleithner N, Wingo CS. Glucocorticoid
H, Schlatter e ects on
E, et al. Chloride Na-K-ATPase
activity in cells ofin
is regulated
nism by alphaof1A-
for coupling waterandand alpha 1B-adrenergic
solute transport receptor
in epithelia. subtypes.
J GenMol Phar-
Physiol. rabbit nephron
isolated perfusedsegments.
cortical thickAm J Physiol.
ascending 1985;248:F487.
limbs of rabbit kidney. P ugers Arch.
macol. 1997;52:1010.
1967;50:2061. 148. Vrtovsnik F, Jourdain M, Cherqui G, et al. Glucocorticoid inhibition o
1983;399:29.
123. Jacobson
154. Bello-Reuss HR,E.Kokko E ectJP o, Seldin
catecholamines
DW, et al. on Lackf uid reabsorption
of solvent drag ofby the and
NaCl iso- Na-Pi Greger
185. cotransport in renal epithelial
R, Schlatter E. Properties cells of is mediated
the lumenbymembrane
protein kinase of theC. Jcor-
Biol
lated proximal
NaHCO3 in rabbit convoluted
proximaltubule. tubules. AmAm J Physiol.
J Physiol. 1980;238:F347.
1982;243:F342. Chem.thick
tical 1994;269:8872.
ascending limb of Henle’s loop of rabbit kidney. P ugers Arch.
124. Gonzalez
155. Aperia A, E, Ibarra F, SvenssonP,LB,
Carpi-Medina et al. Calcineurin
Whittembury G. Cellmediates
osmoticalpha-adrener-
water perme- http://www.ncbi.nlm.nih.gov/pubmed/8132623
1983;396:315.
gic stimulation
ability of isolated o rabbit
Na+ ,K(+)-ATPase
proximal straight activity in renal
tubules. Amtubule
J Physiol.cells. Proc
1982;242:F321. 149. Molony
186. Ali R, Amlal H, Burnham
DA, Reeves CE, et al.
WB, Andreoli TE. Na :K :2Cl- enhance
Glucocorticoids cotransport the and
expres-
the
Natl Acad
156. VallonSci V USA. 1992;89:7394.
, Verkman AS, Schnermann J. Luminal hypotonicity in proxi- sion oascending
thick the basolateral Na+ :HCO3-
limb. Kidney cotransporter in renal proximal tubules. Kid-
Int. 1989;36:418.
125. tubules
mal Singh H,ofLinas SL. Role o protein kinase
aquaporin-1-knockout mice. CAm in beta 2-adrenoceptor
J Physiol Renal Physiol. unc- ney Int.Hebert
187. 2000;57:1063.
SC, Reeves WB, Molony DA, et al. The medullary thick limb: func-
tion in cultured rat proximal tubule epithelial cells. Am J Physiol. 1997;273:F193.
2000;278:F1030. 150.and
tion Liang M, KnoxofFG.
modulation theProduction
single-effectand unctional
multiplier. rolesInt.
Kidney o nitric oxide in the
1987;31:580.
126. Barfuss
157. Cheng XJ, DW,FisoneSchafer G, JA.
AizmanRate O, of et al. PKA-mediated
formation and composition phosphorylation
of absorbate and proximal
188. Good tubule. Am J Physiol absorption
DW. Bicarbonate Regul Integr by Comp the Physiol. 2000;278:R1117.
thick ascending limb of Henle’s
inhibition
from proximal o Na(+)-K(+)-ATPase
nephron segments.inAm response
J Physiol.to1984;247:F117.
beta-adrenergic hormone. Am J 151. Semin
loop. GarvinNephrol.
JL, Herrera M, Ortiz PA. Regulation o renal NaCl transport by nitric
1990;10:132.
Physiol.
158. 1997;273:C893.
Dantzler WH. Studies on nonmammalian nephrons. Kidney Int. 1982; oxide,Greger
189. endothelin, and ATP:
R. Chloride clinical implications.
reabsorption in the rabbit Annu Rev Physiol.
cortical 2011;73:359.
thick ascending limb
127. Seri I, Kone BC, Gullans SR, et al. Locally ormed dopamine inhibits Na+
22:560. http://www.ncbi.nlm.nih.gov/pubmed/20936940
of the loop of Henle. A sodium dependent process. P ugers Arch. 1981;390:38.
-K+ -ATPase
159. Bennettactivity in rat renal
CM, Brenner BM,cortical
Berliner tubule
RW. cells. Am J Physiol.
Micropuncture 1988;255:F666.
study of nephron 152. Koenig
190. Roczniak B, A, Burns KD.
Ricapito NitricR.oxide
S, Kinne Chloride stimulates guanylate
transport in thecyclase and regu-
thick ascending
128. Bertorello
function in the rhesus A, Aperiamonkey. A. Inhibition
J Clin Invest. o proximal
1968;47:203. tubule Na(+)-K(+)-ATPase lates sodium
limb of Henle’ transport
s loop: in rabbit proximal
potassium dependence tubule.and Amstoichiometry
J Physiol. 1996;270:F106.
of the NaCl
activity
160. requires
Kokko simultaneous
JP. Sodium chloride activation
and water o DA1 and DA2
transport in thereceptors.
descendingAm Jlimb
Physiol.of 153. Diamond
cotransport systemJM,inBossert
plasmaWH. Standing-gradient
membrane vesicles. P ugers osmotic f ow.
Arch. A mechanism
1983;399:173.
1990;259:F924.
Henle. J Clin Invest. 1970;49:1838. or coupling
191. ReevesoWB, waterMolonyand solute DA.transport in epithelia.
The physiology J Gen diuretic
of loop Physiol. 1967;50:2061.
action. Semin
129. Kokko
161. Baumann JP, K, ChanFC
Rector YL,Jr.Bode F, et al. E ect
Countercurrent o parathyroid
multiplication hormone
system withoutand http://www.ncbi.nlm.nih.gov/pubmed/6066064
Nephrol. 1988;8:225.
cyclic transport
active adenosinein3 inner ‘ ,5’-monophosphate
medulla. Kidney on Int.isotonic
1972;2:214. f uid reabsorption: polarity 154. Eveloff
192. Jacobson J, HR,
Kinne Kokko JP, Seldin DW, ettransport
R. Sodium-chloride al. Lack oin solvent drag o NaCl
the medullary thick and
as-
o proximal
162. Nielsentubular
S, Pallone cells.T,Kidney
Smith Int.BL, 1977;11:77.
et al. Aquaporin-1 water channels in short NaHCO3limb
cending in rabbit
of Henle’proximal
s loop:tubules.
evidenceAm forJaPhysiol. 1982;243:F342.
sodium-chloride cotransport system
http://www.ncbi.nlm.nih.gov/pubmed/191680
and long loop descending thin limbs and in descending vasa recta in rat kidney. 155.
in Gonzalez
plasma membrane E, Carpi-Medina
vesicles. J Membr P, Whittembury
Biol. 1983;72:173. G. Cell osmotic water perme-
130.J Physiol.
Am Gesek 1995;268:F1023.
FA, Schoolwerth AC. Hormonal interactions with the proximal abilityGamba
193. o isolated rabbit proximal
G, Miyanoshita straight tubules.
A, Lombardi M, et al.Am J Physiol.cloning,
Molecular 1982;242:F321.
primary
Na(+)-H+
163. Zhai exchanger.
XY, FentonAm RA,J Physiol.
Andreasen 1990;258:F514.
A, et al. Aquaporin-1 is not expressed in 156. Vallon
structure, andV,characterization
Verkman AS, Schnermann of two members J. Luminal hypotonicityelectroneu-
of the mammalian in proxi-
131. Weinman
descending thin limbsEJ, Steplock D, Wang
of short-loop Y, et al. JCharacterization
nephrons. Am Soc Nephrol. o2007;18:2937.
a protein co- malsodium-(potassium)-chloride
tral tubules o aquaporin-1-knockout cotransporter mice.family Am expressed
J Physiol inRenal
kidney.Physiol.
J Biol
actor Imai
164. that mediates
M, Kokkoprotein JP. Sodium kinase A regulation
chloride, urea, oandthewater renaltransport
brush borderin themem-thin 2000;278:F1030.
Chem. 1994;269:17713.
brane Na(+)-H+
ascending limb ofexchanger.
Henle. Generation J Clin Invest. 1995;95:2143.
of osmotic gradients by passive diffusion of 157. Payne
194. Bar ussJA,DW, Xu SchaJC, Haaser JA.M,Rate o Primary
et al. ormation and composition
structure, functionaloexpression,
absorbate
http://www.ncbi.nlm.nih.gov/pubmed/7738182
solutes. J Clin Invest. 1974;53:393. rom chromosomal
and proximal nephron segments.
localization of Am
theJ bumetanide-sensitive
Physiol. 1984;247:F117. Na-K-Cl cotrans-
132. Imai
165. Yun CH, Oh S, Zizak
M. Functional M, et al. cAMP-mediated
heterogeneity of the descending inhibition
limbs of o Henle’
the epithelial
s loop. 158. Dantzler
porter in humanWH. colon. Studies on nonmammalian
J Biol Chem. 1995;270:17977. nephrons. Kidney Int. 1982;
brush
II. border Na+/H+
Interspecies differencesexchanger,
amongNHE3, rabbits, requires
rats, andan associated
hamsters.regulatory
P ugers Arch. pro- 22:560.
195. Yang T, Huang YG, Singh I, et al. Localization of bumetanide- and
tein. Proc Natl Acad Sci USA. 1997;94:3010.
1984;402:393. 159. Bennett CM,Na-K-Cl
thiazide-sensitive Brennercotransporters
BM, Berliner RW. along Micropuncture
the rat nephron. study Amo Jnephron
Physiol.
133. Jamison
166. Alexander RL.RT, Grinstein S. Tethering,
Micropuncture study of segmentsrecycling ofand
thin activation
loop of Henle o the in epi-
the unction in the rhesus monkey. J Clin Invest. 1968;47:203.
1996;271:F931.
thelial
rat. Amsodium-proton
J Physiol. 1968;215:236. exchanger, NHE3. J Exp Biol. 2009;212:1630. http://www.ncbi.nlm.nih.gov/pubmed/16695942
196. Kaplan MR, Plotkin MD, Lee WS, et al. Apical localization of the Na-K-Cl
http://www.ncbi.nlm.nih.gov/pubmed/19448073
167. Imai M, Kokko JP. Mechanism of sodium and chloride transport in the thin 160. Kokko JP
cotransporter, . Sodium
rBSC1, on chloride
rat thick and water transport
ascending limbs. Kidney in the descending
Int. 1996;49:40. limb o
134. Weinman
ascending limb ofEJ, Dubinsky
Henle. WP, Fisher
J Clin Invest. K, et al. Regulation o reconstituted
1976;58:1054. Henle.Simon
197. J Clin DB,
Invest. 1970;49:1838.
Karet FE, Hamdan JM, et al. Bartter’s syndrome, hypokalaemic
renal Y Na+/H+ -
168. oshitomiexchangerK, KondobyY,calcium-dependent
Imai M. Evidence protein kinases. Cl
for conductive J Membr
pathwaysBiol. http://www.ncbi.nlm.nih.gov/pubmed/5460286
alkalosis with hypercalciuria, is caused by mutations in the Na-K-2Cl cotrans-
1988;103:237.
across the cell membranes of the thin ascending limb of Henle’s loop. J Clin 161. Kokko
porter NKCC2. JP,Nat
Rector FC 1996;13:183.
Genet. Jr. Countercurrent multiplication system without ac-
http://www.ncbi.nlm.nih.gov/pubmed/2846848
Invest. 1988;82:866. tive transport
198. MonetteinMY inner medulla.
, Rinehart J, Kidney
Lifton RP Int., et
1972;2:214.
al. Rare mutations in the human
135. Kondo
169. Waugh Y WH. Angiotensin
, Yoshitomi K, Imai II: local renal eof ects
M. Effect Ca2o+ on Cl- transport
physiological increments
in thin http://www.ncbi.nlm.nih.gov/pubmed/4657922
Na-K-Cl cotransporter (NKCC2) associated with lower blood pressure ex-
in concentration.
ascending limb ofCan Henle’ J Physiol
s loop.Pharmacol.
Am J Physiol. 1972;50:711.
1988;254:F232. 162. impaired
hibit Nielsen S,processing
Pallone T, and Smithtransport
BL, et al.function.
Aquaporin-1 Am water
J Physiolchannels
Renal in short
Physiol.
http://www.ncbi.nlm.nih.gov/pubmed/4340507
170. Uchida S, Sasaki S, Furukawa T, et al. Molecular cloning of a chloride and long loop descending thin limbs and in descending vasa recta in rat kidney.
2011;300:F840.
136. Hall
channel thatJE, Guyton AC,
is regulated Trippodo NC,
by dehydration andet expressed
al. Intrarenal control o electrolyte
predominantly in kidney Am J Physiol.
199. 1995;268:F1023.
Krishnamurthy H, Piscitelli CL, Gouaux E. Unlocking the molecular
excretionJby
medulla. Biolangiotensin II. Am J Physiol. 1977;232:F538.
Chem. 1993;268:3821. 163. Zhai
secrets XY, Fenton RA,transporters.
of sodium-coupled Andreasen A,Nature. et al. Aquaporin-1
2009;459:347. is not expressed in
137. Uchida
171. Seely JFS, , Levy
SasakiM.S,Control
Nitta K,oet extracellular
al. Localization f uid
andvolume.
functional In: characteriza-
Brenner BM, descending
200. Ares GR, thinCaceres
limbs oPS,short-loop
Ortiz PA. nephrons. J Am Soc Nephrol.
Molecular regulation of NKCC2 2007;18:2937.
in the thick
Rector
tion FC kidney-speci
of rat Jr, eds. The Kidney. Philadelphia,
c chloride channel,PA: W. B. JSaunders;
ClC-K1. Clin Invest. 1982: 371.
1995;95:104. http://www.ncbi.nlm.nih.gov/pubmed/17942963
ascending limb. Am J Physiol Renal Physiol. 2011;301:F1143.
138. Estevez
Valles P,R,Wysocki -
172. BoettgerJ,T,Batlle SteinD.V, Angiotensin
et al. BarttinIIisand a Clrenal tubular
channel ion trans-
beta-subunit 164. Benziane
201. Imai M, Kokko B, DemaretzJP. Sodium chloride, urea,
S, Defontaine N, et and water transport
al. NKCC2 in the thin
surface expression
port. Scifor World -
J. 2005;5:680.
crucial renal Cl reabsorption and inner ear K secretion. Nature. 2001; ascending
in mammalian limbcells:
o Henle. Generation obyosmotic
down-regulation gradients by
novel interaction passive
with di usion
aldolase o
B. J Biol
http://www.ncbi.nlm.nih.gov/pubmed/16142301
414:558. solutes.
Chem. J Clin Invest. 1974;53:393.
2007;282:33817.
139. Waldegger
173. Geibel J, GiebischS, Jeck N, G,Barth
BoronP, WF . Angiotensin
et al. Barttin increases II stimulates both Na(+)-H+
surface expression and http://www.ncbi.nlm.nih.gov/pubmed/11344552
202. Zaarour N, Defontaine N, Demaretz S, et al. Secretory carrier membrane
exchangecurrent
changes and Na+/HCO3
properties of -
cotransport
ClC-K channels. in the rabbit
P ugersproximal tubule. Proc Natl
Arch. 2002;444:411. 165. Imai
protein M. Functional
2 regulates exocytic heterogeneity
insertion ofoNKCC2 the descending limbs
into the cell o Henle’s Jloop.
membrane. Biol
Acad Sci
174. USA.U,1990;87:7917.
Scholl Hebeisen S, Janssen AG, et al. Barttin modulates traf cking and II. Interspecies
Chem. 2011;286:9489.di erences among rabbits, rats, and hamsters. Pf ugers Arch.
http://www.ncbi.nlm.nih.gov/pubmed/2172967
function of ClC-K channels. Proc Natl Acad Sci USA. 2006;103:11411. 1984;402:393.
203. Parvin MN, Turner RJ. Identi cation of key residues involved in the dimer-
140. Fischer
175. Li L, Wang YP, Capparelli
M, Janssen AG, Fahlke AW,C.etBarttin
al. E activates
ect o luminalClC-K angiotensin
channel function II on http://www.ncbi.nlm.nih.gov/pubmed/6522246
ization of the secretory Na( )-K( )-2Cl(-) cotransporter NKCC1. Biochemistry.
proximal
by modulatingtubule f uid Jtransport:
gating. Am Soc Nephrol. role o 2010;21:1281.
apical phospholipase A2. Am J Physiol. 166. Jamison RL. Micropuncture study o segments o thin loop o Henle in the
2011;50:9857.
1994;266:F202.
176. Matsumura Y, Uchida S, Kondo Y, et al. Overt nephrogenic diabetes insipi- rat. Am
204. J Physiol.
Plata C, Mount 1968;215:236.
DB, Rubio V, et al. Isoforms of the Na-K-2Cl cotransporter
141.in Liu
dus miceFY , Coganthe
lacking MG.CLC-K1Angiotensin
chlorideII channel.
stimulates Natearly
Genet. proximal bicarbonate
1999;21:95. http://www.ncbi.nlm.nih.gov/pubmed/5659337
in murine TAL II. Functional characterization and activation by cAMP. Am J
absorption
177. RochainAS, theKokko
rat byJP decreasing cyclic adenosine
. Sodium chloride and water monophosphate.
transport in theJ medul- Clin In- 167. Imai
Physiol. M, Kokko JP. Mechanism o sodium and chloride transport in the thin
1999;276:F359.
vest.thick
lary 1989;84:83.
ascending limb of Henle. Evidence for active chloride transport. J Clin ascending
205. Carota limb o Henle.
I, Theilig J Clin Invest. M,
F, Oppermann 1976;58:1054.
et al. Localization and functional char-
http://www.ncbi.nlm.nih.gov/pubmed/2544631
Invest. 1973;52:612. http://www.ncbi.nlm.nih.gov/pubmed/993330
acterization of the human NKCC2 isoforms. Acta Physiol (Oxf). 2010;199:327.
142. Burg
178. Katz MB,AI, Lindheimer
Green N. Function MD. Renal of thesodium-
thick and potassium-activated
ascending limb of Henle’sadeno- loop. 168. Castrop
206. Yoshitomi H,K,Schnermann
Kondo Y, Imai M. Evidence
J. Isoforms of renalor conductive Cl- pathways
Na-K-2Cl cotransporter
sineJtriphosphatase
Am Physiol. 1973;224:659. and sodium reabsorption in the hypothyroid rat. J Clin Invest. across theexpression
NKCC2: cell membranes and ofunctional
the thin ascending
signi cance. limbAm o Henle’ s loop.
J Physiol J ClinPhysiol.
Renal Invest.
1973;52:796.
179. Katz AI, Doucet A, Morel F. Na-K-ATPase activity along the rabbit, rat, and 1988;82:866.
2008;295:F859.
143. De
mouse Santo NG,
nephron. Am JCapasso
Physiol. G, Paduano C, et al. Tubular transport processes in
1979;237:F114. http://www.ncbi.nlm.nih.gov/pubmed/2458388
207. Gimenez I, Isenring P, Forbush B. Spatially distributed alternative splice
proximal
180. HeberttubulesSC. oHypertonic
hypothyroid cellrats.
volumeMicropuncture
regulation studiesin mouse on isotonic f uid,
thick limbs. 169. Kondo
variants of theYrenal
, Yoshitomi
Na-K-Cl K,cotransporter
Imai M. E ect o Ca2+
exhibit on Cl” transport
dramatically differentinaf thin
ni-
I.amino
ADHacid and bu er
dependency andreabsorption. Pf ugers Arch.
nephron heterogeneity. Am 1980;384:117.
J Physiol. 1986;250:C907. ascending
ties for thelimb o Henle’
transported s loop.
ions. AmChem.
J Biol J Physiol. 1988;254:F232.
2002;277:8767.
http://www.ncbi.nlm.nih.gov/pubmed/6446079
181. Oberleithner H, Guggino W, Giebisch G. Mechanism of distal tubular chlo- 170. Plata
208. Uchida C, S, SasakiP, S,
Meade Furukawa
Vazquez N, etT, al.
et Functional
al. Molecular cloning of
properties o athechloride
apical
144.transport
ride CapassoinG,Amphiuma Lin JT, Dekidney. Santo NG, Am JetPhysiol.
al. Short term e ect o low doses o
1982;242:F331. channel
Na -K that-2Cl-is regulated
cotransporter by dehydration
isoforms. J Bioland Chem.expressed predominantly in kidney
2002;277:11004.
tri-iodothyronine
182. Hebert SC, Andreoli on proximal tubularofmembrane
TE. Effects antidiuretic Na-K-ATPase
hormone onand potassium
cellular con- medulla.
209. J BiolR,Chem.
Greger Weidtke 1993;268:3821.
C, Schlatter E, et al. Potassium activity in cells of iso-
permeability
ductive pathways in thyroidectomized
in mouse medullary rats. Pf ugersascending
thick Arch. 1985;403:90.
limbs of Henle: II. de- http://www.ncbi.nlm.nih.gov/pubmed/7680033
lated perfused cortical thick ascending limbs of rabbit kidney. P ugers Arch.
http://www.ncbi.nlm.nih.gov/pubmed/2984639
terminants of the ADH-mediated increases in transepithelial voltage and in net 171. Uchida S, Sasaki S, Nitta K, et al. Localization and unctional characteriza-
1984;401:52.
145. Cano A, JBaum
Cl-absorption. MembrM,Biol. Moe OW. Thyroid hormone stimulates the renal Na/H
1984;80:221. tion oGuggino
210. rat kidney-speci
SE, Guggino c chloride
WB, Green channel,
N, etClC-K1. J Clin Invest.K1995;95:104.
al. Ca2+-activated channels in
exchanger
183. NHE3
Schlatter E, by transcriptional
Greger R. cAMP activation.
increases the Am Jbasolateral Cl- conductance
Physiol. 1999;276:C102. http://www.ncbi.nlm.nih.gov/pubmed/7814604
cultured medullary thick ascending limb cells. Am J Physiol. 1987;252:C121.
146.
in the Lynch
isolated RE,perfused
Schneider EG, Willis
medullary LR, ascending
thick et al. Absence limbo of mineralocorticoid-de-
Henle’s loop of the 172. Wang
211. Estevez WH.R, Boettger
Two types T, Stein
of K V,channel
et al. Barttin is a ascending
in thick Cl- channellimb beta-subunit cru-
of rat kidney.
pendentPsodium
mouse. ugers Arch. reabsorption
1985;405:367.in dog proximal tubule. AmJ Physiol. 1972;223:40. cial Jor
Am renal Cl”
Physiol. reabsorption and inner ear K+ secretion. Nature. 2001; 414:558.
1994;267:F599.
http://www.ncbi.nlm.nih.gov/pubmed/5039071 http://www.ncbi.nlm.nih.gov/pubmed/11734858
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1189
89

173. Wang
212. Waldegger WH,S,White Jeck N,S,Barth GeibelP, etJ,al.etBarttin
al. A increases
potassiumsurchannel ace expression
in the api- and 197.
240. Y Simon
ang SS, DB,LoKaret
YF, Wu FE, Hamdan
CC, et al.JM, et al. Bartter’s syndrome,
SPAK-knockout mice manifest hypokalaemic
Gitelman
changes
cal currentofproperties
membrane rabbit thick o ClC-K channels.
ascending limbPfof ugers Arch.
Henle’ 2002;444:411.
s loop. Am J Physiol. alkalosis
syndromewith and hypercalciuria, is caused byJ Am
impaired vasoconstriction. mutations
Soc Nephrol. in the2010;21:1868.
Na-K-2Cl cotrans-
http://www.ncbi.nlm.nih.gov/pubmed/12111250
1990;258:F244. porter
241. Lin NKCC2.
SH, YuNat IS,Genet.
Jiang 1996;13:183.
ST, et al. Impaired phosphorylation of Na( )-K( )-
174. Giebisch
213. Scholl U,G, Hebeisen
Hebert SC, S, Janssen
Wang WH AG, et Newal. aspects
Barttin modulates tra f cking
of renal potassium and
trans- http://www.ncbi.nlm.nih.gov/pubmed/8640224
2Cl(-) cotransporter by oxidative stress-responsive kinase-1 de ciency manifests
unction
port. o ClC-K
P ugers Arch.channels.
2003;446:289. Proc Natl Acad Sci USA. 2006;103:11411. 198. Monette
hypotension andMY , Rinehartsyndrome.
Bartter-like J, Li ton RP , etNatl
Proc al. Acad
Rare Sci mutations in the human
USA. 2011;108:17538.
http://www.ncbi.nlm.nih.gov/pubmed/16849430
214. Lu M, Wang T, Yan Q, et al. ROMK is required for expression of the 70-pS K Na-K-Cl
242. Ra cotransporter
qi FH, Zuber AM, (NKCC2)Gloverassociated
M, et al. Role withoflower blood pressureSPAK
the WNK-activated ex-
175. Fischer
channel in theM, thickJanssen AG, Fahlke
ascending limb. Am C. Barttin
J Physiol activates ClC-K channel
Renal Physiol. unction
2004;286:F490. hibit
kinaseimpaired processing
in regulating and transport
blood pressure. EMBO unction.
Mol Med. Am J Physiol Renal Physiol.
2010;2:63.
by modulating
215. Ho K, Nichols gating.CG, J AmLederer
Soc Nephrol.
WJ, et2010;21:1281.
al. Cloning and expression of an in- 2011;300:F840.
243. Hebert SC, Culpepper RM, Andreoli TE. NaCl transport in mouse medul-
176. Matsumura
wardly Y, Uchida S, Kondo
rectifying ATP-regulated potassiumY, et al. Overt nephrogenic
channel. diabetes insipi-
Nature. 1993;362:31. 199. Krishnamurthy
lary thick ascending limbs. H, Piscitelli
II. ADHCL, Gouaux E. of
enhancement Unlocking
transcellular the NaCl
molecular
cotrans-se-
dus inLu
216. mice M, lacking
Wang T,theYan CLC-K1 chloride
Q, et al. Absence channel.
of small Natconductance
Genet. 1999;21:95. K channel crets
port; oorigin
sodium-coupled
of transepithelial transporters.
voltage. Am Nature. 2009;459:347.
J Physiol. 1981;241:F432.
http://www.ncbi.nlm.nih.gov/pubmed/9916798
(SK) activity in apical membranes of thick ascending limb and cortical collecting http://www.ncbi.nlm.nih.gov/pubmed/19458710
244. Hebert SC, Friedman PA, Andreoli TE. Effects of antidiuretic hormone
177. inRocha
duct ROMK AS, Kokkos) JP
(Bartter’ . Sodiummice.
knockout chlorideJ Bioland water
Chem. transport in the medul-
2002;277:37881. 200. Ares GR,
on cellular Caceres pathways
conductive PS, Ortiz in PA.mouse
Molecularmedullaryregulation thicko ascending
NKCC2 in the
limbs
lary thick
217. Simon ascending
DB, Karet limbFE,o Rodriguez-Soriano
Henle. Evidence orJ, active chlorideheterogeneity
et al. Genetic transport. J Clin of thick ascending
of Henle: I. ADH limb. Am J Physiol
increases Renal Physiol.
transcellular conductance2011;301:F1143.
pathways. J Membr Biol.
Invest. 1973;52:612.
Bartter’ s syndrome revealed by mutations in the K channel, ROMK. Nat Genet. 201. Benziane B, Demaretz S, De ontaine N, et al. NKCC2 sur ace expression
1984;80:201.
http://www.ncbi.nlm.nih.gov/pubmed/4685086
1996;14:152. in
245.mammalian
Ecelbarger cells:
CA,down-regulation
Kim GH, Knepper byMA,
novel interaction
et al. Regulation with aldolase B.chan-
of potassium J Biol
178. Simon
218. Burg MB, DB,Green
LiftonN. RP.Function o the thick
Ion transporter mutationsascending limb o s Henle’
in Gitelman’ s loop.s
and Bartter’ Chem.
nel Kir2007;282:33817.
1.1 (ROMK) abundance in the thick ascending limb of Henle’s loop. J Am
Am J Physiol.Curr
syndromes. 1973;224:659.
Opin Nephrol Hypertens. 1998;7:43. http://www.ncbi.nlm.nih.gov/pubmed/17848580
Soc Nephrol. 2001;12:10.
http://www.ncbi.nlm.nih.gov/pubmed/4691283
219. Schwalbe RA, Bianchi L, Accili EA, et al. Functional consequences of 202. ZaarourWB,
246. Reeves N, De ontaine TE.
Andreoli N, Demaretz
Cl- transport S, et in al. basolateral
Secretory carrier renal membrane
medullary
179. Katz
ROMK AI, Doucet
mutants linked A, to Morel
antenatalF. Na-K-ATPase
Bartter’s syndrome activityandalong the rabbit,for
implications rat,treat-
and protein
vesicles:2II.regulates
Cl- channelsexocytic insertion
in planar lipido bilayers.
NKCC2J into Membr theBiol.
cell 1990;113:57.
membrane. J Biol
mouseHum
ment. nephron. Am J Physiol.
Mol Genet. 1998;7:975.1979;237:F114. Chem. 2011;286:9489.
247. Culpepper RM, Andreoli TE. PGE2, forskolin, and cholera toxin in-
180. Wangemann
220. Hebert SC. HypertonicP, Wittner M,cell Di volume
Stefano A, regulation in mouse blockers
et al. Cl(-)-channel thick limbs. in the I. http://www.ncbi.nlm.nih.gov/pubmed/21205824
teractions in modulating NaCl transport in mouse mTALH. Am J Physiol.
ADH ascending
thick dependency limbandofnephron
the loop heterogeneity.
of Henle. Structure Am J activity
Physiol. relationship.
1986;250:C907. P ugers 203. Parvin MN, Turner RJ. Identif cation o key residues involved in the dimer-
1984;247:F784.
181. 1986;407(Suppl
Arch. Oberleithner H,2):S128. Guggino W, Giebisch G. Mechanism o distal tubular ization o the secretory
248. Escalante B, Erlij D, Na(+)-K(+)-2Cl(-)
Falck JR, et al. cotransporter
Effect of cytochrome NKCC1.P450 Biochemistry
arachi-
chloride
221. transportM,
Yoshikawa in Uchida
Amphiuma S, Ykidney.
amauchiAm A,Jet Physiol. 1982;242:F331.
al. Localization of rat CLC-K2 2011;50:9857.
donate metabolites on ion transport in rabbit kidney loop of Henle. Science.
182. Hebert
chloride channelSC,mRNAAndreoli TE.kidney.
in the E ects Am o antidiuretic hormone on cellular con-
J Physiol. 1999;276:F552. http://www.ncbi.nlm.nih.gov/pubmed/21970294
1991;251:799.
ductive
222. pathwaysK,inUchida
Kobayashi mouseS,medullary
Mizutani thick S, et al.ascending
Intrarenal limbs
and ocellular
Henle: II. de-
localiza- 204.
249. Lu PlataM,C,ZhuMount
Y, BalazyDB, M,Rubioet al.V,Effect
et al. of
Isoangiotensin
orms o theIINa-K-2Cl cotransporter
on the apical K chan-
terminants
tion of CLC-K2 o the ADH-mediated
protein in the mouse increases
kidney.inJ Am transepithelial
Soc Nephrol.voltage and in net
2001;12:1327. in
nelmurine TAL II.
in the thick Functional
ascending limb characterization
of the rat kidney. andJactivation
Gen Physiol. by 1996;108:537.
cAMP. Am J Phys-
Cl-absorption.
223. Kramer BK, J MembrBergler Biol.T,1984;80:221.
Stoelcker B, et al. Mechanisms of disease: the iol.
250. 1999;276:F359.
Harris RC, McKanna JA, Akai Y, et al. Cyclooxygenase-2 is associated with
http://www.ncbi.nlm.nih.gov/pubmed/6094817
kidney-speci c chloride channels ClCKA and ClCKB, the Barttin subunit, and 205. Carotadensa
the macula I, Theilig
of ratF,kidney
Oppermann M, et al. with
and increases Localization and unctional
salt restriction. char-
J Clin Invest.
183. clinical
their Schlatter E, Greger
relevance. NatR. cAMP
Clin Pract increases the basolateral Cl- conductance
Nephrol. 2008;4:38. acterization
1994;94:2504. o the human NKCC2 iso orms. Acta Physiol (Ox ). 2010;199:327.
in theBirkenhager
224. isolated per R, used Otto medullary
E, Schurmannthick ascending
MJ, et al. limb o Henle’
Mutation s loop causes
of BSND o the http://www.ncbi.nlm.nih.gov/pubmed/20146722
251. Cheng HF, Harris RC. Cyclooxygenases, the kidney, and hypertension.
mouse. syndrome
Bartter Pf ugers Arch. with 1985;405:367.
sensorineural deafness and kidney failure. Nat Genet. 206. Castrop2004;43:525.
Hypertension. H, Schnermann J. Iso orms o renal Na-K-2Cl cotransporter
http://www.ncbi.nlm.nih.gov/pubmed/3001635
2001;29:310. NKCC2:
252. Harding expression
P, Sigmon and unctional
DH, Al e signif ME, cance. Am J Physiol Renalmediates
et al. Cyclooxygenase-2 Physiol.
184. Di
225. Greger
StefanoR, Oberleithner
A, Greger R, Des H, Schlatter
eurs E, et E, al.
et A al.Ba(2
Chloride activity inK cells
)-insensitive con-o 2008;295:F859.
increased renal renin content induced by low-sodium diet. Hypertension.
isolated per
ductance in used cortical thick
the basolateral ascending
membrane of limbs
rabbit ocortical
rabbit thick
kidney. Pf ugers Arch.
ascending limb 207. Gimenez I, Isenring P, Forbush B. Spatially distributed alternative splice
1997;29:297.
1983;399:29.
cells. Cell Physiol Biochem. 1998;8:89. variants
253. Molony o the DA,
renalAndreoli
Na-K-Cl TE. cotransporter
Diluting power exhibit of dramatically
thick limbs di oferent a f ni-
Henle. I.
http://www.ncbi.nlm.nih.gov/pubmed/6646997 -
226. Di Stefano A, Jounier S, Wittner M. Evidence supporting a role for KCl co- ties or the transported
Peritubular hypertonicity ions. J Biol basolateral
blocks Chem. 2002;277:8767.
Cl channels. Am J Physiol. 1988;
185. Greger
transporter in R,
theSchlatter
thick ascendingE. Properties
limb ofoHenle’the lumen membrane
s loop. Kidney o the cortical
Int. 2001;60:1809. http://www.ncbi.nlm.nih.gov/pubmed/11815599
255:F1128.
thick ascending
227. Velazquezlimb H, Silvao Henle’ s loop oand
T. Cloning rabbit kidney. Pfofugers
localization KCC4 Arch.
in 1983;396:315.
rabbit kidney: 208. Plata C,SC,
254. Hebert Meade P, Vazquez
Culpepper RM,N, et al. Functional
Andreoli TE. NaClproperties
transport in o mouse
the apical Na+
medul-
http://www.ncbi.nlm.nih.gov/pubmed/6844136
expression in distal convoluted tubule. Am J Physiol Renal Physiol. 2003;285:F49. -K+
lary -2Cl-
thick cotransporter
ascending limbs. iso orms. J Biol Chem. of
III. Modulation 2002;277:11004.
the ADH effect by peritubular
186. Boettger
228. Molony DA, ReevesCA,
T, Hubner WB,Maier
Andreoli
H, et TE. Na+ :K+and
al. Deafness :2Clrenal
-
cotransport and the
tubular acidosis http://www.ncbi.nlm.nih.gov/pubmed/11790783
osmolality. Am J Physiol. 1981;241:F443.
thick
in ascending
mice lacking the limb. K-ClKidney Int. 1989;36:418.
co-transporter Kcc4. Nature. 2002;416:874. 209. Greger R,A.Weidtke
255. Khanna Acquired C, Schlatter
nephrogenic E, et diabetes
al. Potassium activitySemin
insipidus. in cellsNephrol.
o iso-
187. Friedman
229. Hebert SC,PA, Reeves
AndreoliWB,TE. Molony DA, et al. The
CO2-stimulated NaClmedullary
absorption thickin limb:
the mouseunc- lated per used cortical thick ascending limbs o rabbit kidney. Pf ugers Arch.
2006;26:244.
tion and
renal modulation
cortical o the single-e
thick ascending limbect of multiplier.
Henle. Evidence Kidney Int.for 1987;31:580.
synchronous Na 1984;401:52.
256. Takaichi K, Kurokawa K. High Ca2+ inhibits peptide hormone-dependent
http://www.ncbi.nlm.nih.gov/pubmed/3550227
/H and Cl-/HCO3- exchange in apical plasma membranes. J Gen Physiol. http://www.ncbi.nlm.nih.gov/pubmed/6089092
cAMP production speci cally in thick ascending limbs of Henle. Miner Electrolyte
188. Good DW. Bicarbonate absorption by the thick ascending limb o Henle’s
1982;80:683. 210.
Metab.Guggino
1986;12:342.SE, Guggino WB, Green N, et al. Ca2+-activated K+ channels in
loop. Semin
230. KikeriNephrol.
D, Azar 1990;10:132.
S, Sun A, et al. Na( )-H antiporter and Na( )-(HCO3-) cultured medullary
257. Takaichi thick ascending
K, Kurokawa limb cells. Am
K. Inhibitory J Physiol.triphosphate-binding
guanosine 1987;252:C121.
nhttp://www.ncbi.nlm.nih.gov/pubmed/2180016
symporter regulate intracellular pH in mouse medullary thick limbs of Henle. 211. Wang WH. Two
protein-mediated types oofK+
regulation channel inaction
vasopressin thick ascending
in isolatedlimb o rat
single kidney.
medullary
189.J Physiol.
Am Greger1990;258:F445.
R. Chloride reabsorption in the rabbit cortical thick ascending limb Am J Physiol.
tubules 1994;267:F599.
of mouse kidney. J Clin Invest. 1988;82:1437.
o the Amemiya
231. loop o Henle. M, Lof A sodium dependent
ng J, Lotscher M, etprocess. Pf ugersof
al. Expression Arch.
NHE-31981;390:38.
in the api- 212. Wang WH,
258. Riccardi D, White
Lee WS, S, Geibel
Lee K,J, etet al. A potassium of
Localization channel in the api-
the extracellular
http://www.ncbi.nlm.nih.gov/pubmed/6264385
cal membrane of rat renal proximal tubule and thick ascending limb. Kidney Int. cal
Ca(2membrane
)-sensingoreceptor
rabbit thick ascending limb
and PTH/PTHrP receptor o Henle’
in rat skidney.
loop. Am Am J Physiol.
190. Koenig B, Ricapito S, Kinne R. Chloride transport in the thick ascend-
1995;48:1206. 1990;258:F244.
1996;271:F951.
ing limb
232. o Henle’
Laghmani K,s loop: potassium
Borensztein dependence
P, Ambuhl P, et al.andChronic
stoichiometry
metabolic o the NaCl
acidosis 213. Giebisch
259. Wang G, Hebert
W, Lu M, Balazy SC,M,Wang et al.WH New aspects
Phospholipase A2oisrenal potassium
involved trans-
in mediating
cotransport
enhances systemprotein
NHE-3 in plasma membrane
abundance andvesicles.
transport Pf ugers
activityArch. 1983;399:173.
in the rat thick as- port. Pf ugers
the effect Arch. 2003;446:289.
of extracellular Ca2+ on apical K channels in rat TAL. Am J Physiol.
http://www.ncbi.nlm.nih.gov/pubmed/6657458
cending limb by increasing NHE-3 mRNA. J Clin Invest. 1997;99:24. http://www.ncbi.nlm.nih.gov/pubmed/12684792
1997;273:F421.
191. Hebert
233. Reeves SC, WB,Andreoli
MolonyTE. DA.Control
The physiology o loop in
of NaCl transport diuretic
the thickaction. Semin
ascending 214. Lu M, WH,
260. Wang WangLu T, M,YanHebert
Q, et al. SC.ROMK is required
Cytochrome P-450 or metabolites
expression omediate
the 70-pSex-
Nephrol.
limb. Am1988;8:225.
J Physiol. 1984;246:F745. K channel in
tracellular the thick
Ca(2 ascending
)-induced limb. Am
inhibition J Physiol
of apical KRenal Physiol.in2004;286:F490.
channels the TAL. Am J
http://www.ncbi.nlm.nih.gov/pubmed/3059427
234. Mutig K, Paliege A, Kahl T, et al. Vasopressin V2 receptor expression along 215.
Physiol.Ho K, Nichols CG, Lederer WJ, et al. Cloning and expression o an in-
1996;271:C103.
192.mouse,
rat, Evelo and J, Kinne
humanR.renal Sodium-chloride
epithelia withtransportfocus oninTAL. the medullary
Am J Physiol thick as-
Renal wardly recti ying
261. el-Hajj ATP-regulated
Fuleihan G, Seifterpotassium
J, Scott J,channel. Nature. 1993;362:31.
et al. Calcium-regulated renal cal-
cending2007;
Physiol. limb o293:F1166.
Henle’s loop: evidence or a sodium-chloride cotransport system http://www.ncbi.nlm.nih.gov/pubmed/7680431
cium handling in healthy men: relationship to sodium handling. J Clin Endocrinol
in plasma
235. Gimenez membraneI, Forbushvesicles. J Membr Biol.stimulation
B. Short-term 1983;72:173. of the renal Na-K-Cl co- 216.
Metab.Lu M, Wang T, Yan Q, et al. Absence o small conductance K+ channel (SK)
1998;83:2366.
http://www.ncbi.nlm.nih.gov/pubmed/6854622
transporter (NKCC2) by vasopressin involves phosphorylation and membrane activity in apical membranes
262. McCarron DA. Diet and o thick
blood ascending limb andparadigm
pressure—the cortical collecting duct
shift. Science.
193. Gambaof
translocation G,the
Miyanoshita
protein. J BiolA, Lombardi M, et al. Molecular cloning, primary
Chem. 2003;278:26946. in ROMK (Bartter’s) knockout mice. J Biol Chem. 2002;277:37881.
1998;281:933.
structure,
236. and characterization
Gunaratne R, Braucht DW, o two members
Rinschen MM,oetthe al. mammalian
Quantitativeelectroneu-
phospho- http://www.ncbi.nlm.nih.gov/pubmed/12130653
263. Wittner M, Di Stefano A. A hormonal mosaic modulating the function of
tral sodium-(potassium)-chloride
proteomic cotransporter amily expressed
analysis reveals cAMP/vasopressin-dependent signaling in kidney.
pathways J Biol
in 217. Simon
the thick DB, Karet
ascending limbFE, Rodriguez-Soriano
of Henle’ s loop. Exp Nephrol. J, et al.1993;1:285.
Genetic heterogeneity o
Chem. renal
native 1994;269:17713.
thick ascending limb cells. Proc Natl Acad Sci USA. 2010;107:15653. Bartter’ s syndrome
264. Bouby revealed byA,mutations
N, Hus-Citharel Marchetti in theal.K+
J, et channel, of
Expression ROMK.
type 1Nat Genet.
angioten-
http://www.ncbi.nlm.nih.gov/pubmed/8021284
237. McCormick JA, Mutig K, Nelson JH, et al. A SPAK isoform switch modu- 1996;14:152.
sin II receptor subtypes and angiotensin II-induced calcium mobilization along
194. renal
lates Payne saltJA, Xu JC, Haas
transport M, et pressure.
and blood al. Primary Cellstructure, unctional expression,
Metab. 2011;14:352. http://www.ncbi.nlm.nih.gov/pubmed/8841184
the rat nephron. J Am Soc Nephrol. 1997;8:1658.
and chromosomal
238. Richardson C, localization
Sakamoto oK,the bumetanide-sensitive
de los Heros P, et al. Regulation Na-K-Cl of cotrans-por-
the NKCC2 218. Simon DB,
265. Amlal Li ton RP
H, LeGoff C,. Ion transporterC,mutations
Vernimmen et al. ANG in Gitelman’ s and Bartter’
II controls Na( )-s
ter in
ion human colon.
cotransporter byJSPAK-OSR1-dependent
Biol Chem. 1995;270:17977. and -independent pathways. J Cell syndromes.
K (NH4 )-2Cl- Curr Opin Nephrol via
cotransport Hypertens.
20-HETE 1998;7:43.
and PKC in medullary thick ascend-
195.2011;124:789.
Sci. Yang T, Huang YG, Singh I, et al. Localization o bumetanide- and thiazide- http://www.ncbi.nlm.nih.gov/pubmed/9442362
ing limb. Am J Physiol. 1998;274:C1047.
sensitive
239. Na-K-Cl cotransporters
Moriguchi T, Urushiyamaalong the rat nephron.
S, Hisamoto N, et Am al. JWNK1
Physiol. regulates
1996;271:F931. phos- 219. Schwalbe
266. Good DW,RA, George Bianchi L, Accili
T, Wang DH.EA,Angiotensin
et al. Functional consequences
II inhibits HCO-3 ab- o
196. KaplanofMR,
phorylation Plotkin MD, Lee WS, etcotransporters
cation-chloride-coupled al. Apical localization
via the oSTE20-related
the Na-K-Cl ROMK
sorptionmutants linked to antenatal
via a cytochrome Bartter’s syndrome
P-450-dependent pathwayand in implications
MTAL. Am Jor treat-
Physiol.
cotransporter,
kinases, SPAK and rBSC1, OSR1.on rat thick
J Biol ascending
Chem. limbs. Kidney Int. 1996;49:40.
2005;280:42685. ment. Hum Mol Genet. 1998;7:975.
1999;276:F726.
http://www.ncbi.nlm.nih.gov/pubmed/8770947 http://www.ncbi.nlm.nih.gov/pubmed/9580661
190
19 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

220. Kwon
267. WangemannTH, Nielsen P, WittnerJ, Kim M, YH,
Di Ste et ano A, et al. Cl(-)-channel
al. Regulation blockers in the
of sodium transporters in 243. Gamba
298. Hebert SC, Culpepper
G, Saltzberg SN,RM, AndreoliM,
Lombardi TE.et NaCl transport
al. Primary in mouse
structure andmedul-
func-
thick
the ascending
thick ascending limblimb o the ofloop o Henle.
rat kidney: Structure
response to activity
angiotensinrelationship.
II. Am JPf ugers
Physiol lary thick
tional ascending
expression oflimbs.
a cDNA II. ADH
encodingenhancement o transcellular electroneutral
the thiazide-sensitive, NaCl cotrans-
Arch. 1986;407(Suppl
Renal Physiol. 2003;285:F152. 2):S128. port; origin o transepithelial
sodium-chloride cotransporter. voltage.
Proc NatlAmAcadJ Physiol. 1981;241:F432.
Sci USA. 1993;90:2749.
221. Staahltoft
268. YoshikawaD,M,Nielsen UchidaS,S,Janjua YamauchiNR, et A, al.
et Losartan
al. Localization
treatment o rat CLC-K2
normalizes 244. Monroy
299. Hebert SC, Friedman
A, Plata PA, Andreoli
C, Hebert SC, et al. TE.Characterization
E ects o antidiuretic of the hormone
thiazide-
chloride
renal channel
sodium and mRNA in the kidney.
water handling in ratsAm J Physiol.
with 1999;276:F552.
mild congestive heart failure. Am J on cellular
sensitive Na(conductive pathways in amouse
)-Cl(-) cotransporter: new model medullaryfor ionsthickandascending limbs
diuretics inter-
222. Kobayashi
Physiol Renal Physiol. K, Uchida S, Mizutani S, et al. Intrarenal and cellular localiza-
2002;282:F307. o Henle:
action. AmI.J Physiol
ADH increases transcellular
Renal Physiol. conductance pathways. J Membr Biol.
2000;279:F161.
tion oMorel
269. CLC-K2 protein
F. Sites in the mouse
of hormone actionkidney. J Am Soc Nephrol.
in the mammalian nephron.2001;12:1327.
Am J Physiol. 1984;80:201.
300. Gamba G. The thiazide-sensitive Na -Cl- cotransporter: molecular biol-
223. Kramer BK, Bergler T, Stoelcker B, et al. Mechanisms o disease: the kid-
1981;240:F159. http://www.ncbi.nlm.nih.gov/pubmed/6094816
ogy, functional properties, and regulation by WNKs. Am J Physiol Renal Physiol.
ney-speci
270. Summersc chloride
RJ, Kuhar channels ClCKA and ClCKB,
MJ. Autoradiographic the Barttin
localization subunit,
of beta- and their
adrenoceptors 245. Ecelbarger CA, Kim GH, Knepper MA, et al. Regulation o potassium chan-
2009;297:F838.
clinical
in relevance.
rat kidney. Eur J Nat Clin Pract
Pharmacol. Nephrol. 2008;4:38.
1983;91:305. nel KirKunchaparty
301. 1.1 (ROMK) S, abundance
Palcso M, in the thick ascending
Berkman limb o Henle’
J, et al. Defective s loop. Jand
processing Am
http://www.ncbi.nlm.nih.gov/pubmed/18094726
271. DiBona GF, Sawin LL. Effect of renal nerve stimulation on NaCl and H2O Soc Nephrol.of2001;12:10.
expression thiazide-sensitive Na-Cl cotransporter as a cause of Gitelman’s
224. Birkenhager
transport in Henle’sR, Otto
loop E, Schurmann
of the rat. Am J Physiol. MJ, et1982;243:F576.
al. Mutation o BSND causes http://www.ncbi.nlm.nih.gov/pubmed/18265082
syndrome. Am J Physiol. 1999;277:F643.
BartterBencsath
272. syndrome with G,
P, Szenasi sensorineural
Takacs L. Water deaand nesselectrolyte
and kidney transportailure. Nat Genet.
in Henle’ s loop 246. De
302. Reeves
JongWB, JC, Andreoli TE. Cl-
Van Der Vliet WA, transport
Van Den in Heuvel
basolateral LP, renal
et al. medullary
Functionalvesi-ex-
2001;29:310.
and distal tubule after renal sympathectomy in the rat. Am J Physiol. 1985;249:F308. cles: II. Cl-
pression of channels
mutationsininplanar lipid bilayers.
the human J Membr Biol.evidence
NaCl cotransporter: 1990;113:57. for impaired
http://www.ncbi.nlm.nih.gov/pubmed/11687798
273. Bailly C, Imbert-Teboul M, Roinel N, et al. Isoproterenol increases http://www.ncbi.nlm.nih.gov/pubmed/1689386
routing mechanisms in Gitelman’s syndrome. J Am Soc Nephrol. 2002;13:1442.
225.Mg,
Ca, Di and
Ste ano
NaClA,reabsorption
Greger R, Desf eurs E,thick
in mouse et al.ascending
A Ba(2+)-insensitive
limb. Am JK+ con-
Physiol. 247. Needham
303. CulpepperPG, RM, Andreoli
Mikoluk TE. PGE2,P, et
K, Dhakarwal orskolin, and cholera toxinNaCl
al. The thiazide-sensitive in-
ductance in the basolateral membrane o rabbit cortical thick ascending limb
1990;258:F1224. teractions in modulating
cotransporter is targeted for NaClchaperone-dependent
transport in mouse endoplasmic mTALH. Amreticulum- J Physiol.
cells. Cell
274. OrtizPhysiol
PA, GarvinBiochem. 1998;8:89.
JL. Role of nitric oxide in the regulation of nephron trans- 1984;247:F784.
associated degradation. J Biol Chem. 2011;286:43611.
http://www.ncbi.nlm.nih.gov/pubmed/9547022
port. Am J Physiol Renal Physiol. 2002;282:F777. 248. Hadchouel
304. Escalante B, Erlij D,C,Falck
J, Delaloy FaureJR, S, et al.
al.Familial
E ect ohyperkalemic
cytochromehypertension.
P450 arachi-
226. Ortiz
275. Di StePA, ano A, Jounier
Hong NJ, Garvin S, Wittner
JL. NOM.decreases
Evidencethick supporting
ascending a role
limborchloride
KCl co- Jdonate
Am Socmetabolites on ion transport in rabbit kidney loop o Henle. Science.
Nephrol. 2006;17:208.
transporter by
absorption in the thick ascending
reducing Na( )-K(limb o Henle’
)-2Cl(-) s loop. Kidney
cotransporter Int. 2001;60:1809.
activity. Am J Physiol 1991;251:799.
305. Wilson FH, Disse-Nicodeme S, Choate KA, et al. Human hypertension
227. Physiol.
Renal Velazquez H, Silva T. Cloning and localization o KCC4 in rabbit kidney:
2001;281:F819. http://www.ncbi.nlm.nih.gov/pubmed/1846705
caused by mutations in WNK kinases. Science. 2001;293:1107.
expression
276. Ortiz inPA, distal
Garvinconvoluted tubule. Am Jand
JL. Cardiovascular PhysiolrenalRenal Physiol.
control in 2003;285:F49.
NOS-de cient 249. Subramanya
306. Lu M, Zhu Y, AR, BalazyLiuM,J,etEllison
al. E ect DH,o angiotensin
et al. WNK4 II on the apical
diverts the K+ chan-
thiazide-
228. Boettger
mouse models. T, Am Hubner
J Physiol CA, Maier
Regul H, et
Integr al. Dea
Comp ness 2003;284:R628.
Physiol. and renal tubular acidosis nel in theNaCl
sensitive thick cotransporter
ascending limbtoothe thelysosome
rat kidney. and J Gen Physiol. AP-3
stimulates 1996;108:537.
interaction.
in mice
277. lacking
Plato the K-Cl
CF, Shesely EG, co-transporter
Garvin JL. eNOS Kcc4. Nature. L2002;416:874.
mediates -arginine-induced inhibi- Jhttp://www.ncbi.nlm.nih.gov/pubmed/8972391
Biol Chem. 2009;284:18471.
http://www.ncbi.nlm.nih.gov/pubmed/11976689
tion of thick ascending limb chloride ux. Hypertension. 2000;35:319. 250. Golbang
307. Harris RC, APMcKanna
, Cope G,JA,Hamad Akai Y,A,et et al. al.
Cyclooxygenase-2
Regulation of the is associated
expressionwith of
229. Turban
278. Friedman S, WangPA, XY Andreoli
, Knepper TE.MA. CO2-stimulated
Regulation of NHE3, NaCl NKCC2,
absorption andinNCC the the macula densa o rat kidney
Na/Cl cotransporter by WNK4 and increases
and WNK1: with salt restriction.
evidence that Jaccelerated
Clin Invest.
mouse renalincortical
abundance kidney thickduringascending
aldosterone limb o Henle.
escape Evidence role
phenomenon: or synchronous
of NO. Am J 1994;94:2504.
dynamin-dependent endocytosis is not involved. Am J Physiol Renal Physiol.
Na +/H+
Physiol andPhysiol.
Renal Cl-/HCO3- exchange in apical plasma membranes. J Gen Physiol.
2003;285:F843. 251. Cheng HF, Harris RC. Cyclooxygenases, the kidney, and hypertension. Hy-
2006;291:F1369.
1982;80:683.
279. Masilamani S, Wang X, Kim GH, et al. Time course of renal Na-K-ATPase, pertension.
308. Yang2004;43:525.
CL, Angell J, Mitchell R, et al. WNK kinases regulate thiazide-
http://www.ncbi.nlm.nih.gov/pubmed/6816900
NHE3, NKCC2, NCC, and ENaC abundance changes with dietary NaCl restric- http://www.ncbi.nlm.nih.gov/pubmed/14732722
sensitive Na-Cl cotransport. J Clin Invest. 2003;111:1039.
230. Am
tion. Kikeri D, Azar
J Physiol Renal S, Physiol.
Sun A, et al. Na(+)-H+ antiporter and Na(+)-(HCO3-) n
2002;283:F648. 252. San-Cristobal
309. Harding P, Sigmon DH, Al e D,
P, Pacheco-Alvarez ME, et al. Cyclooxygenase-2
Richardson C, et al. Angiotensin medi-II
symporter
280. Songregulate
J, Hu X, intracellular
Shi M, et al. pH in mouse
Effects medullary
of dietary fat, NaCl,thickandlimbs o Henle.
fructose Am
on renal ates increased
signaling increasesrenalactivity
renin content
of the renal induced
Na-Clbycotransporter
low-sodium through diet. Hypertension.
a WNK4-
J Physiol.and
sodium 1990;258:F445.
water transporter abundances and systemic blood pressure. Am J 1997;29:297.
SPAK-dependent pathway. Proc Natl Acad Sci USA. 2009;106:4384.
231. Amemiya
Physiol Renal Physiol. M, Lo ng J, Lotscher M, et al. Expression o NHE-3 in the api-
2004;287:F1204. http://www.ncbi.nlm.nih.gov/pubmed/9039118
310. Welling PA, Chang YP, Delpire E, et al. Multigene kinase network, kidney
cal membrane
281. Na KY, Oh o YK,
rat renal
Han JS, proximal tubule and thick
et al. Upregulation of Naascending
transporterlimb.abundances
Kidney Int. 253. Molony
transport, and saltDA,inAndreoli
essential TE. Diluting power
hypertension. Kidney oInt.thick limbs o Henle. I.
2010;77:1063.
1995;48:1206.
in response to chronic thiazide or loop diuretic treatment in rats. Am J Physiol Peritubular
311. Hoornhypertonicity
EJ, Nelson JH,blocks basolateral
McCormick JA, etClal. channels.
-
The WNKAm J Physiol.
kinase network1988;
re-
http://www.ncbi.nlm.nih.gov/pubmed/8569082
Renal Physiol. 2003;284:F133. 255:F1128.
gulating sodium, potassium, and blood pressure. J Am Soc Nephrol. 2011;22:605.
232. Reilly
282. Laghmani K, Borensztein
RF, Ellison DH. Mammalian P, Ambuhl P, et
distal al. Chronic
tubule: physiology,metabolic acidosis
pathophysio- 254. Wilson
312. Hebert FH, SC, Culpepper
Kahle KT, SabathRM, Andreoli
E, et al.TE. NaCl transport
Molecular pathogenesis in mouse medul-
of inherited
enhances
logy, NHE-3 protein
and molecular anatomy. abundance
Physiol Rev. and2000;80:277.
transport activity in the rat thick as- lary thick ascending
hypertension limbs. III. Modulation
with hyperkalemia: the Na-Clocotransporter
the ADH e ect by peritubular
is inhibited os-
by wild-
cending
283. limb by increasing
Bostanjoglo M, ReevesNHE-3 WB, Reilly mRNA. RF, etJ al.
Clin Invest. 1997;99:24. dehydro-
11Beta-hydroxysteroid molality.
type but notAm mutant
J Physiol.WNK4.1981;241:F443.
Proc Natl Acad Sci USA. 2003;100:680.
http://www.ncbi.nlm.nih.gov/pubmed/9011571
genase, mineralocorticoid receptor, and thiazide-sensitive Na-Cl cotransporter 255. Y
313. Khanna
ang SS, A. AcquiredT, nephrogenic
Morimoto diabetes insipidus.
Rai T, et al. Molecular pathogenesis Semin Nephrol.
of pseudo-
233. Hebert
expression by SC,
distalAndreoli
tubules.TE. J AmControl o NaCl1998;9:1347.
Soc Nephrol. transport in the thick ascending 2006;26:244.
hypoaldosteronism type II: generation and analysis of a Wnk4(D561A/ )
limb. Hierholzer
284. Am J Physiol.K,1984;246:F745.
Wiederholt M. Some aspects of distal tubular solute and http://www.ncbi.nlm.nih.gov/pubmed/16713497
knockin mouse model. Cell Metab. 2007;5:331.
234. transport.
water Mutig K, PaliegeKidney A, Int.Kahl T, et al. Vasopressin V2 receptor expression along
1976;9:198. 256. Richardson
314. Takaichi K, C, Kurokawa
Ra qi FH, K. High Ca2+HK,
Karlsson inhibits
et al.peptide
Activationhormone-dependent
of the thiazide-
rat, mouse, and human renal epitheliaG.with ocus on TAL. -
285. Schnermann J, Briggs J, Schubert In situ studies of theAm J Physiol
distal Renal
convoluted cAMP production
sensitive speci cally in by
Na -Cl cotransporter thick
theascending
WNK-regulated limbs okinases
Henle.SPAK Minerand Electrolyte
OSR1.
Physiol.in2007;
tubule 293:F1166.
the rat. I. Evidence for NaCl secretion. Am J Physiol. 1982;243:F160. JMetab. 1986;12:342.
Cell Sci. 2008;121:675.
235. Khuri
286. Gimenez RN, I,Strieder
Forbush N, B. Short-term
Wiederholt M,stimulation
et al. Effectsoof the gradedrenal Na-K-Cl
solute co-
diuresis http://www.ncbi.nlm.nih.gov/pubmed/3027519
315. McCormick JA, Yang CL, Ellison DH. WNK kinases and renal sodium
transporter
on renal tubular(NKCC2) sodium bytransport
vasopressin in theinvolves
rat. Amphosphorylation and membrane
J Physiol. 1975;228:1262. 257. Takaichi
transport in health K, andKurokawa
disease: K. Inhibitoryview.
an integrated guanosine triphosphate-binding
Hypertension. 2008;51:588.
translocation
287. Malnic G, o theKlose protein. J Biol Chem.
RM, Giebisch G. 2003;278:26946.
Micropuncture study of distal tubular protein-mediated
316. Subramanyaregulation AR, Yang CL, o vasopressin
Zhu X, et al. action in isolated single
Dominant-negative medullary
regulation of
236. Gunaratne
potassium and sodium R, Braucht
transport DW,in Rinschen
rat nephron. MM,Am etJ al. Quantitative
Physiol. phospho-
1966;211:529. tubules by
WNK1 o its mouse kidney. J Clin
kidney-speci Invest. 1988;82:1437.
c kinase-defective isoform. Am J Physiol Renal Physiol.
proteomic
288. Hayslettanalysis reveals cAMP/vasopressin-dependent
JP, Boulpaep EL, Kashgarian M, et al. Electrical signaling pathways of
characteristics in 258. Riccardi D, Lee WS, Lee K, et al. Localization o the extracellular
2006;290:F619.
native
the renal thickdistal
mammalian ascending
tubule:limb cells. ProcofNatl
Comparison Acad Sci USA.
Ling-Gerard and 2010;107:15653.
macroelectrodes. Ca(2+)-sensing
317. Hadchouelreceptor and PTH/PTHrP
J, Soukaseum C, Busst C,receptor in rat kidney.
et al. Decreased ENaC Amexpression
J Physiol.
237. McCormick
Kidney Int. 1977;12:324. JA, Mutig K, Nelson JH, et al. A SPAK iso orm switch modu- 1996;271:F951.
compensates the increased NCC activity following inactivation of the kidney-
lates renal
289. Wright saltFS.transport
Increasing andmagnitude
blood pressure. Cell Metab.
of electrical 2011;14:352.
potential along the renal dis- 259. cWang
speci isoform W, Lu of M,WNK1BalazyandM, prevents
et al. Phospholipase
hypertension. A2 Proc
is involved
Natl Acad in mediating
Sci USA.
http://www.ncbi.nlm.nih.gov/pubmed/21907141
tal tubule. Am J Physiol. 1971;220:624. the e ect o extracellular Ca2+ on apical K+ channels in rat TAL. Am J Physiol.
2010;107:18109.
238. Allen
290. Richardson
GG, BarrattC, Sakamoto
LJ. Origin K, de los Herostransepithelial
of positive P, et al. Regulation o the
potential NKCC2
difference 1997;273:F421.
318. Greger R, Velazquez H. The cortical thick ascending limb and early dis-
ionearly
in cotransporter
distal segments by SPAK-OSR1-dependent
of rat kidney. Kidney Int. and1985;27:622.
-independent pathways. J Cell 260.convoluted
tal Wang WH, Lu M,inHebert
tubule SC. Cytochrome
the urinary concentrating P-450mechanism.
metabolites mediate ex-
Kidney Int.
Sci. 2011;124:789.
291. Barratt LJ, Rector FC Jr, Kokko JP, et al. Transepithelial potential difference tracellular Ca(2+)-induced inhibition o apical K+ channels in the TAL. Am J
1987;31:590.
http://www.ncbi.nlm.nih.gov/pubmed/21321328
pro le of the distal tubule of the rat kidney. Kidney Int. 1975;8:368. Physiol.
319. 1996;271:C103.
Yoshitomi K, Shimizu T, Taniguchi J, et al. Electrophysiological character-
239. Ellison
292. Moriguchi DH,T,Velazquez
Urushiyama S, Hisamoto
H, Wright N, et al. WNK1sodium
FS. Thiazide-sensitive regulates phos-
chloride 261. el-Hajj
ization of rabbit Fuleihan G, Sei ter J, tubule
distal convoluted Scott J,cell.
et al.P Calcium-regulated
ugers Arch. 1989;414:457. renal calcium
phorylation in
cotransport o early
cation-chloride-coupled
distal tubule. Am J Physiol. cotransporters via the STE20-related ki-
1987;253:F546. handling
320. in healthy
Velazquez men: relationship
H, Greger R. Electricaltoproperties
sodium handling.
of the early J Clin Endocrinol
distal convolutedMe-
nases,Velazquez
293. SPAK and H, OSR1.Good J Biol
DW,Chem.
Wright 2005;280:42685.
FS. Mutual dependence of sodium and tab. 1998;83:2366.
tubules of the rabbit kidney. Renal Physiol. 1986;9:55.
http://www.ncbi.nlm.nih.gov/pubmed/16263722
chloride absorption by renal distal tubule. Am J Physiol. 1984;247:F904. http://www.ncbi.nlm.nih.gov/pubmed/9661610
321. Bockenhauer D, Feather S, Stanescu HC, et al. Epilepsy, ataxia, sensorineu-
240. Boron
294. Yang SS, WF,Lo YF, Wu
Sackin CC, et al. SPAK-knockout
H. Measurement of intracellularmice ionicmani est Gitelman
composition and 262.
ral McCarron
deafness, DA. Dietandand
tubulopathy, bloodmutations.
KCNJ10 pressure—the N Englparadigm shi t. Science.
J Med. 2009;360:1960.
syndromeinand
activities renalimpaired
tubules.vasoconstriction.
Annu Rev Physiol.J Am Soc Nephrol. 2010;21:1868.
1983;45:483. 1998;281:933.
322. Scholl UI, Choi M, Liu T, et al. Seizures, sensorineural deafness, ataxia,
http://www.ncbi.nlm.nih.gov/pubmed/20813865
295. Costanzo LS. Localization of diuretic action in microperfused rat distal http://www.ncbi.nlm.nih.gov/pubmed/9722467
mental retardation, and electrolyte imbalance (SeSAME syndrome) caused by
241. LinCaSH,
tubules: andYNa u IS, Jiang ST,Am
transport. et Jal. Impaired
Physiol. phosphorylation o Na(+)-K(+)-
1985;248:F527. 263. Wittner
mutations M, Di Ste
in KCNJ10. anoNatl
Proc A. A hormonal
Acad Sci USA.mosaic modulating the unction o
2009;106:5842.
2Cl(-)Beaumont
296. cotransporter by oxidative
K, Vaughn stress-responsive
DA, Healy DP. Thiazide kinase-1
diureticdereceptors:
ciency mani ests
autora- the thick
323. ascending
Reichold limb o AA,
M, Zdebik Henle’ s loop.E,Exp
Lieberer et Nephrol.
al. KCNJ10 1993;1:285.
gene mutations caus-
hypotensionlocalization
diographic and Bartter-likein rat syndrome.
kidney withProc Natl Acad Sci USA.
[3H]metolazone. 2011;108:17538.
J Pharmacol Exp Ther. http://www.ncbi.nlm.nih.gov/pubmed/8081979
ing EAST syndrome (epilepsy, ataxia, sensorineural deafness, and tubulopathy)
http://www.ncbi.nlm.nih.gov/pubmed/21972418
1989;250:414. 264. Bouby
disrupt channel N, function.
Hus-Citharel ProcA,Natl
Marchetti
Acad SciJ, USA.
et al. 2010;107:14490.
Expression o type 1 angioten-
242. Ellison
297. Ra qi FH, DH, Zuber AM, Glover
Biemesderfer M, et al.J, Role
D, Morrisey et al. oImmunocytochemical
the WNK-activated char- SPAK sin II Pacheco-Alvarez
324. receptor subtypesD,and angiotensin
Cristobal PS, MeadeII-induced
P, et al. calcium :Cl- cotransporter
The Na mobilization along
kinase in regulating
acterization of the high-af blood pressure.
nity thiazide EMBO Mol Med.
diuretic 2010;2:63.
receptor in rabbit renal cortex. theactivated
is rat nephron. J Am Soc Nephrol.
and phosphorylated 1997;8:1658.
at the amino-terminal domain upon intracellular
http://www.ncbi.nlm.nih.gov/pubmed/20091762
Am J Physiol. 1993;264:F141. http://www.ncbi.nlm.nih.gov/pubmed/9355068
chloride depletion. J Biol Chem. 2006;281:28755.
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1191
91

265. Wiederholt
325. Amlal H, LeGo C, Vernimmen
M, Hansen L, Giebisch C, etG. al. Dependence
ANG II controls Na(+)-K+
of distal (NH4+
peritubular 291. Barratt PM,
353. Snyder LJ, Rector
OlsonFC DR,Jr,Kabra
Kokko R, JP
et, al.
et al. Transepithelial
cAMP and serum and potential di erence
glucocorticoid-
)-2Cl- cotransport
membrane potential via on
20-HETE
luminaland PKC inand
sodium medullary thick potassium.
peritubular ascend ing limb. Am
P ugers pro le o the
inducible distal
kinase tubule
(SGK) o the rat
regulate thekidney.
epithelial Kidney
Na( Int. 1975;8:368.
) channel through conver-
J Physiol.
Arch. 1998;274:C1047.
1975;355:R55. http://www.ncbi.nlm.nih.gov/pubmed/1206853
gent phosphorylation of Nedd4-2. J Biol Chem. 2004;279:45753.
266. Costanzo
326. Good DW, LS.George
Comparison T, Wang DH. Angiotensin
of calcium and sodiumIItransportinhibits in HCO-3 ab-
early and 292. Ellison DH,EE,
354. Windhager Velazquez
GiebischH,G.Wright FS. Thiazide-sensitive
Micropuncture study of renalsodium tubular chloride
transfer
sorption
late via atubules:
rat distal cytochromeeffect of P-450-dependent
amiloride. Am J pathway in MTAL. Am J Physiol.
Physiol. 1984;246:F937. cotransport in earlyin
of sodium chloride distal tubule.
the rat. Am Am J Physiol.
J Physiol. 1987;253:F546.
1961;200:581.
1999;276:F726.
327. Lof ng J, Pietri L, Aregger F, et al. Differential subcellular localization of 293. VelazquezEL.
355. Boulpaep H, Recent
Good DW, Wright
advances in FS. Mutual dependence
electrophysiology o sodiumAnnu
of the nephron. and
267. Kwon
ENaC subunits TH,inNielsen
mouse J,kidneyKim YH, et al. Regulation
in response to high-o and sodiumlow-Na transporters
diets. AminJ chloride
Rev Physiol.absorption
1976;38:21. by renal distal tubule. Am J Physiol. 1984;247:F904.
the thick
Physiol Renalascending
Physiol. limb o rat kidney: response to angiotensin II. Am J Physiol
2000;279:F252. 294. Boron WF
356. Sansom , Sackin
SC, O’NeilH.RG Measurement
Mineralocorticoid o intracellular
regulation ionic composition
of apical cell mem-and
Renal Physiol.
328. Schmitt 2003;285:F152.
R, Ellison DH, Farman N, et al. Developmental expression of activities
brane Nain renal and tubules.
K transport Annu Rev of Physiol.
the cortical1983;45:483.
collecting duct. Am J Physiol.
268. Staahlto
sodium t D, Nielsen
entry pathways in ratS,nephron.
Janjua NR, Amet al. Losartan
J Physiol. treatment normalizes
1999;276:F367. http://www.ncbi.nlm.nih.gov/pubmed/6342522
1985;248:F858.
renal sodium
329. Biner HL, andArpin-Bott
water handling MP, Lof in rats
ng J,with mild
et al. congestive
Human cortical heart
distalailure. Am J
nephron: 295. Costanzo
357. Sansom SC,LS. Localization
Weinman o diuretic
EJ, O’Neil action in microper
RG. Microelectrode assessment used rat distal
of chloride-
Physiol Renal of
distribution Physiol. 2002;282:F307.
electrolyte and water transport pathways. J Am Soc Nephrol. tubules:
conductive Ca properties
and Na transport.of cortical Amcollecting
J Physiol. duct.
1985;248:F527.
Am J Physiol. 1984;247:F291.
269. Morel F. Sites o hormone action in the mammalian nephron. Am J Physiol.
2002;13:836. 296. Beaumont
358. Stokes JB. K, Vaughn secretion
Potassium DA, HealybyDPcortical . Thiazide diuretictubule:
collecting receptors: autora-
relation to
1981;240:F159.
330. Ellison DH, Velazquez H, Wright FS. Adaptation of the distal convoluted diographic localization
sodium absorption, in rat sodium
luminal kidney with [3H]metolazone.
concentration, J Pharmacol Exp
and transepithelial Ther.
voltage.
270. Summers
tubule of the rat. RJ,Structural
Kuhar MJ.and Autoradiographic
functional effects localization
of dietary o beta-adrenocep-
salt intake and 1989;250:414.
Am J Physiol. 1981;241:F395.
tors in rat
chronic kidney.infusion.
diuretic Eur J Pharmacol.
J Clin Invest.1983;91:305.
1989;83:113. http://www.ncbi.nlm.nih.gov/pubmed/2746509
359. O’Neil RG, Sansom SC. Characterization of apical cell membrane Na and
http://www.ncbi.nlm.nih.gov/pubmed/6311583
331. Chen ZF, Vaughn DA, Beaumont K, et al. Effects of diuretic treatment 297. Ellison DH, Biemesder
K conductances of cortical ercollecting
D, Morrisey ductJ,using
et al. Immunocytochemical
microelectrode techniques. char-
271.ofDiBona
and dietaryGF , Sawinon
sodium LL.renal
E ectbinding
o renalofnerve stimulation on
3H-metolazone. J Am NaCl
Socand H2O
Nephrol. acterization
Am J Physiol.o1984;247:F14.
the high-a nity thiazide diuretic receptor in rabbit renal cortex.
transport in Henle’s loop o the rat. Am J Physiol. 1982;243:F576.
1990;1:91. Am
360.J Physiol.
Sansom1993;264:F141.
SC, Agulian S, Muto S, et al. K activity of CCD principal cells from
272. Lof
332. Bencsath
ng J, P , Szenasi
Lof ng-Cueni G, Takacs
D, Hegyi L. Water
I, et al.and electrolyte
Thiazide transport
treatment in Hen-
of rats pro- 298.
normalGamba G, Saltzberg rabbits.
and DOCA-treated SN, Lombardi M, et al.
Am J Physiol. Primary structure and unc-
1989;256:F136.
le’s loop
vokes and distal
apoptosis tubule
in distal a ter cells.
tubule renalKidney
sympathectomy in the rat. Am J Physiol.
Int. 1996;50:1180. tional expression
361. Garty o a cDNA
H, Palmer encodingsodium
LG. Epithelial the thiazide-sensitive,
channels: function, electroneutral
structure,sodi-and
1985;249:F308.
333. Fanestil DD, Vaughn DA, Blakely P. Dietary NaCl and KCl do not regulate um-chloride cotransporter.
regulation. Physiol Proc Natl Acad Sci USA. 1993;90:2749.
Rev. 1997;77:359.
273. density
renal Bailly C, Imbert-Teboul
of the thiazide diuretic M, Roinelreceptor.N, Am et al. Isoproterenol
J Physiol. increases Ca,
1997;273:R1241. 299. Monroy A,
362. Canessa CM, Plata C, Hebert SC,
Horisberger JD, etRossier
al. Characterization
BC. Epithelialo sodium the thiazide-sen-
channel
Mg, and
334. KnepperNaCl MA, reabsorption
Kim GH, in mouse thick
Masilamani ascending
S. Renal tubule limb.
sodium Amtransporter
J Physiol. sitive
relatedNa(+)-Cl(-)
to proteinscotransporter: a new model or ions
involved in neurodegeneration. and 1993;361:467.
Nature. diuretics interaction.
1990;258:F1224.
abundance pro ling in rat kidney: response to aldosterone and variations in Am
363.J Physiol
Canessa Renal
CM,Physiol.
Schild2000;279:F161.
L, Buell G, et al. Amiloride-sensitive epithelial Na
274. intake.
NaCl Ortiz PA, AnnGarvin
NY Acad JL. Role o nitric oxide in the regulation o nephron trans-
Sci. 2003;986:562. 300.
channel Gamba
is made G. ofThe thiazide-sensitive
three homologous subunits. Na+ -Cl-Nature.
cotransporter:
1994;367:463.molecular biol-
port. Am
335. J Physiol
Shimizu T, Y Renal Physiol.
oshitomi 2002;282:F777.
K, Taniguchi J, et al. Effect of high NaCl intake on ogy,
364. unctional
Palmer LG, properties,
Frindt G. andAmiloride-sensitive
regulation by WNKs. NaAm J Physiolfrom
channels Renalthe Physiol.
api-
275. and
Na OrtizKPA,transport
Hong NJ, in Garvin JL. NOdistal
the rabbit decreases thick ascending
convoluted tubule. Plimb chloride
ugers Arch. 2009;297:F838.
cal membrane of the rat cortical collecting tubule. Proc Natl Acad Sci USA.
absorption by reducing Na(+)-K(+)-2Cl(-) cotransporter activity. Am J Physiol Re-
1989;414:500. 301. Kunchaparty S, Palcso M, Berkman J, et al. De ective processing and ex-
1986;83:2767.
nal Physiol.
336. Farman 2001;281:F819.
N, Bonvalet JP. Aldosterone binding in isolated tubules. III. Auto- pression
365. Canessao thiazide-sensitive
CM, Merillat AM, Na-Cl cotransporter
Rossier BC. Membrane as a cause
topologyo Gitelman’ s syn-
of the epithe-
276. Ortiz PA,
radiography along Garvin
the ratJL. Cardiovascular
nephron. Am J Physiol. and renal control in NOS-de cient
1983;245:F606. drome. Am Jchannel
lial sodium Physiol. in 1999;277:F643.
intact cells. Am J Physiol. 1994;267:C1682.
mouseFarman
337. models.N,Am J Physiol
Oblin ME, Regul
Lombes IntegrM, Comp Physiol. 2003;284:R628.
et al. Immunolocalization of gluco- and 302. De JongPM,
366. Snyder JC, Van
McDonald Der Vliet FJ, WA,
StokesVanJB, Den et Heuvel LP, et al.topology
al. Membrane Functional ex-
of the
277. Plato CF, Shesely
mineralocorticoid receptors EG, Garvinkidney.
in rabbit JL. eNOS mediates
Am J Physiol. L-arginine-induced
1991;260:C226. pression o mutations
amiloride-sensitive in the human
epithelial sodium NaCl channel. cotransporter:
J Biol Chem.evidence or impaired
1994;269:24379.
inhibition
338. o thick ascending
El Mernissi G, Doucetlimb chloride
A. Speci f ux. Hypertension.
c activity of Na-K-ATPase 2000;35:319.
after adre- routing
367. Jastimechanisms
J, Furukawa in H, Gitelman’
Gonzales s syndrome. J Am Soc Nephrol.
EB, et al. Structure 2002;13:1442.
of acid-sensing ion chan-
http://www.ncbi.nlm.nih.gov/pubmed/10642318
nalectomy and hormone replacement along the rabbit nephron. P ugers Arch. 303.
nel 1 atNeedham PG, Mikoluk
1.9 A resolution and low K, Dhakarwal
pH. Nature.P,2007;449:316.
et al. The thiazide-sensitive NaCl
278. Turban S, Wang XY, Knepper MA. Regulation o NHE3, NKCC2, and NCC
1984;402:258. cotransporter
368. Hughey isRPtargeted
, Bruns JB, or Kinlough
chaperone-dependent endoplasmic
CL, et al. Epithelial sodium reticulum-asso-
channels are
abundance
339. in kidney
El Mernissi duringA.aldosterone
G, Doucet Short-termescape effectsphenomenon:
of aldosteronerole and odexametha-
NO. Am J ciated degradation.
activated J Biol Chem.
by furin-dependent 2011;286:43611.
proteolysis. J Biol Chem. 2004;279:18111.
Physiol
sone onRenal Physiol. 2003;285:F843.
Na-K-ATPase along the rabbit nephron. P ugers Arch. 1983;399:147. http://www.ncbi.nlm.nih.gov/pubmed/22027832
369. Hughey RP, Mueller GM, Bruns JB, et al. Maturation of the epithelial Na
279. Stanton
340. Masilamani BA. S,Regulation
Wang X, Kim GH, et al.
by adrenal Time course of
corticosteroids o renal
sodium Na-K-ATPase,
and potas- 304.
channel Hadchouel J, Delaloy processing
involves proteolytic C, Faure S,ofetthe al.alpha-
Familial andhyperkalemic
gamma-subunits. hyperten-
J Biol
NHE3,transport
sium NKCC2,inNCC, loopand ENaC and
of Henle abundance changes
distal tubule ofwith dietary NaCl
rat kidney. J Clin restric-
Invest. sion.
Chem.J 2003;278:37073.
Am Soc Nephrol. 2006;17:208.
tion. Am J Physiol Renal Physiol. 2002;283:F648.
1986;78:1612. http://www.ncbi.nlm.nih.gov/pubmed/16221868
370. Kleyman TR, Carattino MD, Hughey RP. ENaC at the cutting edge: regula-
280. Velazquez
341. Song J, Hu H, X, Shi M, etA,
Bartiss al. Bernstein
E ects o dietary P, et al.at, Adrenal
NaCl, andsteroids
ructosestimulate
on renal 305.
tion ofWilson
epithelialFH,sodiumDisse-Nicodeme
channels byS,proteases.
Choate KA, J BioletChem.
al. Human hypertension
2009;284:20447.
sodium and waterNaCl
thiazide-sensitive transporter
transport abundances
by rat renal and systemic blood pressure.
distal tubules. Am J
Am J Physiol. caused by mutations
371. Carattino MD, in ShengWNKS,kinases.
Kleyman Science. 2001;293:1107.
TR. Epithelial Na channels are acti-
Physiol Renal Physiol. 2004;287:F1204.
1996;270:F211. http://www.ncbi.nlm.nih.gov/pubmed/11498583
vated by laminar shear stress. J Biol Chem. 2004;279:4120.
281. Chen
342. Na KY , Oh
Z, YK, Han
Vaughn DA,JS, et al. PUpregulation
Blakely o Na+ transporter
, et al. Adrenocortical abundances
steroids increase renal 306. Subramanya T,
372. Abi-Antoun AR,Shi LiuS,J,Tolino
EllisonLA, DH,etetal.al.Second
WNK4 transmembrane
diverts the thiazide-sen-
domain
in response
thiazide to chronic
diuretic receptor thiazide
densityorand loop diureticJ treatment
response. Am Soc Nephrol.in rats.1994;5:1361.
Am J Physiol sitive NaCl epithelial
modulates cotransporter sodium to the lysosome
channel and in
gating stimulates
responseAP-3 interaction.
to shear stress. AmJ BiolJ
Renal Physiol.
343. Kim GH, 2003;284:F133.
Masilamani S, Turner R, et al. The thiazide-sensitive Na-Cl Chem.
Physiol2009;284:18471.
Renal Physiol. 2011;300:F1089.
282. Reilly RF,isEllison
cotransporter DH. Mammalian distal
an aldosterone-induced tubule:Proc
protein. physiology,
Natl Acad pathophysio-
Sci USA. http://www.ncbi.nlm.nih.gov/pubmed/19401467
373. Morimoto T, Liu W, Woda C, et al. Mechanism underlying ow stimula-
logy, and molecular anatomy. Physiol Rev. 2000;80:277.
1998;95:14552. 307.
tion ofGolbang
sodium AP , Cope G, in
absorption Hamad A, et al. Regulation
the mammalian collecting o the
duct.expression
Am J Physiolo the Na/
Renal
http://www.ncbi.nlm.nih.gov/pubmed/10617770
344. Rozansky DJ, Cornwall T, Subramanya AR, et al. Aldosterone mediates Cl cotransporter
Physiol. by WNK4 and WNK1: evidence that accelerated dynamin-de-
2006;291:F663.
283. Bostanjoglo
activation M, Reeves WB, Reilly
of the thiazide-sensitive Na-ClRF , et al. 11Beta-hydroxysteroid
cotransporter through an SGK1dehy- and pendent
374. Schildendocytosis
L. The is not involved.
epithelial sodium Amchannel:
J Physiol Renal Physiol. 2006;291:F1369.
from molecule to disease. Rev
dro-genase,
WNK4 mineralocorticoid
signaling pathway. J Clinreceptor, and thiazide-sensitive Na-Cl cotrans-
Invest. 2009;119:2601. 308.
PhysiolYBiochem
ang CL, Pharmacol.
Angell J, Mitchell R, et al. WNK kinases regulate thiazide-sensi-
2004;151:93.
porterArroyo
345. expression by distal
JP, Lagnaz D,tubules.
RonzaudJ Am C, Soc
et al.Nephrol.
Nedd4-2 1998;9:1347.
modulates renal Na - tive
375.Na-Cl cotransport.
Shimkets RA, Warnock J Clin Invest. 2003;111:1039.
DG, Bositis CM, et al. Liddle’s syndrome: heritable
http://www.ncbi.nlm.nih.gov/pubmed/9697656
Cl- cotransporter via the aldosterone-SGK1-Nedd4-2 pathway. J Am Soc Nephrol. http://www.ncbi.nlm.nih.gov/pubmed/12671053
human hypertension caused by mutations in the beta subunit of the epithelial
284. Hierholzer K, Wiederholt M. Some aspects o distal tubular solute and
2011;22:1707. 309.
sodiumSan-Cristobal
channel. Cell.P, 1994;79:407.
Pacheco-Alvarez D, Richardson C, et al. Angiotensin II
water Chiga
346. transport. M, Rai Kidney Int.SS,
T, Yang 1976;9:198.
et al. Dietary salt regulates the phosphorylation of signaling
376. Hansson increases activity o the renal
JH, Nelson-Williams Na-Cl cotransporter
C, Suzuki through caused
H, et al. Hypertension a WNK4- by
http://www.ncbi.nlm.nih.gov/pubmed/940262
OSR1/SPAK kinases and the sodium chloride cotransporter through aldosterone. SPAK-dependent
a truncated epithelial pathway.sodium Procchannel
Natl Acad Sci USA.
gamma 2009;106:4384.
subunit: genetic heterogeneity of
285. Schnermann
Kidney Int. 2008;74:1403. J, Briggs J, Schubert G. In situ studies o the distal convoluted http://www.ncbi.nlm.nih.gov/pubmed/19240212
Liddle syndrome. Nat Genet. 1995;11:76.
tubuleVallon
347. in the V rat. I. Evidence
, Schroth J, Lang or FNaCl
, et al.secretion.
Expression AmandJ Physiol. 1982;243:F160.
phosphorylation of the 310. Welling H,
377. Tamura PA,Schild
Chang L,YP , DelpireN,
Enomoto E,etetal.al.Liddle
Multigene
diseasekinase
causednetwork, kidney
by a missense
286. -Cl-
Na Khuri RN, Strieder
cotransporter NCC N, Wiederholt M, et al. Ebyects
in vivo is regulated o graded
dietary solute diuresis
salt, potassium, and transport,
mutation of andbeta
saltsubunit
in essential hypertension.
of the epithelial sodiumKidney Int. 2010;77:1063.
channel gene. J Clin Invest.
on renalAm
SGK1. tubular
J Physiol sodium
Renaltransport in the rat. Am J Physiol. 1975;228:1262.
Physiol. 2009;297:F704. 311. Hoorn EJ, Nelson JH, McCormick JA, et al. The WNK kinase network re-
1996;97:1780.
287. Chen
348. MalnicZ,G, Klose DA,
Vaughn RM, Fanestil
GiebischDD. G. Micropuncture
In uence of gender studyon o renal
distalthiazide
tubular gulating
378. Snydersodium, PM,potassium,
Steines JC, and bloodDR.
Olson pressure.
RelativeJ Am Soc Nephrol.of2011;22:605.
contribution Nedd4 and
potassium
diuretic and sodium
receptor densitytransport
and response.in rat nephron. Am J Physiol.
J Am Soc Nephrol. 1966;211:529.
1994;5:1112. 312.
Nedd4-2 Wilson FH, Kahle
to ENaC regulation KT, Sabath E, et al.
in epithelia Molecularbypathogenesis
determined RNA interference. o inherited
J Biol
http://www.ncbi.nlm.nih.gov/pubmed/5927880
349. Verlander JW, Tran TM, Zhang L, et al. Estradiol enhances thiazide- hypertension with hyperkalemia: the Na-Cl cotransporter is inhibited by wild-
Chem. 2004;279:5042.
288. Hayslett
sensitive JP, Boulpaep EL,
NaCl cotransporter Kashgarian
density M, etplasma
in the apical al. Electrical
membrane characteristics o
of the distal type
379. but not mutant
Chang SS, Grunder WNK4. S, Proc Natl Acad
Hanukoglu A,Sciet USA. 2003;100:680.
al. Mutations in subunits of the
the mammalian
convoluted tubule distal tubule: Comparison
in ovariectomized rats. JoClin Ling-Gerard and macroelectrodes.
Invest. 1998;101:1661. http://www.ncbi.nlm.nih.gov/pubmed/12515852
epithelial sodium channel cause salt wasting with hyperkalaemic acidosis, pseu-
KidneyPedersen
350. Int. 1977;12:324.
NB, Hofmeister MV, Rosenbaek LL, et al. Vasopressin induces 313. Yang SS, Morimoto
dohypoaldosteronism typeT,1.Rai NatT,Genet.
et al. 1996;12:248.
Molecular pathogenesis o pseudo-hy-
http://www.ncbi.nlm.nih.gov/pubmed/604621
phosphorylation of the thiazide-sensitive sodium chloride cotransporter in the poaldosteronism
380. Terada Y, Knepper type II: generation and analysis NaCl
MA. Thiazide-sensitive o a Wnk4(D561A/+)
absorption in ratknockin cortical
289. convoluted
distal Wright FS. Increasing
tubule. Kidney magnitude o electrical potential along the renal dis-
Int. 2010;78:160. mouse model.
collecting duct.Cell
AmMetab.
J Physiol. 2007;5:331.
1990;259:F519.
tal tubule.
351. MutigAm K,JSaritas
Physiol.T,1971;220:624.
Uchida S, et al. Short-term stimulation of the thiazide- http://www.ncbi.nlm.nih.gov/pubmed/17488636
381. Leviel F, Hubner CA, Houillier P, et al. The Na -dependent chloride-
http://www.ncbi.nlm.nih.gov/pubmed/5545669
sensitive Na -Cl- cotransporter by vasopressin involves phosphorylation and 314. Richardson
bicarbonate exchanger C, RaSLC4A8 qi FH, Karlsson
mediates HK, et al. Activation
an electroneutral Na o the thiazide-
reabsorption
290. Allentranslocation.
membrane GG, Barratt LJ. AmOrigin
J Physiolo positive transepithelial
Renal Physiol. 2010;298:F502.potential di erence sensitive
process inNa+ the -Cl -
renal cotransporter
cortical collecting by theductsWNK-regulated
of mice. J Clin kinases
Invest.SPAK and OSR1.
2010;120:1627.
in early
352. distal segments
Nedvetsky PI, Tammao ratG, kidney. Kidney Int.
Beulshausen S, et1985;27:622.
al. Regulation of aquaporin-2 J382.
Cell Sci. 2008;121:675.
Eladari D, Hubner CA. Novel mechanisms for NaCl reabsorption in the
http://www.ncbi.nlm.nih.gov/pubmed/4010149
traf cking. Handb Exp Pharmacol. 2009:133. http://www.ncbi.nlm.nih.gov/pubmed/18270262
collecting duct. Curr Opin Nephrol Hypertens. 2011;20:506.
192
19 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

315. Tomaschitz
383. McCormickA,JA, PilzYang CL,E,Ellison
S, Ritz DH. WNK kinases
et al. Aldosterone and hypertension.
and arterial renal sodium 411. El Mernissi
339. Blazer-Y ost BL, G,Paunescu
Doucet A. TG,Short-term
Helman SI,eetects o aldosterone and
al. Phosphoinositide dex-
3-kinase
transport
Nat in health and
Rev Endocrinol. disease: an integrated view. Hypertension. 2008;51:588.
2010;6:83. ametha-sone
is required for on aldosterone-regulated
Na-K-ATPase along sodium the rabbit nephron. Am
reabsorption. Pf ugers Arch.
J Physiol.
http://www.ncbi.nlm.nih.gov/pubmed/18212265
384. Wang WH, Giebisch G. Regulation of potassium (K) handling in the renal 1983;399:147.
1999;277:C531.
316. Subramanya
collecting duct. P ugers AR, Y ang CL,
Arch. Zhu X, et al. Dominant-negative regulation o
2009;458:157. http://www.ncbi.nlm.nih.gov/pubmed/6139788
412. Park J, Leong ML, Buse P, et al. Serum and glucocorticoid-inducible
WNK1Naray-Fejes-Toth
385. by its kidney-speci A, cFejes-Toth
kinase-de ective iso orm. Am J Physiol
G. Glucocorticoid Renalmediate
receptors Physiol. 340. Stanton
kinase (SGK) isBA. Regulation
a target of the PI by3-kinase-stimulated
adrenal corticosteroids o sodium
signaling pathway. andEmbo
potas- J.
2006;290:F619.
mineralocorticoid-like effects in cultured collecting duct cells. Am J Physiol. sium transport in loop o Henle and distal tubule o rat kidney. J Clin Invest.
1999;18:3024.
317. Hadchouel J, Soukaseum C, Busst C, et al. Decreased ENaC expression
1990;259:F672. 1986;78:1612.
413. Sansom SC, O’Neil RG. Effects of mineralocorticoids on transport properties
compensates
386. Funder JW, the Pearce
increased NCC activity
PT, Smith ollowing inactivation
R, et al. Mineralocorticoid action:o target
the kidney-
tissue 341.
of Velazquez
cortical collecting H, duct
Bartiss A, Bernstein
basolateral membrane.P, et Am al. Adrenal
J Physiol. steroids stimulate
1986;251:F743.
speci city
c isoisorm
enzyme,o WNK1 and prevents
not receptor, mediated. hypertension. Proc Natl Acad Sci USA.
Science. 1988;242:583. thiazide-sensitive
414. Barlet-Bas C,NaCl transport
Khadouri C, Marsyby ratS, etrenal distal tubules.
al. Enhanced Am J sodium
intracellular Physiol.
2010;107:18109.
387. Pearce D, Yamamoto KR. Mineralocorticoid and glucocorticoid receptor 1996;270:F211.
concentration in kidney cells recruits a latent pool of Na-K-ATPase whose size is
318. Greger
activities R, Velazquez
distinguished H. The cortical
by nonreceptor factorsthickat aascending
compositelimb and early
response element.dis- 342. Chenby
modulated Z, corticosteroids.
Vaughn DA, Blakely J BiolPChem.
, et al. 1990;265:7799.
Adrenocortical steroids increase renal
tal convoluted
Science. tubule in the urinary concentrating mechanism. Kidney Int.
1993;259:1161. thiazide
415. diuretic
Verrey receptor density
F, Kraehenbuhl and response.
JP, Rossier J Am Socinduces
BC. Aldosterone Nephrol.a 1994;5:1361.
rapid increase
1987;31:590.
388. Bonvalet JP, Doignon I, Blot-Chabaud M, et al. Distribution of 11 beta- 343.
in Kim ofGH,
the rate Masilamani
Na,K-ATPase geneS,transcription
Turner R, et al. The kidney
in cultured thiazide-sensitive
cells. Mol Endo- Na-
http://www.ncbi.nlm.nih.gov/pubmed/3550228
hydroxysteroid dehydrogenase along the rabbit nephron. J Clin Invest. 1990;86:832. Cl cotransporter
crinol. 1989;3:1369. is an aldosterone-induced protein. Proc Natl Acad Sci USA.
319. Naray-Fejes-Toth
389. Yoshitomi K, Shimizu T, Taniguchi
A, Watlington CO,J,Fejes-Toth
et al. Electrophysiological character-
G. 11 beta-Hydroxysteroid 1998;95:14552.
416. Palmer LG, Antonian L, Frindt G. Regulation of the Na-K pump of the rat
ization o rabbitactivity
dehydrogenase distal convoluted
in the renal tubule
target cell.cells
Pf ugers Arch. 1989;414:457.
of aldosterone. Endocrinology. http://www.ncbi.nlm.nih.gov/pubmed/9826738
cortical collecting tubule by aldosterone. J Gen Physiol. 1993;102:43.
http://www.ncbi.nlm.nih.gov/pubmed/2477793
1991;129:17. 344. Frindt
417. Rozansky DJ, Cornwall
G, Sackin H, Palmer T, LG.
Subramanya
Whole-cell AR, et al. Aldosterone
currents in rat corticalmediates
collect-
320. Mune
390. Velazquez H, Greger
T, Rogerson FM,R. Nikkila
ElectricalH,properties
et al. Human o thehypertension
early distal convoluted
caused by activation
ing tubule:olow-Na the thiazide-sensitive Na-Cl cotransporter
diet increases amiloride-sensitive through an
conductance. AmSGK1 and
J Physiol.
tubules o in
mutations thetherabbit
kidneykidney. RenalofPhysiol.
isozyme 1986;9:55.
11 beta-hydroxysteroid dehydrogenase. Nat WNK4 signaling pathway. J Clin Invest. 2009;119:2601.
1990;258:F562.
321. Bockenhauer
Genet. 1995;10:394.D, Feather S, Stanescu HC, et al. Epilepsy, ataxia, sensorineu- http://www.ncbi.nlm.nih.gov/pubmed/19690383
418. Pacha J, Frindt G, Antonian L, et al. Regulation of Na channels of the rat
ral deaStanton
391. ness, tubulopathy,
B, Janzen A,and KCNJ10 mutations.
Klein-Robbenhaar G, etNal.Engl J Med. 2009;360:1960.
Ultrastructure of rat ini- 345. Arroyo
cortical JP, Lagnaz
collecting tubuleD, byRonzaud C, etJal.
aldosterone. GenNedd4-2
Physiol. modulates
1993;102:25. renal Na+ -Cl-
322.collecting
tial Scholl UI, Choi Effect
tubule. M, LiuofT,adrenal
et al. Seizures, sensorineural
corticosteroid treatment. deaJness, ataxia,
Clin Invest. cotransporter
419. May A, Puoti via the aldosterone-SGK1-Nedd4-2
A, Gaeggeler HP, et al. Early effect pathway. J Am Soc
of aldosterone on Nephrol.
the rate
mental retardation, and electrolyte imbalance (SeSAME syndrome) caused by
1985;75:1327. 2011;22:1707.
of synthesis of the epithelial sodium channel alpha subunit in A6 renal cells. J Am
mutations
392. Palmerin LG,
KCNJ10.Li JH,Proc Natl Acad
Lindemann B, Sci USA.
et al. 2009;106:5842.
Aldosterone control of the density of http://www.ncbi.nlm.nih.gov/pubmed/21852580
Soc Nephrol. 1997;8:1813.
http://www.ncbi.nlm.nih.gov/pubmed/19289823
sodium channels in the toad urinary bladder. J Membr Biol. 1982;64:91. 346. Boyd
420. Chiga C, M, Rai T, Yang SS, et al.A.Dietary
Naray-Fejes-Toth Gene salt regulatesof
regulation theENaC
phosphorylation
subunits by o
323. Kemendy
393. Reichold M, AE,Zdebik
Kleyman AA, Lieberer
TR, Eaton E, etDC. al. KCNJ10
Aldosterone genealters
mutationsthe opencaus- OSR1/SPAK
serum- and kinases and the sodium chloride
glucocorticoid-inducible cotransporter
kinase-1. through
Am J Physiol aldosterone.
Renal Physiol.
ing EAST syndrome
probability (epilepsy, ataxia,
of amiloride-blockable sodiumsensorineural
channels indea A6ness, and tubulopathy)
epithelia. Am J Physiol. Kidney Int. 2008;74:1403.
2005;288:F505.
disrupt channel unction. Proc Natl Acad Sci USA. 2010;107:14490.
1992;263:C825. 347. Reif
421. Vallon MC, V, Troutman
Schroth J, SL, Lang F, et al.JA.Expression
Schafer and phosphorylation
Sodium transport by rat corticalo col- the
324. Kleyman
394. Pacheco-Alvarez D, Cristobal PS,B,Meade
TR, Coupaye-Gerard ErnstP,SA. et al.Aldosterone
The Na+ :Cldoes -
cotransporter
not alter Na+ -Cl-tubule.
lecting cotransporter
Effects of NCC in vivo is and
vasopressin regulated by dietary salt, potassium,
desoxycorticosterone. and
J Clin Invest.
is activated
apical and phosphorylated
cell-surface expression of atepithelial
the amino-terminal
Na channels domain
in theupon intracellular
amphibian cell SGK1. Am J Physiol Renal Physiol. 2009;297:F704.
1986;77:1291.
chloride
line A6. Jdepletion.
Biol Chem.J1992;267:9622.
Biol Chem. 2006;281:28755. 348. Schlatter
422. Chen Z, Vaughn E, Schafer DA, JA.Fanestil DD. Inf uence ostudies
Electrophysiological genderinonprincipal
renal thiazide
cells
http://www.ncbi.nlm.nih.gov/pubmed/16887815
395. Chen SY, Bhargava A, Mastroberardino L, et al. Epithelial sodium chan- diuretic
of receptor
rat cortical density and
collecting response.
tubules. ADHJ Am Soc Nephrol.
increases 1994;5:1112.
the apical membrane Na -
325.regulated
nel Wiederholt M, Hansen L, Giebisch
by aldosterone-induced G. Dependence
protein sgk. Proc Natl o distal
Acadperitubular
Sci USA. http://www.ncbi.nlm.nih.gov/pubmed/7849251
conductance. P ugers Arch. 1987;409:81.
membrane potential on luminal sodium and peritubular potassium. Pf ugers
1999;96:2514. 349. Schafer
423. Verlander JA,JW, Tran TM,SL.Zhang
Troutman cAMPL,mediates
et al. Estradiol enhances
the increase thiazide-sen-
in apical mem-
Arch. 1975;355:R55.
396. Alvarez de la Rosa D, Coric T, Todorovic N, et al. Distribution and regu- sitive NaCl
brane Na cotransporter
conductance produced density in in theratapical
CCDplasma membraneAm
by vasopressin. o the distal
J Physiol.
326. Costanzo
lation of expression LS. Comparison
of serum- and o calcium and sodium transport
glucocorticoid-induced kinase-1in inearly the andrat convoluted tubule in ovariectomized rats. J Clin Invest. 1998;101:1661.
1990;259:F823.
late rat distal
kidney. tubules:
J Physiol. e ect o amiloride. Am J Physiol. 1984;246:F937.
2003;551:455. http://www.ncbi.nlm.nih.gov/pubmed/9541496
424. Marunaka Y, Eaton DC. Effects of vasopressin and cAMP on single
327. Helms
397. Lo ngMN, J, Pietri L, Aregger
Fejes-Toth F, et al. Di erential
G, Naray-Fejes-Toth subcellular localization
A. Hormone-regulated trans- o 350. Pedersen NB, Na
amiloride-blockable Hochannels.
meister MV Am, Rosenbaek LL, et al. Vasopressin induces
J Physiol. 1991;260:C1071.
ENaC subunits
epithelial in mouse inkidney
Na transport in response
mammalian CCD to high-
cells and low-Na
requires diets. Am J
SGK1 expression. phosphorylation
425. Ismailov, II,o McDuf the thiazide-sensitive
e JH, Benos DJ. sodium
Protein chloride
kinase cotransporter
A phosphorylation in the
Physiol
Am RenalRenal
J Physiol Physiol. 2000;279:F252.
Physiol. 2003;284:F480. distalGconvoluted
and tubule. Kidney
protein regulation Int. ed
of puri 2010;78:160.
renal Na channels in planar bilayer
328. Wulff
398. Schmitt R, Ellison
P, Vallon DH, Farman
V, Huang DY, et al. N, Impaired
et al. Developmental
renal Na( )expression
retention in o theso- http://www.ncbi.nlm.nih.gov/pubmed/20445498
membranes. J Biol Chem. 1994;269:10235.
dium entry pathways
sgk1-knockout mouse.inJ ratClinnephron. Am J Physiol. 1999;276:F367.
Invest. 2002;110:1263. 351. Djelidi
426. Mutig K, S, Saritas
Fay M,T,Cluzeaud
Uchida S,F,etetal.al.Short-term
Transcriptionalstimulation o theofthiazide-
regulation sodium
329. Alvarez
399. Biner HL, de Arpin-Bott
la Rosa D, ZhangMP, Lo P, ng J, et al. HumanA,cortical
Naray-Fejes-Toth distal
et al. The nephron:
serum and sensitive Na+
transport -Cl- cotransporter
by vasopressin by vasopressin
in renal cells. J Biol Chem.involves phosphorylation and
1997;272:32919.
distribution o kinase
glucocorticoid electrolyte and water
sgk increases transport pathways.
the abundance of epithelialJ sodium
Am Socchannels
Nephrol. membrane
427. Proudtranslocation.
D, Nakamura AmS,J Physiol
CaroneRenal FA, Physiol. 2010;298:F502. and renin-
et al. Kallikrein-kinin
2002;13:836.
in the plasma membrane of Xenopus oocytes. J Biol Chem. 1999;274:37834. 352. Nedvetsky
angiotensin systems PI, inTamma
rat renalG, lymph.
BeulshausenKidneyS,Int.
et al. Regulation o aquaporin-2
1984;25:880.
http://www.ncbi.nlm.nih.gov/pubmed/11912242
400. Alvarez de la Rosa D, Paunescu TG, Els WJ, et al. Mechanisms of regulation tra cking.
428. Figueroa Handb CD,Exp Pharmacol.
Gonzalez CB, 2009:133.
Grigoriev S, et al. Probing for the bradykinin
330.
of Ellisonsodium
epithelial DH, Velazquez
channel by H,SGK1
Wright FS. cells.
in A6 Adaptation o the 2004;124:395.
J Gen Physiol. distal convolut- 353.
B2 SnyderinPM,
receptor Olson DR,
rat kidney Kabra R, et al.
by anti-peptide cAMP
and and serum
anti-ligand and glucocorticoid-
antibodies. J Histochem
ed tubule
401. Snydero thePM,rat. Structural
Olson and unctional
DR, Thomas BC. Serum e ects
and oglucocorticoid-regulated
dietary salt intake and inducible kinase
Cytochem. (SGK) regulate the epithelial Na(+) channel through convergent
1995;43:137.
chronicmodulates
kinase diuretic inNedd4-2-mediated
usion. J Clin Invest.inhibition
1989;83:113. of the epithelial Na channel. phosphorylation
429. Tomita K, Pisano o Nedd4-2. J Biol MA.
JJ, Knepper Chem. 2004;279:45753.
Control of sodium and potassium trans-
Jhttp://www.ncbi.nlm.nih.gov/pubmed/2910903
Biol Chem. 2002;277:5. 354. inWindhager
port the corticalEE, Giebischduct
collecting G. Micropuncture
of the rat. Effects study o renal tubular
of bradykinin, trans er
vasopressin,
331. Bhalla
402. Chen V ZF , Vaughn
, Daidie D, Li DA,
H, Beaumont
et al. Serum- K, andet al. E ects o diuretic treatment
glucocorticoid-regulated kinase o sodium
and chloride in the Jrat.
deoxycorticosterone. ClinAm J Physiol.
Invest. 1961;200:581.
1985;76:132.
1and o dietary
regulates sodiumligase
ubiquitin on renal
neural binding
precursoro 3H-metolazone.
cell-expressed, J developmentally
Am Soc Nephrol. http://www.ncbi.nlm.nih.gov/pubmed/13785775
430. Zaika O, Mamenko M, O’Neil RG, et al. Bradykinin acutely inhibits activity
1990;1:91.
down-regulated protein 4-2 by inducing interaction with 14-3-3. Mol Endocrinol. 355.
of the Boulpaep
epithelial EL. Na Recentchannel advances in electrophysiology
in mammalian o the nephron.
aldosterone-sensitive Annu
distal neph-
http://www.ncbi.nlm.nih.gov/pubmed/1966553
2005;19:3073. Rev Physiol.
ron. 1976;38:21.
Am J Physiol Renal Physiol. 2011;300:F1105.
332. Ichimura
403. Lo ng J,T,LoYamamura ng-Cueni H,D, Hegyi I,K,et etal.al.Thiazide
Sasamoto treatment
14-3-3 proteins o rats pro-
modulate the http://www.ncbi.nlm.nih.gov/pubmed/769655
431. Nonoguchi H, Knepper MA, Manganiello VC. Effects of atrial natriuretic
vokes apoptosis
expression in distalNa
of epithelial tubule cells. Kidney
channels Int. 1996;50:1180.
by phosphorylation-dependent interac- 356. Sansom
factor on cyclicSC, O’Neil RG
guanosine Mineralocorticoid
monophosphate and regulation o apicalmonophos-
cyclic adenosine cell mem-
http://www.ncbi.nlm.nih.gov/pubmed/8887276
tion with Nedd4-2 ubiquitin ligase. J Biol Chem. 2005;280:13187. brane accumulation
phate Na+ and K+intransport o thenephron
microdissected cortical segments
collectingfrom duct.
rats.Am J Physiol.
J Clin Invest.
333. Soundararajan
404. Fanestil DD, Vaughn R, Zhang DA,TT, Blakely
WangPJ, . Dietary
et al. ANaClnovelandroleKCl do not regulate
for glucocorticoid- 1985;248:F858.
1987;79:500.
renal density
induced leucineo the thiazide
zipper diuretic
protein receptor.sodium
in epithelial Am J Physiol. 1997;273:R1241.
channel-mediated sodium 357. Dillingham
432. Sansom SC, MA,Weinman
Anderson EJ, O’Neil RG.
RJ. Inhibition Microelectrode
of vasopressin actionassessment
by atrial
334. Knepper
transport. MA, Kim
J Biol Chem. GH, Masilamani S. Renal tubule sodium transporter
2005;280:39970. o chloride-conductive
natriuretic factor. Science.properties
1986;231:1572.o cortical collecting duct. Am J Physiol.
abundance
405. pro ling R,
Soundararajan in Wang
rat kidney: response
J, Melters D, et al. to Differential
aldosteroneactivities
and variations
of gluco- in 1984;247:F291.
433. Wang W, Li C, Nejsum LN, et al. Biphasic effects of ANP infusion in con-
NaCl intake. Ann NY
corticoid-induced Acadzipper
leucine Sci. 2003;986:562.
protein isoforms. J Biol Chem. 2007;282:36303. 358. Stokes
scious, euvolumicJB. Potassium
rats: rolessecretion
of AQP2byand cortical
ENaCcollecting
traf cking.tubule:
Am Jrelation
Physiol to so-
Renal
http://www.ncbi.nlm.nih.gov/pubmed/12763890
406. Soundararajan R, Pearce D, Ziera T. The role of the ENaC-regulatory complex dium absorption,
Physiol. 2006;290:F530. luminal sodium concentration, and transepithelial voltage. Am
335.
in Shimizu T, Yoshitomi
aldosterone-mediated sodium K, transport.
TaniguchiMol J, etCellal.Endocrinol.
E ect o 2011;350:242.
high NaCl intake J Physiol.
434. Rouch 1981;241:F395.
AJ, Chen L, Troutman SL, et al. Na transport in isolated rat CCD:
on Na+
407. and K+B,transport
Boldyreff Wehling M. in the rabbit distal
Non-genomic convoluted
actions tubule. Pf
of aldosterone: ugers Arch.
mechanisms 359. O’Neil
effects RG, Sansom
of bradykinin, ANP SC. Characterization
, clonidine, o apical cell membrane
and hydrochlorothiazide. Am JNa+ and
Physiol.
1989;414:500.
and consequences in kidney cells. Nephrol Dial Transplant. 2003;18:1693. K+ conductances o cortical collecting duct using microelectrode techniques. Am
1991;260:F86.
http://www.ncbi.nlm.nih.gov/pubmed/2550887
408. Stockand JD, Edinger RS, Eaton DC, et al. Toward understanding the J Physiol.
435. Gudme 1984;247:F14.
CN, Larsen AL, Nielsen R. Alpha2-adrenoceptors inhibit antidi-
336.ofFarman
role methylationN, Bonvalet JP. Aldosterone binding
in aldosterone-sensitive Na( )intransport.
isolated tubules. III. Auto-
News Physiol Sci. 360. Sansom
uretic SC, Agulian S,Na
hormone-stimulated Mutoabsorption
S, et al. K activity o CCD
across tight principal
epithelia cellsescu-
(Rana rom
radiography along the rat nephron. Am J Physiol. 1983;245:F606.
2000;15:161. normalPand
lenta). DOCA-treated
ugers rabbits. Am J Physiol. 1989;256:F136.
Arch. 2001;442:346.
337. Rokaw
409. FarmanMD, N, Oblin
WangME, JM, Lombes
Edinger M, RS,etetal.al.Immunolocalization
Carboxylmethylation o ofgluco-
the betaand 361. Stokes
436. Garty H, JB,Palmer
KokkoLG. Epithelial sodium
JP. Inhibition of sodium channels:
transport unction, structure, and
by prostaglandin E2
mineralocorticoid
subunit receptors channel
of xENaC regulates in rabbitactivity.
kidney.JAm BiolJChem.
Physiol.1998;273:28746.
1991;260:C226. regulation.
across Physiol Rev.
the isolated, 1997;77:359.
perfused rabbit collecting tubule. J Clin Invest. 1977;59:1099.
338. Stockand
410. El Mernissi JD,G,SpierDoucetBJ, A. Speci RT,
Worrell c activity o Na-K-ATPase
et al. Regulation of Na(a ter adre-
) reab- http://www.ncbi.nlm.nih.gov/pubmed/9114818
437. Hebert RL, Jacobson HR, Breyer MD. Prostaglandin E2 inhibits sodium
nalectomybyand
sorption thehormone replacement small
aldosterone-induced along G theprotein
rabbit nephron.
K-Ras2A. Pf ugersChem.
J Biol Arch. 362. Canessa
transport in rabbitCM, cortical
Horisberger JD, Rossier
collecting duct by BC. Epithelialintracellular
increasing sodium channel calcium. re-
1984;402:258.
1999;274:35449. Jlated
Clin to proteins
Invest. involved in neurodegeneration. Nature. 1993;361:467.
1991;87:1992.
http://www.ncbi.nlm.nih.gov/pubmed/6097867 http://www.ncbi.nlm.nih.gov/pubmed/8381523
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1193
93

363. Guan
438. Canessa CM, Schild
Y, Zhang L, Buell
Y, Breyer RM,G, et etal.al.Prostaglandin
Amiloride-sensitive epithelial
E2 inhibits renal Na+
col- 387. Pearce WL,
459. Clapp D, Yamamoto
Madsen KM, KR.Verlander
Mineralocorticoid
JW, et al.and glucocorticoid
Morphologic receptor
heterogeneity
channelduct
lecting is made Na o three homologous
absorption subunits.the
by activating Nature.
EP1 1994;367:463.
receptor. J Clin Invest. activities
along thedistinguished
rat inner medullaryby nonreceptor
collecting actors
duct. Labat aInvest.
composite response element.
1989;60:219.
http://www.ncbi.nlm.nih.gov/pubmed/8107805
1998;102:194. Science.
460. Sands1993;259:1161.
JM, Nonoguchi H, Knepper MA. Vasopressin effects on urea and
364. Warden
439. Palmer LG, DH, Frindt
Stokes G. JB. Amiloride-sensitive
EGF and PGE2 inhibit Na rabbit
channels CCDrom Na thetrans-
api- 388. Bonvalet JP
H2O transport in, Doignon I, Blot-Chabaud
inner medullary collecting M,ductet al.subsegments.
Distribution oAm 11Jbeta-hy-
Physiol.
cal membrane
port by differento mechanisms:
the rat cortical PGE2 collecting
inhibits tubule.
Na( )-K Procpump.
Natl AcadAm JSci USA.
Physiol. droxysteroid
1987;253:F823. dehydrogenase along the rabbit nephron. J Clin Invest. 1990;86:832.
1986;83:2767.
1993;264:F670. 389. Naray-Fejes-Toth
461. Stanton A, Watlington
BA. Characterization CO, Fejes-Toth
of apical and basolateral G. 11membrane
beta-Hydroxyster-
conduc-
365. Sakairi
440. CanessaY,CM, MerillatHR,
Jacobson AM, RossierTD,
Noland BC.etMembrane
al. Luminal topology o the epithe-
prostaglandin E re- oid dehydrogenase
tances activity in collecting
of rat inner medullary the renal target
duct. Am cellsJ Physiol.
o aldosterone. Endocrinology.
1989;256:F862.
lial sodium
ceptors channel
regulate salt in
andintact
water cells. Am J Physiol.
transport in rabbit1994;267:C1682.
cortical collecting duct. Am J 1991;129:17.
462. Imai M, Yoshitomi K. Electrophysiological study of inner medullary
366. Snyder
Physiol. PM, McDonald FJ, Stokes JB, et al. Membrane topology o the
1995;269:F257. http://www.ncbi.nlm.nih.gov/pubmed/2055180
collecting duct of hamsters. P ugers Arch. 1990;416:180.
amiloride-sensitive
441. Breyer MD, Jacobson epithelialHR,sodium
Breyerchannel. J Biol Chem.
JA. Epidermal growth 1994;269:24379.
factor inhibits the 390. Mune T, Rogerson
463. Sonnenberg H, HonrathFM, Nikkila
U, Wilson H, DR.
et al.Effects
Human hypertension
of amiloride in thecaused
medul- by
http://www.ncbi.nlm.nih.gov/pubmed/7929098
hydroosmotic effect of vasopressin in the isolated perfused rabbit cortical collect- mutations in the
lary collecting ductkidney
of ratisozyme
kidney. oKidney
11 beta-hydroxysteroid
Int. 1987;31:1121. dehydrogenase. Nat
367.tubule.
ing Jasti J,J Clin
Furukawa
Invest. H, Gonzales EB, et al. Structure o acid-sensing ion chan-
1988;82:1313. Genet. 1995;10:394.
464. Sands JM, Nonoguchi H, Knepper MA. Hormone effects on NaCl perme-
nel 1 at
442. 1.9 A
Muto S, resolution
Furuya H, and Tabei lowK,pH.
et al.Nature.
Site and2007;449:316.
mechanism of action of epider- http://www.ncbi.nlm.nih.gov/pubmed/7670488
ability of rat inner medullary collecting duct. Am J Physiol. 1988;255:F421.
http://www.ncbi.nlm.nih.gov/pubmed/17882215
mal growth factor in rabbit cortical collecting duct. Am J Physiol. 1991;260:F163. 391. StantonDB,
465. Light B, Janzen
McCann A, Klein-Robbenhaar
FV, Keller TM, etG,al. et al. Ultrastructure o rat
Amiloride-sensitive ini-
cation
368. Levchenko
443. Hughey RP,V,Bruns JB, Kinlough
Zheleznova NN, PavlovCL, etTS,al.etEpithelial
al. EGF and sodium channels
its related are
growth tial collecting
channel tubule.
in apical E ect oof adrenal
membrane corticosteroid
inner medullary treatment.
collecting duct. AmJ Clin Invest.
J Physiol.
activated
factors by urin-dependent
mediate sodium transport proteolysis. J Biol Chem.
in mpkCCDc14 2004;279:18111.
cells via ErbB2 (neu/HER-2) 1985;75:1327.
1988;255:F278.
369. Hughey
receptor. J Cell RP , Mueller
Physiol. GM, Bruns JB, et al. Maturation o the epithelial Na+
2010;223:252. http://www.ncbi.nlm.nih.gov/pubmed/2985657
466. Vandorpe DH, Ciampolillo F, Green RB, et al. Cyclic nucleotide-gated
channel
444. involves
Kohan DE. proteolytic
Endothelinprocessing
and collectingo theductalpha- and gamma-subunits.
sodium and water transport. J Biol 392.
cation Palmer
channels LG,mediate
Li JH, Lindemann B, et al.by
sodium absorption Aldosterone control o the
IMCD (mIMCD-K2) density
cells. Am J
Chem. 2003;278:37073.
Contrib Nephrol. 2011;172:94. oPhysiol.
sodium channels in the toad urinary bladder. J Membr Biol. 1982;64:91.
1997;272:C901.
http://www.ncbi.nlm.nih.gov/pubmed/12871941
445. Stoos BA, Garcia NH, Garvin JL. Nitric oxide inhibits sodium reabsorption http://www.ncbi.nlm.nih.gov/pubmed/6276550
467. Wilson DR, Honrath U, Sonnenberg H. Furosemide action on collect-
370.
in the Kleyman TR, Carattino
isolated perfused cortical MD, Hugheyduct.
collecting RP. ENaC at the
J Am Soc cutting1995;6:89.
Nephrol. edge: regula- 393. Kemendy
ing ducts: effectAE, of Kleyman TR, Eaton
prostaglandin DC. Aldosterone
synthesis inhibition. alters
Am J the open 1983;
Physiol. prob-
tion o Sun
446. epithelial sodiumJA.
D, Schafer channels
Dopamineby proteases.
inhibitsJ Biol Chem. 2009;284:20447.
AVP-dependent Na trans- ability
244:F666.o amiloride-blockable sodium channels in A6 epithelia. Am J Physiol.
http://www.ncbi.nlm.nih.gov/pubmed/19401469
port and water permeability in rat CCD via a D4-like receptor. Am J Physiol. 1992;263:C825.
468. Wilson DR, Honrath U, Sonnenberg H. Thiazide diuretic effect on medul-
371. Carattino MD, Sheng S, Kleyman TR. Epithelial Na+ channels are activated
1996;271:F391. 394. Kleymanduct
lary collecting TR, Coupaye-Gerard
function in the rat. B, Kidney
Ernst SA. Int.Aldosterone
1983;23:711. does not alter api-
by laminar
447. Saito shear
O, Ando stress.
Y, JKusano
Biol Chem. E, et2004;279:4120.
al. Functional characterization of basola- cal
469.cell-sur
Zeidelace ML, expression
Kikeri D,oSilva epithelial
P, et al.Na+ channels
Atrial in thepeptides
natriuretic amphibian cellcon-
inhibit line
http://www.ncbi.nlm.nih.gov/pubmed/14625286
teral and luminal dopamine receptors in rabbit CCD. Am J Physiol Renal Physiol. A6. J Biolsodium
ductive Chem. 1992;267:9622.
uptake by rabbit inner medullary collecting duct cells. J Clin
372. Abi-Antoun T, Shi S, Tolino LA, et al. Second transmembrane domain
2001;281:F114. http://www.ncbi.nlm.nih.gov/pubmed/1315763
Invest. 1988;82:1067.
modulates
448. epithelial
Peti-Peterdi sodium channel
J, Warnock DG, Bellgating in response II
PD. Angiotensin to directly
shear stress. Am J
stimulates 395. Chen SY
470. Rocha AS,, Bhargava
Kudo LH. A, Factors
Mastroberardino
governingL,sodium et al. Epithelial
and chloridesodium chan-
transport
Physiol activity
ENaC Renal Physiol.
in the2011;300:F1089.
cortical collecting duct via AT(1) receptors. J Am Soc nel regulated
across the innerbymedullary
aldosterone-induced
collecting duct. protein
Kidneysgk. Proc Natl Acad Sci USA.
Int. 1990;38:654.
373. Morimoto
Nephrol. 2002;13:1131.T, Liu W Woda C, et al. Mechanism underlying f ow stimula- 1999;96:2514.
471. Delpire E, Rauchman MI, Beier DR, et al. Molecular cloning and chro-
tion oBeutler
449. sodiumKT, absorption
Masilamani in the mammalian
S, Turban collecting
S, et al. Long-term duct.regulation
Am J Physiol Renal
of ENaC http://www.ncbi.nlm.nih.gov/pubmed/10051674
mosome localization of a putative basolateral Na( )-K( )-2Cl- cotransporter
Physiol. 2006;291:F663.
expression in kidney by angiotensin II. Hypertension. 2003;41:1143. 396. Alvarezinner
from mouse de la Rosa D, Coric
medullary T, Todorovic
collecting N, et al. Distribution
duct (mIMCD-3) andChem.
cells. J Biol regu-
374. Sun
450. Schild P, YL.
ue The
P, Wangepithelial sodium channel:
WH. Angiotensin rom molecule
II stimulates epithelialtosodium
disease. Rev
chan- lation o expression o serum- and glucocorticoid-induced kinase-1 in the rat
1994;269:25677.
Physiol
nels Biochem
(ENaC) in Pharmacol.
the cortical2004;151:93.
collecting duct of the rat kidney. Am J Physiol Renal kidney. J Physiol. 2003;551:455.
472. Praetorius J, Kim YH, Bouzinova EV, et al. NBCn1 is a basolateral Na -
http://www.ncbi.nlm.nih.gov/pubmed/15146350
Physiol. 2011 Dec 14 [Epub ahead of print]. http://www.ncbi.nlm.nih.gov/pubmed/12816971
HCO3- cotransporter in rat kidney inner medullary collecting ducts. Am J Physiol
375. Hallows
451. ShimketsKR, RA, Wang
Warnock H, DG, Bositis
Edinger RS,CM, et al.
et al. Liddle’s syndrome:
Regulation of epithelial heritable
Na 397. Helms MN,
Renal Physiol. Fejes-Toth G, Naray-Fejes-Toth A. Hormone-regulated trans-
2004;286:F903.
human hypertension
transport caused by
by soluble adenylyl mutations
cyclase in kidney in the beta subunit
collecting o the
duct cells. epithelial
J Biol Chem. epithelial
473. Zeidel Na+ML. transport in mammalian
Renal actions of atrial CCD cells requires
natriuretic peptide:SGK1 expression.
regulation Am
of collect-
sodium channel. Cell. 1994;79:407.
2009;284:5774. Jing
Physiol
duct Renal
sodium Physiol. 2003;284:F480.
and water transport. Annu Rev Physiol. 1990;52:747.
http://www.ncbi.nlm.nih.gov/pubmed/7954808
452. Pech V, Pham TD, Hong S, et al. Pendrin modulates ENaC function by 398. Wul PML,
474. Zeidel , VallonSilvaV P
Huang
, BrennerDY, BM,
et al.etImpaired
al. cGMP renal Na(+) retention
mediates effects of inatrial
the
376. Hansson
changing luminal JH,HCO3.
Nelson-Williams C, Suzuki
J Am Soc Nephrol. H, et al. Hypertension caused by
2010;21:1928. sgk1-knockout
peptides on medullarymouse. collecting
J Clin Invest. 2002;110:1263.
duct cells. Am J Physiol. 1987;252:F551.
a truncated
453. Edinger epithelial sodium
RS, Lebowitz J, channel
Li H, et al.gamma subunit:
Functional genetic heterogeneity
regulation of the epithelialo 399. Alvarez
475. Light DB,deSchwiebert
la Rosa D, EM, Zhang P, Naray-Fejes-Toth
Karlson KH, et al. AtrialA,natriuretic
et al. The peptide
serum and in-
Liddlechannel
Na syndrome. Nat Genet.
by IkappaB 1995;11:76.
kinase-beta occurs via phosphorylation of the ubiqui- glucocorticoid
hibits a cation kinase
channel sgkinincreases the abundance
renal inner o epithelial
medullary collecting sodium
duct cells.channels
Science.
http://www.ncbi.nlm.nih.gov/pubmed/7550319
tin ligase Nedd4-2. J Biol Chem. 2009;284:150. in the plasma membrane o Xenopus oocytes. J Biol Chem. 1999;274:37834.
1989;243:383.
377. Carattino
454. Tamura H,MD, Schild L, Enomoto
Edinger N, et al.
RS, Grieser HJ,Liddle
et al.disease caused
Epithelial by a missense
sodium channel http://www.ncbi.nlm.nih.gov/pubmed/10608847
476. Rocha AS, Kudo LH. Atrial peptide and cGMP effects on NaCl transport in
mutation obybeta
inhibition subunit o the
AMP-activated epithelial
protein kinase sodium channeland
in oocytes gene. J Clin Invest.
polarized renal 400. Alvarez de collecting
inner medullary la Rosa D, duct.
Paunescu
Am JTG, Els WJ,
Physiol. et al. Mechanisms o regulation
1990;259:F258.
1996;97:1780.
epithelial cells. J Biol Chem. 2005;280:17608. o477.
epithelial
Kudo LH, sodium van channel
Baak AA,byRochaSGK1AS. in A6 cells.
Effect of Jvasopressin
Gen Physiol.on 2004;124:395.
sodium trans-
http://www.ncbi.nlm.nih.gov/pubmed/8601645
455. Bhalla V, Hallows KR. Mechanisms of ENaC regulation and clinical impli- http://www.ncbi.nlm.nih.gov/pubmed/15452200
port across inner medullary collecting duct. Am J Physiol. 1990;258:F1438.
378. Snyder
cations. J Am Soc PM,Nephrol.
Steines2008;19:1845.
JC, Olson DR. Relative contribution o Nedd4 and 401. Snyder KJ,
478. Ullrich PM,Papavassiliou
Olson DR, Thomas
F. Sodium BC. Serum andinglucocorticoid-regulated
reabsorption the papillary collecting
Nedd4-2
456. to ENaC
Koeppen BM.regulation
Conductive in properties
epithelia determined
of the rabbitby RNA
outer inter erence.
medullary J Biol
collecting kinase
duct ofmodulates
rats. EffectNedd4-2-mediated
of adrenalectomy, low inhibition
Na diet, o the epithelial Na+
acetazolamide, channel. J
HCO-3-free
Chem.outer
duct: 2004;279:5042.
stripe. Am J Physiol. 1986;250:F70. Biol Chem.and
solutions 2002;277:5.
of amiloride. P ugers Arch. 1979;379:49.
http://www.ncbi.nlm.nih.gov/pubmed/14645220
457. Stokes JB. Na and K transport across the cortical and outer medullary col- http://www.ncbi.nlm.nih.gov/pubmed/11696533
479. Husted RF, Stokes JB. Separate regulation of Na and anion transport by
379. Chang
lecting tubuleSS, of Grunder
the rabbit: S, evidence
Hanukoglu for A, et al. Mutations
diffusion across theinoutersubunits o the
medullary 402.
IMCD:Bhalla V, Daidie
location, D, Li H,hypertonicity,
aldosterone, et al. Serum- TGF-beta
and glucocorticoid-regulated
1, and cAMP. Am J Physiol. kinase
epithelialAm
portion. sodium channel
J Physiol. cause salt wasting with hyperkalaemic acidosis, pseu-
1982;242:F514. 11996;271:F433.
regulates ubiquitin ligase neural precursor cell-expressed, developmentally
dohypoaldosteronism
458. Koeppen BM. Conductive type 1. Natproperties
Genet. 1996;12:248.
of the rabbit outer medullary collect- down-regulated
480. Terada Y, protein Knepper4-2 MA.by inducing
Na -K interaction with 14-3-3.
-ATPase activities Moltubule
in renal Endocrinol.
seg-
http://www.ncbi.nlm.nih.gov/pubmed/8589714
ing duct: inner stripe. Am J Physiol. 1985;248:F500. 2005;19:3073.
ments of rat inner medulla. Am J Physiol. 1989;256:F218.
380. Terada Y, Knepper MA. Thiazide-sensitive NaCl absorption in rat cortical 403. Ichimura T, Yamamura H, Sasamoto K, et al. 14-3-3 proteins modulate the
collecting duct. Am J Physiol. 1990;259:F519. expression o epithelial Na+ channels by phosphorylation-dependent interaction
381. Leviel F, Hubner CA, Houillier P, et al. The Na +-dependent chloride-bicar- with Nedd4-2 ubiquitin ligase. J Biol Chem. 2005;280:13187.
bonate exchanger SLC4A8 mediates an electroneutral Na+ reabsorption process http://www.ncbi.nlm.nih.gov/pubmed/15677482
in the renal cortical collecting ducts o mice. J Clin Invest. 2010;120:1627. 404. Soundararajan R, Zhang TT, Wang J, et al. A novel role or glucocorticoid-
382. Eladari D, Hubner CA. Novel mechanisms or NaCl reabsorption in the induced leucine zipper protein in epithelial sodium channel-mediated sodium
collecting duct. Curr Opin Nephrol Hypertens. 2011;20:506. transport. J Biol Chem. 2005;280:39970.
http://www.ncbi.nlm.nih.gov/pubmed/21610493 http://www.ncbi.nlm.nih.gov/pubmed/16216878
383. Tomaschitz A, Pilz S, Ritz E, et al. Aldosterone and arterial hypertension. 405. Soundararajan R, Wang J, Melters D, et al. Di erential activities o gluco-
Nat Rev Endocrinol. 2010;6:83. corticoid-induced leucine zipper protein iso orms. J Biol Chem. 2007;282:36303.
http://www.ncbi.nlm.nih.gov/pubmed/20027193 http://www.ncbi.nlm.nih.gov/pubmed/17956870
384. Wang WH, Giebisch G. Regulation o potassium (K) handling in the renal 406. Soundararajan R, Pearce D, Ziera T. The role o the ENaC-regulatory com-
collecting duct. Pf ugers Arch. 2009;458:157. plex in aldosterone-mediated sodium transport. Mol Cell Endocrinol. 2011;350:242.
http://www.ncbi.nlm.nih.gov/pubmed/18839206 407. Boldyre B, Wehling M. Non-genomic actions o aldosterone: mechanisms
385. Naray-Fejes-Toth A, Fejes-Toth G. Glucocorticoid receptors mediate and consequences in kidney cells. Nephrol Dial Transplant. 2003;18:1693.
mineralocorticoid-like e ects in cultured collecting duct cells. Am J Physiol. http://www.ncbi.nlm.nih.gov/pubmed/12937210
1990;259:F672. 408. Stockand JD, Edinger RS, Eaton DC, et al. Toward understanding the
386. Funder JW, Pearce PT, Smith R, et al. Mineralocorticoid action: target tis- role o methylation in aldosterone-sensitive Na(+) transport. News Physiol Sci.
sue speci city is enzyme, not receptor, mediated. Science. 1988;242:583. 2000;15:161.
http://www.ncbi.nlm.nih.gov/pubmed/2845584 http://www.ncbi.nlm.nih.gov/pubmed/11390901
(contd.)
194
1932
19 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

409. Tomaschitz
383. Rokaw MD,A,Wang Pilz S,JM,Ritz
Edinger RS,Aldosterone
E, et al. et al. Carboxylmethylation o the beta
and arterial hypertension. 411. Dillingham
432. Blazer-Y ost BL,MA, Anderson
Paunescu TG,RJ.Helman
InhibitionSI, etoal.vasopressin action 3-
Phosphoinositide bykinase
atrial
subunit
Nat o xENaC regulates
Rev Endocrinol. 2010;6:83. channel activity. J Biol Chem. 1998;273:28746. natriuretic
is required actor. for Science. 1986;231:1572.sodium reabsorption. Am J Physiol.
aldosterone-regulated
410. Wang
384. Stockand WH,JD, Spier G.
Giebisch BJ,Regulation
Worrell RT, et al. Regulation
of potassium (K) handling o Na(+) reab-
in the renal http://www.ncbi.nlm.nih.gov/pubmed/3006248
1999;277:C531.
sorption by
collecting duct.theP aldosterone-induced
ugers Arch. 2009;458:157. small G protein K-Ras2A. J Biol Chem. 433. Park
412. WangJ,W,Leong Li C, Nejsum
ML, Buse LN,P,etetal.al.Biphasic
Serum eand ectsglucocorticoid-inducible
o ANP in usion in con-
1999;274:35449.
385. Naray-Fejes-Toth A, Fejes-Toth G. Glucocorticoid receptors mediate scious, (SGK)
kinase euvolumic rats: of
is a target roles
theoPIAQP2 and ENaC tra signaling
3-kinase-stimulated f cking. Am J Physiol
pathway. EmboRenal
J.
http://www.ncbi.nlm.nih.gov/pubmed/10585415
mineralocorticoid-like effects in cultured collecting duct cells. Am J Physiol. Physiol. 2006;290:F530.
1999;18:3024.
411. Blazer-Yost BL, Paunescu TG, Helman SI, et al. Phosphoinositide 3-ki-
1990;259:F672. 434. Sansom
413. Rouch AJ, SC,Chen
O’Neil L,RG.
Troutman
Effects ofSL,mineralocorticoids
et al. Na+ transport on in isolatedproperties
transport rat CCD:
nase is
386. required
Funder or aldosterone-regulated
JW, Pearce sodium reabsorption.
PT, Smith R, et al. Mineralocorticoid action:Am J Physiol.
target tissue e ects
of o bradykinin,
cortical collecting duct ANP , clonidine,
basolateral and hydrochlorothiazide.
membrane. Am J Physiol.
Am J Physiol. 1986;251:F743.
1999;277:C531.
speci city is enzyme, not receptor, mediated. Science. 1988;242:583. 1991;260:F86.
414. Barlet-Bas C, Khadouri C, Marsy S, et al. Enhanced intracellular sodium
412. Pearce
387. Park J,D, Leong
YamamotoML, Buse KR.P,Mineralocorticoid
et al. Serum and and glucocorticoid-inducible
glucocorticoid receptor ki- 435. Gudme in
concentration CN, Larsen
kidney AL,recruits
cells Nielsen R. Alpha2-adrenoceptors
a latent pool of Na-K-ATPaseinhibit whose antidi-
size is
nase (SGK)
activities is a target obythe
distinguished PI 3-kinase-stimulated
nonreceptor factors at a composite signalingresponse
pathway.element.
Embo J. uretic hormone-stimulated
modulated by corticosteroids. Na+ absorption
J Biol across tight epithelia (Rana escu-
Chem. 1990;265:7799.
1999;18:3024.
Science. 1993;259:1161. lenta).VPf
415. ugers
errey Arch. 2001;442:346.
F, Kraehenbuhl JP, Rossier BC. Aldosterone induces a rapid increase
413. Bonvalet
388. Sansom JP SC, O’Neil I,RG.
, Doignon E ects o M,
Blot-Chabaud mineralocorticoids
et al. Distributiononof transport11 beta- http://www.ncbi.nlm.nih.gov/pubmed/11484764
in the rate of Na,K-ATPase gene transcription in cultured kidney cells. Mol Endo-
properties o dehydrogenase
hydroxysteroid cortical collecting alongduct basolateral
the rabbit nephron.membrane.
J Clin Invest.Am J Physiol.
1990;86:832. 436. Stokes
crinol. JB, Kokko JP. Inhibition o sodium transport by prostaglandin E2
1989;3:1369.
1986;251:F743.
389. Naray-Fejes-Toth A, Watlington CO, Fejes-Toth G. 11 beta-Hydroxysteroid acrossPalmer
416. the isolated, per usedL,rabbit
LG, Antonian Frindt collecting tubule.ofJ Clin
G. Regulation Invest.pump
the Na-K 1977;59:1099.
of the rat
414. Barlet-Basactivity
dehydrogenase C, Khadouriin theC,renal Marsy S, etcells
target al. Enhanced
of aldosterone. intracellular sodium
Endocrinology. 437. Hebert
cortical collectingRL, tubule
Jacobson by HR, Breyer MD.
aldosterone. J GenProstaglandin E2 inhibits sodium
Physiol. 1993;102:43.
concentration in kidney cells recruits a latent pool o Na-K-ATPase whose size is
1991;129:17. transport
417. Frindtin rabbit
G, Sackin cortical collecting
H, Palmer LG. duct by increasing
Whole-cell currentsintracellular
in rat cortical calcium.
collect-J
modulated
390. Muneby T, corticosteroids.
Rogerson FM, Nikkila J Biol Chem.H, et1990;265:7799.
al. Human hypertension caused by Clintubule:
ing Invest. 1991;87:1992.
low-Na diet increases amiloride-sensitive conductance. Am J Physiol.
http://www.ncbi.nlm.nih.gov/pubmed/2159458
mutations in the kidney isozyme of 11 beta-hydroxysteroid dehydrogenase. Nat 438. Guan Y, Zhang Y, Breyer RM, et al. Prostaglandin E2 inhibits renal collecting
1990;258:F562.
415. Verrey
Genet. F, Kraehenbuhl JP, Rossier BC. Aldosterone induces a rapid increase
1995;10:394. duct Na+
418. Pacha absorption
J, Frindt by G,activating
Antonian the EP1
L, et al. receptor.
Regulation J Clin Invest.
of Na 1998;102:194.
channels of the rat
in theStanton
391. rate o Na,K-ATPase
B, Janzen A, gene transcription inG,cultured
Klein-Robbenhaar kidney cells. of
et al. Ultrastructure MolratEndo-
ini- http://www.ncbi.nlm.nih.gov/pubmed/9649573
cortical collecting tubule by aldosterone. J Gen Physiol. 1993;102:25.
crinol.
tial 1989;3:1369.
collecting tubule. Effect of adrenal corticosteroid treatment. J Clin Invest. 439. May
419. Warden DH, A,
A, Puoti Stokes JB. EGF
Gaeggeler HP, etandal. PGE2 inhibitofrabbit
Early effect CCD on
aldosterone Na+thetrans-
rate
http://www.ncbi.nlm.nih.gov/pubmed/2558308
1985;75:1327. port
of by di of
synthesis erent mechanisms:
the epithelial sodium PGE2 inhibits
channel alpha Na(+)-K+
subunit in pump. Amcells.
A6 renal J Physiol.
J Am
416. Palmer
392. PalmerLG, LG,LiAntonian L, FrindtB,G.
JH, Lindemann Regulation
et al. Aldosterone o the Na-Kof
control pump o the rat
the density of 1993;264:F670.
Soc Nephrol. 1997;8:1813.
cortical channels
sodium collectingintubule the toadby aldosterone.
urinary bladder. J GenJ Membr
Physiol.Biol.
1993;102:43.
1982;64:91. 440. Boyd
420. SakairiC,Y, Naray-Fejes-Toth
Jacobson HR, Noland A. GeneTD, et al. Luminal
regulation prostaglandin
of ENaC subunitsE re- by
http://www.ncbi.nlm.nih.gov/pubmed/8397277
393. Kemendy AE, Kleyman TR, Eaton DC. Aldosterone alters the open ceptors regulate
serum- salt and water transportkinase-1.
and glucocorticoid-inducible in rabbit Am cortical collecting
J Physiol Renalduct. Am J
Physiol.
417. Frindt
probability ofG, Sackin H, Palmer LG.
amiloride-blockable sodiumWhole-cell
channelscurrents in rat cortical
in A6 epithelia. collect-
Am J Physiol. Physiol. 1995;269:F257.
2005;288:F505.
ing tubule: low-Na diet increases amiloride-sensitive conductance. Am J Physiol.
1992;263:C825. 441. Reif
421. Breyer MC, MD, JacobsonSL,
Troutman HR, BreyerJA.
Schafer JA.Sodium
Epidermal growthbyactor
transport inhibitscol-
rat cortical the
1990;258:F562.
394. Kleyman TR, Coupaye-Gerard B, Ernst SA. Aldosterone does not alter hydroosmotic
lecting tubule.e Effects
ect o vasopressin
of vasopressin in theand isolated per used rabbit cortical
desoxycorticosterone. J Clin collect-
Invest.
418. Pacha
apical J, Frindt
cell-surface G, Antonian
expression L, et al.Na
of epithelial Regulation
channels o Na channels
in the o thecell
amphibian rat ing tubule. J Clin Invest. 1988;82:1313.
1986;77:1291.
cortical
line A6. collecting
J Biol Chem. tubule by aldosterone. J Gen Physiol. 1993;102:25.
1992;267:9622. http://www.ncbi.nlm.nih.gov/pubmed/2844852
422. Schlatter E, Schafer JA. Electrophysiological studies in principal cells
http://www.ncbi.nlm.nih.gov/pubmed/8397276
395. Chen SY, Bhargava A, Mastroberardino L, et al. Epithelial sodium chan- 442.
of rat Muto
cortical S, Furuya
collecting H, Tabei
tubules. K, etADHal. Site and mechanism
increases the apicalomembrane
action o epider-
Na -
419.regulated
nel May A, Puoti A, Gaeggeler HP, et al.
by aldosterone-induced Early esgk.
protein ect Proc
o aldosterone
Natl AcadonScitheUSA. rate mal growth actor
conductance. in rabbit
P ugers Arch. cortical collecting duct. Am J Physiol. 1991;260:F163.
1987;409:81.
o synthesis o the epithelial sodium channel alpha subunit in A6 renal cells. J Am
1999;96:2514. 443. Schafer
423. Levchenko JA, VTroutman
, Zheleznova SL.NN, cAMPPavlov TS, et al.
mediates theEGF and itsinrelated
increase apical growth
mem-
Soc Nephrol.
396. Alvarez1997;8:1813.
de la Rosa D, Coric T, Todorovic N, et al. Distribution and regu- actorsNa
brane mediate sodium transport
conductance produced in in
mpkCCDc14
rat CCD bycells via ErbB2 Am
vasopressin. (neu/HER-2)
J Physiol.
420. Boyd
lation C, Naray-Fejes-Toth
of expression of serum- and A. glucocorticoid-induced
Gene regulation o ENaC subunits
kinase-1 by rat
in the se- receptor. J Cell Physiol. 2010;223:252.
1990;259:F823.
rum- and
kidney. glucocorticoid-inducible
J Physiol. 2003;551:455. kinase-1. Am J Physiol Renal Physiol. http://www.ncbi.nlm.nih.gov/pubmed/20049896
424. Marunaka Y, Eaton DC. Effects of vasopressin and cAMP on single
2005;288:F505.
397. Helms MN, Fejes-Toth G, Naray-Fejes-Toth A. Hormone-regulated trans- 444. Kohan DE. Endothelin
amiloride-blockable Na channels. and Am collecting
J Physiol.duct sodium and water transport.
1991;260:C1071.
421. Rei Na
epithelial MC, transport
Troutman inSL,mammalian
Scha er JA.CCD Sodium cellstransport
requires by SGK1rat cortical col-
expression. Contrib
425. Nephrol.II,
Ismailov, 2011;172:94.
McDuf e JH, Benos DJ. Protein kinase A phosphorylation
lecting
Am tubule.
J Physiol Renal E Physiol.
ects o 2003;284:F480.
vasopressin and desoxycorticosterone. J Clin Invest. http://www.ncbi.nlm.nih.gov/pubmed/21893992
and G protein regulation of puri ed renal Na channels in planar bilayer
1986;77:1291.
398. Wulff P, Vallon V, Huang DY, et al. Impaired renal Na( ) retention in the 445. Stoos BA,
membranes. J BiolGarcia
Chem.NH, Garvin JL. Nitric oxide inhibits sodium reabsorption
1994;269:10235.
http://www.ncbi.nlm.nih.gov/pubmed/2420830
sgk1-knockout mouse. J Clin Invest. 2002;110:1263. in theDjelidi
426. isolatedS,per FayusedM, cortical
Cluzeaud collecting
F, et al. duct. J Am Soc Nephrol.
Transcriptional regulation1995;6:89.
of sodium
422. Alvarez
399. SchlatterdeE,laScha RosaerD, JA.Zhang
Electrophysiological
P, Naray-Fejes-Toth studies A, inet principal cells oand
al. The serum rat http://www.ncbi.nlm.nih.gov/pubmed/7579075
transport by vasopressin in renal cells. J Biol Chem. 1997;272:32919.
cortical collecting
glucocorticoid kinasetubules. ADH increases
sgk increases the apical
the abundance membrane
of epithelial Na+ -conduct-
sodium channels 446. Proud
427. Sun D,D, Scha Nakamuraer JA. S,Dopamine
Carone FA, inhibits
et al. AVP-dependent
Kallikrein-kinin Na+ trans-
and renin-
ance.
in the Pf ugers membrane
plasma Arch. 1987;409:81.
of Xenopus oocytes. J Biol Chem. 1999;274:37834. port and water
angiotensin systemspermeability
in rat renal in lymph.
rat CCD via aInt.
Kidney D4-like receptor. Am J Physiol.
1984;25:880.
http://www.ncbi.nlm.nih.gov/pubmed/2441357
400. Alvarez de la Rosa D, Paunescu TG, Els WJ, et al. Mechanisms of regulation 1996;271:F391.
428. Figueroa CD, Gonzalez CB, Grigoriev S, et al. Probing for the bradykinin
423.
of Scha er
epithelial JA, Troutman
sodium channel by SL.SGK1cAMP in mediates
A6 cells. Jthe Genincrease in apical mem-
Physiol. 2004;124:395. 447.
B2 Saito in
receptor O, ratAndo
kidneyY, Kusano E, et al. and
by anti-peptide Functional characterization
anti-ligand o basola-
antibodies. J Histochem
brane Snyder
401. Na+ conductance
PM, Olson DR, produced
Thomas in BC.
rat Serum
CCD by andvasopressin. Am J Physiol.
glucocorticoid-regulated teral and luminal
Cytochem. 1995;43:137.dopamine receptors in rabbit CCD. Am J Physiol Renal Physiol.
1990;259:F823.
kinase modulates Nedd4-2-mediated inhibition of the epithelial Na channel. 2001;281:F114.
429. Tomita K, Pisano JJ, Knepper MA. Control of sodium and potassium trans-
J424. Marunaka
Biol Chem. Y, Eaton DC. E ects o vasopressin and cAMP on single ami-
2002;277:5. 448. inPeti-Peterdi
port the cortical J,collecting
Warnockduct DG,ofBell
the PD. Angiotensin
rat. Effects II directly
of bradykinin, stimulates
vasopressin,
loride-blockable
402. Bhalla V, Daidie Na channels.
D, Li H, Am et al.J Physiol.
Serum- 1991;260:C1071.
and glucocorticoid-regulated kinase ENaC
and activity in the cortical
deoxycorticosterone. collecting
J Clin duct via AT(1) receptors. J Am Soc Nephrol.
Invest. 1985;76:132.
1425. Ismailov,
regulates II, McDu
ubiquitin f e JH,
ligase Benos
neural DJ. Protein
precursor kinase A phosphorylation
cell-expressed, developmentally and 2002;13:1131.
430. Zaika O, Mamenko M, O’Neil RG, et al. Bradykinin acutely inhibits activity
G protein regulation
down-regulated proteino purif
4-2 by edinducing
renal Na+interaction
channels in planar
with bilayer
14-3-3. Molmembranes.
Endocrinol. 449.
of the Beutler
epithelial KT,NaMasilamani
channel S, in Turban
mammalian S, et aldosterone-sensitive
al. Long-term regulation o neph-
distal ENaC
J Biol Chem. 1994;269:10235.
2005;19:3073. expression
ron. in kidney
Am J Physiol RenalbyPhysiol.
angiotensin II. Hypertension. 2003;41:1143.
2011;300:F1105.
http://www.ncbi.nlm.nih.gov/pubmed/8144604
403. Ichimura T, Yamamura H, Sasamoto K, et al. 14-3-3 proteins modulate the http://www.ncbi.nlm.nih.gov/pubmed/12682079
431. Nonoguchi H, Knepper MA, Manganiello VC. Effects of atrial natriuretic
426. Djelidi
expression of S, Fay M, Na
epithelial Cluzeaud F, et by
channels al. Transcriptional
phosphorylation-dependent regulation o interac-
sodium 450. Sun
factor P, Yue guanosine
on cyclic P, Wang WH. Angiotensin II and
monophosphate stimulates
cyclic epithelial
adenosinesodium chan-
monophos-
transport
tion by vasopressin
with Nedd4-2 in renal
ubiquitin ligase.cells. J Biol
J Biol Chem. Chem. 1997;272:32919.
2005;280:13187. nels (ENaC)
phate in the cortical
accumulation collecting duct
in microdissected nephron o the rat kidney.
segments fromAm J Physiol
rats. Renal
J Clin Invest.
427. Soundararajan
404. Proud D, Nakamura R, Zhang S, Carone
TT, Wang FA, J,etetal.al.Kallikrein-kinin
A novel role forand renin-angio-
glucocorticoid- Physiol. 2011 Dec 14 [Epub ahead o print].
1987;79:500.
tensin systems
induced leucineinzipper
rat renal lymph.
protein in Kidney Int. sodium
epithelial 1984;25:880. channel-mediated sodium 451. Dillingham
432. Hallows KR,MA, Wang H, Edinger
Anderson RS, et al.ofRegulation
RJ. Inhibition vasopressino action
epithelial Na+
by atrial
428. Figueroa
transport. CD, Gonzalez
J Biol Chem. CB, Grigoriev S, et al. Probing or the bradykinin
2005;280:39970. transport by
natriuretic soluble
factor. adenylyl
Science. cyclase in kidney collecting duct cells. J Biol Chem.
1986;231:1572.
B2 receptor
405. in rat kidney
Soundararajan by anti-peptide
R, Wang J, Melters D, andet anti-ligand
al. Differential antibodies.
activitiesJ Histochem
of gluco- 2009;284:5774.
433. Wang W, Li C, Nejsum LN, et al. Biphasic effects of ANP infusion in con-
Cytochem. 1995;43:137.
corticoid-induced leucine zipper protein isoforms. J Biol Chem. 2007;282:36303. http://www.ncbi.nlm.nih.gov/pubmed/19126549
scious, euvolumic rats: roles of AQP2 and ENaC traf cking. Am J Physiol Renal
http://www.ncbi.nlm.nih.gov/pubmed/7822771
406. Soundararajan R, Pearce D, Ziera T. The role of the ENaC-regulatory complex 452. Pech
Physiol. V, Pham TD, Hong S, et al. Pendrin modulates ENaC unction by
2006;290:F530.
429.
in Tomita K, Pisano JJ,
aldosterone-mediated Knepper
sodium MA. Control
transport. Mol Cell o Endocrinol.
sodium and2011;350:242.
potassium trans- changing
434. Rouch luminal
AJ, ChenHCO3. J Am Soc Nephrol.
L, Troutman SL, et al.2010;21:1928.
Na transport in isolated rat CCD:
port inBoldyreff
407. the cortical collecting
B, Wehling M. duct o the rat.actions
Non-genomic E ectsofo aldosterone:
bradykinin, mechanisms
vasopressin, 453. Edinger
effects RS, Lebowitz
of bradykinin, ANP, J, Li H, et al.
clonidine, and Functional regulation o Am
hydrochlorothiazide. the Jepithelial
Physiol.
deoxycorticosterone.
and consequences in kidney J Clin Invest.
cells. 1985;76:132.
Nephrol Dial Transplant. 2003;18:1693. Na+ channel by IkappaB kinase-beta occurs via phosphorylation o the ubiqui-
1991;260:F86.
http://www.ncbi.nlm.nih.gov/pubmed/4019771
408. Stockand JD, Edinger RS, Eaton DC, et al. Toward understanding the tin ligase
435. Gudme Nedd4-2. J Biol Chem.
CN, Larsen 2009;284:150.
AL, Nielsen R. Alpha2-adrenoceptors inhibit antidi-
430.ofZaika
role O, Mamenko
methylation M, O’Neil RG, et al.Na(
in aldosterone-sensitive Bradykinin acutely
) transport. inhibits
News activity
Physiol Sci. http://www.ncbi.nlm.nih.gov/pubmed/18981174
uretic hormone-stimulated Na absorption across tight epithelia (Rana escu-
o the epithelial Na+ channel in mammalian aldosterone-sensitive distal neph-
2000;15:161. 454. Carattino
lenta). P ugers Arch. MD,2001;442:346.
Edinger RS, Grieser HJ, et al. Epithelial sodium channel
ron. Am
409. RokawJ Physiol
MD,RenalWangPhysiol. 2011;300:F1105.
JM, Edinger RS, et al. Carboxylmethylation of the beta inhibition
436. Stokes byJB,AMP-activated
Kokko JP. Inhibitionprotein kinase
of sodium in oocytes andby
transport polarized renal epi-
prostaglandin E2
431. Nonoguchi
subunit of xENaC H, Knepper
regulates MA, Manganiello
channel activity. J BiolVC. Chem. E ects o atrial natriuretic
1998;273:28746. thelial the
across cells. J Biol Chem.
isolated, perfused2005;280:17608.
rabbit collecting tubule. J Clin Invest. 1977;59:1099.
actor Stockand
410. on cyclic JD, guanosine
Spier BJ, monophosphate
Worrell RT, etand al. cyclic
Regulationadenosineof Na( monophos-
) reab- http://www.ncbi.nlm.nih.gov/pubmed/15753079
437. Hebert RL, Jacobson HR, Breyer MD. Prostaglandin E2 inhibits sodium
phate accumulation
sorption in microdissected nephron
by the aldosterone-induced small G segments
protein K-Ras2A. rom rats.JJBiolClin Chem.
Invest. 455. Bhalla
transport V, Hallows
in rabbit KR. collecting
cortical Mechanisms duct o by
ENaC regulation
increasing and clinical
intracellular impli-
calcium.
1987;79:500.
1999;274:35449. Jcations. J Am1991;87:1992.
Clin Invest. Soc Nephrol. 2008;19:1845.
http://www.ncbi.nlm.nih.gov/pubmed/3027127 http://www.ncbi.nlm.nih.gov/pubmed/18753254
(contd.)
CHAPTER 5 TUBULAR SODIUM TRANSPORT 1195
193
93

456. Guan
438. Koeppen BM. Conductive
Y, Zhang Y, Breyer RM, properties
et al. of the rabbit outer
Prostaglandin medullary
E2 inhibits collect-
renal col- 470. Rocha WL,
459. Clapp AS, Kudo
Madsen LH.KM,
Factors governing
Verlander JW, etsodium and chloride
al. Morphologic transport
heterogeneity
ing duct:duct
lecting outerNastripe. Am J Physiol.
absorption by 1986;250:F70.
activating the EP1 receptor. J Clin Invest. across
along thethe rat
inner medullary
inner medullary collecting
collecting duct. Kidney
duct. Int. 1990;38:654.
Lab Invest. 1989;60:219.
457. Stokes JB. Na and K transport across the cortical and outer medullary col-
1998;102:194. http://www.ncbi.nlm.nih.gov/pubmed/2146432
460. Sands JM, Nonoguchi H, Knepper MA. Vasopressin effects on urea and
lectingWarden
439. tubule DH,of theStokes
rabbit:JB.evidence
EGF andforPGE2 diffusion
inhibitacross theCCD
rabbit outerNamedullary
trans- 471. Delpire E,inRauchman
H2O transport MI, Beier
inner medullary DR, et duct
collecting al. Molecular cloning
subsegments. Amand chro-
J Physiol.
portion.
port Am J Physiol.
by different 1982;242:F514.
mechanisms: PGE2 inhibits Na( )-K pump. Am J Physiol. mosome localization of a putative basolateral Na(+)-K(+)-2Cl- cotransporter
1987;253:F823.
458. Koeppen BM. Conductive properties of the rabbit outer medullary collect-
1993;264:F670. from
461. mouse
Stanton inner medullary collecting
BA. Characterization of apical duct
and(mIMCD-3) cells. J Biol
basolateral membrane Chem.
conduc-
ing duct:
440. innerY,stripe.
Sakairi JacobsonAm JHR,Physiol.
Noland1985;248:F500.
TD, et al. Luminal prostaglandin E re- 1994;269:25677.
tances of rat inner medullary collecting duct. Am J Physiol. 1989;256:F862.
459. Clapp
ceptors regulateWL,salt
Madsen KM, transport
and water VerlanderinJW, et al.cortical
rabbit Morphologic
collectingheterogeneity
duct. Am J http://www.ncbi.nlm.nih.gov/pubmed/7929272
462. Imai M, Yoshitomi K. Electrophysiological study of inner medullary
along the
Physiol. rat inner medullary collecting duct. Lab Invest. 1989;60:219.
1995;269:F257. 472. Praetorius
collecting duct ofJ,hamsters.
Kim YH, PBouzinova
ugers Arch. EV1990;416:180.
, et al. NBCn1 is a basolateral Na+
http://www.ncbi.nlm.nih.gov/pubmed/2915516
441. Breyer MD, Jacobson HR, Breyer JA. Epidermal growth factor inhibits the -HCO3- cotransporter
463. Sonnenberg in rat kidney
H, Honrath inner DR.
U, Wilson medullary
Effectscollecting
of amilorideducts. Ammedul-
in the J Phys-
460. Sands JM,
hydroosmotic Nonoguchi
effect H, Knepper
of vasopressin MA. Vasopressin
in the isolated effects
perfused rabbit on urea
cortical and
collect- iol
laryRenal Physiol.
collecting 2004;286:F903.
duct of rat kidney. Kidney Int. 1987;31:1121.
H2Otubule.
ing transport
J ClininInvest.
inner1988;82:1313.
medullary collecting duct subsegments. Am J Physiol. 473. Zeidel JM,
464. Sands ML.Nonoguchi
Renal actionsH, of atrial natriuretic
Knepper MA. Hormone peptide: regulation
effects on NaCl of collect-
perme-
1987;253:F823.
442. Muto S, Furuya H, Tabei K, et al. Site and mechanism of action of epider- ing duct
ability ofsodium
rat innerand water transport.
medullary collectingAnnuduct.RevAmPhysiol. 1990;52:747.
J Physiol. 1988;255:F421.
461.growth
mal Stanton BA.inCharacterization
factor of apical and
rabbit cortical collecting duct.basolateral membrane
Am J Physiol. conduc-
1991;260:F163. http://www.ncbi.nlm.nih.gov/pubmed/2139559
465. Light DB, McCann FV, Keller TM, et al. Amiloride-sensitive cation
tancesLevchenko
443. of rat innerV,medullary
Zheleznova collecting
NN, Pavlovduct.TS,AmetJal.
Physiol.
EGF and1989;256:F862.
its related growth 474.
channel Zeidel ML, Silva
in apical P, Brenner
membrane BM, etmedullary
of inner al. cGMP collecting
mediates effects
duct. Amof atrial pep-
J Physiol.
462. Imai
factors M, Ysodium
mediate oshitomitransport
K. Electrophysiological
in mpkCCDc14study of inner
cells via ErbB2medullary
(neu/HER-2)col- tides on medullary collecting duct cells. Am J Physiol. 1987;252:F551.
1988;255:F278.
lecting duct
receptor. of Physiol.
J Cell hamsters. Pf ugers Arch. 1990;416:180.
2010;223:252. 475.
466. V Light DB, Schwiebert
andorpe DH, CiampolilloEM, Karlson
F, Green KH,RB,et al.
et Atrial natriuretic
al. Cyclic peptide in-
nucleotide-gated
http://www.ncbi.nlm.nih.gov/pubmed/2162031
444. Kohan DE. Endothelin and collecting duct sodium and water transport. hibits
cation achannels
cation channel
mediateinsodium
renal inner medullary
absorption by IMCDcollecting duct cells.
(mIMCD-K2) Science.
cells. Am J
463. Sonnenberg
Contrib H, Honrath U, Wilson DR. Effects of amiloride in the medul-
Nephrol. 2011;172:94. 1989;243:383.
Physiol. 1997;272:C901.
lary collecting
445. Stoos BA,duct Garciaof rat
NH,kidney.
GarvinKidney Int. 1987;31:1121.
JL. Nitric oxide inhibits sodium reabsorption http://www.ncbi.nlm.nih.gov/pubmed/2463673
467. Wilson DR, Honrath U, Sonnenberg H. Furosemide action on collect-
464.
in the Sands
isolatedJM, Nonoguchi
perfused H, collecting
cortical Knepper MA. duct.Hormone
J Am Soc effects
Nephrol. on1995;6:89.
NaCl perme- 476. Rochaeffect
ing ducts: AS, Kudo LH. Atrial peptide
of prostaglandin and cGMP
synthesis effects Am
inhibition. on NaCl transport
J Physiol. 1983;in
abilitySun
446. of ratD,inner medullary
Schafer collecting duct.
JA. Dopamine Am JAVP-dependent
inhibits Physiol. 1988;255:F421.
Na trans- inner medullary collecting duct. Am J Physiol. 1990;259:F258.
244:F666.
465. and
port Light DB,permeability
water McCann FV in, Keller
rat CCD TM,via et al. Amiloride-sensitive
a D4-like cation
receptor. Am J Physiol. 477. Kudo LH,
468. Wilson DR,van Baak AA,
Honrath Rocha AS. Effect
U, Sonnenberg of vasopressin
H. Thiazide diureticon sodium
effect trans-
on medul-
channel in apical membrane of inner medullary collecting duct. Am J Physiol.
1996;271:F391. port across inner
lary collecting medullary
duct functioncollecting
in the rat.duct.
KidneyAmInt.
J Physiol. 1990;258:F1438.
1983;23:711.
1988;255:F278.
447. Saito O, Ando Y, Kusano E, et al. Functional characterization of basola- 478. UllrichML,
469. Zeidel KJ, Papavassiliou
Kikeri D, Silva F. P
Sodium reabsorption
, et al. Atrial in the
natriuretic papillary
peptides collecting
inhibit con-
466. and
teral Vandorpe
luminalDH, Ciampolillo
dopamine receptors F, Green
in rabbitRB,CCD.
et al.Am Cyclic nucleotide-gated
J Physiol Renal Physiol. duct
ductiveof rats.
sodiumEffect of adrenalectomy,
uptake by rabbit inner lowmedullary
Na+ diet, collecting
acetazolamide,
duct HCO-3-free
cells. J Clin
cation channels mediate sodium absorption by IMCD (mIMCD-K2) cells. Am J
2001;281:F114. solutions and of amiloride. Pf ugers Arch. 1979;379:49.
Invest. 1988;82:1067.
Physiol.
448. 1997;272:C901.
Peti-Peterdi J, Warnock DG, Bell PD. Angiotensin II directly stimulates http://www.ncbi.nlm.nih.gov/pubmed/571103
470. Rocha AS, Kudo LH. Factors governing sodium and chloride transport
467. Wilson
ENaC activityDR, Honrath
in the U, Sonnenberg
cortical collecting duct H. Furosemide action on Jcollect
via AT(1) receptors. Am Socing 479.
across Husted
the inner RFmedullary
, Stokes JB.collecting
Separate duct.
regulation
KidneyofInt.
Na+ and anion transport by
1990;38:654.
ducts: effect
Nephrol. of prostaglandin synthesis inhibition. Am J Physiol. 1983; 244:F666.
2002;13:1131. IMCD: location,
471. Delpire E, aldosterone,
Rauchman MI, hypertonicity,
Beier DR, TGF-beta 1, and cAMP
et al. Molecular . Amand
cloning J Physiol.
chro-
468. Beutler
449. Wilson KT,DR, Masilamani
Honrath U, S, Sonnenberg
Turban S, H. Thiazide
et al. diuretic
Long-term effect onofmedul-
regulation ENaC 1996;271:F433.
mosome localization of a putative basolateral Na( )-K( )-2Cl- cotransporter
lary collecting
expression duct function
in kidney in the rat.
by angiotensin II. Kidney Int. 1983;23:711.
Hypertension. 2003;41:1143. 480. Terada inner
from mouse Y, Knepper MA. Na
medullary +-K +-ATPase
collecting activities incells.
duct (mIMCD-3) renalJ tubule
Biol Chem.seg-
http://www.ncbi.nlm.nih.gov/pubmed/6876566
450. Sun P, Yue P, Wang WH. Angiotensin II stimulates epithelial sodium chan- ments of rat inner medulla. Am J Physiol. 1989;256:F218.
1994;269:25677.
469.(ENaC)
nels Zeidel in ML,theKikeri D, collecting
cortical Silva P, et duct
al. Atrial
of thenatriuretic
rat kidney.peptides inhibitRenal
Am J Physiol con- 472. Praetorius J, Kim YH, Bouzinova EV, et al. NBCn1 is a basolateral Na -
ductive 2011
Physiol. sodium Decuptake
14 [Epub by rabbit
ahead of inner medullary collecting duct cells. J Clin
print]. HCO3- cotransporter in rat kidney inner medullary collecting ducts. Am J Physiol
Invest.Hallows
451. 1988;82:1067.
KR, Wang H, Edinger RS, et al. Regulation of epithelial Na Renal Physiol. 2004;286:F903.
transport by soluble adenylyl cyclase in kidney collecting duct cells. J Biol Chem. 473. Zeidel ML. Renal actions of atrial natriuretic peptide: regulation of collect-
2009;284:5774. ing duct sodium and water transport. Annu Rev Physiol. 1990;52:747.
452. Pech V, Pham TD, Hong S, et al. Pendrin modulates ENaC function by 474. Zeidel ML, Silva P, Brenner BM, et al. cGMP mediates effects of atrial
changing luminal HCO3. J Am Soc Nephrol. 2010;21:1928. peptides on medullary collecting duct cells. Am J Physiol. 1987;252:F551.
453. Edinger RS, Lebowitz J, Li H, et al. Functional regulation of the epithelial 475. Light DB, Schwiebert EM, Karlson KH, et al. Atrial natriuretic peptide in-
Na channel by IkappaB kinase-beta occurs via phosphorylation of the ubiqui- hibits a cation channel in renal inner medullary collecting duct cells. Science.
tin ligase Nedd4-2. J Biol Chem. 2009;284:150. 1989;243:383.
454. Carattino MD, Edinger RS, Grieser HJ, et al. Epithelial sodium channel 476. Rocha AS, Kudo LH. Atrial peptide and cGMP effects on NaCl transport in
inhibition by AMP-activated protein kinase in oocytes and polarized renal inner medullary collecting duct. Am J Physiol. 1990;259:F258.
epithelial cells. J Biol Chem. 2005;280:17608. 477. Kudo LH, van Baak AA, Rocha AS. Effect of vasopressin on sodium trans-
455. Bhalla V, Hallows KR. Mechanisms of ENaC regulation and clinical impli- port across inner medullary collecting duct. Am J Physiol. 1990;258:F1438.
cations. J Am Soc Nephrol. 2008;19:1845. 478. Ullrich KJ, Papavassiliou F. Sodium reabsorption in the papillary collecting
456. Koeppen BM. Conductive properties of the rabbit outer medullary collecting duct of rats. Effect of adrenalectomy, low Na diet, acetazolamide, HCO-3-free
duct: outer stripe. Am J Physiol. 1986;250:F70. solutions and of amiloride. P ugers Arch. 1979;379:49.
457. Stokes JB. Na and K transport across the cortical and outer medullary col- 479. Husted RF, Stokes JB. Separate regulation of Na and anion transport by
lecting tubule of the rabbit: evidence for diffusion across the outer medullary IMCD: location, aldosterone, hypertonicity, TGF-beta 1, and cAMP. Am J Physiol.
portion. Am J Physiol. 1982;242:F514. 1996;271:F433.
458. Koeppen BM. Conductive properties of the rabbit outer medullary collect- 480. Terada Y, Knepper MA. Na -K -ATPase activities in renal tubule seg-
ing duct: inner stripe. Am J Physiol. 1985;248:F500. ments of rat inner medulla. Am J Physiol. 1989;256:F218.
C H A P T E R

6 Tubular Potassium Transport


David B. Mount

This chapter reviews the renal and extrarenal mecha-


TUBULAR POTASSIUM TRANSPORT nisms of K homeostasis, with primary emphasis on the
Potassium (K ) functions in a diversity of physiologic pro- physiology of renal K transport.
cesses. Changes in intracellular K affect cell volume, in-
tracellular pH, enzymatic function, protein synthesis, DNA
synthesis, and apoptosis. Changes in the ratio of intracellular Extrarenal Potassium Homeostasis
to extracellular K affect the cellular resting membrane po- The intracellular accumulation of K against its electro-
tential, causing depolarization in hyperkalemia and hyper- chemical gradient is an energy-consuming process, mediat-
polarization in hypokalemia; as a consequence, potassium ed by the ubiquitous Na /K –ATPase. The Na /K –ATPase
disorders primarily affect excitable tissues, chie y heart and functions as an electrogenic pump, with a transport stoichi-
muscle. Hypokalemia and hyperkalemia also have a variety ometry of three intracellular Na ions to two extracellular
of renal and cardiovascular consequences. A large body of K ions. The enzyme complex is made up of a tissue-speci c
experimental and epidemiologic evidence thus implicates combination of multiple , , and subunits, which are fur-
hypokalemia or reduced K intake in the pathogenesis of ther subject to tissue-speci c patterns of regulation. Cardiac
hypertension, heart failure, and stroke.1 Hypokalemia also glycosides (i.e., digoxin and ouabain) bind to the subunits
causes a host of structural and functional changes in the kid- of Na /K –ATPase at an exposed extracellular hairpin loop
ney, whereas hyperkalemia in turn has a signi cant effect on that also contains the major binding sites for extracellular
the ability to excrete an acid urine due to interference with K .2 The binding of digoxin and K to the Na /K –ATPase
the urinary excretion of ammonium (NH4 ). complex is thus mutually antagonistic, explaining in part the
Potassium is almost exclusively an intracellular cation, potentiation of digoxin toxicity by hypokalemia.3 Although
with only 2% of total body K contained within the extracel- the four subunits have equivalent af nity for ouabain, they
lular uid. Extracellular K is maintained within a very nar- differ signi cantly in the intrinsic K /ouabain antagonism.4
row range by three major mechanisms. First, the distribution Ouabain binding to isozymes containing the ubiquitous 1
of K between the intracellular and extracellular space is de- subunit is relatively insensitive to K concentrations within
termined by the activity of a number of widely expressed the physiologic range, such that this isozyme is protected
and/or ubiquitous transport pathways. A net increase in cel- from digoxin under conditions wherein cardiac 2 and 3
lular uptake can thus cause transient hypokalemia, whereas subunits, the therapeutic targets of digoxin, are inhibited.4
impairment of cellular uptake can lead to hyperkalemia. Sec- Notably, the digoxin/ouabain binding site of subunits is
ond, the colon has the ability to absorb and secrete K , with highly conserved, suggesting a potential role in the physi-
signi cant mechanistic and regulatory similarities to renal ologic response to endogenous ouabain/digoxinlike com-
K transport. However, the colon has a relatively limited ca- pounds. Consistent with this hypothesis, a mouse strain that
pacity for K excretion, and a third mechanism, changes in expresses 2 subunits with engineered resistance to ouabain
renal K excretion, plays the dominant role in responding to is strikingly resistant to ouabain-induced hypertension and
changes in K intake. Regulated increases in K secretion by to adrenocorticotrophic hormone-dependent hypertension,5
the connecting tubule (CNT) and the cortical collecting duct the latter of which is known to involve an increase in circu-
(CCD) play a critical role in the response to hyperkalemia lating ouabainlike glycosides.
and K loading, whereas inhibition of K secretion and in- Potassium can also accumulate in cells by coupling to
creases in the reabsorption of K by the CCD and the outer the gradient for Na entry, entering via the electroneutral
medullary collecting duct (OMCD) function in the response Na -K -2Cl cotransporters NKCC1 and NKCC2. The
to hypokalemia or K deprivation. NKCC2 protein is found only at the apical membrane of
19 4
CHAPTER 6 TUBULAR POTASSIUM TRANSPORT 1195
95

the thick ascending limb (TAL) and the macula densa cells, K by the liver and muscles is stimulated via 2 receptors.
where it functions in transepithelial salt transport and tubu- This hypokalemic effect of catecholamines is independent of
lar regulation of renin release (see Potassium Transport in changes in circulating insulin and has been reported in ne-
the Thick Ascending Limb).6 NKCC1 is widely expressed in phrectomized animals.17 The cellular mechanisms whereby
multiple tissues,6 including muscle, where it modulates ex- catecholamines induce K uptake in muscles include an ac-
trarenal K homeostasis.7 The cotransport of K -Cl by the tivation of the Na /K –ATPase, likely via increases in cyclic-
four K -Cl cotransporters (KCC1 to 4) can also function AMP (cAMP).18 Skeletal muscle -adrenergic receptors also
in the transfer of K across membranes; although the KCCs activate NKCC1, which may account for as much as one
typically function as ef ux pathways, they can mediate in- third of the uptake response.7 In contrast to -adrenergic
ux when extracellular K increases.6 stimulation, -adrenergic agonists impair the ability to buf-
Skeletal muscle contains approximately 75% of the fer increases in K induced via intravenous loading or by ex-
body’s potassium and exerts considerable in uence on extra- ercise19; the transport mechanisms whereby this occurs are
cellular K . Skeletal muscle Na /K –ATPase activity in par- not known. -adrenergic stimulation increases K uptake
ticular is a major determinant of the capacity for extrarenal during exercise to lessen exercise-induced hyperkalemia,
K homeostasis. Hypokalemia induces a marked decrease in whereas -adrenergic mechanisms help blunt the ensuing
muscle K content and Na /K –ATPase activity, an altruis- postexercise nadir.19
tic8 mechanism to regulate plasma K . This adaptation is pri- The ef ux of K out of cells is primarily mediated by
marily mediated by dramatic decreases in the protein abun- K channels, which comprise the largest group of ion chan-
dance of the -2 subunit of Na /K –ATPase. In contrast, nels in the human genome. There are three major subclasses
hyperkalemia due to potassium loading is associated with of mammalian K channels: the six-transmembrane domain
adaptive increases in muscle K content and Na /K –ATPase (TMD) family, which encompasses both the voltage-sensitive
activity.9 These interactions are re ected in the relationship and Ca2 -activated K channels; the two-pore, four TMD
between physical activity and the ability to regulate extra- family; and the two TMD family of inward rectifying K (Kir)
cellular K during exercise10; exercise training is associated channels. There is tremendous genomic variety in human
with increases in muscle Na /K –ATPase concentration and K channels. Further complexity is generated by the pres-
activity, with reduced interstitial K in trained muscles and ence of multiple accessory subunits and alternative patterns
an enhanced recovery of plasma K after de ned amounts of mRNA splicing. Not surprisingly, an increasing number
of exercise.10 and variety of K channels have been implicated in the con-
Several hormones have been implicated in the control of trol of K homeostasis and the membrane potential of excit-
extrarenal K homeostasis. In particular, increases in plasma able cells such as muscle and heart, with important, evolving
K have a stimulatory effect on insulin levels,11 which in turn roles in the pathophysiology of potassium disorders. Adrenal
stimulates the uptake of K by muscles, the liver, and other K channels have also been implicated in the adrenal release
tissues. In contrast, insulin-stimulated K uptake is rapidly of aldosterone induced by hyperkalemia.20,21 The emphasis
reduced by 2 days of K depletion, prior to a modest drop in this chapter is on renal K channels, particularly those in
in plasma K ,12 and in the absence of a change in plasma K the distal nephron that mediate K secretion.
in rats subject to a lesser K restriction for 14 days.13 Insulin
activates muscle Na /K –ATPase, inducing the translocation
of the Na /K –ATPase -2 subunit to the plasma membrane Potassium Transport in the Proximal Tubule
with a lesser effect on the 1 subunit.14 This translocation The proximal tubule reabsorbs 50% to 70% of ltered K
is dependent on the activity of phosphoinositide-3 (PI-3) (Fig. 6.1). Proximal tubules generate minimal transepithelial
kinase,14 which itself also binds to a proline-rich motif in K gradients, and the fractional reabsorption of K by this
the N-terminus of the subunit; the activation of PI3-ki- nephron segment is similar to that of Na .22 K absorption
nase by insulin induces phosphatase enzymes to dephos- thus follows that of uid, Na , and other solutes,23 such that
phorylate a speci c serine residue adjacent to the PI3-kinase proximal tubules do not play a direct role in regulated renal
binding domain. Traf cking of Na /K –ATPase to the cell K excretion. However, changes in Na -Cl reabsorption by
surface also requires phosphorylation of an adjacent tyro- the proximal tubule have considerable effects on distal tubu-
sine residue, perhaps catalyzed by the tyrosine kinase activ- lar ow and distal tubular Na delivery, with attendant effects
ity of the insulin receptor itself.15 In addition, the serum and on the excretory capacity for K . In addition, K loading has
glucocorticoid-induced kinase 1 (SGK1) plays a critical role signi cant effects on proximal tubular Na -Cl reabsorption.
in insulin-stimulated K uptake via stimulatory effects on Thus, the intravenous infusion of K -Cl or increases in the
Na /K –ATPase activity and/or the Na -K –2Cl cotrans- peritubular K concentration causes an inhibition of proxi-
port.16 The hypokalemic effect of insulin plus glucose is thus mal tubular Na -Cl reabsorption 24; this serves to increase
blunted in SGK1 knockout mice, with a marked reduction both distal tubular ow and distal tubular Na delivery, thus
in hepatic insulin-stimulated K uptake.16 increasing distal K secretion after K loading.
The sympathetic nervous system also affects the balance The mechanisms involved in transepithelial K transport
between extracellular and intracellular K . The uptake of by the proximal tubule are not completely clear, although
196
19 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

apical and basolateral membrane. Therefore, the apparent


convective transport of K would have to constitute pseu-
dosolvent drag, with uncharacterized, coordinating interac-
tions between water traversing the transcellular route and
the diffusion of K along the claudin-dependent paracellular
pathway.29

The Loop of Henle and Medullary


Potassium Recycling
Transport by the loop of Henle functions in medullary K
recycling (Fig. 6.2). A considerable fraction of K secreted by
the CCD is reabsorbed by the medullary collecting ducts and is
then secreted into the late proximal tubule and/or the descending
thin limbs of long-looped nephrons.30 In potassium-loaded
rats there is thus a doubling of luminal K in terminal thin
descending limbs, with a sharp drop after the inhibition of
CCD K secretion by amiloride.31 Enhancement of CCD K
secretion with the treatment of 1-desamino-8-D-arginine
FIGURE 6.1 Potassium transport along the nephron. Approxi- vasopressin (dDAVP) also results in an increase in luminal K
mately 90% of ltered K is reabsorbed by the proximal tubule in descending thin limbs.32 This recycling pathway (i.e., secre-
and the loop of Henle. K is secreted in the connecting tubule tion in CCD, absorption in the OMCD and the inner medullary
and the cortical collecting duct; net reabsorption occurs in collecting duct (IMCD), secretion in the descending thin limb)
response to K depletion, primarily within the medullary collect-
ing duct. PCT, proximal tubule; TAL, thick ascending limb; DCT,
distal convoluted tubule; CNT, connecting tubule; CCD, cortical
collecting duct; S, secretion; R, reabsorption; ALDO, aldosterone;
ADH, antidiuretic hormone; MCD, medullary collecting duct.

active transport does not appear to play a major role.23,25


Luminal barium has modest effects on transepithelial K
transport, suggesting a component of transcellular transport
via barium-sensitive K channels.26 However, the bulk of
K transport is thought to occur via the paracellular path-
way,26,27 driven by the lumen-positive potential difference in
the mid-to-late proximal tubule. The total K permeability
of the proximal tubule is thus quite high due to the high per-
meability of the paracellular pathway.26, 27 The combination
of luminal K concentrations that are 10% higher than that
of plasma, a lumen-positive potential difference of 2 mV,
and high paracellular permeability leads to considerable
paracellular absorption in the proximal tubule. The tight
junction protein claudin-2 plays a key role in paracellular
cation transport by the proximal tubule; targeted deletion in
knockout mice generates a “tight” epithelium in the proxi-
mal tubule, with a reduction in Na , Cl , and uid absorp- FIGURE 6.2 A schematic representation of medullary K recy-
tion.28 The loss of claudin-2 expression does not affect the cling. Medullary interstitial K increases considerably after di-
ultrastructure of tight junctions, but does lead to a reduction etary K loading due to the combined effects of secretion in the
in paracellular cation permeability.28 cortical collecting duct (CCD), absorption in the outer medullary
The absorption of K across the paracellular pathway collecting duct (OMCD), the thick ascending limb (TAL), and the
is thought to occur via convective transport—solvent drag inner medullary collecting duct (IMCD), and secretion in the
due to frictional interactions between water and K —rather descending thin limb (see text for details). (From Stokes JB. Con-
than diffusional transport.29 Notably, however, the primary sequences of potassium recycling in the renal medulla. Effects of
pathway for water movement in the proximal tubule is ion transport by the medullary thick ascending limb of Henle’s
quite conclusively transcellular, via water channels in the loop. J Clin Invest. 1982;70:219–229.)
CHAPTER 6 TUBULAR POTASSIUM TRANSPORT 1197
97

is associated with a marked increase in medullary interstitial K


concentration. Passive transepithelial K absorption by the thin
ascending limb and active absorption by the thick ascending
limb (TAL)33 also contribute to this increase in interstitial K
(Fig. 6.2). Speci cally, the absorption of K by the ascending
thin limb, TAL, and OMCD exceeds the secretion by descending
thin limbs, thus concentrating K within the interstitium.
K is secreted into descending thin limbs by passive
diffusion, driven by the high medullary interstitial K con-
centration. Descending thin limbs thus have a very high K
permeability, without evidence for active transepithelial K
transport.34 Transepithelial K transport by ascending thin
limbs has not been measured; however, as is the case for
Na -Cl transport,35 the absorption of K by thin ascending
limbs is presumably passive, via the paracellular pathway.
The physiologic signi cance of medullary K recycling
is not completely clear. However, an increase in interstitial
K from 5 to 25 mM dramatically inhibits Cl transport by
perfused thick ascending limbs.33 By inhibiting Na -Cl FIGURE 6.3 The potassium transport pathways in the thick
absorption by the TAL, increases in interstitial K would ascending limb (TAL); see text for details. NKCC2, Na -K -2Cl–
increase Na delivery to the CNT and CCD, thus enhancing cotransporter 2; ROMK, renal outer medullary K channel;
the lumen-negative potential difference (PD) in these tubules CLC-NKB, human Cl– channel; Barttin, Cl– channel subunit;
and increasing K secretion.33 A high K diet also reduces KCC4, K -Cl– cotransporter 4.
the absorption of K by the TAL36 (i.e., there are direct effects
on K secretion by the TAL). The marked increase in medul-
lary interstitial K after dietary K loading is also postulated logic subtypes, a rough-surfaced cell type (R cells) with
to limit the difference between luminal and peritubular K prominent apical microvilli and a smooth-surfaced cell type
in the collecting duct, thus minimizing passive K loss from (S cells) with an abundance of subapical vesicles.37,40 In the
the collecting duct. hamster TAL, cells can also be separated into those with
high apical and low basolateral K conductance and a low
basolateral Cl conductance (LBC cells), versus a second
Potassium Transport in the Thick population with low apical and high basolateral K con-
Ascending Limb ductance, combined with high basolateral Cl conductance
Active transepithelial K transport across the TAL includes (HBC) cells.39,41 The relative frequency of the morphologic
both a transcellular component, via the apical Na -K -2Cl and functional subtypes in the cortical and medullary TAL
cotransporter NKCC2, and a paracellular pathway (Fig. 6.3). suggests that HBC cells correspond to S cells and LBC cells
Potassium transport appears to differ in the major morpho- correspond to R cells.39
logic subtypes of TAL cells (i.e., rough- and smooth-sur- Morphologic and functional heterogeneity notwith-
faced cells). Morphologically, the TAL begins abruptly after standing, the cells of the medullary TAL, the cortical TAL,
the thin ascending limb of long-looped nephrons and after and the macula densa are presumed to share the same basic
the aquaporin-negative segment of short-limbed nephrons. or at least composite transport mechanisms (see Fig. 6.3).
The TAL then extends into the renal cortex, where it meets Na -Cl reabsorption by the TAL is a secondarily active
its parent glomerulus at the vascular pole; the plaque of cells process, driven by the favorable electrochemical gradient for
at this junction forms the macula densa, which functions as Na established by the basolateral Na /K –ATPase.42 The
the tubular sensor for both tubuloglomerular feedback and Na , K , and Cl ions are cotransported across by the api-
the tubular regulation of renin release by the juxtaglomeru- cal membrane by an electroneutral Na -K -2Cl cotrans-
lar apparatus. Cells in the medullary TAL are 7 to 8 m in porter; this transporter generally requires the simultaneous
height, with extensive invaginations of the basolateral plasma presence of all three ions, such that the transport of Na
membrane and interdigitations between adjacent cells37; and Cl across the epithelium is mutually codependent
cells in the cortical TAL are considerably shorter, about and dependent on the luminal presence of K . An api-
2 m in height at the end of the cortical TAL in rabbits, with cal Na -K -2Cl cotransport is mediated by the cation–
less mitochondria and a simpler basolateral membrane.37 chloride cotransporter NKCC2, encoded by the SLC12A1
Macula densa cells also lack the lateral cell processes and in- gene.6 Functional expression of NKCC2 in Xenopus laevis
terdigitations that are characteristic of medullary TAL cells.37 oocytes yields Cl -dependent and Na -dependent uptake of
86
Scanning electron microscopy has revealed that the Rb (a radioactive substitute for K ) and Cl -dependent
TAL of both rats38 and hamsters39 contains two morpho- and K -dependent uptake of 22Na .6 NKCC2 is sensitive to
198
19 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

micromolar concentrations of furosemide, bumetanide, and K channels in Na -Cl absorption by the TAL. Clearance
other loop diuretics.6 studies in ROMK-de cient mice also indicate that 80% of
Immuno uorescence indicates the expression of the NKCC2 activity is dependent on expression of the ROMK
NKCC2 protein along the entire length of the TAL.6 In par- K channel with the TAL.47
ticular, immunoelectron microscopy reveals the expression Three types of apical K channels have been identi-
in both rough (R; see previous) and smooth (S) cells of the ed in the TAL, a 30 pS channel, a 70 pS channel, and a
TAL (see previous).40 NKCC2 expression in subapical vesi- high-conductance, calcium-activated maxi K channel.48–50
cles is particularly prominent in smooth cells,40 suggesting The higher open probability and greater density of the 30
a role for vesicular traf cking in the regulation of NKCC2. pS and 70 pS channels, versus the maxi K channel, sug-
NKCC2 is also expressed in macula densa cells,40 which gest that these are the primary routes for K recycling across
have been shown to possess apical Na -K -2Cl cotrans- the apical membrane; the 70 pS channel in turn appears to
port activity. Both tubuloglomerular feedback (TGF) and re- mediate 80% of the apical K conductance of TAL cells.51
nal renin secretion are controlled by NKCC2 in the macula The low conductance 30 pS channel shares several electro-
densa; luminal loop diuretics block both TGF and the sup- physiologic and regulatory characteristics with ROMK, the
pression of renin release by the luminal Cl .6 cardinal inward-rectifying K channel (KIR 1.1) that was
Microperfused TALs develop a lumen-positive PD during initially cloned from the renal outer medulla.52,53 ROMK
perfusion with Na -Cl .43 This lumen-positive PD plays a protein has been identi ed at the apical membrane of the
critical role in the physiology of the TAL, driving the para- medullary TAL, the cortical TAL, and the macula densa.54
cellular transport of Na , K , Ca2 , and Mg2 (see Fig. 6.3). Furthermore, the 30 pS channel is also absent from the api-
Originally attributed to electrogenic Cl transport,43 the cal membrane of knockout mice with a homozygous deletion
lumen-positive, transepithelial PD in the TAL is generated by of the gene encoding ROMK.55 Notably, not all cells in the
the combination of apical K channels and basolateral Cl TAL are labeled with anti-ROMK antibodies,54,56 suggesting
channels.42 The conductivity of the apical membrane of TAL that ROMK might be absent in the HBC cells with high baso-
cells is predominantly, if not exclusively, K selective. Lumi- lateral Cl conductance and low apical/high basolateral K
nal recycling of K via Na -K -2Cl cotransport and apical conductance (see also previous).39,41 HBC cells are thought
K channels, along with basolateral depolarization due to to correspond to the smooth-surfaced morphologic subtype
Cl exit through Cl channels, results in the lumen-positive of TAL cells (S cells)39; however, distribution of the ROMK
transepithelial PD.42 protein by immunoelectron microscopy has not as yet been
Several lines of evidence indicate that apical K chan- published, hence correlation cannot be made as of yet be-
nels are required for transepithelial Na -Cl transport by tween morphology and ROMK expression.
the TAL.42 First, the removal of K from luminal perfusates Alternative splicing and alternative promoter use of the
results in a marked decrease in Na -Cl reabsorption by KCNJ1 gene encoding ROMK/KIR1.1 generates three differ-
the TAL, as measured by a short circuit current; the residual ent protein isoforms, differing in the extreme aminotermi-
Na -Cl transport in the absence of luminal K is sustained nal amino acid sequence: ROMK1, ROMK2, and ROMK3.57
by the exit of K via apical K channels, because the com- Although the functional signi cance of this variation is not
bination of K removal and a luminal K channel inhibitor clear, these isoforms are differentially distributed along
(barium) almost abolishes the short-circuit current.44 Apical the nephron. ROMK1 is exclusive to the CNT, CCD, and
K channels are thus required for the activity of NKCC2, the OMCD; ROMK2 is expressed from the TAL to the CCD; and
apical Na -K -2Cl cotransporter; the low luminal concen- ROMK3 is expressed in the TAL and the distal convoluted
tration of K in this nephron segment would otherwise be- tubule (DCT).57 Apical ROMK channels in the TAL are thus
come limiting for transepithelial Na -Cl transport. Second, a mixture of ROMK2 and ROMK3 proteins.
the net transport of K across perfused TAL is 10% that of ROMK clearly plays a critical role in Na -Cl absorp-
Na and Cl 33; 90% of the K transported by NKCC2 is tion by the TAL, given that loss-of-function mutations in
recycled across the apical membrane via K channels, result- this gene are associated with Bartter syndrome.46 The role of
ing in minimal net K absorption by the TAL.42 Third, the in- ROMK in Bartter syndrome was initially discordant with the
tracellular K activity of perfused TAL cells is 15 to 20 mV data indicating that the 70 pS K channel is the dominant
above equilibrium, due to furosemide-sensitive entry of K conductance at the apical membrane of TAL cells51; heterolo-
via NKCC2.45 Given an estimated apical K conductivity of gous expression of the ROMK protein in Xenopus oocytes had
12 m per square centimeter, this intracellular K activ- yielded a channel with a conductance of 30 pS, suggesting
ity yields a calculated K current of 200 A per square that the 70 pS channel was distinct from ROMK. This para-
centimeter; this corresponds quantitatively to the uptake of dox was resolved by the observation that the 70 pS channel
K by the apical Na -K -2Cl cotransporter. Finally, the is absent from the TAL of ROMK knockout mice, indicating
observation that Bartter syndrome can be caused by mu- that ROMK proteins form a subunit of the 70 pS channel.58
tations in ROMK (renal outer medullary K channel),46 a ROMK activity in the TAL is clearly modulated by association
critical component of apical K channels in the TAL (see with other proteins. That coassociation with other subunits
the following), provides genetic proof for the importance of generates the 70 pS channel and is perfectly compatible with
CHAPTER 6 TUBULAR POTASSIUM TRANSPORT 1199
99

the known physiology of this channel protein. ROMK thus permeability of the CNT is considerably lower than that of
associates with scaffolding proteins NHERF-1 and NHERF-2 the CCD,66 with a signi cant number of early (CNT1) CNT
via the C-terminal PDZ-binding motif of ROMK; NHERF-2 cells that do not express aquaporin-2 in the absence of vaso-
is coexpressed with ROMK in the TAL.59 The association of pressin. Aquaporin-2 expression is also low or undetectable
ROMK with NHERFs brings ROMK into closer proximity in CNT1 cells on a high K diet,56 further optimizing these
to the cystic brosis transmembrane regulator (CFTR) pro- early CNT segments for K secretion.
tein.59 This ROMK-CFTR interaction is in turn required for The apical membrane of CNT cells and principal cells
reconstitution of the native ATP and glibenclamide sensitiv- contain prominent Na and K conductances,63,65,67 without
ity of apical K channels in the TAL.60 a measurable apical conductance for Cl .68 The entry of Na
TAL cells are phenotypically de ned by the apical ex- occurs via the highly selective ENaC, which is sensitive to mi-
pression of uromodulin or Tamm-Horsfall glycoprotein cromolar concentrations of amiloride (Fig. 6.4). This selective
(THP), a TAL-speci c, GPI-linked membrane protein that is absorption of a positive charge generates a lumen-negative
shed into the tubular lumen as the most abundant protein PD, the magnitude of which varies considerably as a function
in normal human urine. THP interacts with ROMK protein of mineralocorticoid status and other factors. This lumen-
in yeast two-hybrid screens and activates the channel in negative PD serves to drive the following critical processes:
coexpression experiments.61 THP also exhibits functional (1) K secretion via apical K channels, (2) paracellular Cl
interactions with NKCC2, activating the cotransporter in transport through the adjacent tight junctions, and (3) elec-
coexpression experiments.62 Membrane traf cking of both trogenic H secretion via adjacent type A intercalated cells.
ROMK and NKCC2 proteins is affected in THP knockout The three ENaC subunits are detectable at the api-
mice, with greater accumulation of both transport proteins cal membrane of CNT cells and principal cells within the
in subapical vesicles. THP knockout mice also exhibit a CCD, OMCD, and IMCD. Notably, however, several lines of
blunted natriuretic and kaliuretic response to furosemide, evidence support the hypothesis that the CNT makes the
which is consistent with a partial de ciency in TAL func-
tion.62 Mutations of the THP gene cause the allelic disorders
familial juvenile hyperuricemic nephropathy, medullary cys-
tic kidney disease type II, and glomerulocystic kidney dis-
ease. All three disorders share reduced expression of THP at
the apical membrane with retention in the endoplasmic re-
ticulum. The associated defects in NKCC2 and ROMK may
explain the renal salt wasting and hyperuricemia that can be
associated with these genetic disorders.61,62

Potassium Secretion by the Distal


Convoluted Tubule, Connecting Tubule,
and Cortical Collecting Duct
Approximately 90% of ltered K is reabsorbed by the prox-
imal tubule and the loop of Henle (Fig. 6.1); the nal adjust-
ments in renal K excretion occur in the downstream distal
nephron. The bulk of regulated secretion occurs in the CNT
and CCD, whereas K reabsorption primarily occurs in the
OMCD (see the following). K secretion is initially detect-
able in the early DCT, wherein cells expressing the thiazide-
sensitive Na -Cl cotransporter (NCC) express ROMK, the
apical K secretory channel (see the following).54 More re- FIGURE 6.4 The transport pathways for potassium in principal/
cent studies in rat kidneys have localized the protein to both connecting tubule (CNT) cells and intercalated cells. Potassium
DCT1 and DCT2 segments.56 Classically, the CCD is con- secretion occurs in CNT and principal cells, driven by the lumen-
sidered the primary site for distal K secretion, largely due negative potential difference generated by the epithelial Na
to the greater ease with which this segment can be perfused channel (ENaC). In intercalated cells, potassium reabsorbed by
and studied. However, as in Na absorption (see below),63,64 apical H /K -ATPase recycles across the apical membrane via K
the bulk of distal K secretion appears to occur prior to the channels; alternatively, in the setting of hypokalemia or potassium
CCD,22 within the CNT.65 In addition to a lesser endowment deprivation, it exits the cell via basolateral K channels. Potassium
with absorptive apical epithelial Na channels (ENaC) and that is secreted across intercalated cells enters at the basolateral
secretory K channels, water absorption in the vasopressin- membrane via NKCC1 and exits via apical BKand Kv1.3 K chan-
responsive CCD limits K secretion by allowing the concen- nels. BK, BKK channel; ROMK, renal outer medullary K channel;
tration of luminal K to rise toward equilibrium.66 Water Kv1.3, Kv1.3 K channel; NKCC1, Na -K -2Cl cotransporter 1.
200
20 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

dominant contribution to amiloride-sensitive Na reabsorp- BK channels (see the following).77 Loss-of-function muta-
tion by the distal nephron. First, amiloride-sensitive Na tions in human KCNJ1 are associated with Bartter syndrome;
currents in the CNT are two- to fourfold higher than in the ROMK expression is critical for the 30 pS and 70 pS chan-
CCD; the maximal capacity of the CNT for Na reabsorption nels that generate the lumen-positive PD in the TAL (see Fig.
is estimated to be 10 times higher than that of the CCD.63 6.3).55,58 These patients typically have slightly higher serum
Second, targeted deletion of -ENaC in the collecting duct K than the other genetic forms of Bartter syndrome,46 and
abolishes amiloride-sensitive currents in CCD principal affected patients with severe neonatal hyperkalemia have
cells, but does not affect Na or K homeostasis. Thus, the also been described; this neonatal hyperkalemia is presum-
residual ENaC expression in the late DCT and CNT of these ably the result of a transient developmental de cit in apical
knockout mice easily compensates for the loss of the chan- BK channel activity (see the following).
nel in CCD.69 In contrast, the more extensive deletion of The apical Ca2 -activated BK channel plays a critical
-ENaC in all aquaporin-2–positive cells in both CNT and role in ow-dependent K secretion by the CNT and CCD.75
CCD causes a profound impairment in Na and K homeo- BK channels have a heteromeric structure, with -subunits
stasis.64 Third, Na /K –ATPase activity in the CCD is con- that form the ion channel pore and modulatory -subunits
siderably less than that of the DCT70; this speaks to a greater that affect the biophysical, regulatory, and pharmacologic
capacity for transepithelial Na -Cl absorption by the DCT characteristics of the channel complex.75 BK -subunit tran-
and CNT. Fourth, the apical recruitment of ENaC subunits scripts are expressed in multiple nephron segments, and chan-
in response to dietary Na restriction begins in the CNT, nel protein is detectable at the apical membrane of principal
with progressive recruitment of subunits in the downstream and intercalated cells in the CCD and CNT.75 The subunits
CCD at lower levels of dietary Na 71; under conditions of are differentially expressed within the distal nephron. Thus
high Na -Cl and low K intake, the bulk of aldosterone- 1 subunits are restricted to the CNT,75 with no expression
stimulated Na transport likely occurs prior to the entry of in intercalated cells,78 whereas 4 subunits are detectable at
tubular uid into the CCD. the apical membranes of TAL, DCT, and intercalated cells.78
In principal cells and CNT cells, the lumen-negative po- Increased distal ow has a well-established stimulatory
tential difference generated by Na entry via ENaC drives effect on K secretion, due in part to both the enhanced
passive K exit through apical K channels. Although there delivery and absorption of Na and to the increased removal
is evolving evidence for ENaC and Na -independent se- of secreted K .73 The pharmacology of ow-dependent K
cretion (see also the following),72 distal K secretion is criti- secretion in the CCD is consistent with BK channels,79 and
cally dependent on delivery of adequate luminal Na to the ow-dependent K secretion is reduced in mice with targeted
CNT and CCD,73 essentially ceasing when luminal Na deletion of the 1 and 1 subunits.75,80,81 Both mice strains
drops below 8 mmol/L.74 Dietary Na intake also in uences develop hyperaldosteronism that is exacerbated by high-K
K excretion, such that excretion is enhanced by excess Na diet,81 leading to hypertension in the 1 subunit knockout.81
intake and reduced by Na restriction.73 Secreted K enters One enigma has been the greater density of BK channels
principal cells via the basolateral Na /K –ATPase, which in intercalated cells, in both the CCD82 and the CNT.83 This
also generates the gradient that drives apical Na entry via has suggested a major role for intercalated cells in K secre-
ENaC (Fig. 6.4). tion; however, the much lower density of Na /K –ATPase
Two major subtypes of apical K channels function in se- activity in intercalated cells has been considered inadequate
cretion by the CNT and CCD, /– DCT; a small-conductance to support K secretion across the apical membrane.84 More
(SK) 30 pS channel55,65 and a large-conductance, Ca2 - recent evidence reveals a major role for the basolateral
activated 150 pS (“maxi-K” or BK) channel.65,75 The den- Na -K -2Cl cotransporter NKCC1 in K secretion medi-
sity and high open probability of the SK channel indicates ated by apical BK channels (see also Fig. 6.4)85; NKCC1 is
that this pathway alone is suf cient to mediate the bulk of expressed almost exclusively at the basolateral membrane of
K secretion in the CCD under baseline conditions,76 hence intercalated cells,86 providing an alternative entry pathway
its designation as the “secretory” K channel. Notably, SK for basolateral K secreted at the apical membrane. This still
channel density is considerably higher in the CNT than begs the question of how basolateral Na recycles across the
in the CCD,65 consistent with the greater capacity for Na basolateral membrane, in the absence of signi cant Na /K –
absorption 63 and K secretion in the CNT. The character- ATPase activity; one possibility is an alternative basolateral
istics of the SK channel are similar to those of the ROMK Na pump, the ouabain-insensitive furosemide-sensitive
K channel,53 and ROMK protein has been localized at the Na –ATPase, a transport activity that has been detected in
apical membrane of principal cells.54 SK channel activity is cell culture models of intercalated cells.85 At the apical mem-
absent from apical membranes of the CCD in homozygous brane, BK-mediated K secretion is only partially dependent
ROMK knockout mice, de nitive proof that ROMK is the on luminal Na 87; K secretion would eventually hyper-
SK channel.55 The observation that these knockout mice are polarize the membrane in the absence of apical Na entry,
normokalemic with an increased excretion of K illustrates which is mediated by ENaC in principal cells. An intriguing
the considerable redundancy in distal K secretory path- possibility is that apical Cl channels88 allow for a parallel
ways55; distal K secretion in these mice is mediated by apical secretion of K and Cl in intercalated cells (see Fig. 6.4).
CHAPTER 6 TUBULAR POTASSIUM TRANSPORT 2201
01

BK channels also play a critical role in cell volume reg- the apical membrane, the latter via K recycling at the baso-
ulation by intercalated cells, with indirect, ow-mediated lateral membrane to maintain activity of the Na /K –ATPase.
in uences on distal K secretion. MDCK-C11 cells have an A variety of different K channels have been described in the
intercalated cell phenotype and express BK and 4 sub- electrophysiologic characterization of the basolateral mem-
units, as do intercalated cells78; shear stress activates BK brane of principal cells, which has a number of technical
channels in these cells, leading to a loss of K and cell shrink- barriers (reviewed by Gray et al.94). However, a single pre-
age.89 Mice with a targeted deletion of the 4 subunit exhibit dominant activity can be identi ed in principal cells from
normal K excretion on a normal diet.84 However, when fed the rat CCD, using whole-cell recording techniques under
a high K diet, which increases urinary and tubular ow conditions in which ROMK is inhibited (low intracellular pH
rates and tubular shear stress, the 4-knockout mice develop or the presence of the ROMK inhibitor tertiapin-Q).94 This
hyperkalemia with a blunted increase in both K excretion basolateral current is tetraethylammonium (TEA) insensitive,
and urinary ow rates. Intercalated cells from 4 knockout barium sensitive, and acid sensitive (pKa 6.5), with a con-
may fail to signi cantly decrease cell volume in response ductance of 17 pS and weak inward recti cation. These
to a high K diet. Intercalated cells thus function as “speed properties do not correspond exactly to speci c characterized
bumps” that protrude into the lumen of distal tubules; ow- K channels, or combinations thereof. However, candidate
activated BK channels reduce the cell volume of intercalated inward-rectifying K channel subunits that have been local-
cells after K loading, reducing tubular resistance, increasing ized at the basolateral membrane of the CCD include KIR4.1,
tubular ow rates, and increasing distal K secretion.84 KIR5.1, KIR7.1, and KIR2.3.94 A more recent report suggests
The physiologic rationale for the presence of two apical that KIR4.1/KIR5.1 channels generate a predominant 40 pS
secretory K channels—ROMK/SK and BK channels—is not basolateral K channel in murine principal cells.95 Notably,
completely clear. However, the high density and higher open basolateral K channel activity increases on a high K diet,
probability of SK/ROMK channels is perhaps better suited suggesting a role in transepithelial K secretion.94
for a role in basal K secretion, with additional recruitment Similar K channels are found at the basolateral mem-
of the higher capacity, ow-activated BK channels when ad- brane of DCT cells. Cell-attached patches in the basolateral
ditional K secretion is required.75 The evolving evidence membranes of microdissected DCTs detect an inward recti-
also indicates that BK channels function in partially Na - fying K channel with characteristics similar to heteromeric
independent K secretion by intercalated cells, with ROMK KIR4.1/KIR5.1 and KIR4.2/KIR5.1 channels.96 Basolateral
functioning in ENaC- and Na -dependent K excretion by membranes of the DCT express immunoreactive KIR4.197 and
DCT, CNT, and CCD cells. Regardless, at the whole organ KIR5.198 proteins, and DCT cells express KIR4.2 mRNA.96
level, the two K channels can substitute for one another, Patients with loss-of-function mutations in the KCNJ10 gene
with BK-dependent K secretion in ROMK knockout mice77 that encodes KIR4.1 develop a syndrome of epilepsy, ataxia,
and an upregulation of ROMK in the distal nephron of 1 sensorineural deafness, and renal tubulopathy.97,99 The tu-
subunit BK knockout mice.80 bulopathy phenotype encompasses hypokalemia, metabolic
Other K channels reportedly expressed at the luminal alkalosis, hypocalciuria, and hypomagnesemia97,99; KIR4.1
membranes of the CNT and CCD include voltage-sensitive knockout mice demonstrate a greater natriuresis than lit-
channels such as Kv1.3,90 double-pore K channels such as termate controls, in addition to hypocalciuria.97 As in prin-
TWIK-1,91 and KCNQ1.92 KCNQ1 mediates K secretion in cipal cells, KIR4.1/KIR5.1 and KIR4.2/KIR5.1 channels at
the inner ear and is expressed at the apical membrane of the basolateral membrane of DCT cells are hypothesized to
principal cells in the CCD,92 whereas TWIK-1 is expressed function in basolateral K recycling, maintaining adequate
at the apical membrane of intercalated cells.91 The roles of Na /K –ATPase activity for Na -Cl absorption and other
these channels in renal K secretion or absorption are not aspects of DCT function.
fully characterized. However, Kv1.3 may play a role in distal In addition to apical K channels, considerable evi-
K secretion by intercalated cells (see Fig. 6.4) in that lumi- dence implicates apical K -Cl cotransport (or functionally
nal margatoxin, a speci c blocker of this channel, reduces equivalent pathways)100 in distal K secretion.101 In per-
K secretion in CCDs of rat kidneys from animals on a high fused rat distal tubules, a reduction in luminal Cl mark-
K diet (see also previous).90 Other apical K channels in edly increases K secretion 102; the replacement of luminal
the distal nephron subserve other physiologic functions. Cl with SO4 or gluconate has an equivalent stimulatory
For example, the apical Kv1.1 channel is critically involved effect on K secretion. This anion-dependent component
in Mg2 transport by the DCT, likely by hyperpolarizing of K secretion is not in uenced by luminal Ba2 ,102 sug-
the apical membrane and increasing the driving force for gesting that it does not involve apical K channel activity.
Mg2 in ux via transient receptor potential cation channel Perfused surface distal tubules are a mixture of the DCT, the
6 (TRPM6); missense mutations in Kv1.1 are a cause of ge- connecting segment, and the initial collecting duct; how-
netic hypomagnesemia.93 ever, Cl -coupled K secretion is detectable in both the
K channels present at the basolateral membrane of prin- DCT and in the early CNT.103 In addition, similar pathways
cipal cells set the resting potential of the basolateral mem- are detectable in rabbit CCD, where a decrease in luminal
brane and function in K secretion and Na absorption at Cl from 112 mmol per liter to 5 mmol per liter increases
202
20 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

K secretion by 48%.104 A reduction in basolateral Cl also subunit is usually sensitive to ouabain and resistant to
decreases K secretion without an effect on transepithelial SCH-28080.110 Within the kidney, the HK -1 subunit is ex-
voltage or Na transport, and the direction of K ux can pressed at the apical membrane of at least a subset of type A
be reversed by a lumen-to-bath Cl gradient, resulting in intercalated cells in the distal nephron.111 HK -2 distribu-
K absorption.104 In perfused CCDs from rats treated with tion in the distal nephron is more diffuse, with robust ex-
a mineralocorticoid, vasopressin increases K secretion.105 pression at the apical membrane of type A and B intercalated
Because this increase in K secretion is resistant to lumi- cells and connecting segment cells, and a lesser expression
nal Ba2 (2 mmol per liter), vasopressin may stimulate in principal cells.112
Cl -dependent K secretion.106 Recent pharmacologic HK -1 and HK -2 are both constitutively expressed in
studies of perfused tubules are consistent with K -Cl co- the distal nephron. However, tubule perfusion of K -replete
transport mediated by the KCCs101; however, of the three animals suggests a functional dominance of omeprazole/SCH-
renal KCCs, only KCC1 is apically expressed in the distal 28080-sensitive, ouabain-resistant H -K –ATPase activity
nephron. Other functional possibilities for Cl -dependent that is consistent with holoenzymes containing HK -1.113 K
K secretion include the parallel operation of apical H -K - deprivation increases the overall activity of H -K –ATPase
exchange and Cl -HCO3 exchange in type B intercalated in the collecting duct, with the emergence of a ouabain-
cells100 and parallel K and Cl channels in type A interca- sensitive H -K –ATPase activity.114 This is consistent with
lated cells (see Fig. 6.4).88 a relative dominance of HK -2 during K -restricted condi-
A provocative study by Frindt and Palmer72 under- tions. Consistent with this pharmacology, K -restriction also
scores the importance of ENaC-independent K excretion, induces a dramatic upregulation of HK -2 transcript and
be it mediated by apical K -Cl cotransport and/or by protein in the outer and inner medulla during K depletion;
other mechanisms (see also Integrated Regulation of Distal HK -1 expression is unaffected.115,116 Mice with a targeted
Sodium and Potassium Transport). Rats were infused with deletion of HK -2 exhibit lower plasma and muscle K than
amiloride via osmotic minipumps, generating urinary con- wild-type littermates when maintained on a K -de cient
centrations considered suf cient to inhibit 98% of ENaC diet. However, this appears to be due to a marked loss of K
activity. Whereas amiloride almost abolished K excretion in the colon rather than the kidney, because renal K excre-
in rats on a normal K intake, acute and long-term high K tion is appropriately reduced in the K -depleted knockout
diets led to an increasing fraction of K excretion that was mice.110 Presumably, the lack of an obvious renal phenotype
independent of ENaC activity ( 50% after 7 to 9 days on a in either HK -1110 or HK -2110 knockout mice re ects the
high K diet). marked redundancy in the expression of HK subunits in
the distal nephron. Indeed, collecting ducts from the HK -
1 knockout mice have signi cant residual ouabain-resistant
Potassium Reabsorption by the and SCH-28080-sensitive H -K –ATPase activities, consis-
Collecting Duct tent with the expression of other HK subunits that con-
In addition to K secretion, the distal nephron is capable of fer characteristics similar to the “gastric” H -K –ATPase.110
considerable K reabsorption, primarily during the restric- However, data from HK -1 and HK -2 knockout mice sug-
tion of dietary K .22,107,108 This reabsorption is accomplished gest that compensatory mechanisms in these mice are not
in large part by intercalated cells in the OMCD, via the ac- accounted for by ATPase-type mechanisms.117
tivity of apical H /K –ATPase pumps. Under K -replete The importance of K reabsorption mediated by the
conditions, apical H /K –ATPase activity recycles K with collecting duct is dramatically illustrated by the pheno-
an apical K channel, without effect on transepithelial K type of transgenic mice with generalized overexpression
absorption (see Figure 6.4). Under K -restricted conditions, of a gain-of-function mutation in H -K –ATPase; this
K absorbed via apical H /K –ATPase appears to exit inter- effectively bypasses the redundancy and complexity of
calated cells via a basolateral K channel, thus achieving the this reabsorptive pathway. This mutant HK subunit has
transepithelial transport of K .109 a tyrosine-to-alanine mutation within the C-terminal tail
H -K –ATPase holoenzymes are members of the P-type that abrogates regulated endocytosis from the plasma mem-
family of ion transport ATPases, which also includes subunits brane; these mice have higher plasma K than their wild-
of the basolateral Na -K –ATPase.110 HK -1 and HK -2 are type littermates, with approximately half the fractional
also referred to as the gastric and colonic subunits, respec- excretion of K .118
tively. A speci c HK subunit interacts with the HK sub- Finally, it should be noted that considerable evidence
units to ensure delivery to the cell surface and a complete implicates the H -K –ATPases in renal acid secretion.110 In
expression of H -K –ATPase activity; HK -2 subunits are particular, acid extrusion from intercalated cells is markedly
also capable of interaction with Na -K –ATPase subunits. reduced in doubly de cient HK -1/HK -2 knockout
The pharmacology of H -K –ATPase holoenzymes differs mice.110 In addition, the H -K –ATPases may function di-
considerably, such that the gastric HK -1 is classically rectly in Na balance, collaborating, for example, with api-
sensitive to the H -K –ATPase inhibitors SCH-28080 and cal Cl :HCO3 exchangers in the CCD in apical Na -Cl
omeprazole and is resistant to ouabain; the colonic HK -2 uptake.110
CHAPTER 6 TUBULAR POTASSIUM TRANSPORT 2203
03

The Regulation of Distal Tubular Aldosterone also has clinically relevant effects on K
Potassium Transport homeostasis, with a clear relationship at all levels of serum
K between circulating levels of the hormone and the ability
Modulation of ROMK Activity to excrete K .
ROMK and other KIR channels are inward rectifying (i.e., K Renin released from the kidney stimulates aldosterone
ows inward more readily than outward). Even though out- release from the adrenal via angiotensin-II (AT-II). Renin
ward conductance is usually less than inward conductance, secretion by juxtaglomerular cells within the afferent ar-
K ef ux through ROMK predominates in the CNT and teriole is initiated in response to a signal from the macula
CCD because the membrane potential is more positive than densa, speci cally a decrease in luminal chloride transported
the equilibrium potential for K . Intracellular magnesium through the Na -K -2Cl cotransporter (NKCC2) at the
(Mg2 )119 and polyamines120 play key roles in inward recti- apical membrane of macula densa cells.6 In addition to this
cation, binding and blocking the pore of the channel from macula densa signal, decreased renal perfusion pressure and
the cytoplasmic side.121 A single transmembrane residue, as- renal sympathetic tone stimulate renal renin secretion.
paragine-171 in ROMK1, controls the af nity and the block- Hyperkalemia is also an independent and synergistic
ing effect of both Mg2 and polyamines.119,120 Intracellular stimulus for aldosterone release from the adrenal gland,129 al-
Mg2 in TAL, DCT, CNT, and principal cells is thought to though dietary K loading is less potent than dietary Na -Cl
have a signi cant effect on ROMK activity, because it inhib- restriction in increasing circulating aldosterone.130 The rest-
its outward ROMK-dependent currents in principal cells.122 ing membrane potential of adrenal glomerulosa cells is hy-
The blocking af nity of Mg2 is enhanced at lower extracel- perpolarized due to the activity of the “leak” K channels
lular K concentrations,122 which should aid in reducing K TASK-1 and TASK-3; the combined deletion of genes encod-
secretion during hypokalemia and K de ciency. A reduc- ing these channels leads to baseline depolarization of adre-
tion of this intracellular Mg2 block may also explain the hy- nal glomerulosa cells and an increase in plasma aldosterone
pokalemia associated with hypomagnesemia, wherein distal that is resistant to dietary sodium loading.20 The KCNJ5 K
K secretion is enhanced.121 channel also plays a role, in that mutations that produce a
In addition to inward recti cation, the endogenous depolarizing acquisition of KCNJ5-mediated Na conduc-
ROMK channels in the TAL and principal cells exhibit a very tance are associated with adrenal adenomas.21 AT-II and K
high channel open probability (Po). The high Po of ROMK both activate Ca2 entry in glomerulosa cells via voltage-
is maintained by the combined effects of the binding of sensitive T-type Ca2 channels, primarily Cav3.2.131 Eleva-
phosphatidylinositol-4,5-bisphosphate (PIP2) to the channel tions in extracellular K thus depolarize glomerulosa cells
protein, direct channel phosphorylation by protein kinase and activate these Ca2 channels, which are independently
A (PKA), ATP binding to the ROMK-CFTR complex, and and synergistically activated by AT-II. The calcium-depen-
cytoplasmic pH. PIP2 binding to ROMK is thus required to dent activation of calcium-calmodulin (CaM)-dependent
maintain the channel in an open state,123 whereas cytoplas- protein kinase, in turn, activates the synthesis and release
mic acidi cation inhibits the channel. PKA phosphorylates of aldosterone via the induction of aldosterone synthase. K
ROMK protein at one N-terminal serine and two C-terminal and AT-II also enhance the transcription of the Cav3.2 Ca2
serines124: S25, S200, and S294 in the ROMK2 isoform. channel by abrogating repression of this gene by the neuron
Phosphorylation of all three sites is required for full channel restrictive silencing factor (NRS); this ultimately ampli es
function. The phosphorylation of the N-terminal site over- the induction of aldosterone synthase.131
rides the effect of a carboxy-terminal endoplasmic reticulum The adrenal release of aldosterone due to increased K
retention signal, thus increasing the expression of the chan- is dependent on an intact adrenal renin–angiotensin system,
nel protein at the cell membrane.125 The phosphorylation of particularly during Na -Cl restriction. Angiotensin con-
S200 and S294 maintains the channel in a high Po state, in verting enzyme (ACE) inhibitors and angiotensin–receptor
part by modulating the effects of PIP2,126 ATP,60 and pH.127 blockers (ARBs) completely abrogate the effect of high K on
Because ROMK channels exhibit such a high Po, the salt-restricted adrenals.132 Direct, G protein–dependent acti-
physiologic regulation of the channel is primarily achieved vation of the TASK-1 and/or TASK-3 K channels by AT1A
by regulated changes in the number of active channels on or AT1B receptors is thought to underlie the effect of AT-II on
the plasma membrane. The associated mechanisms are dis- adrenal aldosterone release,20 with abrogation of this effect by
cussed in the context of the adaptation to K loading/hyper- ARBs or ACE inhibitors. Other clinically relevant activators
kalemia and K deprivation/hypokalemia. of adrenal aldosterone release include prostaglandins and cat-
echolamines via increases in cAMP.133 Finally, atrial natriuretic
Aldosterone and Potassium Secretion peptide (ANP) exerts a potent negative effect on aldosterone
Aldosterone has a potent kaliuretic effect,128 with impor- release induced by K and other stimuli,134 at least in part by
tant interrelationships between circulating K and aldoste- inhibiting early events in aldosterone synthesis.135 ANP inhib-
rone. Aldosterone release by the adrenal is thus induced by its both renal renin release and adrenal aldosterone release,
hyperkalemia and/or a high K diet,129 suggesting an im- functions that may be central to the roles of this hormone in
portant feedback effect of aldosterone on K homeostasis.130 the pathophysiology of hyporeninemic hypoaldosteronism.134
204
20 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Within the kidney, aldosterone has no effect on the den- SGK1 modulates membrane expression of ENaC by in-
sity of apical SK channels in the CCD136; it does however in- terfering with regulated endocytosis of its channel subunits.
duce a marked increase in the density of apical Na channels Speci cally, the kinase interferes with interactions between
in the CNT and CCD.136 Aldosterone activates ENaC via in- ENaC subunits and the ubiquitin-ligase Nedd4-2.142 PPxY
terrelated effects on the transcription, synthesis, traf cking, domains in the C-termini of all three ENaC subunits bind to
and membrane-associated activity of the subunits encoding WW domains of Nedd4-2. Coexpression of Nedd4-2 with
the channel. Aldosterone is thus induced by a high K diet a wild-type ENaC channel results in a marked inhibition of
and strongly stimulates apical ENaC activity, which gener- channel activity due to retrieval from the cell membrane;
ates the lumen-negative PD that stimulates K secretion by Nedd4-2 is thought to ubiquitinate ENaC subunits, result-
principal cells. The hormone also has signi cant effects on ing in the removal of channel subunits from the cell mem-
the basolateral membrane of principal cells, with dramatic brane and degradation in lysosomes and the proteosome.142
changes in cellular morphology and the length of basolateral A PPxY domain in SGK1 also binds to Nedd4-2, which
membranes.137 This is accompanied by an increase in baso- is a phosphorylation substrate for the kinase; the phos-
lateral Na -K –ATPase activity, although it has been dif - phorylation of Nedd4-2 by SGK1 abrogates its inhibitory
cult to determine how much of these cellular and functional effect on ENaC subunits.145,146 Aldosterone also stimulates
changes are due to enhanced Na entry via apical ENaC. It Nedd4-2 phosphorylation in vivo147 and Nedd4-2 phos-
is, however, known that aldosterone increases the expression phorylation, in turn, results in the ubiquitin-mediated deg-
of the Na -K –ATPase 1 and 1 subunits in the CCD138; radation of SGK1.148
these effects are evidently independent of ENaC activity.139 Finally, aldosterone indirectly activates ENaC channels
Transcriptional effects of aldosterone also include the through the induction of channel activating proteases, which
induction of the -ENaC subunit via a glucocorticoid- increase open channel probability by the cleavage of the extra-
response element in the promoter of the SCNN1A gene. Al- cellular domain of - and -ENaC. Proteases that have been
dosterone also relieves a tonic inhibition of the SCNN1A gene implicated in the processing of ENaC include furin, elastase,
by a complex that includes the Dot1a (disruptor of telomere plasmin, kallikrein, and three novel, membrane-associated
silencing splicing variant a) and the AF9 and AF17 tran- proteases called channel activating proteases 1, 2, and
scription factors.140 This transcriptional activation results 3(CAP1, 2, and 3).149–151 Proteolytic cleavage of ENaC ap-
in an increased abundance of -ENaC protein in response pears to activate the channel by removing the self-inhibitory
to either exogenous aldosterone or dietary Na -Cl restric- effect of external Na 150; in furin-mediated proteolysis of
tion 141; the response to Na -Cl restriction is blunted by ENaC, this appears to involve the removal of an inhibi-
spironolactone, indicating involvement of the mineralocorti- tory domain from within the extracellular loop.152 The struc-
coid receptor. At the baseline, -ENaC transcripts in the kid- tures of the extracellular domains of ENaC and the related
ney are less abundant than those encoding - and -ENaC. channels resemble an outstretched hand holding a ball, with
All three subunits are required for the ef cient processing de ned subdomains termed the wrist, nger, thumb, palm,
of heteromeric channels in the endoplasmic reticulum and -ball, and knuckle; functionally relevant proteolytic events
traf cking to the plasma membrane, such that the induction target the nger domains of ENaC subunits.151 Unprocessed
of -ENaC is thought to relieve a major bottleneck in the channels at the plasma membrane are thought to function
processing and traf cking of active ENaC complexes.142 as a reserve pool, capable of rapid activation by membrane-
Aldosterone also plays an indirect role in the regulated associated luminal proteases.150
traf cking of ENaC subunits to the plasma membrane via
the regulation of accessory proteins that interact with pre- Adaptation to High Potassium Intake
existing ENaC subunits. Aldosterone rapidly induces the Much of the renal adaptation to high K intake is aldoste-
expression of a serine-threonine kinase denoted SGK1.143 rone independent. For example, a high K diet increases
The coexpression of SGK1 with ENaC subunits in Xenopus apical Na reabsorption and K secretion in the CCD in
oocytes results in a dramatic activation of the channel due to adrenalectomized animals that lack aldosterone.153 At the
increased expression at the plasma membrane.141 Analogous tubular level, when basolateral K is increased, there is a sig-
in vivo redistribution of ENaC subunits occurs in the CNT ni cant activation of the Na /K –ATPase, accompanied by
and early CCD from a largely cytoplasmic location during a secondary activation of apical Na and K channels.154 In-
dietary Na -Cl excess to a purely apical distribution after creased dietary K also markedly increases the density of SK
aldosterone or Na -Cl restriction.71,141 Furthermore, there channels in the CCD (Fig. 6.5), along with a modest increase
is a temporal correlation between the appearance of induced in ENaC density.136 This is associated with changes in the
SGK1 protein in the CNT and the redistribution of ENaC subcellular distribution of the ROMK protein, with an in-
protein to the plasma membrane.141 The amiloride-sensitive, crease in apical expression.155 Notably, this increase in ENaC
lumen-negative potential difference generated by ENaC is and SK density in the CCD occurs within hours of assuming
reduced in SGK1 knockout mice,144 resulting in a decreased a high K diet, with a minimal associated increase in cir-
driving force for distal K secretion and marked susceptibil- culating aldosterone.156 In contrast, a week of low Na -Cl
ity to hyperkalemia. intake, with almost a 1000-fold increase in aldosterone, has
CHAPTER 6 TUBULAR POTASSIUM TRANSPORT 2205
05

these channels are also activated by dietary K loading.


Flow-stimulated K secretion by the CCD of both mice77
and rats158 is enhanced on a high-K diet, with an absence
of ow-dependent K secretion in rats on a low K diet.158
This is accompanied by commensurate changes in transcript
levels for - and 2–4-subunits of the BK channel proteins
in microdissected CCDs ( 1 subunits are restricted to the
CNT75). Traf cking of BK subunits is also affected by di-
etary K , with largely intracellular distribution of -subunits
in K -restricted rats and prominent apical expression in
K -loaded rats.158 Aldosterone does not contribute to the
regulation of BK channel activity or expression in response
to a high K diet.159
Regulatory information is notably more extensive for
ROMK/SK channels than for BK channels. ROMK channels
exhibit a high Po; physiologic regulation of SK channels is
primarily achieved by regulated changes in the number of
active channels on the plasma membrane. The changes in
traf cking and/or activity of the ROMK channel that are in-
duced by dietary K restriction appear to involve tyrosine
phosphorylation of the ROMK protein (see the following);
K loading reverses this tyrosine phosphorylation. The for-
ward traf cking of ROMK channels to the apical membrane
of CNT and principal cells after K loading is also phosphory-
lation dependent. PKA phosphorylates ROMK protein at
one N-terminal serine and two C-terminal serines.124 Phos-
phorylation of the N-terminal serine overrides the effect of
a carboxy-terminal endoplasmic reticulum retention signal,
thus inducing exit from the Golgi apparatus and expression
of the channel protein at the cell membrane.125 This explains
in large part the induction of SK channel density by vaso-
pressin 160 and cAMP.76 Notably, this N-terminal serine is also
FIGURE 6.5 A high K diet rapidly activates small conductance a substrate for SGK1, the aldosterone-induced kinase that
(SK) channels in the cortical collecting duct (CCD), which is activates ENaC, which then activates ROMK by increasing
mediated by the renal outer medullary K channel (ROMK) membrane expression.161 Thus, although aldosterone does
(Kir 1.1) K channel. Histograms of N (channels/patch) are shown not appear to have signi cant effects on SK density,156 SGK1
for rats on a control diet (A), on a high Kdiet for 6 hours (B), and is conceivably involved; alternatively, other antagonistic
on a high Kdiet for 48 hours (C). Each determination of N repre- effects of SGK1 on ROMK162 traf cking abrogate the effect
sents a single cell-attached patch. The high K diet results in a mediated by N-terminal phosphorylation.
progressive recruitment of SK channels at the apical membrane. A recent series of reports have linked changes in the ex-
(From Palmer LG, Frindt G. Regulation of apical K channels in rat pression of WNK1 kinase subunits in the response to high K
cortical collecting tubule during changes in dietary Kintake. diet. WNK1 and WNK4 were initially identi ed as causative
Am J Physiol. 1999;277:F805–812.) genes for familial hypertension with hyperkalemia (FHHt),
also known as Gordon syndrome or pseudohypoaldosteron-
ism type II.163 The regulation of NCC, the thiazide-sensitive
no effect on SK channel density, nor for that matter does Na -Cl cotransporter in the DCT, is a major role of the
2 days of aldosterone infusion, despite the development WNK kinases.164–166 NCC activity is a major determinant of
of hypokalemia (Table 6.1).156 Of note, unlike the marked Na delivery to the downstream CNT, with potent effects
increase seen in the CCD,136,156 the density of SK channels on the ability to secrete K (see also Integrated Regulation
in the CNT is not increased by high dietary K .65 This ap- of Distal Sodium and Potassium Transport). However, the
pears to be due to dif culties in estimating channel densities WNK kinases also directly regulate traf cking and activity of
in small membrane patches, because the measurement of both ROMK and BK channels.
whole cell currents indicates an upregulation of ROMK ac- ROMK expression at the membrane of Xenopus oocytes
tivity in the CNT of animals maintained on a high K diet.157 is dramatically reduced by the coexpression of WNK4;
BK channels in the CNT and CCD play an important FHHt-associated mutations dramatically increase this effect,
role in the ow-activated component of distal K excretion 75; suggesting a direct inhibition of SK channels in FHHt.167
206
20 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

TA B L E

6.1 The Effect of a High K Diet, Aldosterone, and/or Na -Cl Restriction on


Small Conductance Channel Density in the Rat Cortical Collecting Duct
K Channel Plasma Aldo
Condition Density/ m2 (ng/dL) Plasma K (mM)

Control 0.41 15 3.68


High K diet, 6 h 1.51 36 NM
High K diet, 48 h 2.13 98 4.37
Low Na diet, 7 d 0.48 1260 NM
Aldo infusion, 48 h 0.44 550 2.44

Aldo high K diet 0.32 521 3.80

K, potassium; h, hour; Na, sodium; d, day; Aldo, aldosterone; NM, not measured.
Modi ed from Palmer LG, Frindt G. Regulation of apical K channels in rat cortical collecting tubule during changes in
dietary K intake. Am J Physiol. 1999;277:F805–F812.

The coexpression of WNK4 also inhibits BK channels in


a heterologous expression system, apparently by inducing
lysosomal degradation.168 In the case of WNK1, the analysis
is complicated dramatically by the transcriptional complex-
ity of its gene, which has at least three separate promoters
and a number of alternative isoforms. In particular, the pre-
dominant intrarenal WNK1 isoform is generated by a distal
nephron transcriptional site that bypasses the N-terminal
exons that encode the kinase domain, thus yielding a
kinase-de cient short form of the protein (“WNK1-S”).169
Full-length WNK1 (WNK1-L) inhibits ROMK activity by
inducing endocytosis of the channel protein 170–172; kinase ac-
tivity and/or the N-terminal kinase domain of WNK1 appear
to be required for this effect,171,172 although Cope et al.170
have reported that a kinase-dead mutant of WNK1 is un-
impaired. WNK1 and WNK4 induce endocytosis of ROMK
via an interaction with intersectin, a multimodular endocytic
scaffold protein.173 The additional binding of ROMK to the
clathrin adaptor protein autosomal recessive hypercholester-
olemia (ARH) is required for basal and WNK1-stimulated FIGURE 6.6 The balance between the major short (WNK1-S)
endocytosis of the channel protein.174 The ubiquitination of and long (WNK1-L) isoforms of the WNK1 kinases affects K
the ROMK protein is also involved in clathrin-dependent en- secretion. The WNK1-S isoform lacks a kinase domain and in-
docytosis, requiring an interaction between the channel and hibits the effects of the long isoform. Potassium loading causes
the U3 ubiquitin ligase POSH (plenty of SH domains).175 a reduction in the ratio of WNK1-L to WNK1-S, resulting in an
The shorter WNK1-S isoform, which lacks the kinase increased expression of the renal outer medullary K channel
domain, appears to inhibit the effect of WNK1-L.171,172 The (ROMK) at the plasma membrane of connecting tubule (CNT)
ratio of WNK1-S to WNK1-L transcripts is reduced by K cells and principal cells, with a concomitant reduction in the api-
restriction (greater endocytosis of ROMK)172,176 and is in- cal expression of the thiazide-sensitive Na -Cl cotransporter
creased by K loading (reduced endocytosis of ROMK),171,176 (NCC) in the distal convoluted tubule (DCT) and the furosemide-
thus suggesting that this ratio between WNK1-S and WNK1- sensitive Na -K -2Cl cotransporter NKCC2 in the thick ascend-
L functions as a “switch” to regulate distal K secretion ing limb (TAL). The net result is increased Na delivery to the
(Fig. 6.6). The inhibitory effect of WNK1-S tracks to the rst CNT and principal cells with enhanced apical secretory K chan-
253 amino acids of the protein, encompassing the initial 30 nel activity, resulting in an enhanced K secretion.
CHAPTER 6 TUBULAR POTASSIUM TRANSPORT 2207
07

amino acids unique to this isoform and an adjacent auto- reducing the distal tubular K secretion.189 IGF-1 is produced
inhibitory domain.177 Transgenic mice that overexpress this in the kidney and upregulated by dietary K restriction190.
inhibitory domain of WNK1-S have lower serum K con- The downstream activation of PI3-kinase by insulin and IGF-
centrations, higher fractional excretion of K , and increased 1 results in the Akt1- or SGK1-dependent phosphorylation of
expression of the ROMK protein at the apical membrane of WNK1, leading to endocytosis of ROMK.162
CNT and CCD cells, all of which are consistent with an im- Reports of transient postprandial kaliuresis in sheep, in-
portant inhibitory effect of WNK1-S.177 dependent of changes in plasma K or aldosterone, suggest
that an enteric or hepatoportal K sensor controls kaliure-
Potassium Deprivation sis via a sympathetic re ex191; tissue kallikrein has recently
A reduction in dietary K leads, within 24 hours, to a dramatic emerged as a candidate mediator for this postprandial kaliure-
drop in urinary K excretion.176,178 This drop in excretion sis (see the following). Regardless of the signaling involved,
is due to both an induction of reabsorption by intercalated changes in dietary K absorption have a direct “anticipatory”
cells in the OMCD107,108 and to a reduction in SK channel effect on K homeostasis in the absence of changes in plasma
activity in principal cells.179 The mechanisms involved in K K . Such a feed-forward control has the theoretical advan-
reabsorption by intercalated cells were previously discussed. tage of greater stability, because it operates prior to changes
Notably, H /K –ATPase activity in the collecting duct does in plasma K .192 Notably, changes in ROMK phosphoryla-
not appear to be regulated by aldosterone180; stimulatory tion status and insulin-sensitive muscle uptake can be seen
effects of mineralocorticoid on H /K –ATPase activity are in K -de cient animals in the absence of a change in plasma
abrogated by K loading, indicating a primary role for K ,13 suggesting that the upstream activation of the major
hypokalemia.181 mechanisms that serve to reduce K excretion (reduced K
Dietary K intake modulates traf cking of the ROMK secretion in the CNT/CCD, decreased peripheral uptake, and
channel protein to the plasma membrane of principal cells, increased K reabsorption in the OMCD) does not require
with a marked increase in the relative proportion of intracel- changes in the plasma K . Consistent with this hypothesis,
lular channel protein in K -depleted animals155 and clearly moderate K restriction—without an associated drop in
de ned expression at the plasma membrane of CCD cells plasma K —is suf cient to induce AT-II-dependent superox-
from animals on a high K diet.155 The membrane insertion ide generation and c-src activation, leading to the inhibition
and activity of ROMK is modulated by tyrosine phosphory- of ROMK channel activity.189
lation of the channel protein, such that the phosphoryla-
tion of tyrosine residue 337 stimulates endocytosis and Vasopressin
dephosphorylation induces exocytosis.182,183 This tyrosine
Vasopressin has a stimulatory effect on K secretion by the
phosphorylation appears to play a key role in the regula-
distal nephron.32 This serves to preserve K secretion during
tion of ROMK by dietary K .184 Whereas the levels of pro-
dehydration and extracellular volume depletion, when cir-
tein tyrosine phosphatase-1D do not vary with K intake,
culating levels of vasopressin are high and tubular delivery
intrarenal activity of the cytoplasmic tyrosine kinases c-src
of Na and uid is reduced. The stimulation of basolateral
and c-yes are inversely related to dietary K intake, with a
V2 receptors results in the activation of ENaC, which in-
decrease under high K conditions and a marked increase
creases the driving force for K secretion by CNT cells and
after several days of K restriction.179,185 Immunolocaliza-
principal cells.193 In addition, vasopressin activates SK chan-
tion indicates coexpression of c-src with ROMK in the TAL
nels directly in the CCD,160 as does cAMP.76 The ROMK
and the principal cells of the CCD.155 The inhibition of pro-
protein is directly phosphorylated by PKA on three serine
tein tyrosine phosphatase activity, leading to a dominance of
residues (S25, S200, and S294 in the ROMK2 isoform), with
tyrosine phosphorylation, dramatically increases the propor-
phosphorylation of all three sites required for full activity
tion of intracellular ROMK in the CCD of animals on a high
(see also the previous discussion). Finally, the stimulation of
K diet.155
luminal V1 receptors also increases K secretion in the CCD,
The neurohumoral factors that induce the K -dependent
apparently via the activation of BK channels.194
traf cking and expression of apical ROMK155 and BK chan-
nels158 have only recently been identi ed. Several studies have
implicated the intrarenal generation of superoxide anions Tissue Kallikrein
in the activation of cytoplasmic tyrosine kinases and in the The serine protease tissue kallikrein (TK) is involved in
downstream phosphorylation of the ROMK channel protein the generation of kinins, which ultimately stimulates the
by K depletion.186,187 Candidates for the upstream kaliuretic formation of bradykinin.195 Within the kidney, TK is synthe-
factor include AT-II and growth factors such as insulin-like sized within CNT cells and is released into the tubular lumen
growth factor-1 (IGF-1).162 AT-II inhibits ROMK activity in and the peritubular interstitium. Although TK-induced bra-
K -restricted rats, but not in rats on a normal K diet.188 This dykinin has a number of effects on distal tubular physiol-
inhibition involves the downstream activation of superoxide ogy,195 more recent data have revealed a provocative role in
production and c-src activity, such that the well-known induc- postprandial kaliuresis. Thus, oral K -Cl loading leads to
tion of AT-II by a low K diet appears to play a major role in a spike in urinary K and TK excretion in rats, mice, and
208
20 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

humans.195 The increase in urinary TK after K loading is in certain circumstances associated with a marked induction
not accompanied by changes in urinary aldosterone and can of aldosterone, such as dietary sodium restriction, sodium
be detected in aldosterone synthase knockout mice.149 Mice balance is maintained without affecting K homeostasis.
de cient in TK demonstrate postprandial hyperkalemia, in- This aldosterone paradox—the independent regulation of
dicating a role for the protease in postprandial kaliuresis. Na -Cl and K handling by the aldosterone-sensitive distal
This transient hyperkalemia is accompanied by a marked nephron—is only recently beginning to yield to investiga-
increase in K reabsorption by perfused CCDs due to an tive efforts. The major factors in the integrated control of
upregulation of H /K –ATPase activity and an increase in Na -Cl and K transport appear to include electroneutral,
HK -2 transcript. The addition of luminal but not basolat- NCC-independent, and thiazide-sensitive Na -Cl transport
eral TK inhibits the activated CCD H /K –ATPase activity within the CCD196,197,199; K -dependent changes in NCC
in TK knockout mice, which is consistent with direct pro- activity within the DCT; ENaC-independent K excretion
teolytic activation. There is also a marked increase in Na within the distal nephron 72; and the differential regulation
reabsorption by perfused CCDs from TK knockout mice, of various transport and signaling pathways by aldosterone,
without the development of a lumen-negative PD. This is AT-II, TK, and dietary K .195,200,201
consistent with an increased activity of the electroneutral Thiazide-sensitive electroneutral Na -Cl transport
Na -Cl cotransport mediated by the Na -driven SLC4A8 within the CCD is mediated by parallel activity of the Na -
Cl -HCO3 exchanger and the SLC26A4 Cl -HCO3 ex- driven SLC4A8 Cl -HCO3 exchanger and the SLC26A4
changer (see also the following text).196 This electroneu- Cl -HCO3 exchanger,196 expressed in intercalated cells.
tral transport pathway had previously been shown to be The molecular identity of this transport mechanism has only
inhibited by bradykinin 197; hence, the activation by TK recently emerged 196; hence, regulatory in uences are not ful-
deletion presumably re ected a loss of tonic inhibition by ly characterized. However, electroneutral Na -Cl transport
TK-generated bradykinin. Prior data had indicated that TK within the CCD is evidently induced by both volume deple-
mediates proteolytic cleavage of the subunit of ENaC, with tion and mineralocorticoid treatment.196,197,199 This mecha-
reduced ENaC activity in TK-de cient mice198; net Na bal- nism mediates 50% of Na reabsorption in the CCD under
ance is thus neutral in these mice. these conditions, all without affecting the lumen potential
In summary, TK secretion from CNT cells is induced difference and thus without in uencing K excretion. A high
by oral K -Cl loading, causing proteolytic activation of K diet also increases the fraction of ENaC-independent,
ENaC198 and thus an increase in ENaC-driven K secre- amiloride-resistant K excretion to 50%; again, this elec-
tion and the bradykinin-dependent inhibition of electroneu- troneutral, aldosterone-independent pathway for K excre-
tral Na -Cl cotransport in the CCD.196,197 Thus, a further tion uncouples distal tubular Na and K excretion.72
augmentation of electrogenic Na transport (favoring K In a landmark study, Kahle et al.167 established in 2003
secretion) and a direct luminal inhibition of H /K –ATPase that the WNK4 kinase, encoded by a disease gene for FHHt,
activity causes a decrease or tonic inhibition of K reabsorp- inhibits ROMK activity in Xenopus oocytes. This identi ed
tion. TK may very well be the postprandial factor191 that WNK-dependent signaling as a major pathway for integrat-
functions in the feed-forward control of plasma K .192 ing Na -Cl and K transport within the distal nephron.
Details of the relevant effects of WNK kinases on NCC and
ROMK are discussed earlier and elsewhere.202–204 However,
The Integrated Regulation of Distal Sodium
key ndings include the differential in uence of K intake
and Potassium Transport on circulating AT-II, ROMK activity (i.e., K secretory capac-
In CNT and principal cells, the lumen-negative potential dif- ity), the ratio of WNK1 isoforms, and the activity of NCC
ference generated by Na entry via ENaC induces the exit in the DCT. AT-II activates NCC via WNK4 and the down-
of K via apical K -selective channels. This arrangement stream SPAK kinase,202–204 thus reducing delivery of Na to
explains much of the known physiology and pathophysiol- the CNT and limiting K secretion. AT-II also inhibits ROMK
ogy of renal K secretion, yet has several key consequences activity via several mechanisms, including the downstream
that bear emphasis. First, enhanced Na -Cl reabsorption activation of c-src tyrosine kinases.187–189 Whereas K re-
upstream of the CNT and CCD will reduce the delivery of striction induces renin and circulating AT-II, increases in
luminal Na to the CNT and CCD,73 decrease the lumen- dietary K suppress AT-II levels.189,205 A decrease in circulat-
negative potential difference, and thus decrease K secretion; ing and local AT-II explains why NCC phosphorylation and
K secretion by the CCD essentially stops when luminal Na activity is downregulated by a high K diet (Fig. 6.7).56,206
drops below 8 mmol per liter.74 In this respect, the increas- Teleologically, this serves to increase the delivery of Na to
ingly re ned phenotypic understanding of FHHt, caused by the CNT, thus increasing K secretion. Finally, within CNT
kinase-induced gain-of-function of the DCT, has served to cells and principal cells, increases in aldosterone induce the
underline that a variation in NCC-dependent Na -Cl ab- SGK1 kinase, which phosphorylates WNK4 and attenuates
sorption, just upstream of the CNT, has major effects on the effect of WNK4 on ROMK,207 while activating ENaC via
the ability to excrete dietary K .166 Second, aldosterone is Nedd4-2-dependent effects. However, when dietary K in-
a kaliuretic hormone, induced by hyperkalemia. However, take is reduced, c-src tyrosine kinase activity increases under
CHAPTER 6 TUBULAR POTASSIUM TRANSPORT 2209
09

FIGURE 6.7 The differential effects of a high


and low K diet on the membrane expres-
sion of Na -Cl cotransporter (NCC) and
renal outer medullary K channel (ROMK) in
late distal convoluted tubule (DCT) and the
connecting tubule (CNT). A: Whereas NCC
expression at the plasma membrane of DCT2
cells is enhanced in rats treated with a low
K diet and is reduced by a high K diet, the
opposite occurs for ROMKprotein. B: This A
dichotomy persists at the junction between
DCT2 and the early CNT (CNT1: the juncture
is denoted by a dashed white line). However,
within CNT2 segments, which lack NCC, apical
labeling of ROMK under high K conditions is
less striking. (From Wade JB, et al. Differential
regulation of ROMK (Kir1.1) in distal nephron
segments by dietary potassium. Am J Physiol.
2011;300(6):F1385–1393.)

the in uence of increased AT-II, causing direction inhibition and hypotension due to marked suppression of membrane-
of ROMK activity via tyrosine phosphorylation of the chan- associated total and phosphorylated NCC in the DCT and
nel.182–184 The increase in c-src tyrosine kinase activity also NKCC2 in the TAL.209 In contrast, selective knockout of the
abrogates the effect of SGK1 on WNK4201; this latter mecha- WNK1-S exons leads to a major gain in function for NCC
nism can also be induced by AT-II.208 and NKCC2.209 Therefore, K loading will lead to an increase
The ratio of WNK1 isoforms is also a critical determinant in the ratio of WNK1-S to WNK1-L, reduced endocytosis of
in the balance between distal Na and K transport. The ROMK, and enhanced endocytosis of NCC; this will have
shorter WNK1-S isoform, which lacks the kinase domain, the effect of reducing Na absorption in the DCT and thus
appears to inhibit the effect of WNK1-L.171,172 The ratio of increasing Na delivery and tubular ow rate in the CNT,
WNK1-S to WNK1-L transcripts is reduced by K restriction where ROMK /– BK channel activity is maximized (see also
(greater endocytosis of ROMK)172,176 and is increased by K Fig. 6.6). The converse cascade occurs in K restriction.
loading (reduced endocytosis of ROMK).171,176 This suggests
that the ratio between WNK1-S and WNK1-L functions as
REFERENCES
a switch to regulate distal K secretion (see also Fig. 6.6).
Transgenic mice that overexpress this inhibitory domain 1. Coca
Coca SG,SG, Perazella
Perazella MA, Buller
Buller GK.
GK. The
The cardiovascular
cardiovascular implications of
o
hypokalemia. Am JJKidney
KidneyDis.
Dis. 2005;45(2):233–247.
2005;45(2):233–247.
of WNK1-S have lower serum K concentrations, a higher http://www.ncbi.nlm.nih.gov/pubmed/15685500
2. Lingrel JB, Croyle ML, Woo AL, Arguello JM. Ligand binding sites of
fractional excretion of K , and an increased expression of 2. Lingrel JB,
Na,K-ATPase. ActaCroyle
PhysiolML, Woo
Scand AL, 1998;643:69–77.
Suppl. Arguello JM. Ligand binding sites o Na,
ROMK protein at the apical membrane of CNT and CCD K-ATPase. Acta Physiol
3. McDonough AA,Scand
WangSuppl. 1998;643:69–77.
J, Farley RA. Signi cance of sodium pump isoforms
http://www.ncbi.nlm.nih.gov/pubmed/9789548
in digitalis therapy. J Mol Cell Cardiol. 1995;27(4):1001–1009.
cells, all of which are consistent with an important inhibitory 3. McDonough
4. Crambert G, AA, Wang
Hasler U, J,Beggah
Farley RA.
AT, Signif cance o sodium
et al. Transport pump iso orms
and pharmacological
effect of WNK1-S.177 In contrast, WNK1-L activates NCC in digitalis therapy.
properties of nineJ Mol Cell Cardiol.
different human1995;27(4):1001–1009.
Na, K-ATPase isozymes. J Biol Chem.
by enhancing expression at the cell membrane; this acti- 4. Crambert G, Hasler U, Beggah AT, et al. Transport and pharmacological
2000;275(3):1976–1986.
properties
5. LorenzoJN,nine di erent
Loreaux human Na, K-ATPase
EL, Dostanic-Larson isozymes. J Biol
I, et al. ACTH-induced Chem.
hyperten-
vation is blocked by WNK1-S. Again, transgenic WNK1-S 2000;275(3):1976–1986.
sion is dependent on the ouabain-binding site of the alpha2-Na -K -ATPase
mice have the expected phenotype, with renal salt wasting subunit. Am J Physiol Heart Circ Physiol. 2008;295(1):H273–280.
210
21 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

5. Hebert
6. Lorenz SC, JN, Mount
LoreauxDB, EL,Gamba
Dostanic-Larson
G. Molecular I, etphysiology
al. ACTH-induced hyperten-
of cation-coupled 29. Tabei
34. Wareing M, Wilson
K, Imai RW, Kibble
M. K transport in JD,
upperGreenportionR. Estimated
of descendingpotassiumlimbsref ofec tion
long-
sioncotransport:
Cl- is dependent the on the family.
SLC12 ouabain-binding
P ugers Arch. site2004;447(5):580–593.
o the alpha2-Na+-K+-ATPase coe nephron
loop cient in per fromused proximal
hamster. Am Jconvoluted tubules oPtthe
Physiol. 1987;252(3 anaesthetized rat in
2):F387–392.
subunit.
7. GosmanovAm J Physiol AR,Heart
WongCirc JA,Physiol.
Thomason2008;295(1):H273–280.
DB. Duality of G protein-coupled vivo.
35. JLiuPhysiol. 1995;488 T,
W, Morimoto (PtKondo
1):153–161.
Y, et al. Analysis of NaCl transport in thin
mechanisms
6. Hebertfor SC,beta-adrenergic
Mount DB, Gamba activation of NKCCphysiology
G. Molecular activity inoskeletal muscle.
cation-coupled http://www.ncbi.nlm.nih.gov/pubmed/8568651
ascending limb of Henle’s loop in CLC-K1 null mice. Am J Physiol Renal Physiol.
Am
Cl- Jcotransport:
Physiol Cell the Physiol.
SLC12 2002;283(4):C1025–1032.
amily Pf ugers Arch. 2004;447(5):580–593. 30. Johnston PA, Battilana CA, Lacy FB, Jamison RL. Evidence or a concentra-
2002;282(3):F451–457.
8. McDonough AA, Thompson CB, Youn JH. Skeletal muscle regulates extra-
http://www.ncbi.nlm.nih.gov/pubmed/12739168 tion
36.gradient
Unwin avoringR, Capasso outward G, movement
Giebisch G. o sodium
Potassium romand the thin
sodiumloop transport
o Henle.
cellular potassium.AR,
7. Gosmanov AmWong
J PhysiolJA,Renal Physiol. DB.
Thomason 2002;282(6):F967–974.
Duality o G protein-coupled J Clin Invest.
along the loop 1977;59(2):234–240.
of Henle: effects of altered dietary potassium intake. Kidney Int.
9. Bundgaard
mechanisms H, Schmidt TA,activation
or beta-adrenergic Larsen JS, o Kjeldsen
NKCC activity K. K insupplementation
skeletal muscle. http://www.ncbi.nlm.nih.gov/pubmed/833272
1994;46(4):1092–1099.
increases
Am J Physiol muscle [Na -K
Cell Physiol. -ATPase] and improves extrarenal K homeostasis
2002;283(4):C1025–1032. 31. Madsen
37. BattilanaKM, CA,Tishler
Dobyan CC. DC, Lacy FB,
Anatomy of et
theal.kidney.
E ect In:o chronic
Brenner potassium
BM, ed. Brennerload-
in rats. J Appl Physiol.
8. McDonough AA,1997;82(4):1136–1144.
Thompson CB, Youn JH. Skeletal muscle regulates extra- ing on
and potassium
Rector’s secretion
The Kidney. 7th by
ed.the pars recta or
Philadelphia, PA:descending
W. B. Saunders limb Co.;
o the juxtamed-
2004:3–72.
10. McKenna
cellular potassium. MJ.Am Effects of training
J Physiol on potassium
Renal Physiol. homeostasis during exercise.
2002;282(6):F967–974. ullary nephron
38. Allen in the rat.
F, Tisher CC.J Morphology
Clin Invest. 1978;62(5):1093–1103.
of the ascending thick limb of Henle.
J Mol
9. Cell Cardiol. H,
Bundgaard 1995;27(4):941–949.
Schmidt TA, Larsen JS, Kjeldsen K. K+ supplementation in- 32. Elalou
Kidney JM, Roinel N, de Rou gnac C. E ects o dDAVP on rat juxta-
Int. 1976;9(1):8–22.
11. Dluhy
creases muscle RG,[Na+Axelrod L, Williams
-K+-ATPase] GH. Serum
and improves immunoreactive
extrarenal K+ homeostasis insulin and
in rats. medullary
39. Tsuruoka nephrons:
S, Koseki stimulation
C, Muto o S,medullary
Tabei K, KImai recycling.
M. Axial Amheterogeneity
J Physiol. 1985; of
growth hormone
J Appl Physiol. response to potassium infusion in normal man. J Appl Physiol.
1997;82(4):1136–1144. 249(2 Pt 2):F291–298.
potassium transport across hamster thick ascending limb of Henle’s loop. Am J
1972;33(1):22–26.
http://www.ncbi.nlm.nih.gov/pubmed/9104850 33. Stokes
Physiol. JB. Consequences
1994;267(1 Pt 2):F121–129. o potassium recycling in the renal medulla. E -
12.
10. Choi CS, Thompson ects
40. oNielsen
ion transport by the medullary
S, Maunsbach thick ascending
AB, Ecelbarger CA, Knepper limb oMA. Henle’ s loop. J Clin
Ultrastructural
McKenna MJ. E ectsCB, Leong PK,
o training McDonough
on potassium AA, Youn during
homeostasis JH. Short-term
exercise.
K( ) deprivation provokes insulin resistance of cellular K( ) uptake revealed Invest. 1982;70(2):219–229.
localization of Na-K-2Cl cotransporter in thick ascending limb and macula densa
J Mol Cell Cardiol. 1995;27(4):941–949.
with the K( ) clamp. Am J Physiol Renal Physiol. 2001;280(1):F95–F102. http://www.ncbi.nlm.nih.gov/pubmed/6284797
of rat kidney. Am J Physiol. 1998;275(6 Pt 2):F885–893.
http://www.ncbi.nlm.nih.gov/pubmed/7563106
13. Chen P, Guzman JP, Leong PK, et al. Modest dietary K restriction 34. Y
41. Tabei
oshitomi K, Imai K, M.Kondo K transport
Y, ImaiinM. upper portionforo conductive
Evidence descending Cl- limbs o long-
pathways
11. Dluhy RG, Axelrod L, Williams GH. Serum immunoreactive insulin and
provokes insulin resistance of cellular K uptake and phosphorylation of renal loop nephron
across the cell rom hamster. of
membranes Amthe J Physiol. 1987;252(3
thin ascending Pt 2):F387–392.
limb of Henle’s loop. J Clin
growth hormone response to potassium in usion in normal man. J Appl Physiol.
outer medulla K channel without fall in plasma K concentration. Am J Physiol 35. Liu
Invest. W, Morimoto T, Kondo Y, et al. Analysis o NaCl transport in thin as-
1988;82(3):866–871.
1972;33(1):22–26.
Cell Physiol. 2006;290(5):C1355–1363. cending
42. Hebert limb SC, o Henle’
Andreoli s loop
TE. in CLC-K1
Control null mice.
of NaCl transportAm in J Physiol
the thick Renal Physiol.
ascending
12. Choi CS, Thompson CB, Leong PK, McDonough AA, Youn JH. Short-term
14. Al-Khalili L, Yu M, Chibalin AV. Na( ),K( )-ATPase traf cking in skeletal 2002;282(3):F451–457.
limb. Am J Physiol. 1984;246(6 Pt 2):F745–756.
K(+) deprivation provokes insulin resistance o cellular K(+) uptake revealed with 36. Rocha
UnwinAS, R, Kokko
Capasso G, Giebisch G. and Potassium and sodium
muscle: insulin stimulates translocation of both alpha(1)- and alpha(2)-subunit 43. JP. Sodium chloride water transport in thetransport
medul-
the K(+) clamp. Am J Physiol Renal Physiol. 2001;280(1):F95–F102. alongthick
the ascending
loop o Henle:
isoforms. FEBS Lett. 2003;536(1–3):198–202. lary limb eof ects
Henle. o altered
Evidence dietary potassium
for active chloride intake. Kidney
transport. Int.
J Clin
13. Feraille
15. Chen P,E,Guzman CarranzaJPML,, Leong Gonin PK,S, et
et al. Modest dietary stimulation
al. Insulin-induced K+ restriction of 1994;46(4):1092–1099.
Invest. 1973;52(3):612–623.
provokes insulin resistance o cellular K+ uptake
Na ,K( )-ATPase activity in kidney proximal tubule cells depends on phos- and phosphorylation o renal http://www.ncbi.nlm.nih.gov/pubmed/7861703
44. Greger R, Schlatter E. Presence of luminal K , a prerequisite for active
outer medulla
phorylation K+ alpha-subunit
of the channel without all in plasma
at Tyr-10. Mol BiolK+ concentration.
Cell. Am J Physiol
1999;10(9):2847–2859. 37. transport
NaCl Madsen KM, Tishler
in the CC. Anatomy
cortical o the kidney
thick ascending limbIn: ofBrenner
Henle’s BM, looped. ofBrenner
rabbit
Cell
16.Physiol.
Boini KM, 2006;290(5):C1355–1363.
Graf D, Kuhl D, Haussinger D, Lang F. SGK1 dependence of and Rector
kidney. ’ s TheArch.
P ugers Kidney. 7th ed. Philadelphia, PA: W. B. Saunders Co.; 2004:3–72.
1981;392(1):92–94.
14. Al-Khalili L,
insulin induced hypokalemia. Yu M, Chibalin P ugers AVArch.
Na(+),K(+)-ATPase
2009;457(4):955–961. tra cking in skeletal http://www.ncbi.nlm.nih.gov/pubmed/15548171
45. Greger R, Weidtke C, Schlatter E, Wittner M, Gebler B. Potassium activ-
muscle:
17. Olsson insulinAM,stimulates
Persson S,translocation
Schoder R. Effectso both alpha(1)- and
of terbutaline and alpha(2)-subunit
isoproterenol on ity38. AllenofF, isolated
in cells Tisher CC. Morphology
perfused corticalo thick
the ascending
ascendingthick limbs limb o Henle.
of rabbit Kid-
kidney.
iso orms. FEBS Lett. 2003;536(1–3):198–202.
hyperkalemia in nephrectomized rabbits. Scand J Urol Nephrol. 1978;12(1):35–38. neyugers
P Int. 1976;9(1):8–22.
Arch. 1984;401(1):52–57.
http://www.ncbi.nlm.nih.gov/pubmed/12586363
18. Scheid CR, Fay FS. Beta-adrenergic stimulation of 42K in ux in isolated 39. Simon
46. Tsuruoka DB,S,Karet
Koseki FE,C,Rodriquez-Soriano
Muto S, Tabei K, Imai J, et M. Axial heterogeneity
al. Genetic heterogeneity o
15. Feraille
smooth muscleE,cells. Carranza ML, Gonin
Am J Physiol. S, et al.Pt Insulin-induced
1984;246(5 1):C415–421. stimulation o potassium
of Bartter’s transport
syndromeacross revealed hamster thick ascending
by mutations in the Klimb o Henle’
channel, ROMK.s loop.Nature
Am J
Na19.+,K(+)-ATPase
Williams ME,activity GervinoinEV kidney
, Rosa proximal
RM, et al.tubule cells depends
Catecholamine on phos-
modulation of Physiol. 1994;267(1
Genetics. 1996;14:152–156. Pt 2):F121–129.
phorylation
rapid potassium o the alpha-subunit
shifts at Tyr-10.
during exercise. MolJBiol
N Engl Med.Cell. 1999;10(9):2847–2859.
1985;312(13):823–827. 40. Cantone
47. Nielsen S,A,Maunsbach
Yang X, Yan AB,Q, Ecelbarger
et al. Mouse CA, Knepper
model ofMA. typeUltrastructural
II Bartter’s syn- lo-
16. Davies
20. Boini KM,LA, Hu GraC, Guagliardo
D, Kuhl D,NA, Haussinger
et al. TASK D,channel
Lang F.deletion
SGK1 dependence
in mice causes o calizationI. oUpregulation
drome. Na-K-2Cl cotransporter in thick ascending
of thiazide-sensitive limb and macula
Na-Cl cotransport activity.densa
AmoJ
insulin induced
primary hypokalemia.Proc
hyperaldosteronism. Pf ugers Arch.Sci
Natl Acad 2009;457(4):955–961.
USA. 2008;105(6):2203–2208. rat kidney
Physiol Renal AmPhysiol.
J Physiol. 1998;275(6 Pt 2):F885–893.
2008;294(6):F1366–1372.
http://www.ncbi.nlm.nih.gov/pubmed/18665390
21. Choi M, Scholl UI, Yue P, et al. K channel mutations in adrenal 41. Taniguchi
48. Yoshitomi J,K,Guggino Kondo Y , Imai
WB. M. Evidence
Membrane stretch: or aconductive
physiological Cl-stimulator
pathways
17. Olsson AM, Persson
aldosterone-producing S, Schoder
adenomas andR. hereditary
E ects o terbutaline
hypertension. and isoproterenol
Science. 2011; on across
of Ca2the-activated
cell membranes K channels o the thin ascending
in thick ascendinglimb olimb. Henle’ Ams loop. J Clin 1989;
J Physiol. Invest.
331(6018):768–772.
hyperkalemia in nephrectomized rabbits. Scand J Urol Nephrol. 1978;12(1):35–38. 1988;82(3):866–871.
257(3 Pt 2):F347–352.
22. Malnic G, Klose RM, Giebisch G. Micropuncture study of renal potassium
http://www.ncbi.nlm.nih.gov/pubmed/635484 42. Bleich
49. HebertM, SC,Schlatter
AndreoliE, TE.Greger
Control R. oTheNaCl transport
luminal K inchannelthe thick of ascending
the thick
excretion
18. Scheid in the CR,rat.Fay
AmFS.J Physiol. 1964;206(4):674–686.
Beta-adrenergic stimulation o 42K inf ux in isolated limb. Am Jlimb
ascending Physiol. 1984;246(6
of Henle’ s loop.Pt P 2):F745–756.
ugers Arch. 1990;415(4):449–460.
23. Kaufman
smooth JS, Hamburger
muscle cells. Am J Physiol. RJ.1984;246(5
Passive potassium transport in the proximal
Pt 1):C415–421. 43. Wang
50. RochaWH. AS, Kokko
Two types JP Sodium chlorideinand
of K channel water
thick transport
ascending in the
limb medul
of rat lary
kidney.
convoluted
19. Williams tubule.ME,Am J Physiol.
Gervino EV,1985;248(2
Rosa RM, etPtal. 2):F228–232.
Catecholamine modulation o thickJ Physiol.
Am ascending limb o Henle.
1994;267(4 Evidence or active chloride transport. J Clin Invest.
Pt 2):F599–605.
24. Brandis
rapid potassium M, shi
Keyes J, Windhager
ts during exercise.EE. Potassium-induced
N Engl inhibition of proxi-
J Med. 1985;312(13):823–827. 1973;52(3):612–623.
51. Wang W, Lu M. Effect of arachidonic acid on activity of the apical K channel
mal tubular uid reabsorption in rats. Am J Physiol. 1972;222(2):421–427.
http://www.ncbi.nlm.nih.gov/pubmed/2858053 in44. Greger
the thick R, Schlatter
ascending limb E. Presence
of the o luminal
rat kidney. K +, a1995;106(4):727–743.
J Gen Physiol. prerequisite or active
25.
20. Wilson
Davies RW, LA, Wareing M, Green R.
Hu C, Guagliardo NA,Theetrole
al. of active
TASK transport
channel in potassium
deletion in mice NaCl
52. transport
Ho K, Nichols in theCG, cortical
LedererthickWJ,ascending
et al. Cloning limband o Henle’ s loopofo anrabbit
expression kid-
inwardly
reabsorption
causes primary in the proximal convolutedProc
hyperaldosteronism. tubuleNatlof the
Acad anaesthetized rat. J Physiol.
Sci USA. 2008;105(6): ney Pf ugers
rectifying Arch. 1981;392(1):92–94.
ATP-regulated potassium channel. Nature. 1993;362(6415):31–38.
1997;500
2203–2208. (Pt 1):155–164. 45. Palmer
53. Greger LG, R, Weidtke
Choe H,C,Frindt Schlatter
G. IsE,theWittner
secretory M, Gebler
K channel B. Potassium
in the rat activ-
CCT
26.
21. Kibble
Choi M, JD, Wareing
Scholl UI, M, Wilson
Yue P, RW, et al.Green
K+ R.channel
Effect ofmutations
barium on in potassium
adrenal ity in cells
ROMK? AmoJ Physiol.
isolated1997;273(3
per used cortical thick ascending limbs o rabbit kidney.
Pt 2):F404–410.
diffusion
aldosterone-producing adenomas and hereditary hypertension. Science. 2011;Am
across the proximal convoluted tubule of the anesthetized rat. 331J Pf54.
ugersXuArch. 1984;401(1):52–57.
JZ, Hall AE, Peterson LN, et al. Localization of the ROMK protein on apical
Physiol. 1995;268(4 Pt 2):F778–783.
(6018):768–772. 46. SimonofDB,
membranes Karet FE,
rat kidney Rodriquez-Soriano
nephron segments. Am JJ,Physiol. et al. Genetic heterogeneity o
1997(273):F739–F748.
27. Wilson RW, Wareing M, Kibble J, Green R. Potassium permeability in the Bartter’
55. Lu s syndrome
M, Wang T, revealed
Yan Q,by mutations
et al. Absenceinofthe small K+ channel,
conductance ROMK. K Nature
channelGenet-
(SK)
http://www.ncbi.nlm.nih.gov/pubmed/21311022
absence of uid reabsorption in proximal tubule of thestudy anesthetized rat. Am J ics. 1996;14:152–156.
activity in apical membranes of thick ascending limb and cortical collecting duct
22. Malnic G, Klose RM, Giebisch G. Micropuncture o renal potassium
Physiol. 1998;274(6 Pt 2):F1109–1112. http://www.ncbi.nlm.nih.gov/pubmed/8841184
in ROMK (Bartter’s) knockout mice. J Biol Chem. 2002;277(40):37881–37887.
excretion in the rat. Am J Physiol. 1964;206(4):674–686.
28. Muto S, Hata M, Taniguchi J, et al. Claudin-2-de cient mice are defective 47. Wade
56. Cantone JB,A,Fang
YangL,X, Coleman
Yan Q, et al. RA,Mouse
et al.model o typeregulation
Differential II Bartter’s syn drome.
of ROMK
http://www.ncbi.nlm.nih.gov/pubmed/14166157
in the leaky and cation-selective paracellular permeability properties of renal I. Upregulation
(Kir1.1) in distal o nephron
thiazide-sensitive
segmentsNa-Cl cotransport
by dietary activityAm
potassium. AmJJPhysiol
Physiol Renal
Renal
23. Kau man JS, Hamburger RJ. Passive potassium transport in the proximal
proximal tubules. Proc Natl Acad Sci USA. 2010;107(17):8011–8016. Physiol. 2008;294(6):F1366–1372.
2011;300(6):F1385–1393.
convoluted tubule. Am J Physiol. 1985;248(2 Pt 2):F228–232.
29. Wareing M, Wilson RW, Kibble JD, Green R. Estimated potassium re ec- 48. Boim
57. TaniguchiMA, Ho J, Guggino
K, ShuckWB. ME,Membrane
et al. ROMK stretch:
inwardlya physiological stimulator o
rectifying ATP-sensitive
24. Brandis M, Keyes J, Windhager EE. Potassium-induced inhibition o proxi- Ca2 channel.
+-activatedII.K+ channels
tion coef cient in perfused proximal convoluted tubules of the anaesthetized rat K Cloning andin distribution
thick ascending limb. Am Jforms.
of alternative Physiol.Am 1989; 257(3
J Physiol.
mal tubular f uid reabsorption in rats. Am J Physiol. 1972;222(2):421–427. Pt 2):F347–352.
in vivo. J Physiol. 1995;488 (Pt 1):153–161. 1995;268(6 Pt 2):F1132–1140.
http://www.ncbi.nlm.nih.gov/pubmed/5058384
30. Johnston PA, Battilana CA, Lacy FB, Jamison RL. Evidence for a concentra- 49. Lu
58. BleichM, M, Wang Schlatter
T, YanE,Q, Greger
et al.R.ROMKThe luminalis requiredK+ channel o the thick
for expression of theas-
25.gradient
tion Wilsonfavoring
RW, WareingoutwardM, movement
Green R. The role o active
of sodium from thetransport in potassium
thin loop of Henle. cendingKlimb
70-pS o Henle’
channel in stheloop. Pf ugers
thick Arch. 1990;415(4):449–460.
ascending limb. Am J Physiol Renal Physiol.
reabsorption in the proximal
J Clin Invest. 1977;59(2):234–240. convoluted tubule o the anaesthetized rat. J Physiol. http://www.ncbi.nlm.nih.gov/pubmed/2315005
2004;286(3):F490–495.
1997;500
31. Battilana (Pt 1):155–164.
CA, Dobyan DC, Lacy FB, et al. Effect of chronic potassium 50. Y
59. Wang
oo D,WH. Flagg Two TPtypes
, Olsen o K+O, channel in thick ascending
et al. Assembly and traf cking limb oofrata kidney.
multi-
26. Kibble
loading on potassium JD, Wareing M, Wilson
secretion by the RW,parsGreenrectaR.orE descending
ect o barium on of
limb potas-
the Am J Physiol.
protein ROMK 1994;267(4
(Kir 1.1)Ptchannel 2):F599–605.
complex by PDZ interactions. J Biol Chem.
sium di usion across the proximal convoluted tubule
juxtamedullary nephron in the rat. J Clin Invest. 1978;62(5):1093–1103. o the anesthetized rat. Am 51. Wang W, Lu M. E ect o arachidonic acid on activity o the apical K+channel
2004;279(8):6863–6873.
J 32.
Physiol. 1995;268(4
Elalouf JM, Roinel Pt 2):F778–783.
N, de Rouf gnac C. Effects of dDAVP on rat juxta- in60.
the Lu
thick M,ascending
Leng Q,limb Egano ME,the ratetkidney
al. CFTR J GenisPhysiol.
required 1995;106(4):727–743.
for PKA-regulated
27. Wilson RW, Wareing
medullary nephrons: stimulation M, Kibble J, Green R.
of medullary PotassiumAm
K recycling. permeability in the
J Physiol. 1985; http://www.ncbi.nlm.nih.gov/pubmed/8576704
ATP sensitivity of Kir1.1 potassium channels in mouse kidney. J Clin Invest.
absence
249(2 Pt o2):F291–298.
f uid reabsorption in proximal tubule o the anesthetized rat. Am J 52. Ho K, Nichols CG, Lederer WJ, et al. Cloning and expression o an inwardly
2006;116(3):797–807.
Physiol.
33. Stokes1998;274(6 Pt 2):F1109–1112.
JB. Consequences of potassium recycling in the renal medulla. recti
61. ying ATP-regulated
Renigunta A, Renigunta potassium channel.
V, Saritas T, etNature. 1993;362(6415):31–38.
al. Tamm-Horsfall glycoprotein in-
28. Muto
Effects of ionS,transport
Hata M, Taniguchi J, et al. Claudin-2-de
by the medullary thick ascending cientlimb
miceofare de ective
Henle’ s loop.in http://www.ncbi.nlm.nih.gov/pubmed/7680431
teracts with renal outer medullary potassium channel ROMK2 and regulates its
Jthe leaky
Clin and1982;70(2):219–229.
Invest. cation-selective paracellular permeability properties o renal proxi- 53. Palmer
function. J BiolLG,Chem.Choe H, Frindt G. Is the secretory K channel in the rat CCT
2011;286(3):2224–2235.
mal tubules. Proc Natl Acad Sci USA. 2010;107(17):8011–8016. ROMK? Am J Physiol. 1997;273(3 Pt 2):F404–410.
CHAPTER 6 TUBULAR POTASSIUM TRANSPORT 2211
11

54. Mutig
62. Xu JZ,K, HallKahlAE,T,Peterson
Saritas T, LN,et etal.al.Activation
Localization of theo the ROMK protein on
bumetanide-sensitive 80. Rieg T, Vallon V, Sausbier
90. Carrisoza-Gaytan R, SalvadorM, C, et al. TheLM,
Satlin roleeto al.thePotassium
BK channel in potas-
secretion by
apical,Kmembranes
Na o rat kidney
,2Cl- cotransporter (NKCC2) nephron segments.
is facilitated Am J Physiol. 1997(273):
by Tamm-Horsfall protein in sium homeostasis
voltage-gated and fchannel
potassium ow-inducedKv1.3renal in thepotassium
rat kidney. excretion. KidneyRenal
Am J Physiol Int.
aF739–F748.
chloride-sensitive manner. J Biol Chem. 2011;286(34):30200–30210. 2007;72(5):566–573.
Physiol. 2010;299(1):F255–264.
55. Frindt
63. Lu M, Wang G, PalmerT, YanLG. Q, etNaal. channels
Absence oinsmall conductance
the rat connecting K+tubule.
channelAm (SK)J http://www.ncbi.nlm.nih.gov/pubmed/17579662
91. Lesage F, Lazdunski M. Molecular and functional properties of two-pore-
activityRenal
Physiol in apical membranes
Physiol. o thick ascending limb and cortical collecting duct
2004;286(4):F669–674. 81. Grimm
domain PR, Irsik
potassium DL, Settles
channels. DC, Holtzclaw
Am J Physiol JD, Sansom
Renal Physiol. SC. Hypertension
2000;279(5):F793–801.
in64.
ROMK (Bartter’s)BM,
Christensen knockout
Perriermice.
R, Wang J Biol Q,
Chem. 2002;277(40):37881–37887.
et al. Sodium and potassium bal- o 92. Zheng W,
Kcnmb1-/- Verlander
is linked to deJW, Lynch
cient IJ, et al.and
K secretion Cellular distribution
aldosteronism. ProcofNatl
theAcad
po-
http://www.ncbi.nlm.nih.gov/pubmed/12130653
ance depends on alphaENaC expression in connecting tubule. J Am Soc Nephrol. tassium
Sci USA. channel KCNQ1 in normal mouse kidney. Am J Physiol Renal Physiol.
2009;106(28):11800–11805.
56. Wade JB, Fang L, Coleman RA, et al. Di erential regulation o ROMK
2010;21(11):1942–1951. 2007;292(1):F456–466.
http://www.ncbi.nlm.nih.gov/pubmed/19556540
(Kir1.1)
65. Frindtin distal nephron
G, Palmer LG.segments by dietarychannels
Apical potassium potassium. Am rat
in the J Physiol Renal
connecting 93. Glaudemans
82. Pacha J, Frindt B, van der Wijst
G, Sackin H, J, Scola LG.
Palmer RH, Apical
et al. Amaxi
missense mutation
K channels in in the
inter-
Physiol. Am
tubule. 2011;300(6):F1385–1393.
J Physiol Renal Physiol. 2004;287(5):F1030–1037. Kv1.1 voltage-gated
calated cells o CCT. Am potassium
J Physiol.channel-encoding gene KCNA1 is linked to hu-
1991;261(4 Pt 2):F696–705.
57. Weinstein
66. Boim MA, AM. Ho K,A Shuck ME, et al.
mathematical ROMK
model of inwardly
rat distal recti ying ATP-sensi-
convoluted tubule. man
83. autosomal
Palmer LG, dominant
Frindt G.hypomagnesemia.
High-conductance J Clin Invest. 2009;119(4):936–942.
K channels in intercalated cells o
tivePotassium
II. K+ channel. II. Cloning
secretion along and distributionsegment.
the connecting o alternative orms. Renal
Am J Physiol Am J Physiol.
Physiol. 94.ratGray
the distalDA, Frindt Am
nephron. G, Zhang
J PhysiolYY , Palmer
Renal LG. 2007;292(3):F966–973.
Physiol. Basolateral K conductance in
1995;268(6 Pt 2):F1132–1140.
2005;289(4):F721–741. principal cells of rat
84. Holtzclaw JD, CCD.
Grimm AmPR,J Physiol
Sansom Renal SC.Physiol. 2005;288(3):F493–504.
Intercalated cell BK-alpha/beta4
58. Frindt
67. Lu M, G, Wang T, Yan
Palmer LG.Q,Low-conductance
et al. ROMK is required K channels or expression o the 70-pS
in apical membrane of 95. Lachheb
channels modulate S, Cluzeaud
sodium and F, Bens M, et al.
potassium Kir4.1/Kir5.1
handling during channel
potassium forms the
adapta-
K channel
rat cortical in the thicktubule.
collecting ascending
Am Jlimb.
Physiol. Am1989;256(1
J Physiol Renal Physiol. 2004;286(3):
Pt 2):F143–151. majorJ Am
tion. K Soc channel
Nephrol.in the basolateral membrane of mouse renal collecting duct
2010;21(4):634–645.
F490–495.
68. Palmer LG, Frindt G. Cl- channels of the distal nephron. Am J Physiol Renal principal
85. Liu cells. Am J Physiol
W, Schreck Renal Physiol.
C, Coleman RA, et al. 2008;294(6):F1398–1407.
Role o NKCC in BK channel-medi-
59. Yoo
Physiol. D, Flagg TP, Olsen O, et al. Assembly and tra cking o a multi-
2006;291(6):F1157–F1168. 96. net
ated Lourdel S, Paulais
K secretion in theM,CCD.
Cluzeaud F, et al.Renal
Am J Physiol An inward
Physiol.recti er K( ) channel
2011;301(5):F1088–
protein ROMK
69. Rubera (Kirng
I, Lof 1.1) channel
J, Palmer LG,complex by PDZduct-speci
et al. Collecting interactions. J Biol
c gene Chem.
inactiva- at the basolateral membrane of the mouse distal convoluted tubule: similarities
1097.
2004;279(8):6863–6873.
tion of alphaENaC in the mouse kidney does not impair sodium and potassium with
86. Kir4-Kir5.1
Ginns SM, heteromeric
Knepper MA,channels.
Ecelbarger J Physiol.
CA, et2002;538(Pt 2):391–404.o the
al. Immunolocalization
http://www.ncbi.nlm.nih.gov/pubmed/14604981
balance. J Clin Invest. 2003;112(4):554–565. 97. Bockenhauer
secretory D, Feather
iso orm o Na-K-Cl S, Stanescu
cotransporter HC,
in rat et intercalated
renal al. Epilepsy,cells.ataxia,
J Amsen-
Soc
60. Katz
70. Lu M, AI, Leng
Doucet Q,A,Egan
MorelME, et al. CFTRactivity
F. Na-K-ATPase is required
along theor rabbit,
PKA-regulated
rat, and sorineural
Nephrol. deafness, tubulopathy, and KCNJ10 mutations. N Engl J Med.
1996;7(12):2533–2542.
ATP sensitivity
mouse nephron.oAmKir1.1 potassium
J Physiol. channels in mouse kidney J Clin Invest.
1979;237(2):F114–120. 2009;360(19):1960–1970.
87. Muto S, Tsuruoka S, Miyata Y, et al. Basolateral Na+/H+ exchange main tains
2006;116(3):797–807.
71. Lof ng J, Pietri L, Aregger F, et al. Differential subcellular localization of 98. Tanemoto
potassium secretionM, Abe T, Onogawa
during diminished T, Ito S. PDZ
sodium bindinginmotif-dependent
transport the rabbit cortical lo-
61. Renigunta
ENaC subunits inA,mouse ReniguntakidneyV, Saritas T, et al.toTamm-Hors
in response all glycoprotein
high- and low-Na diets. Amin-J calization duct.
collecting of K KidneychannelInt. on the basolateral side in distal tubules. Am J Physiol
2009;75(1):25–30.
teracts Renal
Physiol with renal
Physiol.outer medullary potassium channel ROMK2 and regulates its
2000;279(2):F252–258. Renal
88. Physiol.
Fernandez 2004;287(6):F1148–1153.
R, Bosqueiro JR, Cassola AC, Malnic G. Role o Cl- in electro-
unction.
72. Frindt J BiolG,Chem.
Palmer 2011;286(3):2224–2235.
LG. K secretion in the rat kidney: Na channel- 99. Scholl UI, Choi
genic H+ secretion by cortical M, Liu distal
T, et tubule.
al. Seizures,
J Membr sensorineural deafness, ataxia,
Biol. 1997;157(2):193–201.
62. Mutig and
dependent K, Kahl T, Saritas T,mechanisms.
-independent et al. Activation Amo Jthe bumetanide-sensitive
Physiol Renal Physiol. mental retardation, and electrolyte imbalance (SeSAME syndrome) caused by
http://www.ncbi.nlm.nih.gov/pubmed/9151660
Na+, K+, 2Cl- cotransporter (NKCC2) is acilitated by Tamm-Hors all protein in a
2009;297(2):F389–396. mutations in KCNJ10.
89. Holtzclaw JD, Liu ProcL, Natl
GrimmAcadPR,Sci USA.
Sansom 2009;106(14):5842–5847.
SC. Shear stress-induced vol-
chloride-sensitive
73. Muto S. Potassium manner.transport
J Biol Chem.in the 2011;286(34):30200–30210.
mammalian collecting duct. Physiol Rev. 100. decrease
ume Zhou X,inXia SL, Wingo
C11-MDCK CS.
cells by Chloride transport
BK-alpha/beta4. Amby the rabbit
J Physiol Renal cortical
Physiol.
63. Frindt G, Palmer LG. Na channels in the rat connecting tubule. Am J Physiol
2001;81(1):85–116. collecting duct:
2010;299(3):F507–516. dependence on H,K-ATPase. J Am Soc Nephrol. 1998;9(12):
Renal
74. Physiol.
Stokes JB. 2004;286(4):F669–674.
Potassium secretion by cortical collecting tubule: relation to 2194–2202.
90. Carrisoza-Gaytan R, Salvador C, Satlin LM, et al. Potassium secretion by
64. Christensen
sodium absorption,BM, Perrier
luminal R, Wang
sodium Q, et al. Sodium
concentration, and potassium
and transepithelial bal-
voltage. 101. Amorimpotassium
JB, Baileychannel
MA, Musa-Aziz
voltage-gated Kv1.3 inR,the Giebisch
rat kidney.G, Malnic G. Role
Am J Physiol of lumi-
Renal Phys-
anceJ Physiol.
Am depends1981;241(4):F395–402.
on alphaENaC expression in connecting tubule. J Am Soc Nephrol. nal anion and pH in distal tubule potassium secretion. Am J Physiol Renal Physiol.
iol. 2010;299(1):F255–264.
2010;21(11):1942–1951.
75. Pluznick JL, Sansom SC. BK channels in the kidney: role in K( ) se- 2003;284(2):F381–388.
91. Lesage F, Lazdunski M. Molecular and unctional properties o two-pore-
http://www.ncbi.nlm.nih.gov/pubmed/20947633
cretion and localization of molecular components. Am J Physiol Renal Physiol. 102. Ellison DH, Velazquez H, JWright
domain potassium channels. Am Physiol FS.
RenalUnidirectional potassium uxes in
Physiol. 2000;279(5):F793–801.
65. Frindt G, Palmer LG. Apical potassium channels in the rat connecting tu-
2006;291(3):F517–529. renal distal tubule: effects of chloride and barium. Am J Physiol. 1986;250(5 Pt 2):
92. Zheng W, Verlander JW, Lynch IJ, et al. Cellular distribution o the po-
bule.
76. AmGray J Physiol
DA, Frindt Renal G,Physiol.
Palmer 2004;287(5):F1030–1037.
LG. Quanti cation of K secretion through F885–894.
tassium channel KCNQ1 in normal mouse kidney. Am J Physiol Renal Physiol.
66. Weinstein
apical low-conductance AM. A mathematical
K channels in model o rat distal
the CCD. Am J convoluted
Physiol Renal tubule. II.
Physiol. 103. Velazquez H, Ellison DH, Wright FS. Chloride-dependent potassium
2007;292(1):F456–466.
Potassium secretion along the connecting segment. Am J Physiol Renal Physiol.
2005;289(1):F117–126. secretion in early and late renal distal tubules. Am J Physiol. 1987;253(3 Pt 2):
93. Glaudemans B, van der Wijst J, Scola RH, et al. A missense mutation in the
2005;289(4):F721–741.
77. Bailey MA, Cantone A, Yan Q, et al. Maxi-K channels contribute to urinary F555–F562.
Kv1.1 voltage-gated potassium channel-encoding gene KCNA1 is linked to hu-
67. Frindt
potassium G, Palmer
excretion in LG.
the Low-conductance
ROMK-de cient mouse K channelsmodel in apical
of Type membrane
II Bartter’ os 104. Wingo CS. Potassium secretion by the cortical collecting tubule: effect of
man autosomal dominant hypomagnesemia. J Clin Invest. 2009;119(4):936–942.
rat corticaland
syndrome collecting tubule. to
in adaptation Ama Jhigh-K
Physiol.diet.
1989;256(1
Kidney Int. Pt 2006;70(1):51–59.
2):F143–151. C1 gradients and ouabain. Am J Physiol. 1989;256(2 Pt 2):F306–313.
http://www.ncbi.nlm.nih.gov/pubmed/19307729
68. Grimm
78. Palmer LG, Frindt RM,
PR, Foutz G. Cl- channels
Brenner o the distal
R, Sansom SC. nephron.
Identi cationAm JandPhysiol Renal
localiza- 105. Schafer JA, Troutman SL. Potassium transport in cortical collecting tubules
94. Gray DA, Frindt G, Zhang YY, Palmer LG. Basolateral K+ conductance in
Physiol.
tion 2006;291(6):F1157–F1168.
of BK-beta subunits in the distal nephron of the mouse kidney. Am J Physiol from mineralocorticoid-treated rat. Am J Physiol. 1987;253(1 Pt 2):F76–88.
69. Physiol.
Rubera 2007;293(1):F350–359.
I, Lo ng J, Palmer LG, et al. Collecting duct-speci c gene inactiva- principal cells o rat CCD. Am J Physiol Renal Physiol. 2005;288(3):F493–504.
Renal 106. Wingo CS. Reversible chloride-dependent potassium ux across the rabbit
tion 95. Lachheb S, Cluzeaud F, Bens M, et al. Kir4.1/Kir5.1 channel orms the ma-
79. oWoda
alphaENaC
CB, Bragin in theA,mouse
Kleyman kidney TR, does
Satlinnot LM.impair sodium and K
Flow-dependent potassium
secre- cortical collecting tubule. Am J Physiol. 1989;256(4 Pt 2):F697–704.
balance. J Clin Invest. 2003;112(4):554–565. jor K+ channel in the basolateral membrane o mouse renal collecting duct prin-
tion in the cortical collecting duct is mediated by a maxi-K channel. Am J Physiol 107. Wingo CS, Armitage FE. Rubidium absorption and proton secretion by
http://www.ncbi.nlm.nih.gov/pubmed/12925696 cipal cells. Am J Physiol Renal Physiol. 2008;294(6):F1398–1407.
Renal Physiol. 2001;280(5):F786–793. rabbit outer medullary collecting duct via H-K-ATPase. Am J Physiol. 1992;
263(5 Lourdel
70. Rieg
Katz T, AI,Vallon
DoucetV,A, Morel F.M, Na-K-ATPase activity along 96. S, Paulais M, Cluzeaud F, et al. An inward recti er K(+) channel at
80. Sausbier et al. The role of the BKthe rabbit,inrat,
channel and
potas- Pt 2):F849–857.
mousehomeostasis
sium nephron. Amand J Physiol. 1979;237(2):F114–120.
ow-induced renal potassium excretion. Kidney Int. the
108.basolateral
Okusa MD, membrane
Unwin RJ, o the mouse distal
Velazquez convoluted
H, Giebisch tubule:
G, Wright FS.similarities with
Active potassi-
71. Lo ng J, Pietri L, Aregger F, et al. Di erential subcellular localization o
2007;72(5):566–573. Kir4-Kir5.1
um absorption heteromeric
by the renal channels. J Physiol.
distal tubule. Am J2002;538(Pt 2):391–404.
Physiol. 1992;262(3 Pt 2):F488–493.
ENaC subunits
81. Grimm PR,inIrsik
mouse DL,kidney
Settles inDC,response
Holtzclaw to high- and low-Na
JD, Sansom diets. Am J
SC. Hypertension 97. Bockenhauer D, Feather S, Stanescu HC,
109. Zhou X, Lynch IJ, Xia SL, Wingo CS. Activation of H( )-K( et al. Epilepsy, ataxia, sen sorineural
)-ATPase
Physiol
of Renal Physiol.
Kcnmb1-/- is linked 2000;279(2):F252–258.
to de cient K secretion and aldosteronism. Proc Natl Acad dea ness, tubulopathy and KCNJ10 mutations. N Engl
by CO(2) requires a basolateral Ba(2 )-sensitive pathway during K restriction. J Med. 2009;360(19):
Sci72.
USA.Frindt G, Palmer LG. K+ secretion in the rat kidney: Na+ channel dependent
2009;106(28):11800–11805. 1960–1970.
Am J Physiol Renal Physiol. 2000;279(1):F153–160.
and
82.-independent
Pacha J, Frindt mechanisms.
G, SackinAm H, JPalmer
Physiol LG.RenalApical
Physiol. 2009;297(2):F389–396.
maxi K channels in inter- http://www.ncbi.nlm.nih.gov/pubmed/19420365
110. Gumz ML, Lynch IJ, Greenlee MM, Cain BD, Wingo CS. The renal H -
73. Muto
calated cells S.
ofPotassium
CCT. Am Jtransport in the mammalian
Physiol. 1991;261(4 collecting duct. Physiol Rev.
Pt 2):F696–705. K98.-ATPases:
Tanemoto M, Abe T,regulation,
physiology, Onogawa T, and Itostructure.
S. PDZ binding moti -dependent
Am J Physiol Renal Physiol. lo-
2001;81(1):85–116.
83. Palmer LG, Frindt G. High-conductance K channels in intercalated cells of calization o K+ channel on the basolateral side in distal tubules. Am J Physiol Renal
2010;298(1):F12–21.
the74.ratStokes JB. Potassium
distal nephron. Am Jsecretion
Physiol Renal by cortical
Physiol.collecting tubule: relation to so-
2007;292(3):F966–973. Physiol.
111. Kraut2004;287(6):F1148–1153.
JA, Helander KG, Helander HF, et al. Detection and localiza-
dium
84. absorption,
Holtzclaw JD, luminal
Grimm sodium
PR, concentration,
Sansom SC. Intercalatedand transepithelial voltage. Am J
cell BK-alpha/beta4 99.
tion of H -KUI,-ATPase
Scholl Choi M,isoforms
Liu T, etin al. Seizures,
human kidney.sensorineural
Am J Physiol deaRenal
ness,Physiol.
ataxia,
Physiol. 1981;241(4):F395–402.
channels modulate sodium and potassium handling during potassium adapta- mental retardation, and electrolyte imbalance (SeSAME syndrome) caused by mu-
2001;281(4):F763–768.
75. J Pluznick
tion. JL, Sansom
Am Soc Nephrol. SC. BK channels in the kidney: role in K(+) se-
2010;21(4):634–645. tations in KCNJ10.
112. Fejes-Toth G,Proc Natl Acad Sci USA.
Naray-Fejes-Toth 2009;106(14):5842–5847.
A. Immunohistochemical localization of
cretion
85. Liuand localization
W, Schreck o molecular
C, Coleman RA, et components.
al. Role of NKCC AminJ BK Physiol Renal Physiol.
channel-mediated http://www.ncbi.nlm.nih.gov/pubmed/19289823
colonic H-K-ATPase to the apical membrane of connecting tubule cells. Am J
2006;291(3):F517–529.
net K secretion in the CCD. Am J Physiol Renal Physiol. 2011;301(5):F1088–1097. 100.
PhysiolZhou
RenalX,Physiol.
Xia SL,2001;281(2):F318–325.
Wingo CS. Chloride transport by the rabbit cortical collect-
76. Ginns
86. Gray DA, SM, Frindt
Knepper G, MA,
Palmer LG. Quanti
Ecelbarger CA, etcation o K+ secretion through
al. Immunolocalization of the ing
113.duct: dependence
Silver RB, Soleimanion H,K-ATPase.
M. H -K J Am Soc Nephrol.
-ATPases: 1998;9(12):
regulation and role 2194–2202.
in patho-
apical low-conductance
secretory isoform of Na-K-Cl K channels
cotransporter in the CCD.
in rat renalAm J Physiol cells.
intercalated RenalJ AmPhysiol.
Soc 101. Amorimstates.
physiological JB, Bailey
Am JMA, Musa-Aziz
Physiol. 1999;276(6 R, Giebisch G, Malnic G. Role o lumi-
Pt 2):F799–811.
2005;289(1):F117–126.
Nephrol. 1996;7(12):2533–2542. nal
114.anion and pH S,
Nakamura in Wang
distal tubule
Z, Gallapotassium secretion.
JH, Soleimani M.Am K J Physiol
depletionRenal Physiol.
increases
77. Muto
87. BaileyS,MA, Cantone
Tsuruoka S, A, Yan Q,
Miyata Y, et al.al. Maxi-K
Basolateralchannels
Na /H contribute
exchangeto urinary
main- 2003;284(2):F381–388.
HCO3- reabsorption in OMCD by activation of colonic H( )-K( )-ATPase. Am
potassium
tains excretion
potassium in theduring
secretion ROMK-de cient mouse
diminished sodium model o Type
transport in II
theBartter’
rabbits syn-
cor- 102. Ellison
J Physiol. DH, Velazquez
1998;274(4 H, Wright FS. Unidirectional potassium f uxes in
Pt 2):F687–692.
drome
tical and in adaptation
collecting duct. Kidney to aInt.
high-K diet. Kidney Int. 2006;70(1):51–59.
2009;75(1):25–30. renal
115. distal
Krauttubule:
JA, Hiurae ects o chloride
J, Besancon M,and barium.
et al. EffectAm J Physiol. 1986;250(5
of hypokalemia Pt 2):
on the abun-
78. Fernandez
88. Grimm PR,R,Foutz RM, Brenner
Bosqueiro JR, Cassola R, Sansom
AC, MalnicSC. Identi
G. Role cation
of Cl-and
in localiza-
electro- F885–894.
dance of HK alpha 1 and HK alpha 2 protein in the rat kidney. Am J Physiol.
tion oHBK-beta
genic secretionsubunits in thedistal
by cortical distaltubule.
nephron o theBiol.
J Membr mouse kidney. Am J Physiol
1997;157(2):193–201. 103. Velazquez
1997;272(6 H, Ellison DH, Wright FS. Chloride-dependent potassium se-
Pt 2):F744–750.
Renal
89. Physiol.
Holtzclaw 2007;293(1):F350–359.
JD, Liu L, Grimm PR, Sansom SC. Shear stress-induced vol- cretion in early
116. Codina and late renal
J, Delmas-Mata JT,distal
DuBose tubules.
TD Jr. AmThe Jalpha-subunit
Physiol. 1987;253(3 Pt 2):
of the colonic
ume79. decrease
Woda CB, Bragin A, Kleyman
in C11-MDCK cells byTR, Satlin LM. Flow-dependent
BK-alpha/beta4. Am J Physiol RenalK+ secre tion
Physiol. F555–F562.
H ,K -ATPase assembles with beta1-Na ,K -ATPase in kidney and distal
in the cortical collecting duct is mediated by a maxi-K channel. Am J Physiol Renal
2010;299(3):F507–516. 104.
colon. Wingo CS. Potassium
J Biol Chem. secretion by the cortical collecting tubule: e ect o
1998;273(14):7894–7899.
Physiol. 2001;280(5):F786–793. C1 gradients and ouabain. Am J Physiol. 1989;256(2 Pt 2):F306–313.
212
21 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

105. Dherbecourt
117. Scha er JA, Troutman
O, ChevalSL. L, Potassium
Bloch-Faure transport
M, Menetonin cortical
P, Doucetcollecting tubules
A. Molecular 130. Chen
143. PalmerSYLG, Frindt G.
, Bhargava A, Aldosterone
Mastroberardino and potassium secretion sodium
L, et al. Epithelial by the cortical
chan-
rom mineralocorticoid-treated
identi cation of Sch28080-sensitive rat. AmK-ATPase
J Physiol. 1987;253(1
activities in Pt the2):F76–88.
mouse kidney. collecting
nel regulatedduct.byKidney Int. 2000;57(4):1324–1328.
aldosterone-induced protein sgk. Proc Natl Acad Sci USA.
P106.
ugersWingo CS. Reversible chloride-dependent potassium f ux across the rabbit
Arch. 2006;451(6):769–775. 131. Somekawa S, Imagawa K, Naya N, et al. Regulation o aldosterone and cor-
1999;96(5):2514–2519.
cortical
118. collectingN,tubule.
Abuladze Lee I,Am J Physiol.D,1989;256(4
Newman et al. Axial Pt 2):F697–704.
heterogeneity of sodium- tisol production
144. Huang DY,by the transcriptional
Wulff P, Volkl H, et al.repressor
Impairedneuron
regulationrestrictive
of renalsilencer
K elimina-actor.
107. Wingocotransporter
bicarbonate CS, Armitageexpression
FE. Rubidium in the absorption and proton
rabbit proximal secretion
tubule. by rab-
Am J Physiol. Endocrinology.
tion 2009;150(7):3110–3117.
in the sgk1-knockout mouse. J Am Soc Nephrol. 2004;15(4):885–891.
bit outer medullary
1998;274(3 collecting duct via H-K-ATPase. Am J Physiol. 1992; 263(5 Pt 2):
Pt 2):F628–633. 145.
132. Snyder
Mazzocchi PM,G,Olson DR, Thomas
Malendowicz BC. Serum A,
LK, Markowska and glucocorticoid-regulated
Albertin G, Nussdor er GG.
F849–857.
119. Lu Z, MacKinnon R. Electrostatic tuning of Mg2 af nity in an inward- kinase
Role o modulates Nedd4-2-mediated
adrenal renin-angiotensin system inhibition of theoepithelial
in the control aldosteroneNa secretion
channel. in
108.Okusa
recti er K MD, Unwin
channel. RJ, Velazquez
Nature. H, Giebisch G, Wright FS. Active potassium
1994;371(6494):243–246. Jsodium-restricted
Biol Chem. 2002;277(1):5–8.
rats. Am J Physiol Endocrinol Metab. 2000;278(6): E1027–1030.
absorption
120. Lopatin by the
AN,renal distal tubule.
Makhina Am J Physiol.
EN, Nichols CG. 1992;262(3
Potassium channel Pt 2):F488–493.
block by 146.
133. Debonneville
Csukas S, Hanke C, FloresCJ, SY , KamyninaD,E,Campbell
Rewolinski et al. Phosphorylation of Nedd4-2
WB. Prostaglandin E2-
109. Zhou X,
cytoplasmic Lynch IJ, Xia
polyamines SL, mechanism
as the Wingo CS. Activationof intrinsic o recti
H(+)-K(+)-ATPase
cation. Nature. by by Sgk1 aldosterone
induced regulates epithelial
release Na( ) channel
is mediated by cell
an surface expression.
EP2 receptor. Embo J.
Hypertension.
CO(2) requires a basolateral Ba(2+)-sensitive pathway during K restriction. Am J
1994;372(6504):366–369. 2001;20(24):7052–7059.
1998;31(2):575–581.
PhysiolHuang
121. Renal Physiol.
CL, Kuo2000;279(1):F153–160.
E. Mechanism of hypokalemia in magnesium de ciency. 147. Flores SY, Lof ng-Cueni D, Kamynina E, et al. Aldosterone-induced serum
http://www.ncbi.nlm.nih.gov/pubmed/9461224
J110.
Am SocGumz ML, 2007;18(10):2649–2652.
Nephrol. Lynch IJ, Greenlee MM, Cain BD, Wingo CS. The renal H+- and
134. glucocorticoid-induced
Clark BA, Brown RS, kinase Epstein 1FH. expression
E ect o isatrialaccompanied
natriureticbypeptide
Nedd4-2 on
K+-ATPases:
122. Yang L,physiology,
Frindt G, regulation,
Palmer LG.and structure.modulates
Magnesium Am J Physiol ROMK Renalchannel-
Physiol. phosphorylation
potassium-stimulated and increased
aldosterone Nasecretion:
transport in cortical
potential collecting
relevance duct cells.
to hypoaldoste-
2010;298(1):F12–21.
mediated potassium secretion. J Am Soc Nephrol. 2010;21(12):2109–2116. Jronism
Am SocinNephrol. 2005;16(8):2279–2287.
man. J Clin Endocrinol Metab. 1992;75(2):399–403.
111. Huang
123. Kraut JA, CL, Helander KG, Helander
Feng S, Hilgemann HF, etactivation
DW. Direct al. Detection and localiza-
of inward recti er 148. Zhou R, Snyder PM. Nedd4-2 phosphorylation induces serum and gluco-
http://www.ncbi.nlm.nih.gov/pubmed/1386372
tion o H+-K+-ATPase
potassium channels by iso PIP2ormsandinitshuman kidney. by
stabilization AmGJbeta Physiol Renal Nature.
gamma. Physiol. corticoid-regulated kinase (SGK) ubiquitination
135. Cherradi N, Brandenburger Y, Rossier MF, and et al.degradation. J Biolpeptide
Atrial natriuretic Chem.
2001;281(4):F763–768.
1998;391(6669):803–806. 2005;280(6):4518–4523.
inhibits calcium-induced steroidogenic acute regulatory protein gene transcrip-
112. Xu
124. Fejes-Toth
ZC, YangG,Y, Naray-Fejes-Toth
Hebert SC. PhosphorylationA. Immunohistochemical
of the ATP-sensitive, localization
inwardly o 149.
tion inEladrenal
Moghrabi S, Houillier
glomerulosa P, Picard
cells. N, et al. Tissue
Mol Endocrinol. kallikrein permits early renal
1998;12(7):962–972.
colonic H-K-ATPase
rectifying K channel, to ROMK,
the apical by membrane o connecting
cyclic AMP-dependent tubule
protein cells. JAm
kinase. BiolJ adaptation to potassium load. Proc Natl Acad Sci USA. 2010;107(30):13526–13531.
http://www.ncbi.nlm.nih.gov/pubmed/9658401
Physiol1996;271(16):9313–9319.
Chem. Renal Physiol. 2001;281(2):F318–325. 150.
136. Kleyman
Palmer LG, TR,Antonian
MyerburgL,MM, FrindtHughey RP. Regulation
G. Regulation o apicalof ENaCs
K and by Na prote-
chan-
113. O’Connell
125. Silver RB, AD, Soleimani
Leng Q, M.Dong H+-K+-ATPases: regulation and role in patho-
K, et al. Phosphorylation-regulated endo- ases: An increasingly complex story. Kidney Int. 2006;70(8):1391–1392.
nels and Na/K pumps in rat cortical collecting tubule by dietary K. J Gen Physiol.
physiological
plasmic states.retention
reticulum Am J Physiol.
signal 1999;276(6
in the renal Pt 2):F799–811.
outer-medullary K channel 151. Kleyman TR, Carattino MD, Hughey RP. ENaC at the cutting edge: regu-
1994;104(4):693–710.
114. Nakamura
(ROMK). Proc NatlS,Acad Wang Z, Galla
Sci USA. JH, Soleimani M. K+ depletion increases
2005;102(28):9954–9959. lation of epithelial sodium channels by proteases. J Biol Chem. 2009;284(31):
http://www.ncbi.nlm.nih.gov/pubmed/7836937
HCO3-
126. reabsorption
Liou HH, Zhou inSS,OMCD HuangbyCL. activation
Regulation o colonic
of ROMK1 H(+)-K(+)-ATPase.
channel by protein Am J 20447–20451.
137. Stanton B, Janzen A, Klein-Robbenhaar G, et al. Ultrastructure o rat ini-
Physiol.A
kinase 1998;274(4 Pt 2):F687–692. 4,5-bisphosphate-dependent mechanism.
via a phosphatidylinositol 152. Carattinotubule.
tial collecting MD, ShengE ect S,o Bruns
adrenal JB, corticosteroid
et al. The epithelial Na J Clin
treatment. channel is
Invest.
115.Natl
Proc Kraut
Acad JA,SciHiura
USA.J,1999;96(10):5820–5825.
Besancon M, et al. E ect o hypokalemia on the abun- inhibited by a peptide derived from proteolytic processing of its alpha subunit.
1985;75(4):1327–1334.
danceLeipziger
127. o HK alpha 1 and HK GG,
J, MacGregor alphaCooper
2 protein GJ, inet the
al. rat
PKA kidney. Am J Physiol.
site mutations of J138.
Biol Chem.
Welling 2006;281(27):18901–18907.
PA, Caplan M, Sutters M, Giebisch G. Aldosterone-mediated Na/ K-
1997;272(6
ROMK2 Pt 2):F744–750.
channels shift the pH dependence to more alkaline values. Am J Physiol 153.
ATPaseMuto S, Sansom
expression S, Giebisch
is alpha 1 iso orm G. speci
Effectsc of
in athe
high
renalpotassium diet on electri-
cortical collecting duct.
116. Physiol.
Renal Codina2000;279(5):F919–926.
J, Delmas-Mata JT, DuBose TD Jr. The alpha-subunit o the colonic cal properties of cortical collecting
J Biol Chem. 1993;268(31):23469–23476. ducts from adrenalectomized rabbits. J Clin
H +, K+-ATPase
128. August JT,assembles
Nelson DH, withThorn
beta1-NaGW.+,K+-ATPase
Response ofinnormal kidneysubjects
and distal tocolon.
large Invest. 1988;81(2):376–380.
139. Summa V, Mordasini D, Roger F, et al. Short term e ect o aldosterone on
J Biol Chem.
amounts 1998;273(14):7894–7899.
of aldosterone. J Clin Invest. 1958;37(11):1549–1555. 154. Muto S, Asano Y, Seldin D, Giebisch G.collecting
Basolateral Nacells.pump
Na,K-ATPase cell sur ace expression in kidney duct J Biolmodu-
Chem.
117. Dluhy
129. Dherbecourt O, Cheval
RG, Axelrod L, L, Bloch-FaureRH,
Underwood M, Williams
Meneton PGH. , Doucet A. Molecu-
Studies of the lates apical Na and K conductances in rabbit cortical collecting ducts. Am J
2001;276(50):47087–47093.
lar identiofcation
control plasma o Sch28080-sensitive
aldosterone concentration K-ATPaseinactivities
normalinman. the mouse kidney.
II. Effect of Physiol. 1999;276(1 Pt 2):F143–158.
http://www.ncbi.nlm.nih.gov/pubmed/11598118
Pf ugers Arch.
dietary 2006;451(6):769–775.
potassium and acute potassium infusion. J Clin Invest. 1972;51(8): 155.
140. Lin DH, Sterling
Reisenauer H, Yang B,M,etHuang
MR, Anderson al. Protein
L, ettyrosine
al. AF17kinase is expressed
competes with AF9and or
http://www.ncbi.nlm.nih.gov/pubmed/16208521
1950–1957. regulates ROMK1 location in the cortical collecting duct. Am J Physiol Renal
binding to Dot1a to up-regulate transcription o epithelial Na+ channel alpha. J
118. Palmer
130. Abuladze LG,N, Lee G.
Frindt I, Newman
Aldosterone D, andet al. Axial heterogeneity
potassium secretion by the o sodium-
cortical Physiol. 2004;286(5):F881–892.
Biol Chem. 2009;284(51):35659–35669.
bicarbonate
collecting cotransporter
duct. Kidney Int.expression in the rabbit proximal tubule. Am J Physiol.
2000;57(4):1324–1328. 156. Palmer LG, Frindt G. Regulation of apical K channels in rat cortical collecting
http://www.ncbi.nlm.nih.gov/pubmed/19864429
1998;274(3
131. Somekawa Pt 2):F628–633.
S, Imagawa K, Naya N, et al. Regulation of aldosterone and tubule during changes in dietary K intake. Am J Physiol. 1999;277(5 Pt 2):F805–812.
141. Lo ng J, Zecevic M, Feraille E, et al. Aldosterone induces rapid apical
119. Luproduction
cortisol Z, MacKinnon by the R. transcriptional
Electrostatic tuning o Mg2+
repressor neuron a nity in an silencer
restrictive inward- 157. Frindt G, Shah A, Edvinsson J, Palmer LG. Dietary K regulates ROMK
translocation o ENaC in early portion o renal collecting system: possible role o
recti erEndocrinology.
factor. K+ channel. Nature. 1994;371(6494):243–246.
2009;150(7):3110–3117. channels in connecting tubule and cortical collecting duct of rat kidney. Am J
SGK. Am J Physiol Renal Physiol. 2001;280:F675–682.
http://www.ncbi.nlm.nih.gov/pubmed/7915826
132. Mazzocchi G, Malendowicz LK, Markowska A, Albertin G, Nussdorfer Physiol Renal Physiol. 2009;296(2):F347–354.
142. Snyder PM. Minireview: regulation o epithelial Na+ channel tra cking.
120. Role
GG. Lopatin AN, Makhina
of adrenal EN, Nichols
renin-angiotensin systemCG. inPotassium
the control channel block by
of aldosterone 158. Najjar F, Zhou H, Morimoto T, et al. Dietary K regulates apical membrane
Endocrinology. 2005;146(12):5079–5085.
cytoplasmic
secretion polyamines as the
in sodium-restricted rats.mechanism
Am J Physiol o Endocrinol
intrinsic Metab.
recti cation. Nature.
2000;278(6): expression of maxi-K channels in rabbit cortical collecting duct. Am J Physiol
1994;372(6504):366–369. 143. Chen SY, Bhargava A, Mastroberardino L, et al. Epithelial sodium chan-
E1027–1030. Renal Physiol. 2005;289(4):F922–932.
http://www.ncbi.nlm.nih.gov/pubmed/7969496 nel regulated by aldosterone-induced protein sgk. Proc Natl Acad Sci USA.
133. Csukas S, Hanke CJ, Rewolinski D, Campbell WB. Prostaglandin E2- 159. Estilo G, Liu W, Pastor-Soler N, et al. Effect of aldosterone on BK chan-
121. Huang CL, KuoreleaseE. Mechanism o hypokalemia 1999;96(5):2514–2519.
induced aldosterone is mediated by an EP2inreceptor.magnesium de ciency.
Hypertension. nel expression in mammalian cortical collecting duct. Am J Physiol Renal Physiol.
J Am Soc Nephrol. 2007;18(10):2649–2652. 144. Huang DY, Wul P, Volkl H, et al. Impaired regulation o renal K+ elimina-
1998;31(2):575–581. 2008;295(3):F780–788.
122. Clark
Yang L, Frindt G, Palmer LG. Magnesium tion in the sgk1-knockout mouse. J Am Soc Nephrol. 2004;15(4):885–891.
134. BA, Brown RS, Epstein FH. Effect modulates ROMK channel-medi-
of atrial natriuretic peptide on 160. Cassola AC, Giebisch G, Wang W. Vasopressin increases density of apical low-
ated potassium secretion.
potassium-stimulated J Am Soc secretion:
aldosterone Nephrol. 2010;21(12):2109–2116.
potential relevance to hypoaldoste- 145. SnyderKPM,channels
conductance Olson DR, in ratThomas
CCD. Am BC. Serum 1993;264(3
J Physiol. and glucocorticoid-regulated
Pt 2):F502–509.
123. Huang
ronism in man.CL, Feng
J Clin S, Hilgemann
Endocrinol DW Direct activation o inward recti-
Metab. 1992;75(2):399–403. 161. Yoo D, Kim BY, Campo C, et al. Cell surface expression Na+
kinase modulates Nedd4-2-mediated inhibition o the epithelial of thechannel.
ROMKJ
er potassium
135. Cherradi channels by PIP2 and
N, Brandenburger its stabilization
Y, Rossier MF, et al.by G beta
Atrial gamma.peptide
natriuretic Nature. Biol Chem.
(Kir 2002;277(1):5–8.
1.1) channel is regulated by the aldosterone-induced kinase, SGK1, and
1998;391(6669):803–806.
inhibits calcium-induced steroidogenic acute regulatory protein gene transcrip- 146. Debonneville
protein kinase A. J Biol C, Chem.
Flores 2003;278(25):23066–23075.
SY, Kamynina E, et al. Phosphorylation o Nedd4-
http://www.ncbi.nlm.nih.gov/pubmed/9486652
tion in adrenal glomerulosa cells. Mol Endocrinol. 1998;12(7):962–972. 2 by Cheng
162. Sgk1 regulates
CJ, Huang epithelial Na(+) channel
CL. Activation cell surstimulates
of PI3-kinase ace expression. EmboofJ.
endocytosis
124. Palmer
136. Xu ZC,LG, Yang Y, HebertL,SC.
Antonian Phosphorylation
Frindt G. Regulationoofthe ATP-sensitive,
apical inwardly
K and Na channels 2001;20(24):7052–7059.
ROMK via Akt1/SGK1-dependent phosphorylation of WNK1. J Am Soc Nephrol.
recti Na/K
and ying K+ pumpschannel,
in ratROMK,
corticalbycollecting
cyclic AMP-dependent
tubule by dietary protein
K. Jkinase. J Biol
Gen Physiol. 147. Flores SY, Lo ng-Cueni D, Kamynina E, et al. Aldosterone-induced se-
2011;22(3):460–471.
Chem. 1996;271(16):9313–9319.
1994;104(4):693–710. rum and
163. glucocorticoid-induced
Wilson FH, Disse-Nicodeme kinase 1 expression
S, Choate KA, et is accompanied by Nedd4-2
al. Human hypertension
http://www.ncbi.nlm.nih.gov/pubmed/8621594
137. Stanton B, Janzen A, Klein-Robbenhaar G, et al. Ultrastructure of rat ini- phosphorylation
caused by mutationsand increased Na+ transport
in WNK kinases. Science.in2001;293(5532):1107–1112.
cortical collecting duct cells. J Am
125.collecting
tial O’Connell AD, Leng
tubule. EffectQ,ofDongadrenal K, corticosteroid
et al. Phosphorylation-regulated
treatment. J Clin Invest. endo- Soc Nephrol.
164. 2005;16(8):2279–2287.
Ko B, Hoover RS. Molecular physiology of the thiazide-sensitive sodium-
plasmic reticulum retention signal in the renal outer-medullary K+ channel
1985;75(4):1327–1334. chloride cotransporter. PM.
148. Zhou R, Snyder CurrNedd4-2
Opin Nephrolphosphorylation induces serum and gluco-
Hypertens. 2009;18(5):421–427.
(ROMK).
138. Proc PA,
Welling NatlCaplan
Acad SciM,USA. 2005;102(28):9954–9959.
Sutters M, Giebisch G. Aldosterone-mediated Na/ corticoid-regulated
165. Gamba G. Thekinase (SGK) ubiquitination
thiazide-sensitive and degradation.
Na -Cl- cotransporter: J Biol Chem.
molecular biol-
http://www.ncbi.nlm.nih.gov/pubmed/15987778
K-ATPase expression is alpha 1 isoform speci c in the renal cortical collecting 2005;280(6):4518–4523.
ogy, functional properties, and regulation by WNKs. Am J Physiol Renal Physiol.
126. JLiou
duct. HH, Zhou
Biol Chem. SS, Huang CL. Regulation o ROMK1 channel by protein
1993;268(31):23469–23476. http://www.ncbi.nlm.nih.gov/pubmed/15576372
2009;297(4):F838–848.
kinaseSumma
139. A via a phosphatidylinositol
V, Mordasini D, Roger 4,5-bisphosphate-dependent
F, et al. Short term effect ofmechanism. aldosteroneProc on 149. Lalioti
166. El Moghrabi S, Houillier
MD, Zhang P, Picard
J, Volkman HM,N,et etal.al.Wnk4
Tissuecontrols
kallikrein permits
blood ear-
pressure
Natl Acad Sci USA.
Na,K-ATPase 1999;96(10):5820–5825.
cell surface expression in kidney collecting duct cells. J Biol Chem. ly renal
and adaptation
potassium to potassium
homeostasis load. Proc
via regulation Natl Acad
of mass Sci USA.
and activity 2010;107(30):
of the distal con-
127. Leipziger J, MacGregor GG, Cooper GJ, et al. PKA site mutations o ROMK2
2001;276(50):47087–47093. 13526–13531.
voluted tubule. Nat Genet. 2006;38(10):1124–1132.
channels
140. shi t theMR,
Reisenauer pHAnderson
dependence to more
M, Huang L, alkaline
et al. AF17values. Am J with
competes Physiol
AF9Renal
for http://www.ncbi.nlm.nih.gov/pubmed/20624970
167. Kahle KT, Wilson FH, Leng Q, et al. WNK4 regulates the balance between
Physiol. 2000;279(5):F919–926.
binding to Dot1a to up-regulate transcription of epithelial Na channel alpha. 150. Kleyman
renal NaCl TR, Myerburg
reabsorption and KMM,secretion.
Hughey RP NatRegulation o ENaCs by proteases:
Genet. 2003;35(4):372–376.
J128. August
Biol Chem. JT, Nelson DH, Thorn GW Response o normal subjects to large
2009;284(51):35659–35669. An increasingly
168. Zhuang J,complex
Zhang X,story.Wang Kidney
D, etInt.
al.2006;70(8):1391–1392.
WNK4 kinase inhibits Maxi K chan-
amounts
141. Lofongaldosterone.
J, Zecevic JM, Clin Invest. E,
Feraille 1958;37(11):1549–1555.
et al. Aldosterone induces rapid apical 151. Kleyman TR, Carattino MD,
nel activity by a kinase-dependent mechanism. Hughey RP ENaC AmatJthe cutting
Physiol edge:
Renal regu-
Physiol.
http://www.ncbi.nlm.nih.gov/pubmed/13587664
translocation of ENaC in early portion of renal collecting system: possible role of lation o epithelial sodium channels by proteases. J Biol Chem. 2009;284(31):
2011;301(2):F410–419.
129. Am
SGK. Dluhy RG, Axelrod
J Physiol L, Underwood
Renal Physiol. RH, Williams GH. Studies o the control
2001;280:F675–682. 20447–20451.
169. Delaloy C, Lu J, Houot AM, et al. Multiple promoters in the WNK1 gene:
o plasma
142. Snyderaldosterone concentration
PM. Minireview: in normal
regulation man. II. E
of epithelial Naect channel
o dietarytraf potassium
cking. 152.controls
one Carattino MD, Sheng
expression of S, Bruns JB, et al.c The
a kidney-speci epithelial Na+isoform.
kinase-defective channel Molis inhib-
Cell
and acute potassium
Endocrinology. in usion. J Clin Invest. 1972;51(8): 1950–1957.
2005;146(12):5079–5085. ited by
Biol. a peptide derived rom proteolytic processing o its alpha subunit. J Biol
2003;23(24):9208–9221.
http://www.ncbi.nlm.nih.gov/pubmed/5054456 Chem. 2006;281(27):18901–18907.
CHAPTER 6 TUBULAR POTASSIUM TRANSPORT 2213
13

153. Cope
170. Muto G, S, Sansom
MurthyS,M,Giebisch
Golbang G.AP
E , ects
et al.o WNK1
a high potassium diet on
affects surface electrical
expression http://www.ncbi.nlm.nih.gov/pubmed/19244242
190. Flyvbjerg A, Marshall SM, Frystyk J, et al. Insulin-like growth factor
properties
of the ROMK o cortical collecting
potassium channelductsindependent
rom adrenalectomized
of WNK4. rabbits.J Am Soc J Clin Invest.
Nephrol. 178. Ornt renal
I in initial DB, Tannen
hypertrophyRL. Demonstration o an intrinsic
in potassium-depleted renalJ Physiol.
rats. Am adaptation 1992; or
1988;81(2):376–380.
2006;17(7):1867–1874. K+conservation in short-term K+ depletion. Am J Physiol. 1983;245(3):F329–338.
262(6 Pt 2):F1023–1031.
154. Wade
171. Muto JB, S, Asano
Fang L, Y, Liu
Seldin
J, etD,al.Giebisch
WNK1 kinase G. Basolateral
isoform Na+switch pump modurenal
regulates lates 179. Wang W, Lerea
191. Rabinowitz L. KM, Chan M,and
Aldosterone Giebisch G. Protein
potassium tyrosine kinase
homeostasis. Kidneyregu-Int.
apical Na+ excretion.
potassium and K+ conductances
Proc Natl Acad in Sci
rabbit
USA.cortical collecting ducts. Am J Physiol.
2006;103(22):8558–8563. lates the number o renal secretory K channels. Am J Physiol Renal Physiol.
1996;49(6):1738–1742.
1999;276(1
172. LazrakPtA,2):F143–158.
Liu Z, Huang CL. Antagonistic regulation of ROMK by long and 2000;278(1):F165–171.
192. McDonough AA, Youn JH. Role of muscle in regulating extracellular [K ].
155. Lin DH,c Sterling
kidney-speci H, Yang B,
WNK1 isoforms. Procet Natl
al. Protein
Acad Scityrosine kinase is expressed and
USA. 2006;103(5):1615–1620. 180.
Semin Eiam-Ong S, Kurtzman NA, Sabatini S. Regulation o collecting tubule
Nephrol. 2005;25(5):335–342.
regulates
173. He ROMK1
G, Wanglocation
HR, Huang in theSK, cortical
Huangcollecting duct. Am
CL. Intersectin J Physiol
links WNKRenal kinasesPhys-
to adenosine
193. Bugaj triphosphatases
V, PochynyukbyO, aldosterone
Stockand andJD. potassium.
ActivationJ Clinof theInvest. 1993;91(6):
epithelial Na
iol. 2004;286(5):F881–892.
endocytosis of ROMK1. J Clin Invest. 2007;117(4):1078–1087. 2385–2392.
channel in the collecting duct by vasopressin contributes to water reabsorption.
156. Fang
174. Palmer LG, Frindt
L, Garuti R, Kim G. BYRegulation
, Wade JB,o Welling
apical KPA. channels
The ARH in adaptor
rat cortical col-
protein http://www.ncbi.nlm.nih.gov/pubmed/8390478
Am J Physiol Renal Physiol. 2009;297(5):F1411–1418.
lecting tubule
regulates during of
endocytosis changes
the ROMK in dietary K intake.
potassium Am J Physiol.
secretory channel1999;277(5
in mouse 181. GreenleeJB,
194. Amorim MM, Lynch IJ,R,Gumz
Musa-Aziz ML, Cain
Mello-Aires BD, Wingo
M, Malnic CS. Mineralocorti-
G. Signaling path of the
Pt 2):F805–812.
kidney. J Clin Invest. 2009;119(11):3278–3289. coids
actionstimulate
of AVP onthe activity
distal and expression
K secretion. KidneyoInt. renal H +, K+-ATPases. J Am Soc
2004;66(2):696–704.
157. Lin
175. FrindtDH, G, Y Shah
ue P,A,PanEdvinsson
CY, et J, al.Palmer
POSHLG. Dietary Kthe
stimulates regulates ROMK chan-
ubiquitination and Nephrol. 2011;22(1):49–58.
195. Chambrey R, Picard N. Role of tissue kallikrein in regulation of tubule
nels in
the connecting tubule and
clathrin-independent cortical collecting
endocytosis of ROMK1 duct ochannels.
rat kidneyJ Am BiolJ Physiol
Chem. http://www.ncbi.nlm.nih.gov/pubmed/21164026
function. Curr Opin Nephrol Hypertens. 2011;20(5):523–528.
Renal Physiol. 2009;296(2):F347–354.
2009;284(43):29614–29624. 182. Lin DH,
196. Leviel F, Sterling
Hubner H, CA,Lerea KM, etP, al.
Houillier et Kal.depletion
The Na increases
-dependent protein tyro-
chloride-
158. O’Reilly
176. Najjar F, M, Zhou H, Morimoto
Marshall T, et al. Dietary
E, Macgillivray T, et K+al. regulates apical membrane
Dietary electrolyte- driven sine kinase-mediated
bicarbonate exchangerphosphorylation
SLC4A8 mediates o ROMK. Am J Physiol
an electroneutral NaRenal Physiol.
reabsorp-
expression oin maxi-K
responses the renal channels
WNKin kinaserabbit cortical
pathway collecting
in vivo. duct.J Am JSoc Physiol Renal
Nephrol. 2002;283(4):F671–677.
tion process in the renal cortical collecting ducts of mice. J Clin Invest.
Physiol. 2005;289(4):F922–932.
2006;17(9):2402–2413. 183. Sterling H, Lin DH, Gu RM, et al. Inhibition o protein-tyrosine phos-
2010;120(5):1627–1635.
159. Liu
177. Estilo G, LiuHR,
Z, Wang W, Huang
Pastor-Soler N, et al. of
CL. Regulation E ROMK
ect o aldosterone
channel andon K BK chan-
homeo- phatase
197. Tomitastimulates the dynamin-dependent
K, Pisano JJ, Burg MB, Knepperendocytosis
MA. EffectsoofROMK1.vasopressinJ Biol
andChem.
bra-
nel expression
stasis in mammalian
by kidney-speci c WNK1cortical
kinase.collecting
J Biol Chem. duct. Am J Physiol Renal Physiol.
2009;284(18):12198–12206. 2002;277(6):4317–4323.
dykinin on anion transport by the rat cortical collecting duct. Evidence for an elec-
2008;295(3):F780–788.
178. Ornt DB, Tannen RL. Demonstration of an intrinsic renal adaptation for K 184. Lin sodium
troneutral DH, Sterlingchloride H, transport
Wang WH. The protein
pathway. tyrosine
J Clin Invest. kinase-dependent
1986;77(1):136–141.
160.Cassola AC,
conservation Giebisch G,
in short-term KWang W Vasopressin
depletion. increases
Am J Physiol. density o apical low-
1983;245(3):F329–338. pathway
198. Picard mediates the e D,
N, Eladari ect El
o Moghrabi
K intake onS,renal
et al.KDefective
secretion.ENaCPhysiology (Bethesda).
processing and
conductance
179. Wang W, K+ Lerea
channels
KM,inChanrat CCD. Am J Physiol.
M, Giebisch 1993;264(3
G. Protein Pt 2):F502–509.
tyrosine kinase regu- 2005;20:140–146.
function in tissue kallikrein-de cient mice. J Biol Chem. 2008;283(8):4602–4611.
161. the
lates Yoonumber
D, Kim BY of, renal
Camposecretory
C, et al. K Cell sur ace expression
channels. Am J Physiol o the ROMK
Renal (Kir
Physiol. 185. Wei Y, Bloom
199. Terada P, Lin MA.
Y, Knepper D, Gu R, Wang WH. E NaCl
Thiazide-sensitive ect o absorption
dietary K intakein ratoncortical
apical
1.1) channel is regulated by the aldosterone-induced kinase, SGK1, and protein
2000;278(1):F165–171. small-conductance
collecting duct. AmKJ channel in CCD: rolePt
Physiol. 1990;259(3 o 2):F519–528.
protein tyrosine kinase. Am J Physiol
kinaseEiam-Ong
180. A. J Biol Chem. 2003;278(25):23066–23075.
S, Kurtzman NA, Sabatini S. Regulation of collecting tubule Renal
200. Physiol.
Welling2001;281(2):F206–212.
PA, Chang YP, Delpire E, Wade JB. Multigene kinase network,
162. Cheng
adenosine CJ, Huang CL.
triphosphatases Activation oandPI3-kinase
by aldosterone potassium.stimulates
J Clin Invest.endocytosis
1993;91(6): o 186.
kidneyBabilonia
transport, E, and
Lin D,saltZhang Y, et al.hypertension.
in essential Role o gp91phox-containing NADPH
Kidney Int. 2010;77(12):
ROMK via Akt1/SGK1-dependent phosphorylation o WNK1. J Am Soc Nephrol.
2385–2392. oxidase
1063–1069. in mediating the e ect o K restriction on ROMK channels and renal K
2011;22(3):460–471.
181. Greenlee MM, Lynch IJ, Gumz ML, Cain BD, Wingo CS. Mineralocorti- excretion.
201. Yue JP,Am LinSocDH,Nephrol.
Pan CY 2007;18(7):2037–2045.
, et al. Src family protein tyrosine kinase (PTK)
http://www.ncbi.nlm.nih.gov/pubmed/21355052
coids stimulate the activity and expression of renal H ,K -ATPases. J Am Soc 187. Wangthe
modulates ZJ,effect
Sun of P, Xing
SGK1W, andet WNK4
al. Decrease
on ROMK in dietary K intake
channels. Procstimulates
Natl Acad the Sci
163. Wilson
Nephrol. FH, Disse-Nicodeme S, Choate KA, et al. Human hypertension
2011;22(1):49–58. generation o superoxide anions in the kidney and inhibits K secretory channels
USA. 2009;106(35):15061–15066.
causedLin
182. by DH,
mutations
Sterlingin WNK
H, Lereakinases.
KM, Science.
et al. K2001;293(5532):1107–1112.
depletion increases protein tyro- in the San-Cristobal
202. CCD. Am J Physiol Renal Physiol. 2010;298(6):F1515–1522.
P, Pacheco-Alvarez D, Richardson C, et al. Angiotensin II
http://www.ncbi.nlm.nih.gov/pubmed/11498583
sine kinase-mediated phosphorylation of ROMK. Am J Physiol Renal Physiol. 188. Weiincreases
signaling Y, ZavilowitzactivityB, of
Satlin LM, Wang
the renal Na-Cl WH. Angiotensin
cotransporter II inhibits
through a WNK4- the
164. Ko B, Hoover RS. Molecular physiology o the thiazide-sensitive sodium-
2002;283(4):F671–677. ROMK-like
SPAK-dependent small pathway.
conductance ProcKNatlchannel in renal
Acad Sci USA.cortical collecting duct during
2009;106(11):4384–4389.
chloride
183. cotransporter.
Sterling H, Lin DH, Curr GuOpinRM, Nephrol
et al.Hypertens.
Inhibition2009;18(5):421–427.
of protein-tyrosine phos- dietary potassium
203. Sandberg MB,restriction.
Riquier AD, J Biol Chem. 2007;282(9):6455–6462.
Pihakaski-Maunsbach K, McDonough AA, Mauns-
165. Gamba
phatase G. The
stimulates the thiazide-sensitive
dynamin-dependent Na+-Cl- cotransporter:
endocytosis of ROMK1. molecular biol-
J Biol Chem. 189.
bach AB.Jin ANG
Y, Wang Y, Wangacute
II provokes ZJ, LintrafDH, Wang
cking WH.tubule
of distal Inhibition
Na o-Cl(-) angiotensin type
cotransporter
ogy, unctional properties, and regulation by WNKs. Am J Physiol Renal Physiol.
2002;277(6):4317–4323. 1toreceptor impairs renal
apical membrane. Am Jability
PhysioloRenal
K conservation in response to K restriction. Am
Physiol. 2007;293(3):F662–669.
2009;297(4):F838–848.
184. Lin DH, Sterling H, Wang WH. The protein tyrosine kinase-dependent J204.
Physiol Renal
Yang LE,Physiol.
Sandberg 2009;296(5):F1179–1184.
MB, Can AD, Pihakaski-Maunsbach K, McDonough
166. Lalioti
pathway MD, Zhang
mediates the J, Volkman
effect of KHM, et al.on
intake Wnk4renalcontrols blood pressure
K secretion. and
Physiology 190. Flyvbjerg
AA. Effects A, Marshall
of dietary salt onSM,renal
Frystyk
Na J, transporter
et al. Insulin-like growth
subcellular actor I in
distribution,
potassium homeostasis
(Bethesda). 2005;20:140–146. via regulation o mass and activity o the distal con voluted initial renal hypertrophy
abundance, in potassium-depleted
and phosphorylation rats. Am
status. Am J Physiol J Physiol.
Renal Physiol.1992; 262(6 Pt
2008;295(4):
tubule.Wei
185. NatY,Genet.
Bloom2006;38(10):1124–1132.
P, Lin D, Gu R, Wang WH. Effect of dietary K intake on api- 2):F1023–1031.
F1003–1016.
167.small-conductance
cal Kahle KT, WilsonKFH, Leng in
channel Q, CCD:
et al. WNK4
role of regulates the balance
protein tyrosine kinase.between
Am J 191. Rabinowitz
205. Sealey JE, ClarkL. I,Aldosterone
Bull MB, Laragh and JH.
potassium
Potassium homeostasis.
balance and Kidney
the controlInt.
renal NaCl
Physiol Renal reabsorption and K+ secretion. Nat Genet. 2003;35(4):372–376.
Physiol. 2001;281(2):F206–212. 1996;49(6):1738–1742.
of renin secretion. J Clin Invest. 1970;49(11):2119–2127.
http://www.ncbi.nlm.nih.gov/pubmed/14608358
186. Babilonia E, Lin D, Zhang Y, et al. Role of gp91phox-containing NADPH http://www.ncbi.nlm.nih.gov/pubmed/8743488
206. Vallon V, Schroth J, Lang F, Kuhl D, Uchida S. Expression and phosphory-
168. Zhuang
oxidase J, Zhang
in mediating theX,effect
Wang of D, et al. WNK4
K restriction on kinase
ROMKinhibits
channelsMaxi and K chan-
renal K 192.
lation McDonough
of the Na -Cl- AA, cotransporter
Youn JH. RoleNCC o muscle
in vivoin regulating
is regulated extracellular
by dietary[K+].
salt,
nel activityJ Am
excretion. by Soca kinase-dependent mechanism. Am J Physiol Renal Physiol.
Nephrol. 2007;18(7):2037–2045. Semin Nephrol.
potassium, and2005;25(5):335–342.
SGK1. Am J Physiol Renal Physiol. 2009;297(3):F704–712.
2011;301(2):F410–419.
187. Wang ZJ, Sun P, Xing W, et al. Decrease in dietary K intake stimulates the http://www.ncbi.nlm.nih.gov/pubmed/16139689
207. Ring AM, Leng Q, Rinehart J, et al. An SGK1 site in WNK4 regulates Na
169. Delaloy
generation C, Lu J, Houot
of superoxide anionsAM, in et
theal.kidney
Multiple andpromoters
inhibits Kinsecretory
the WNK1 gene:
channels 193.
channel Bugaj
andVK , Pochynyuk O, Stockand
channel activity JD. Activation
and has implications o the
for epithelial
aldosterone Na+channel
signaling
onethecontrols
in CCD. Am expression
J Physiol oRenala kidney-specif c kinase-de ective iso orm. Mol Cell
Physiol. 2010;298(6):F1515–1522. in
andtheKcollecting duct by
homeostasis. vasopressin
Proc Natl Acadcontributes to water reabsorption. Am J Phys-
Sci USA. 2007;104(10):4025–4029.
Biol. 2003;23(24):9208–9221.
188. Wei Y, Zavilowitz B, Satlin LM, Wang WH. Angiotensin II inhibits the iol Renal
208. YuePhysiol.
P, Sun2009;297(5):F1411–1418.
P, Lin DH, et al. Angiotensin II diminishes the effect of
http://www.ncbi.nlm.nih.gov/pubmed/14645531
ROMK-like small conductance K channel in renal cortical collecting duct during 194.
SGK1 Amorim
on the JB, Musa-Aziz R, Mello-Aires
WNK4-mediated inhibition M, of Malnic
ROMK1G.channels.
Signaling Kidney
path o Int.the
170. Cope
dietary G, Murthy
potassium M, Golbang
restriction. AP, et2007;282(9):6455–6462.
J Biol Chem. al. WNK1 a ects sur ace expression action o AVP on distal K+ secretion. Kidney Int. 2004;66(2):696–704.
2011;79(4):423–431.
o theJinROMK
189. Y, Wang potassium
Y, Wangchannel
ZJ, Lin independent
DH, Wang WH. o WNK4.InhibitionJ Am of Soc Nephrol.
angiotensin http://www.ncbi.nlm.nih.gov/pubmed/15253724
209. Liu Z, Xie J, Wu T, et al. Downregulation of NCC and NKCC2 cotransporters
2006;17(7):1867–1874.
type 1 receptor impairs renal ability of K conservation in response to K restric- 195. Chambrey R,
by kidney-speci Picard N.revealed
c WNK1 Role o tissue
by genekallikrein in regulation
disruption o tubulemouse
and transgenic unc-
http://www.ncbi.nlm.nih.gov/pubmed/16775035
tion. Am J Physiol Renal Physiol. 2009;296(5):F1179–1184. tion. CurrHum
models. Opin MolNephrol
Genet.Hypertens. 2011;20(5):523–528.
2011;20(5):855–866.
171. Wade JB, Fang L, Liu J, et al. WNK1 kinase iso orm switch regulates renal 196. Leviel F, Hubner CA, Houillier P, et al. The Na+-dependent chloride-bicar-
potassium excretion. Proc Natl Acad Sci USA. 2006;103(22):8558–8563. bonate exchanger SLC4A8 mediates an electroneutral Na+reabsorp tion process in
172. Lazrak A, Liu Z, Huang CL. Antagonistic regulation o ROMK by long and the renal cortical collecting ducts o mice. J Clin Invest. 2010;120(5):1627–1635.
kidney-specif c WNK1 iso orms. Proc Natl Acad Sci USA. 2006;103(5):1615– 197. Tomita K, Pisano JJ, Burg MB, Knepper MA. E ects o vasopressin and bra-
1620. dykinin on anion transport by the rat cortical collecting duct. Evidence or an
173. He G, Wang HR, Huang SK, Huang CL. Intersectin links WNK kinases to elec-troneutral sodium chloride transport pathway. J Clin Invest. 1986;77(1):136–
endocytosis o ROMK1. J Clin Invest. 2007;117(4):1078–1087. 141.
174. Fang L, Garuti R, Kim BY, Wade JB, Welling PA. The ARH adaptor protein 198. Picard N, Eladari D, El Moghrabi S, et al. De ective ENaC processing and
regulates endocytosis o the ROMK potassium secretory channel in mouse kidney. unction in tissue kallikrein-def cient mice. J Biol Chem. 2008;283(8):4602–4611.
J Clin Invest. 2009;119(11):3278–3289. http://www.ncbi.nlm.nih.gov/pubmed/18086683
175. Lin DH, Yue P, Pan CY, et al. POSH stimulates the ubiquitination and 199. Terada Y, Knepper MA. Thiazide-sensitive NaCl absorption in rat cortical
the clathrin-independent endocytosis o ROMK1 channels. J Biol Chem. collecting duct. Am J Physiol. 1990;259(3 Pt 2):F519–528.
2009;284(43):29614–29624. 200.Welling PA, Chang YP, Delpire E, Wade JB. Multigene kinase network, kidney
http://www.ncbi.nlm.nih.gov/pubmed/19710010 transport, and salt in essential hypertension. Kidney Int. 2010;77(12): 1063–1069.
176. O’Reilly M, Marshall E, Macgillivray T, et al. Dietary electrolyte- driv- http://www.ncbi.nlm.nih.gov/pubmed/20375989
en responses in the renal WNK kinase pathway in vivo. J Am Soc Nephrol. 201. Yue P, Lin DH, Pan CY, et al. Src amily protein tyrosine kinase (PTK)
2006;17(9):2402–2413. modulates the e ect o SGK1 and WNK4 on ROMK channels. Proc Natl Acad Sci
http://www.ncbi.nlm.nih.gov/pubmed/16899520 USA. 2009;106(35):15061–15066.
177. Liu Z, Wang HR, Huang CL. Regulation o ROMK channel and K+homeo- 202. San-Cristobal P, Pacheco-Alvarez D, Richardson C, et al. Angiotensin
stasis by kidney-specif c WNK1 kinase. J Biol Chem. 2009;284(18):12198–12206. II signaling increases activity o the renal Na-Cl cotransporter through a
(contd.)
213
21 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

WNK4-SPAK-dependent
117. Dherbecourt O, Cheval pathway. Proc Natl
L, Bloch-Faure Acad SciP, USA.
M, Meneton Doucet2009;106(11):
A. Molecular 206. Chen
143. VallonSYV,, Schroth
Bhargava J, Lang F, Kuhl D, Uchida
A, Mastroberardino L, etS. al.
Expression
Epithelialand phosphory-
sodium chan-
4384–4389.
identi cation of Sch28080-sensitive K-ATPase activities in the mouse kidney. lationregulated
nel o the Na+-Cl– cotransporter NCC
by aldosterone-induced in vivo
protein sgk.is Proc
regulated
Natl by dietary
Acad salt,
Sci USA.
P203.
ugersSandberg MB, Riquier AD, Pihakaski-Maunsbach K, McDonough AA,
Arch. 2006;451(6):769–775. potassium, and SGK1. Am J Physiol Renal Physiol. 2009;297(3):F704–712.
1999;96(5):2514–2519.
Mauns-bach
118. AbuladzeAB.N,ANGLee III,provokes
NewmanacuteD, ettraal.f cking
Axial oheterogeneity
distal tubuleofNa+-Cl(–)
sodium- 207. Huang
144. Ring AM, Leng Q,
DY, Wulff Rinehart
P, Volkl H, et J,
al. et al. An regulation
Impaired SGK1 site of inrenal
WNK4 K regulates
elimina-
cotransporter
bicarbonate to apical membrane.
cotransporter expression inAmtheJ rabbit
Physiolproximal
Renal Physiol.
tubule.2007;293(3):
Am J Physiol. Na+channel
tion and K+ channel
in the sgk1-knockout mouse.activity
J Am Soc andNephrol.
has implications or aldosterone
2004;15(4):885–891.
F662–669. Pt 2):F628–633.
1998;274(3 signaling
145. and K+
Snyder PM,homeostasis. Proc NatlBC.
Olson DR, Thomas AcadSerum
Sci USA.and2007;104(10):4025–4029.
glucocorticoid-regulated
204. Lu
119. Yang
Z, LE, SandbergR. MB,
MacKinnon Can AD,tuning
Electrostatic Pihakaski-Maunsbach
of Mg2 af nityK,inMcDonough
an inward- http://www.ncbi.nlm.nih.gov/pubmed/17360471
kinase modulates Nedd4-2-mediated inhibition of the epithelial Na channel.
AA. Eer ects
recti o dietaryNature.
K channel. salt on renal Na+ transporter subcellular distribution,
1994;371(6494):243–246. J208.
BiolYChem.
ue P, Sun P, Lin DH, et al. Angiotensin II diminishes the e ect o SGK1 on the
2002;277(1):5–8.
abundance,
120. LopatinandAN,
phosphorylation
Makhina EN,status. Am CG.
Nichols J Physiol Renal Physiol.
Potassium 2008;295(4):
channel block by WNK4-mediated
146. Debonneville inhibition
C, Floreso SY
ROMK1
, Kamyninachannels.
E, et Kidney Int. 2011;79(4):423–431.
al. Phosphorylation of Nedd4-2
F1003–1016.polyamines as the mechanism of intrinsic recti cation. Nature.
cytoplasmic 209.Sgk1
by Liu regulates
Z, Xie J, Wu T, et al. Na(
epithelial Downregulation
) channel cell o NCC and NKCC2
surface cotransport-
expression. Embo J.
205. Sealey JE, Clark I, Bull MB, Laragh JH. Potassium balance and the control
1994;372(6504):366–369. ers by kidney-specif c WNK1 revealed by gene disruption and transgenic mouse
2001;20(24):7052–7059.
o renin
121. secretion.
Huang J ClinE.Invest.
CL, Kuo 1970;49(11):2119–2127.
Mechanism of hypokalemia in magnesium de ciency. models.
147. HumSY
Flores Mol Genet.
, Lof 2011;20(5):855–866.
ng-Cueni D, Kamynina E, et al. Aldosterone-induced serum
Jhttp://www.ncbi.nlm.nih.gov/pubmed/4319969
Am Soc Nephrol. 2007;18(10):2649–2652. http://www.ncbi.nlm.nih.gov/pubmed/21131289
and glucocorticoid-induced kinase 1 expression is accompanied by Nedd4-2
122. Yang L, Frindt G, Palmer LG. Magnesium modulates ROMK channel- phosphorylation and increased Na transport in cortical collecting duct cells.
mediated potassium secretion. J Am Soc Nephrol. 2010;21(12):2109–2116. J Am Soc Nephrol. 2005;16(8):2279–2287.
123. Huang CL, Feng S, Hilgemann DW. Direct activation of inward recti er 148. Zhou R, Snyder PM. Nedd4-2 phosphorylation induces serum and gluco-
potassium channels by PIP2 and its stabilization by G beta gamma. Nature. corticoid-regulated kinase (SGK) ubiquitination and degradation. J Biol Chem.
1998;391(6669):803–806. 2005;280(6):4518–4523.
124. Xu ZC, Yang Y, Hebert SC. Phosphorylation of the ATP-sensitive, inwardly 149. El Moghrabi S, Houillier P, Picard N, et al. Tissue kallikrein permits early renal
rectifying K channel, ROMK, by cyclic AMP-dependent protein kinase. J Biol adaptation to potassium load. Proc Natl Acad Sci USA. 2010;107(30):13526–13531.
Chem. 1996;271(16):9313–9319. 150. Kleyman TR, Myerburg MM, Hughey RP. Regulation of ENaCs by prote-
125. O’Connell AD, Leng Q, Dong K, et al. Phosphorylation-regulated endo- ases: An increasingly complex story. Kidney Int. 2006;70(8):1391–1392.
plasmic reticulum retention signal in the renal outer-medullary K channel 151. Kleyman TR, Carattino MD, Hughey RP. ENaC at the cutting edge: regu-
(ROMK). Proc Natl Acad Sci USA. 2005;102(28):9954–9959. lation of epithelial sodium channels by proteases. J Biol Chem. 2009;284(31):
126. Liou HH, Zhou SS, Huang CL. Regulation of ROMK1 channel by protein 20447–20451.
kinase A via a phosphatidylinositol 4,5-bisphosphate-dependent mechanism. 152. Carattino MD, Sheng S, Bruns JB, et al. The epithelial Na channel is
Proc Natl Acad Sci USA. 1999;96(10):5820–5825. inhibited by a peptide derived from proteolytic processing of its alpha subunit.
127. Leipziger J, MacGregor GG, Cooper GJ, et al. PKA site mutations of J Biol Chem. 2006;281(27):18901–18907.
ROMK2 channels shift the pH dependence to more alkaline values. Am J Physiol 153. Muto S, Sansom S, Giebisch G. Effects of a high potassium diet on electri-
Renal Physiol. 2000;279(5):F919–926. cal properties of cortical collecting ducts from adrenalectomized rabbits. J Clin
128. August JT, Nelson DH, Thorn GW. Response of normal subjects to large Invest. 1988;81(2):376–380.
amounts of aldosterone. J Clin Invest. 1958;37(11):1549–1555. 154. Muto S, Asano Y, Seldin D, Giebisch G. Basolateral Na pump modu-
129. Dluhy RG, Axelrod L, Underwood RH, Williams GH. Studies of the lates apical Na and K conductances in rabbit cortical collecting ducts. Am J
control of plasma aldosterone concentration in normal man. II. Effect of Physiol. 1999;276(1 Pt 2):F143–158.
dietary potassium and acute potassium infusion. J Clin Invest. 1972;51(8): 155. Lin DH, Sterling H, Yang B, et al. Protein tyrosine kinase is expressed and
1950–1957. regulates ROMK1 location in the cortical collecting duct. Am J Physiol Renal
130. Palmer LG, Frindt G. Aldosterone and potassium secretion by the cortical Physiol. 2004;286(5):F881–892.
collecting duct. Kidney Int. 2000;57(4):1324–1328. 156. Palmer LG, Frindt G. Regulation of apical K channels in rat cortical collecting
131. Somekawa S, Imagawa K, Naya N, et al. Regulation of aldosterone and tubule during changes in dietary K intake. Am J Physiol. 1999;277(5 Pt 2):F805–812.
cortisol production by the transcriptional repressor neuron restrictive silencer 157. Frindt G, Shah A, Edvinsson J, Palmer LG. Dietary K regulates ROMK
factor. Endocrinology. 2009;150(7):3110–3117. channels in connecting tubule and cortical collecting duct of rat kidney. Am J
132. Mazzocchi G, Malendowicz LK, Markowska A, Albertin G, Nussdorfer Physiol Renal Physiol. 2009;296(2):F347–354.
GG. Role of adrenal renin-angiotensin system in the control of aldosterone 158. Najjar F, Zhou H, Morimoto T, et al. Dietary K regulates apical membrane
secretion in sodium-restricted rats. Am J Physiol Endocrinol Metab. 2000;278(6): expression of maxi-K channels in rabbit cortical collecting duct. Am J Physiol
E1027–1030. Renal Physiol. 2005;289(4):F922–932.
133. Csukas S, Hanke CJ, Rewolinski D, Campbell WB. Prostaglandin E2- 159. Estilo G, Liu W, Pastor-Soler N, et al. Effect of aldosterone on BK chan-
induced aldosterone release is mediated by an EP2 receptor. Hypertension. nel expression in mammalian cortical collecting duct. Am J Physiol Renal Physiol.
1998;31(2):575–581. 2008;295(3):F780–788.
134. Clark BA, Brown RS, Epstein FH. Effect of atrial natriuretic peptide on 160. Cassola AC, Giebisch G, Wang W. Vasopressin increases density of apical low-
potassium-stimulated aldosterone secretion: potential relevance to hypoaldoste- conductance K channels in rat CCD. Am J Physiol. 1993;264(3 Pt 2):F502–509.
ronism in man. J Clin Endocrinol Metab. 1992;75(2):399–403. 161. Yoo D, Kim BY, Campo C, et al. Cell surface expression of the ROMK
135. Cherradi N, Brandenburger Y, Rossier MF, et al. Atrial natriuretic peptide (Kir 1.1) channel is regulated by the aldosterone-induced kinase, SGK1, and
inhibits calcium-induced steroidogenic acute regulatory protein gene transcrip- protein kinase A. J Biol Chem. 2003;278(25):23066–23075.
tion in adrenal glomerulosa cells. Mol Endocrinol. 1998;12(7):962–972. 162. Cheng CJ, Huang CL. Activation of PI3-kinase stimulates endocytosis of
136. Palmer LG, Antonian L, Frindt G. Regulation of apical K and Na channels ROMK via Akt1/SGK1-dependent phosphorylation of WNK1. J Am Soc Nephrol.
and Na/K pumps in rat cortical collecting tubule by dietary K. J Gen Physiol. 2011;22(3):460–471.
1994;104(4):693–710. 163. Wilson FH, Disse-Nicodeme S, Choate KA, et al. Human hypertension
137. Stanton B, Janzen A, Klein-Robbenhaar G, et al. Ultrastructure of rat ini- caused by mutations in WNK kinases. Science. 2001;293(5532):1107–1112.
tial collecting tubule. Effect of adrenal corticosteroid treatment. J Clin Invest. 164. Ko B, Hoover RS. Molecular physiology of the thiazide-sensitive sodium-
1985;75(4):1327–1334. chloride cotransporter. Curr Opin Nephrol Hypertens. 2009;18(5):421–427.
138. Welling PA, Caplan M, Sutters M, Giebisch G. Aldosterone-mediated Na/ 165. Gamba G. The thiazide-sensitive Na -Cl- cotransporter: molecular biol-
K-ATPase expression is alpha 1 isoform speci c in the renal cortical collecting ogy, functional properties, and regulation by WNKs. Am J Physiol Renal Physiol.
duct. J Biol Chem. 1993;268(31):23469–23476. 2009;297(4):F838–848.
139. Summa V, Mordasini D, Roger F, et al. Short term effect of aldosterone on 166. Lalioti MD, Zhang J, Volkman HM, et al. Wnk4 controls blood pressure
Na,K-ATPase cell surface expression in kidney collecting duct cells. J Biol Chem. and potassium homeostasis via regulation of mass and activity of the distal con-
2001;276(50):47087–47093. voluted tubule. Nat Genet. 2006;38(10):1124–1132.
140. Reisenauer MR, Anderson M, Huang L, et al. AF17 competes with AF9 for 167. Kahle KT, Wilson FH, Leng Q, et al. WNK4 regulates the balance between
binding to Dot1a to up-regulate transcription of epithelial Na channel alpha. renal NaCl reabsorption and K secretion. Nat Genet. 2003;35(4):372–376.
J Biol Chem. 2009;284(51):35659–35669. 168. Zhuang J, Zhang X, Wang D, et al. WNK4 kinase inhibits Maxi K chan-
141. Lof ng J, Zecevic M, Feraille E, et al. Aldosterone induces rapid apical nel activity by a kinase-dependent mechanism. Am J Physiol Renal Physiol.
translocation of ENaC in early portion of renal collecting system: possible role of 2011;301(2):F410–419.
SGK. Am J Physiol Renal Physiol. 2001;280:F675–682. 169. Delaloy C, Lu J, Houot AM, et al. Multiple promoters in the WNK1 gene:
142. Snyder PM. Minireview: regulation of epithelial Na channel traf cking. one controls expression of a kidney-speci c kinase-defective isoform. Mol Cell
Endocrinology. 2005;146(12):5079–5085. Biol. 2003;23(24):9208–9221.
C H A P T E R

7 Renal Acid–Base Transport


Thomas D. DuBose Jr. • Amanda K. Goode

CO2 remains as dissolved CO2; only about 1 part in 1,000


ACID–BASE HOMEOSTASIS forms H2CO3, a nonvolatile acid. Because H2CO3 is a weak
The body maintains systemic acid–base homeostasis (a pH acid, it dissociates rapidly to yield H and HCO3 .
in the range of 7.35 to 7.45) in two ways: (1) through chem-
ical buffering in extracellular uid (ECF) and intracellular H2CO3 H HCO3 (2)
uid (ICF), and (2) through physiologic regulation con-
trolled by the metabolic, renal, and respiratory systems. The Because the concentration of H2CO3 remains low and
central nervous and respiratory systems control CO2 tension proportional to the concentration of dissolved CO2, Eqs 1
(PCO2), and the kidneys regulate the plasma HCO3 concen- and 2 can be combined and treated as a single reaction:
tration. Buffers in the ECF and ICF guard against acid and
base retention. These processes serve to dispose of carbonic CO2 H2O H HCO3 (3)
and nonvolatile acids on a daily basis and pathologic quanti-
ties of acid and alkali as needed. This chapter brie y reviews The equilibrium constant for this reaction is given by
the role of chemical buffers, but focuses primarily on the
renal transport and regulatory processes that maintain acid– [H ] [HCO3 ]
K __
(4)
base balance. [CO2] [H2O]
De ning K K[H2O] as the apparent equilibrium con-
Buffer Systems stant and using Eq. 4,
The body’s buffer systems keep the blood from becoming too [H ] [HCO3 ]
K __

(5)
basic or too acidic by combining with or releasing H , and co2 PCO2
thus are comprised of a base (i.e., an H acceptor) and an
Taking logarithms of both sides of Eq. 5 and recogniz-
acid (i.e., an H donor). The most prodigious base is bicar-
ing that pK log10(K ), the familiar Henderson-Hasselbalch
bonate (HCO3 ) and the most common acid is carbonic acid
equation is derived:
(H2CO3). Because buffer systems attenuate system changes,
acid or base loads in the presence of a buffer cause smaller pK log10 [HCO3 ]
pH __

(6)
changes in the pH than if the buffer were absent. During ( co2Pco2)
homeostasis, the extracellular H concentration ([H ]e) is
Using pK 6.1 in Eq. 6, the Henderson equation is
constant. H concentration can be expressed either directly
derived, which may be used in the clinical interpretation of
as [H ] or indirectly as pH.
acid–base data:
When CO2 is dissolved in water, H2CO3 is formed
according to the reaction 24 PCO2 (mm Hg)
[H ](nmol/L) __

(7)
[HCO3 ] (mM)
CO2 H2O H2CO3 (1)

The rate of this reaction, in the absence of the enzyme Physiologic Processes that Protect the
carbonic anhydrase, is slow, with a half-life of about 8 sec- Plasma Bicarbonate
onds at 37°C. The importance of carbonic anhydrase in Three physiologic processes mitigate changes in the HCO3 /
bicarbonate equilibrium in the kidneys and lungs are dis- CO2 ratio: (1) metabolic regulation, (2) respiratory
cussed in the following paragraphs. The major portion of regulation, and (3) renal regulation. Metabolic regulation is

21 4
CHAPTER 7 RENAL ACID–BASE TRANSPORT 2215
15

of minor importance in terms of the overall physiologic reg- Potential sources of bases are also found in the diet
ulation of acid–base balance. Some enzymes are regulated by (e.g., acetate, lactate, citrate) and can be absorbed to neu-
changes in blood pH. For example, the activity of phospho- tralize the acid loads from the three sources just mentioned.
fructokinase, the pivotal enzyme in the glycolytic pathway, is The net base absorbed by the gastrointestinal tract is de-
inhibited by low pH and enhanced by high pH. rived from the anion gap (AG) of the diet minus that of the
Because, under most circumstances, respiratory CO2 stool. Acid production is partially offset by HCO3 , which is
excretion and CO2 production are matched, the usual steady produced when organic anions combine with H and are
state arterial PCO2 (PaCO2) is maintained at 40 mm Hg. oxidized to CO2 and H2O or when dibasic phosphoesters
Underexcretion of CO2 produces hypercapnia, and overexcre- combine with H during hydrolysis. The gastrointestinal
tion produces hypocapnia. Production and excretion are again tract may modify the amount of these potential bases reab-
matched but at a new steady state PCO2. Therefore, PaCO2 sorbed under particular circumstances of acidosis or growth.
is regulated primarily by neurorespiratory factors and is not It has been con rmed in patients ingesting an arti cial diet
subject to regulation by the rate of metabolic CO2 production. that urinary net acid excretion is equal to urinary [(SO24 )
Hypercapnia is primarily the result of hypoventilation, not organic A dietary phosphoester-derived H ].1,2,5 There-
increased CO2 production. Increases or decreases in PCO2 fore, in summary, the metabolism of certain proteins, nucleic
represent derangements of control of neurorespiratory regula- acids, and small fractions of lipids and certain carbohydrates
tion or can result from compensatory changes in response to a generate speci c organic acids that cannot be burned to CO2
primary alteration in the plasma HCO3 concentration. (e.g., uric, oxalic, glucuronic, hippuric acids). In addition,
the inorganic acids H2SO4 and H3PO4, derived respectively
from sulfur-containing amino acids and organophosphates,
Sources of Endogenous Acids are excreted by the kidneys or the gastrointestinal tract.
Pathologically, acid loads may be derived from endogenous
acid production (e.g., generation of ketoacids and lactic
acids) or loss of base (e.g., through diarrhea) or from exog- Impact of Daily Metabolism on
enous sources (e.g., ammonium chloride or toxin ingestion). Acid–Base Balance
Under normal physiologic circumstances, a daily input of Human subjects ingesting a typical Western diet are con-
acid derived from the diet and metabolism confronts the fronted, under most physiologic circumstances, with an acid
body. The net result of these processes amounts to about challenge. Metabolism generates a daily load of relatively
1 mEq of new H per kilogram per day entering the ECF.1–4 strong acids (lactate, citrate, acetate, and pyruvate), but
Sulfuric acid is formed when organic sulfur from under physiologic circumstances, in the steady state, meta-
methionine and cysteine residues of proteins are oxidized bolic production and consumption are matched. However,
to SO24 . The metabolism of sulfur-containing amino acids if production and consumption rates become mismatched,
is the primary source of acid in the usual Western diet, organic acids can accumulate (e.g., lactic acid accumulation
accounting for approximately 50%. The quantity of sulfuric with anoxia or ischemia). These acids are buffered by HCO3
acid generated is equal to the SO24 excreted in the urine. in the ECF, causing a decline in plasma HCO3 concentration
Organic acids are derived from intermediary metabolites as the organic acid concentration increases. During recovery,
formed by the partial combustion of dietary carbohydrates, fats, these organic acids reenter the metabolic pathways to CO2
and proteins as well as from nucleic acids (uric acid). Organic production, the removal of H , and the generation of HCO3
acid generation contributes to net endogenous acid production unless the organic anions are excreted (e.g., ketonuria), and
when the conjugate bases are excreted in the urine as organic thereby are no longer available for regeneration of HCO3 .
anions. If full oxidation of these acids can occur, however, H is Both metabolic and renal regulatory mechanisms protect
reclaimed and is eliminated as CO2 and water. The net amount a normal HCO3 concentration in blood (25 mEq per liter)
of H added to the body from this source can be estimated by despite the daily addition of acid (or alkali) to the ECF. Al-
the amount of organic anions excreted in the urine. though the buffering capacity of the body is magni ed sever-
Phosphoric acid can be derived from hydrolysis of PO34 alfold by respiratory adjustments in PaCO2, primary changes
esters in proteins and nucleic acids if it is not neutralized in PaCO2 may result in acidosis or alkalosis, depending on
by mineral cations (e.g., Na , K , Mg2 ). The contribution whether CO2 is elevated above or depressed below the nor-
of dietary phosphate to acid production is dependent on mal value: 40 mm Hg (respiratory acidosis and respiratory
the kind of protein ingested. Some proteins generate phos- alkalosis), respectively. A primary change in the plasma HC
phoric acid, whereas others generate only neutral phosphate O3 concentration as a result of metabolic production or the
salts.1–4 Hydrochloric acid is generated by the metabolism retention or excretion of an acid or base by the kidney evokes
of cationic amino acids (lysine, arginine, and some histidine a ventilator response because a decrease or increase in pH is
residues) into neutral products. Other potential acid or base sensed by chemoreceptors in the circulation and signals the
sources in the diet can be estimated from the amount of un- respiratory center to increase or decrease minute ventilation.
identi ed cations and anions ingested. The respiratory response to acidemia (increase ventilation
216
21 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

and decrease PCO2) or alkalemia (decrease in ventilation by net acid excretion, metabolic acidosis would ensue.
and increase in PCO2) thereby blunts the change in blood Conversely, an increase in net acid excretion above the level
pH that would occur otherwise. Such respiratory altera- of net acid production results in metabolic alkalosis. Each
tions that adjust blood pH toward normal are referred to as milliequivalent of net acid excreted corresponds to 1 mEq
secondary or compensatory alterations, because they occur of HCO3 returned to the ECF. This process is called HCO3
in response to primary metabolic changes. While helpful, regeneration and is necessary for replacing the HCO3 lost
respiratory compensation to metabolic acidosis or alkalosis by the entry of xed acids into the ECF or, less commonly,
is never suf cient to return blood pH to normal. Therefore, replacing the HCO3 excreted in stool or urine.
the kidneys must play a very signi cant role in adjustments To prevent metabolic acidosis, this excreted acid must
to metabolic disturbances by altering bicarbonate reabsorp- be recovered. The kidney recovers this acid through the
tion and net acid excretion. second process, HCO3 regeneration, which is represented
by the renal output of acid or net acid excretion (NAE), or
the sum of ammonia (NH3)/ammonium (NH4 ) plus excreted
RENAL PARTICIPATION IN ACID–BASE titratable acids (TA) minus any bicarbonate excreted.
HOMEOSTASIS Net acid excretion
Although temporary relief from changes in the pH of body (UNH V) (UTAV) (U HCO V) (9)
(NAE) 4 3

uids may be derived from chemical buffering or respiratory


compensation, the ultimate defense against the addition of where V is urinary volume per 24 hours and U refers to the
nonvolatile acid or of alkali resides in the kidneys. The addition concentration of the moiety in the urine.
of a strong acid (e.g., HA) to the ECF titrates plasma HCO3 : TA excretion in the urine is represented by buffers
that are ltered at the glomerulus and then titrated to acid
HA NaHCO3 NaA H2O CO2 (8) moieties in the tubule lumen. Because renal H -secretory
mechanisms cannot generate such steep pH gradients
The CO2 is expired by the lungs, and body HCO3 between the cell (pH 7.3) and lumen, excretion of 70 mEq
declines. This process occurs constantly as endogenous meta- of acid per day requires that most of this acid be buffered in
bolic acids are generated. To maintain a normal plasma HC the luminal uid.
O3 in the face of the constant accession of metabolic acids, Buffers with a pK between 5.0 and 7.4, the pH range
the kidneys must (1) conserve the HCO3 present in the l- of the luminal uid, are most effective chemically. In this
tered load (through reclamation), and (2) regenerate the HC regard, the most abundant and effective buffer in the urine
O3 decomposed by reaction with metabolic acids (Eq. 8). is the phosphate buffer pair, H2PO4 HPO24 , with a p K of
The rst function of the kidneys in maintaining acid– 6.9. The excretion of phosphate, the major titratable buffer,
base homeostasis is to reclaim or reabsorb ltered HCO3 . is usually regulated in accordance with phosphate homeo-
The kidneys lter approximately 4,000 mEq of bicarbonate stasis. Phosphate excretion increases modestly in meta-
daily (de ned as the product of the glomerular ltration bolic acidosis, but the magnitude of the increase is limited
rate [GFR] and the plasma HCO3 concentration), and the by the ltered load of phosphate and, therefore, is signi -
excretion of any signi cant portion of this leads to meta- cantly regulatable. In general, the buffers responsible are the
bolic acidosis. The renal tubules essentially reabsorb all of NH3 NH4 and titratable buffer, such as HPO24 H2PO4 ,
the ltered HCO3 , or may excrete excessive HCO3 when systems. Physiologically, the most signi cant of these is the
needed, to maintain the normal plasma HCO3 concentration NH3–NH4 system.
of 25 mEq per liter.
HCO3 reclamation is accomplished principally in the
proximal tubule. There is a subsequent contribution by Ammoniagenesis
the loop of Henle and a minor contribution by more distal Ammoniagenesis by protein catabolism is balanced by
nephron segments. Under most circumstances, the ltered the generation of new HCO3 through renal NH4 and TA
load of HCO3 is absorbed almost completely, especially dur- excretion (Fig. 7.1A). The products of these neutral reac-
ing an acid load. Nevertheless, when less acid is generated tions are HCO3 and NH4 . Most ammoniagenesis occurs
or if the plasma HCO3 concentration increases above the in the proximal tubule. NH4 produced by the metabolism
normal value of 25 mEq per liter, HCO3 will be excreted of amino acids reacts with HCO3 or forms urea and thus
ef ciently into the urine. has no impact on acid–base balance. A portion of this NH4
The generation of acid in the body by metabolism is is diverted to glutamine synthesis, the amount of which is
referred to as net acid production. Because a typical Western regulated by pH. Acidemia promotes and alkalemia inhibits
diet generates xed acids at 50 to 70 mEq per day, net acid glutamine synthesis. Each glutamine molecule metabolized
excretion must be affected to maintain acid–base balance. produces two NH4 and two HCO3 molecules (Fig. 7.1A)
Therefore, net acid excretion approximates 50 to 70 mEq (water, CO2, and glucose are also formed). This breakdown
per day. If acid production remained stable and unabated occurs as follows: glutamine is transported into the proximal
CHAPTER 7 RENAL ACID–BASE TRANSPORT 2217
17

FIGURE 7.1 Cell models of ammonia synthesis and excretion pathways. A: A proximal convoluted tubule. Ammonia is derived
from glutamine precursors to produce 2 NH4 and 2 HCO3 molecules through an enzymatic pathway activated by acidemia and
hypokalemia and inhibited by alkalemia and hyperkalemia. B: A type A intercalated cell in the collecting tubule. Ammonium enters
across the basolateral membrane through the substitution of K for NH4 in K conductance and is secreted across the apical
membrane via renal outer medullary potassium (ROMK) or RhCG (see text). In both A and B, NH3 diffusion coupled with H secretion
traps NH4 in the tubule lumen.

tubule across the apical and basolateral membranes and is would negate the new HCO3 realized from -ketoglutarate
then transported into the mitochondria. In the mitochon- in the kidney. Thus, ammoniagenesis does not contribute to
dria, an NH4 molecule is formed when glutamine is deami- acid–base homeostasis until the urine is acidi ed and the
nated by glutaminase to glutamate. When glutamate is then HCO3 produced by glutamine metabolism is added to the
deaminated to -ketoglutarate, a second NH4 molecule is ECF. In order for this to occur, the ammonia synthesized
formed. Finally, the -ketoglutarate molecule is metabolized in the proximal tubule has to be transported to the termi-
in the Krebs cycle to CO2, H2O, glucose, and the two HCO3 nal nephron segments and is then excreted by the kidney
molecules. Glutamine deamination in the kidney is highly (the pathway is described in detail in the following para-
regulated by systemic pH, so that acidemia augments and graphs). This ammonia transport occurs through both non-
alkalemia inhibits NH4 and HCO3 production. Hepatic ionic diffusion (for NH3) and ionic transport (for NH4 ). NH3
regulation of NH4 metabolic pathways appears to facilitate quickly and easily diffuses across the plasma membrane to
glutamine production when NH4 excretion is stimulated by compartments of lower pH, where it is rapidly converted to
acidemia or, conversely, blunts glutamine production when NH4 , thus leading to accumulated NH4 in acidic compart-
excretion is inhibited by alkalemia.5 ments. NH4 can diffuse across tight junctions or can substi-
tute on transporters such as the apical membrane Na –H
Renal Excretion of Ammonia/Ammonium (NHE-3) antiporter (as Na –NH4 exchange).6 In general,
The ultimate control, however, resides in the renal excretion NH3 has a higher membrane permeability than NH4 , but
of NH4 , because the NH4 must be excreted to escape entry the concentration of NH4 exceeds that of NH3 by approx-
into the hepatic urea synthetic pool. Hepatic urea synthesis imately 100-fold at a pH 7.4 (pK 9.3). Thus, the relative
218
21 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

permeability of the two moieties de nes whether either it would return to the distal nephron and diffuse back into
crosses membranes by nonionic NH3 diffusion or ionic N the blood, thereby failing to be excreted in the urine. The
H4 transport; for example, if the NH4 moiety is transported thick ascending limb of the loop of Henle also creates a
in a speci c nephron segment as in the thick ascending limb “corticomedullary concentration gradient” for NH3–NH4
of the loop of Henle by substitution for potassium on the . The NH3–NH4 transported into the medullary intersti-
Na –K –2Cl cotransporter. tium accumulates at a higher concentration by countercur-
The rst step of ammonia transport and urinary excre- rent multiplication and can reenter the collecting tubule
tion following ammoniagenesis in the proximal tubule is for for a nal excretion in a more regulated way. Nonionic dif-
the ammonia to enter the proximal tubule lumen, and it can fusion of NH3 into the medullary interstitium is not pos-
do so in two ways. Because the luminal pH is lower than sible because the thick ascending limb’s apical membrane
cell pH, much of the NH3 is diffused into the tubule uid is highly impermeable to NH3.9 NaCl transport enables
and is converted into NH4 . The apical membrane Na -H the ionic transport of NH4 into the interstitium. NH4 can
antiporter also plays an important role in NH3–NH4 trans- substitute for K on the Na –K –2Cl cotransporter, or it
port (Fig. 7.1A).7,8 Transport mechanisms for NH3–NH4 in can be transported via the renal outer medullary potassium
more distal segments function to ensure that the NH3– NH4 (ROMK) channel for transport across the apical membrane
issuing out of the proximal tubule uid is excreted into the (Fig. 7.1B). Alternatively, lumen-positive voltage can drive
urine in a regulated fashion. NH4 diffusion across the tight junction. NH4 that enters the
From the lumen, ammonia enters the medullary thick ascending limb cell can exit the basolateral membrane
interstitium (Fig. 7.2) via the thick ascending limb of the by nonionic diffusion driven by cell-to-interstitial NH3
loop of Henle. If ammonia did not enter the interstitium, concentration gradient. Once in the interstitium, the NH3

FIGURE 7.2 Nephron sites of ammonia production, secretion, transport, and ultimately, excretion. Note that approximately 75%
of ammonia/ammonium delivered to the loop of Henle is transported into the medullary interstitium and is accumulated through
countercurrent multiplication, thus resulting in higher concentrations from the cortex to the medulla. In acidosis, both the production
of ammonia/ammonium and the transport, multiplication, and excretion of NH4 is augmented. Therefore, the physiologic response of
the kidney to an acid load is to increase bicarbonate reabsorption and ammonia production and excretion. The resulting urine should
demonstrate a pH 5.5 and a higher concentration of ammonium. If these conditions are met, a normal gap metabolic acidosis
would be nonrenal in origin.
CHAPTER 7 RENAL ACID–BASE TRANSPORT 2219
19

will again accept a H , forming NH4 . Furosemide, which observed when RhCG was deleted only from the collect
inhibits the Na –K –2Cl cotransporter and secondarily ducts.17 In summary, that both intercalated and principal
inhibits the lumen-positive voltage, signi cantly inhibits cells express RhCG and that metabolic acidosis increases
all three mechanisms of thick ascending limb NH3–NH4 RhCG in both cell types suggests that RhCG contributes to
absorption.6,10,11 transepithelial ammonia secretion.16
Through these processes, NH3–NH4 accumulates in the Therefore, the sum of TA and ammonia/ammonium
renal medulla (Fig. 7.2). The countercurrent system concen- excretion is represented, as noted previously, as net
trates the solute toward the tip of the medulla and preserves acid excretion (Eq. 9). All three components of net acid
the cortical-to-medullary gradient. Blood in the descending excretion are dependent on the operation of an H secre-
vasa recta has lower NH3–NH4 concentrations than the sur- tory mechanism into the tubule uid. Therefore, increases
rounding interstitium, allowing net entry. Conversely, blood in the rate of H secretion will increase urinary ammo-
in the ascending vasa recta has a higher NH3–NH4 concen- nium and TA excretion and will lower urinary HCO3 ex-
tration than the surrounding interstitium, causing net ef ux. cretion. A protein-rich diet leads to acid production, so
In patients with an intact urinary concentrating system and the kidneys must both reclaim the ltered bicarbonate
medullary architecture, the net result is a steep gradient, and also excrete an acid equivalent to that produced in
with the highest concentrations of NH3–NH4 in the inner the diet. In this way, in a steady state, net acid produc-
medullary interstitium. tion equals net acid excretion, and acid–base balance is
Ultimately, NH3–NH4 is transported from the medul- maintained. In the presence of nonprotein-rich diets, the
lary interstitium into the lumen of the collecting tubule for tubules do not reclaim all of the ltered bicarbonate and
nal urinary excretion (Figs. 7.1B and 7.2). The apical and they decrease production of new bicarbonate in order to
basolateral membranes of the medullary collecting tubule maintain acid–base homeostasis.
are permeable to NH3, allowing nonionic diffusion to be
driven by the low pH of the collecting tubule uid. In ad-
dition, NH4 can be transported from the medullary intersti- SPECIFIC MECHANISMS OF H – HCO3
tium into the collecting tubule cell on the Na , K -ATPase.12 TRANSPORT ALONG THE NEPHRON
Once inside the cell, the dissociation of NH4 into NH3 and How the transport of H and HCO3 in the nephron occurs
H provides a source of H for transport into the luminal is generally de ned by the speci c characteristics of each
uid by the vacuolar H -ATPase. H secreted into the lu- epithelium and cell type within the nephron segments.
minal uid will combine with secreted NH3 to form NH4 , Bicarbonate reabsorption is largely the responsibility
maintaining the gradient for nonionic NH3 diffusion into the of the proximal tubule and net acid secretion is largely
luminal compartment, which has a much lower pH, and this of the distal tubule. A number of transport mechanisms
way NH4 is “trapped” in tubule uid and is swept into the across either the apical (for H ) or basolateral (for alkali)
collecting system. Medullary interstitial NH3–NH4 concen- membranes have been identi ed and are detailed in the
trations are determined by the rate of ammonia synthesis following paragraphs.
in the proximal tubule, the ef ciency of ammonia transport
into the lumen of the proximal tubule and subsequently into
the medullary interstitium in the thick ascending limb, and Apical Membrane H -Transport Mechanisms
the ef ciency of countercurrent trapping of ammonia in the Renal tubule cells use both primary and secondary trans-
medullary interstitium. Both ammonia synthesis and each port mechanisms for active H secretion. A primary trans-
step in the highly integrated response by the nephron to ex- port couples the metabolism to a transport mechanism,
crete ammonia are upregulated by metabolic acidosis and whereas a secondary transport couples the transporter to
chronic hypokalemia, and are inhibited by alkalosis and hy- the metabolism, which generates a concentration gradient
perkalemia (see Chapter 18). for a solute.
The Rh glycoprotein (RhCG/Rhcg), which is expressed There are two mechanisms for primary active H secre-
not only in both principal and intercalated cells but also tion, both of which are directly related to the metabolism
in the distal convoluted tubule, connecting tubule, initial of adenosine triphosphate (ATP) to adenosine diphosphate
collecting tubule, cortical collecting duct, and the outer and (ADP) and inorganic phosphate (Pi). The rst and quanti-
inner collecting ducts, is now thought to be critical for the tatively most important mechanism involves the vacuolar
excretion of ammonia by the kidney (Fig. 7.1B).13,14 RhCG H -ATPase, or V-type ATPase, a multisubunit protein com-
expression is greater in A-type intercalated cells than in prin- plex that secretes H ,18 and is electrogenic, thereby gener-
cipal cells but is not detectable in B-type intercalated cells.15 ating a positive charge in the tubule lumen. The ability of
RhCG is thought to play a role in renal ammonia excretion the H -ATPase to affect H secretion depends on where it
because of some recent genetic studies. Biver and colleagues16 is located; when it is in the proximal tubule, it minimally
found that global RhCG deletion impaired ammonia excre- mediates H secretion into the lumen, but in type A (acid
tion in the urine and decreased basal ammonia excretion secreting) intercalated cells of the collecting tubule, it sig-
in response to metabolic acidosis. Similar ndings were ni cantly mediates H secretion into the lumen. This
220
22 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

mechanism is able to adapt as needed by increasing mRNA


and protein abundance to protect against metabolic and re-
SEGMENTAL CONTRIBUTION TO
spiratory acidosis. BICARBONATE ABSORPTION AND
The second mechanism for primary active H secretion ACIDIFICATION OF THE URINE
involves P-type ATPases. The P-type ATPases, which are
Proximal Tubule
isoforms of the H –K –ATPase (HK 1, the gastric isoform,
and HK 2, the colonic isoform), participate importantly in The proximal tubule reabsorbs approximately 80% of the
H transport by the collecting tubule. These P-type ATPases ltered HCO3 load; the HCO3 concentration at the end of
are coupled to ATP metabolism, have a phosphorylated the proximal tubule is approximately 8 mEq per liter and the
high-energy intermediate, and possess and subunits. pH is 6.7.45,46 The reabsorption rate is a function of proximal
Both the gastric and colonic isoforms are expressed on the tubule segmentation, so the rate of HCO3 reabsorption is
apical membranes of type A intercalated cells (IC) in the highest in the early segment (S1), decreases in the midseg-
cortical collecting tubule, and the outer and inner medul- ment (S2), and is lowest in the terminal segment (S3).
lary collecting tubules.19–24 These transport processes are The speci c mechanisms responsible for H –HCO3
electroneutral, mediating H secretion across the apical transport in the proximal tubule are shown in Figure 7.3.
membrane into the tubule lumen in exchange for K entry Approximately two-thirds of H secretion is mediated by
(into the cell). Both isoforms react to metabolic or respirato- the apical membrane Na –H antiporter, and the remaining
ry acidosis by increasing in abundance and activity. Alterna- one-third is mediated by the vacuolar H –ATPase.47–49
tively, the two types of H -transporters are also located on The active secretion of H into HCO3 -rich glomerular
the basolateral membranes of type B (bicarbonate secreting) ultra ltrate by the NHE-3 results in the formation of H2CO3.
ICs in the cortical and outer collecting tubule.25–29 The Luminal carbonic anhydrase (type IV) facilitates the conversion
H -ATPase or H , K -ATPase in this location mediates of H2CO3 to CO2 and H2O and prevents the generation of lim-
H transport across the basolateral membrane into the iting pH gradients across the proximal convoluted tubule.50
interstitium. CO2 diffuses readily into the cell where, under the in uence of
The secondary active H transporter is an isoform of the cytoplasmic carbonic anhydrase (type II), it forms H2CO3 that
Na -H antiporter, NHE-3. Although there are eight known dissociates rapidly to H and HCO3 , which are transported,
isoforms of the Na -H antiporter, NHE-3 (SLC9A3) is the respectively, across the apical and basolateral membranes.
most important for bicarbonate reabsorption. It is located in Studies from Weinman’s group51 have clearly established that
the S1 and S2 proximal tubule and thick ascending limb of NHERF-1 (sodium-hydrogen exchange factor 1) is required for
the loop of Henle on the apical membrane and mediates H a protein kinase A (PKA)–associated downregulation of NHE-
secretion into the tubule uid.30–33 3 activity. NHE-3 has a speci c requirement for NHERF-1
for cAMP-mediated phosphorylation and inhibition. In addi-
Basolateral Bicarbonate Transport tion to H secretion via NHE-3, an apical H -ATPase is also
Two transporters mediate passive base ef ux along the responsible for a small but signi cant fraction of bicarbonate
nephron. One transporter is the electroneutral Cl /HCO 3 reclamation in the proximal tubule.
exchanger, which carries HCO 3 out and Cl into the cell HCO3 generated in the proximal tubule cell exits passively
across the basolateral membrane in the S3 segment of across the basolateral membrane Na /n-HCO3 cotransporter,
the proximal tubule, the thick ascending limb of the or NBCe1, encoded by SLC4A4 which, as mentioned previ-
loop of Henle, and the cortical and medullary collecting ously, transports the equivalent of three HCO3 ions out in
tubules. This Cl /HCO 3 exchanger is a member of the parallel with one Na out (Fig. 7.3).41–44,52,53 This is a passive
anion exchanger 1 (AE1) family and is a truncated form process because this transporter carries a net of two nega-
of the red cell AE1 Cl /HCO 3 exchanger,34–37 or kAE1 tive charges, and the negative cell voltage provides a strong,
(SLC4A1). The mRNA lacks the rst three exons of red favorable driving force for base ef ux.
cell mRNA,38 but the net result of this truncation is a As noted previously, in addition to bicarbonate re-
protein with similar transport characteristics but altered absorption, a second function of the proximal tubule
cytoskeletal interactions. Cl enters the cell in exchange in acid–base balance is to synthesize the NH3 needed for
for HCO3 and exits the basolateral membrane through a HCO3 regeneration (see Fig. 7.1A) (see previous discussion
Cl conductance.39,40 on ammoniagenesis and ammonia/ammonium transport).
The second is the sodium bicarbonate cotransporter
(NBC), of which there are four isoforms. The NBCe1 (SLC4A4) The Loop of Henle
isoform is the most important for base ef ux, mediating the Approximately 50% to 70% of the HCO3 delivered out
majority of base ef ux ( 80%) out of the cell and into the of the proximal tubule is reabsorbed in the thick ascend-
interstitium.41–44 It is present on the basolateral membrane ing limb of the loop of Henle.54 The secretion of H across
of the proximal tubule and is electrogenic because it trans- the apical membrane is mediated by NHE-3 as in the
ports three base molecules with one Na ion (Na /n HCO3 proximal tubule (Fig. 7.4B).30,54 A low cellular Na con-
or SLC4A4).41–44 centration, as in the proximal tubule, is maintained by the
CHAPTER 7 RENAL ACID–BASE TRANSPORT 2221
21

FIGURE 7.3 A proximal convoluted tubule: the coupled apical and basolateral transporters responsible for bicarbonate reabsorption.

basolateral membrane Na , K -ATPase, to provide the driv- Subsequent segments, including the connecting tubule, the
ing force for operation of the Na /H exchanger. Both AE1 initial cortical collecting tubule, and the cortical collecting
(chloride–bicarbonate exchange) and NBC-2 or -3 (sodi- tubule all possess ICs that mediate H –HCO3 transport.
um–bicarbonate cotransport) are expressed on the basolat- Type A ICs secrete H into the tubule uid and type B ICs
eral membrane, where they mediate bicarbonate exit across secrete HCO3 across the apical membrane (Fig. 7.4D).
the basolateral membrane.55–57 Type A ICs accomplish H ion secretion through a pro-
cess mediated by two ATP-dependent H pumps, a V-type
The Distal Tubule and the Collecting Duct H -ATPase, and an H , K -ATPase (which is a P-type
The distal tubule and the collecting duct are responsible for ATPase) (Fig. 7.5). There is abundant evidence supporting
the nal acidi cation of the urine. This process involves three a role for both transporters. The H -ATPase has been lo-
steps: (1) the residual HCO3 in tubule uid is reabsorbed; calized by (1) the labeling of cells with antibodies against
(2) the nal titration of buffers is accomplished (TA excre- subunits of the vacuolar H -ATPase,25,26 (2) the inhibition
tion); and (3) the H secreted traps ammonium in the lu- of H transport by ba lomycin A1, a speci c inhibitor of
men so that the majority of ammonia produced is excreted vacuolar H -ATPases,27,29 and (3) the electrogenicity of acid-
as NH4 (Fig. 7.2). Stoichiometrically, excretion of NH4 is i cation in this nephron segment.62 With regard to the lat-
necessary to regenerate the HCO3 lost in the buffering of ter, luminal acidi cation is associated with a lumen-positive
acid products of metabolism. Therefore, each NH4 excreted voltage. Generation of a transepithelial voltage requires elec-
replenishes one HCO3 that is returned to the ECF via the re- trogenic or conductive processes on both apical and basolat-
nal vein. If the NH4 is not excreted by the kidney, NH3 is re- eral membranes. Because the IC does not possess signi cant
turned to the liver where each NH3 molecule generates 2H apical membrane conductance,39,40 electrogenic acidi cation
in ureagenesis. Conversely, when the kidney is required to requires an electrogenic H pump.
excrete alkali, the distal nephron is able to secrete HCO3 .58 Immunohistochemical studies show localization of the
The distal nephron, which begins at the macula densa, electroneutral H , K -ATPase to the apical membrane of a
is a short segment that exhibits a small amount of amiloride- type A IC using antibodies against both the gastric (HK 1) and
sensitive HCO3 reabsorption mediated by an apical mem- the colonic (HK 2) H , K -ATPase isoforms (Fig. 7.5).63–65
brane Na -H antiporter isoform NHE2 (Fig. 7.4C).29,59–61 Bicarbonate transport and potassium absorption are both
222
22 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

H+
Na +
Na +
Na + Cl– 2K+
H+ 3HCO 3 – 3Na +

2K+ H+
3Na + Na +
Cl–

2K+
Na + 3Na +

K+ K+
H+ Cl–
Na + K+
2K+ H+ H+ Cl–
3Na +
Na + Cl– H+ Cl–
2Cl– K+ K+
K+ Na +
3HCO 3 –
Cl– HCO 3 – K+
Cl–
HCO 3 –
Cl–
K+
H+

FIGURE 7.4 The H –HCO3 transport in the proximal tubule (A), the thick ascending limb of the loop of Henle (B), the distal convoluted
tubule (C), and the collecting duct (D). Note that three cell types are displayed for the collecting tubule (D): the principal cell that does not
participate directly in H secretion, the type A intercalated or acid-secreting cell, and the type B intercalated or base-secreting cell.

inhibited by SCH 28080, a speci c inhibitor of the gastric Type BICs secrete HCO3 into tubule uid through the cou-
H , K -ATPase.29,66–69 pling of active secretion of H across the basolateral membrane
The colonic isoform of H , K -ATPase, HK 2, is highly into the interstitium (Fig. 7.5) by both a vacuolar H -ATPase
responsive to and upregulated by chronic potassium de- and an H , K -ATPase.66 Therefore, these cells are mirror im-
pletion. The site of regulation is most robust in the outer ages of type A ICs and appear to be involved in protecting
and inner medulla. The H , K -ATPase may play a highly against metabolic alkalosis through a more ef cient excretion
signi cant role in potassium depletion, when its expres- of chronic bicarbonate loads. Type B ICs secrete HCO3 across
sion is signi cantly increased to minimize K loss in the the apical membrane into the collecting duct via a Cl /HCO3
urine.24,70 This regulatory response may explain the role of exchanger. However, this Cl /HCO3 exchanger is not the same
persistent chronic hypokalemia in the maintenance of meta- AE1 Cl /HCO3 exchanger found on the basolateral membrane
bolic alkalosis by maintaining H secretion mediated by the of the proximal tubule. Rather, the anion exchanger pendrin,
H , K -ATPase, to prevent dangerous hypokalemia. In addi- which is encoded by SLC26A4, is localized to the apical mem-
tion, both HK 1 and HK 2 increase abundance and activity brane of nonacid-secreting ICs in the kidney cortical collect-
due to either metabolic or respiratory acidosis. There is no ing duct (CCD) (Fig. 7.5). Cl that enters the cell in exchange
speci c inhibitor for HK 2 but Li and associates71 in the Du- for HCO3 exits the cell through a basolateral membrane Cl
Bose laboratory have shown inhibition by intermediate con- conductance. This process does not generate a transepithelial
centrations of ouabain in vitro. Therefore, in summary, both voltage, because there is no net transcellular current.
the V-type H -ATPase and the H , K -ATPases contribute to The ability to secrete HCO3 disappears in the outer
H secretion, although their relative contributions appear to medullary collecting tubule. Here, the H ion secretion is
be nephron segment–speci c. mediated by cells that are functionally similar to type A ICs.
CHAPTER 7 RENAL ACID–BASE TRANSPORT 2223
23

FIGURE 7.5 The H –HCO3 transport in the collecting tubule, which occurs in type A and type B intercalated cells.

In some cases, these cells are designated as non-A and non-B turn signal the tubules to secrete H more rapidly. If the
intercalated cells. The inner stripe of the outer medullary rate of acid excretion did not change, too little bicarbonate
collecting tubule is a high-capacity segment for H -secre- would be reabsorbed and the luminal pH would decrease
tion. The inner medullary collecting tubule has been dif cult too slowly. Alkaline tubule uid stimulates apical membrane
to study because of the dif culty in isolating this nephron H transporters.72 The tubule ow rate can also directly
segment for tubular perfusion. Nevertheless, studies in our regulate the rate of proximal tubule H secretion, regardless
laboratory provide evidence for a mechanism of H secretion of HCO3 concentration or luminal composition.73
similar to that described for the type A ICs.24,70 Moreover, it
should be emphasized that this key segment is responsible Systemic pH
for the nal acidi cation of the tubule uid and generates the As noted previously, the kidneys respond to changes in
low urinary pH typical of high acid excretion states. systemic arterial pH (PCO2) by either increasing urinary
acidi cation (for acidosis) or by inhibiting acidi cation and
INTEGRATED KIDNEY REGULATION OF increasing bicarbonate excretion (for alkalosis). Because
changes in plasma pH cause changes in renal interstitial
URINARY ACIDIFICATION AND EXCESS
pH, the mechanisms in uencing bicarbonate ef ux on the
BASE EXCRETION basolateral membrane are also affected. Acidosis enhances
The integrated response of heterogeneous nephron segments HCO3 ef ux and alkalosis inhibits basolateral membrane
to regulate acid–base balance requires the regulation of trans- HCO3 ef ux. Thus, in acidosis, cell pH decreases; whereas
port in each of the nephron segment in concert (Fig. 7.4). in alkalosis, it increases.74 It is the change in intracellular pH
In the following sections, the most important regulatory that determines apical membrane H ion secretion; relative
responses of H –HCO3 transport are emphasized. acidity stimulates H secretion, whereas a more alkaline pH
reduces or inhibits H secretion.
Luminal HCO3 Delivery The Na -H antiporter is activated by decreases in
In order to avoid metabolic acidosis or alkalosis, H ion intracellular pH,75 which also stimulates the insertion of H
secretion and HCO3 reabsorption must work in concert. transporters into the apical membrane.76,77 These “reserved”
GFR increases lead to luminal ow rate increases, which in H ions in the apical membrane can enhance the rate of H
224
22 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

transport when needed. Long-term adaptations in tubular func- excretion and a relatively alkaline urine pH, parallel with
tion result from chronic systemic pH changes. In the proximal a spontaneous nongap metabolic acidosis, thus providing
tubule, chronic metabolic and respiratory acidosis enhances evidence for an acidi cation defect in this model. Moreover,
the proximal tubule capacity for H secretion and ammonia- some of the features of the renal acidi cation defect in
genesis.78,79 These changes are mediated by parallel increases in GPR4-/- resemble that which is reported for mice with the
apical membrane Na -H antiporter and electrogenic basolat- deletion of one of the genes that encodes for the rate-limiting
eral membrane Na /n-HCO3 cotransporter activity.80–83 bicarbonate transporter, AE1, in the kidney collecting tu-
Such transporter adaptations are known to be a direct bule. Therefore, GPR4 appears to function as a pH sensor in
effect of pH because they can be experimentally induced by the collecting tubule.
incubating cultured proximal tubule cells in acid media.84 Recent studies have revealed that soluble adenylyl
Enhanced activity of the apical Na -H antiporter is medi- cyclase (sAC) is highly expressed in the collecting tubule and
ated by the traf cking of NHE-3 to the apical membrane, and may colocalize with the H -ATPase in the apical membrane
increasing whole cell NHE-3 abundance.85 Several groups have of type A ICs.100 It has been suggested that sAC-regulated
demonstrated that endothelin-1 (ET-1) increases the activity of cAMP signaling may constitute a general sensing mecha-
NHE-3 and that metabolic acidosis stimulates renal proximal nism for regulating H -ATPase-mediated proton transport.
tubule ET-1 expression.86–89 Studies in ETB receptor knockout Although parallels exist in the male reproductive tract,101
mice demonstrate that the ET-1/ETB signaling pathway medi- how sAC functions to sense bicarbonate, primarily, and how
ates acid stimulation of NHE-3 activity.90,91 In addition, a con- it might function to play a regulatory role in the response to
sensus sequence (KXXXVPKXXXV) in the second intracellular an acid intracellular or extracellular pH, which is pivotal for
loop of the ETB receptor appears to be responsible for ET-1/ETB the renal defense against metabolic acidosis, has not been
stimulation of NHE-3 activity.92 satisfactorily explained to date.
How the extracellular pH and the systemic acid–base
status are sensed by renal epithelial cells and initiate a regu-
latory response has not been elucidated. Factors such as pH, INTEGRATION OF KIDNEY RESPONSE
PCO2 and bicarbonate, along with possible hormonal stim-
uli28 have all been proposed at one time or another. Early
TO DEFEND AGAINST ACID–BASE
studies by Pitts dating to the 1940s indicated that metabolic DISTURBANCES
acidosis induced by the infusion of 0.1 N HCl causes an As noted previously, a high-protein diet, such as that con-
increase in net acid excretion, predominately as ammonium sumed by most Westerners, requires renal reclamation of
excretion.93,94 Schwartz and Al-Awqati77 demonstrated that most of the ltered bicarbonate as well as net acid excre-
an increase in ambient PCO2 stimulated proton secretion tion that is equivalent to net acid production. The ammonia
in isolated perfused proximal tubules and in the collecting produced in the kidney is derived from the synthesis of glu-
ducts of rabbit nephrons. It was later shown that respira- tamine, and the amount produced is regulated by pH. Aci-
tory acidosis was associated with the apical insertion of the demia promotes and alkalemia inhibits glutamine synthesis.
H -ATPase. Recent studies have suggested that the nonre- Long-term changes in dietary acidity can cause chronic
ceptor tyrosine kinases Pyk2 and c-Src may represent a pos- changes in H and HCO3 secretion. For example, in the
sible signaling pathway involved in the increase in NHE-3 cortical collecting tubule, chronic increases in dietary acid
function in the proximal tubule in response to metabolic aci- result in an increased capacity for H secretion into the
dosis.95–97 Although there is preliminary evidence that Pyk2 lumen, whereas chronic increases in dietary alkali lead to
is expressed and may also function in the collecting tubule an increased capacity for HCO3 secretion.102 As noted pre-
as a pH sensor, the signaling pathway has not yet been delin- viously, type A ICs mediate H secretion and type B ICs
eated (K. Fisher, personal communication, February 2012). mediate HCO3 secretion. The elegant studies of Schwartz
Recently, a small number of G protein-coupled receptors and Al-Awqati103 over many years have demonstrated that
(GPCR) have been identi ed, the activation of which is stim- these cells can “switch” phenotypes based on chronic meta-
ulated by a reduction in extracellular pH, prompting them to bolic changes. For example, during metabolic acidosis, the
be termed “proton-sensing” receptors. One of these putative number of type B ICs decreases and the number of type A ICs
proton-sensing GPCRs is the orphan receptor GPR4.98 GPR4 increases, but the total number of intercalated cells does not
is an upstream activator of cAMP-PKA signaling, and our change.104–107 This “switch” could be due to an extracellular
preliminary studies implicate GPR4 in the upregulation of protein called hensin.103 Recent results demonstrate that aci-
proton transporters in the collecting duct of the kidney. dosis induces the polymerization of this novel extracellular
Together with existing literature on the activation of other matrix protein as follows: The deposition of hensin in the
proton translocating ATPases, these ndings indicate the matrix leads to the conversion of a HCO3 -secreting epithe-
presence of an underlying signaling network involved in lium to an acid-secreting epithelium. Hensin is deposited
coordinating the renal adaptive response to acidosis. in the extracellular matrix of H -secreting type A ICs fol-
Petrovic and colleagues99 recently reported that deletion lowing acid incubation.108 Alternatively, this “switch” could
of GPR4 in vivo was associated with a reduction in net acid also be due to the exocytotic insertion of H -ATPase into the
CHAPTER 7 RENAL ACID–BASE TRANSPORT 2225
25

apical membrane of type A ICs and the endocytic retrieval of increases net acid excretion and helps correct the acidosis in
the Cl /HCO3 exchanger from the apical membrane of the disorders associated with hyperkalemia and the metabolic
type B ICs. acidosis of renal origin.20,114
As noted previously, the kidneys eliminate the acid
that is produced daily and have the capacity to increase
urinary net acid excretion (and, hence, HCO3 generation) in
THE EFFECT OF
response to endogenous or exogenous acid loads. The renal MINERALOCORTICOIDS ON URINARY
excretion of acid is usually matched to the net production of ACIDIFICATION
metabolic and dietary acids, so little disturbance in systemic Mineralocorticoids such as aldosterone stimulate urinary
pH occurs. acidi cation through a number of different mechanisms.
As an acid load is incurred, the kidneys respond to Aldosterone binds to its intracellular mineralocorticoid
restore balance by increasing the ammonium excretion (TA receptor in collecting tubule cells and stimulates the rate of
excretion has limited capacity for regulation). With contin- H ion transport119,120 by parallel increases in the activities
ued acid loading, the renal net acid excretion increases over of the apical membrane V-type H -ATPase121 and the baso-
the course of 3 to 5 days (Fig. 7.2) but does not quite achieve lateral membrane Cl /HCO3 exchanger.122 It also has indi-
the level of acid production. Progressive positive acid balance rect effects on H secretion by increasing Na reabsorption
ensues, which is presumably buffered by bone carbonate. in the cortical collecting tubule (CCT) through increasing
the open probability of the apical sodium channel in princi-
pal cells. This secondarily increases the transepithelial nega-
K DEFICIENCY AND HYPERKALEMIA
tive potential difference, which is an important driving force
Chronic K de ciency leads to adaptations in H ion secre- for H secretion.123
tion that are similar to those evoked by chronic metabolic
acidosis. Thus, chronic K de ciency leads to an increased Effective Arterial Volume
capacity for transepithelial H secretion by the proximal
tubule through parallel increases in the activities of NHE-3 Effective arterial volume is an important regulator of renal
and the basolateral membrane Na –3HCO3 cotransporter109 acidi cation and is reviewed in the following paragraphs.
and increases in activities of ammoniagenesis that are also
stimulated by chronic metabolic acidosis (Fig. 7.1A).110 The Extracellular Volume Expansion
similarity between these two responses may be the result of The addition of sodium bicarbonate to the body results in
chronic K de ciency, which decreases intracellular pH and prompt cellular buffering and respiratory compensation.
increases the NHE-3 transport rate.111,112 However, as with an acid load, the kidneys have the ulti-
In the collecting tubule, chronic K de ciency increases mate responsibility for the disposal of base to restore a nor-
H secretion through the insertion of H pumps into the mal serum bicarbonate concentration. The renal response
apical membrane of H -secreting cells (A-type ICs) and is more rapid with the addition of HCO3 than with acid
through a robust increase in the activity of the H , K - ingestion. The speed and ef ciency by which HCO3 can
ATPase (Fig. 7.1B).70,113–115 The result of the latter is to en- be excreted renders it challenging to cause more than mild
hance H ion secretion while, at the same time, to enhance metabolic alkalosis in a patient with normal renal function
K absorption by the distal nephron. The stimulation of re- even with as much as 24 mEq per kilogram per day of so-
nal acidi cation in chronic K de ciency explains the asso- dium bicarbonate over several weeks.124,125 Type B ICs in
ciation of K de ciency with chronic metabolic alkalosis and the collecting tubule are capable of HCO3 /Cl exchange
is primarily an adaptive upregulation of the H , K -ATPase, (bicarbonate secretion). However, the role of HCO3 secre-
thus offering an explanation for the paradoxical increase in tion in chronic alkali loading has never been quantitated
bicarbonate absorption in the face of chronic metabolic alka- precisely in whole organ clearance studies. Presumably,
losis with hypokalemia.116 Furthermore, the metabolic alka- HCO3 secretion by the type B IC moderates the devel-
losis associated with hypokalemia cannot be corrected until opment of a more severe alkalosis, enhancing the HCO3
the potassium de ciency is repaired. excretory response (Fig. 7.5).
Hyperkalemia appears to have the opposite effect on The proximal tubule is principally responsible for HCO3
acid–base balance because hyperkalemia inhibits proximal excretion when the blood HCO3 concentration increases.
tubule ammoniagenesis and ammonium transport. This Absolute proximal HCO3 reabsorption does not increase
decrease in ammonium excretion due to hyperkalemia in proportion to the load of HCO3 because alkalemia sup-
contributes to the acidi cation defect characteristic of type presses proximal acidi cation,126 causing an increase in the
4 renal tubular acidosis.117,118 Hyperkalemia not only sup- HCO3 delivery to the distal nephron. The limited capacity
presses ammoniagenesis, but also inhibits NH4 reabsorption of the distal nephron to secrete H can be overwhelmed eas-
in the thick ascending limb, resulting in low NH3 levels in ily, and bicarbonaturia increases progressively. NH4 and TA
the renal medulla, and, thus, a reduced capacity to buffer H excretion are moderated in response to the increasing urine
secreted in the collecting tubule.118 Correcting hyperkalemia pH.124,126
226
22 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Acute graded HCO3 loads that concomitantly increase activity. When taken together, these factors enhance HCO3
ECF also function in human subjects to progressively reabsorption and regeneration.
increase urinary HCO3 excretion as the plasma HCO3 con- De ciency of both Cl and K is common in metabolic
centration increases.126 In summary, an acute base load is ex- alkalosis because of renal and/or gastrointestinal losses that
creted entirely when the kidney function is normal, and the occur concurrently with the generation of the alkalosis.129,130
blood HCO3 concentration is returned to normal within 12 With Cl depletion, the normal bicarbonaturic response to
to 24 hours because of the depression of fractional proximal an increase in plasma HCO3 is prevented, and metabolic
HCO3 reabsorption. In addition to the suppression of the alkalosis can develop. Potassium depletion, even without
ltered HCO3 load reabsorption, direct HCO3 secretion by mineralocorticoid administration, can cause metabolic alka-
the CCT is another means for HCO3 disposal during meta- losis in rats and humans. When Cl and K depletion co-
bolic alkalosis.124 exist, severe metabolic alkalosis may develop in all species
The increased delivery of HCO3 out of the proximal studied.
tubule in response to an increased blood HCO3 concentra- Two general mechanisms exist by which the bicar-
tion ( ltered HCO3 load) in the setting of ECF expansion bonaturic response to hyperbicarbonatemia can be prevent-
facilitates HCO3 excretion and the return of blood pH to ed by Cl and/or K depletion: (1) As the plasma HCO3
normal. However, other factors may independently enhance concentration increases, there is a reciprocal fall in GFR and,
distal H secretion suf ciently to prevent HCO3 excretion if inversely proportional to the rise in the plasma HCO3
and thus counterbalance the suppressed fractional proximal concentration, the ltered HCO3 load would not exceed
HCO3 reabsorptive capacity. Under these circumstances, the normal level. Accordingly, normal rates of proximal and
the alkalosis is maintained. For example, in the setting of distal HCO3 reabsorption would suf ce to prevent bicar-
primary hyperaldosteronism, despite the expanded ECF, a bonaturia. (2) Cl de ciency or K de ciency increases the
stable mild alkalotic condition persists in most experimental overall renal HCO3 reabsorption in the setting of a normal
models owing to augmented collecting duct H secretion.124 GFR and a high ltered HCO3 load. In this case, overall re-
In such cases, concurrent hypokalemia facilitates the genera- nal HCO3 reabsorption, and therefore acidi cation, would
tion and maintenance of metabolic alkalosis by enhancing N be increased. An increase in renal acidi cation might occur
H4 production and excretion.124,126 Moreover, chronic hy- as a result of an increase in H secretion by the proximal or
pokalemia dramatically enhances the abundance and func- the distal nephron or by both nephron segments.124,127,129
tionality of the H , K -ATPase in the medullary collecting The augmented HCO3 absorption in distal nephron seg-
tubule, thus increasing rather than decreasing bicarbonate ments appears to be due to a primary increase in H secretion
absorption.124,127–129 Enhanced nonreabsorbable anion de- that is independent of the delivered HCO3 load. Chronic hy-
livery, as with drug anions, also increases the net collecting pokalemia dramatically enhances the abundance and function
tubule H secretion by increasing the effective luminal nega- of the colonic isoform of the H , K -ATPase in the medullary
tive potential difference or by suppressing HCO3 secretion collecting tubule, which may serve as a signi cant factor in
in the cortical collecting duct (CCD). the maintenance of chronic metabolic alkalosis.70,115,127
In summary, the physiologic response by the kidney to a
Extracellular Volume Contraction base load associated with volume expansion is to excrete the
base. Base is retained, however, if there is enhanced distal
The renal response to an increase in plasma HCO3 concen- HCO3 reabsorption as a result of K and/or Cl de ciency.
tration can be modi ed signi cantly in the presence of ECF
contraction.128,130 Because the distribution volume of Cl
REFERENCES
is approximately equal to ECF, the depletion of the ECF is
roughly equivalent to the depletion of Cl . The critical role 1. Bidani
Bidani A,
A, DuBose
DuBose TD
TD Jr.
Jr. Acid-base
Acid-base regulation:
regulation: cellular
cellular and
and whole
whole body.
body. In:
In:
AI, DeFronzo
Arieff AI, DeFronzoRA, RA,eds.
eds.Fluid,
Fluid,Electrolyte,
Electrolyte, and
and Acid
Acid Base
Base Disorders.
Disorders. 2nd ed.
ed.
of effective ECF and K stores in modifying net HCO3 reab- New York: Churchill Livingstone; 1995:69.
sorption has been demonstrated in numerous experimental BidaniA,A,Tauzon
2. Bidani TauzonDM,DM,Heming
Heming TA.TA. Regulation
Regulation of whole
of whole bodybody acid-base
acid-base bal-
models. balance.
ance. In: DuBose
In: DuBose TD, Hamm
TD, Hamm LL, eds.LL, eds. Acid-Base
Acid-Base and Electrolyte
and Electrolyte Disorders:
Disorders: A Com-
A Comto
panion panion to and
Brenner Brenner andThe
Rector’s Rector’s
Kidney.The Kidney Philadelphia:
Philadelphia: WB Saunders;WB Saunders;
2002:1–21.
In most cases, metabolic alkalosis is maintained by 2002:1–21.
3. Madias NE, Adrogue HJ. Respiratory alkalosis. In: DuBose TD, Hamm LL,
decreases in effective arterial volume. This is due, in part, to eds.3. Acid-Base
Madias NE,andAdrogue HJ. Disorders:
Electrolyte RespiratoryA alkalosis.
CompanionIn: to
DuBose TD,and
Brenner Hamm LL,
Rector’s
volume-induced decreases in GFR (leading to a decrease in eds. Kidney.
The Acid-Base and Electrolyte
Philadelphia: WBDisorders:
Saunders;A2002:147–164.
Companion to Brenner and Rector’s The
Kidney. Philadelphia:
4. Toews WB Saunders;
GB. Respiratory acidosis.2002:147–164.
In: DuBose TD, Hamm LL, eds. Acid-Base
ltered HCO3 load).131 Moreover, a decrease in effective ar- http://www.ncbi.nlm.nih.gov/pubmed/12198178
and Electrolyte Disorders: A Companion to Brenner and Rector’s The Kidney.
terial volume increases renal acidi cation by (1) decreasing 4. Toews GB.
Philadelphia: WBRespiratory acidosis. In: DuBose TD, Hamm LL, eds. Acid-Base
Saunders; 2002:129–146.
paracellular HCO3 permeability in the proximal tubule, and5.Electrolyte
Gennari Disorders:
FJ, MaddoxA Companion
DA. Renal to Brenner and Rector’s
regulation The Kidney.
of acid-base Philadel-
homeostasis:
phia: WB Saunders;
integrated response.2002:129–146.
In: Seldin DW, Giebisch G, eds. The Kidney: Physiol-
which inhibits HCO3 back leak132; (2) decreasing the dis- ogy5.and Gennari FJ, Maddox
Pathophysiology. DA.Philadelphia:
3rd ed. Renal regulation of acid-base
Lippincott Williams &homeosta-
Wilkins;
tal delivery of Cl , thus inhibiting HCO3 secretion in the sis: integrated response. In: Seldin DW, Giebisch G, eds. The Kidney: Physiol-
2000:2015–2054.
cortical collecting tubule (by limiting the apical mem- ogy6.and Pathophysiology.
Good DW, Knepper3rdMA. ed.Ammonia
Philadelphia: Lippincott
transport in theWilliams
mammalian& Wilkins;
kidney.
2000:2015–2054.
Am J Physiol. 1985;248(4 Pt 2):F459–F471.
brane Cl –HCO3 exchange); and (3) secondarily increas- 6. Kinsella
7. Good DW, KnepperPS.
JL, Aronson MA. Ammonia
Interaction of transport
NH4 andinLithewithmammalian
the renal kidney.
micro-
ing aldosterone and angiotensin II, as well as renal nerve Am J Physiol.
villus membrane1985;248(4
Na -H Ptexchanger.
2):F459–F471.Am J Physiol. 1981;241(5):C220–C226.
CHAPTER 7 RENAL ACID–BASE TRANSPORT 2227
27

7. Preisig
8. KinsellaPA, JL,Alpern
Aronson RJ.PS. Interaction
Pathways o NH4+
for apical andand Li+ with membrane
basolateral the renal micro-NH3 32. OrlowskiVL,
36. Schuster J, Kandasamy
Bonsib SM,RA, Shull GE.
Jennings ML.Molecular
Two typescloning o putaduct
of collecting tive mem-
mito-
villusNH
and membrane
4
movement Na+-H+ in rat exchanger.
proximalAm J Physiol.
tubule. Am 1981;241(5):C220–C226.
J Physiol. 1990;259(4 Pt 2): bers o the Na/H
chondria-rich exchangercells:
(intercalated) genelectin
amily. andcDNAband 3cloning, deducedAm
cytochemistry. amino acid
J Physiol.
8. Preisig PA, Alpern RJ. Pathways or apical and basolateral membrane NH3
F587–F593. sequence,
1986;251(3and mRNA tissue expression o the rat Na/H exchanger NHE-1 and
Pt 1):C347–C355.
and9. NH 4
+
Kikeri movement
D, Sun A,inZeidel rat proximal
ML, Hebert tubule. Am Jmembranes
SC. Cell Physiol. 1990;259(4
impermeable Pt 2):
to two
37.structurally
Wagner S, relatedVogen proteins.
R, LietzkeJ R, BioletChem. 1992;267(13): 9331–9339.
al. Immunochemical characterization of
F587–F593.
NH3. Nature (London). 1989;339(6224):478–480. http://www.ncbi.nlm.nih.gov/pubmed/1577762
a band 3-like anion exchanger in collecting duct of human kidney. Am J Physiol.
9. Good
10. KikeriDW, D, SunKnepperA, Zeidel
MA, ML,BurgHebert
MB. AmmoniaSC. Celland membranes
bicarbonate impermeable
transport by to 33. Tse CM,
1987;253(2 Brant SR, Walker MS, et al. Cloning and sequencing o a rab-
Pt 2):F213–F221.
NH3. ascending
thick Nature (London).limb of1989;339(6224):478–480.
rat kidney. Am J Physiol. 1984;247(1 Pt 2):F35–F44. bit38.cDNA encoding
Kudrycki KE, an intestinal
Shull GE. Ratand kidney-specif
kidney band 3 Cl c –Na +
/HCO/H+–3 exchanger
exchanger iso orm
mRNA
http://www.ncbi.nlm.nih.gov/pubmed/2725680
11. Good DW. Active absorption of NH4 by rat medullary thick ascending (NHE-3).
is transcribed J Biolfrom
Chem.an1992;267(13):9340–9346.
alternative promoter. Am J Physiol. 1993;264(3 Pt 2):
10. inhibition
limb: Good DW,by Knepper
potassium. MA, Am BurgJ Physiol.
MB. Ammonia and bicarbonate
1988;255(1 Pt 2):F78–F87.transport by 34. Brosius FC, III, Alper SL, Garcia AM, et al. The major kidney band 3 gene
F540–F547.
thick
12. ascending
Wall SM. limb o rat kidney.
Mechanisms of NHAm 4
J Physiol.
and NH3 1984;247(1
transport during Pt 2):F35–F44.
hypokalemia. transcript
39. Koeppen predictsBM.anConductive
amino-terminal truncated
properties of theband 3 polypeptide.
rabbit outer medullary J Biolcollect-
Chem.
11. Physiol
Acta Good DW. Scand. Active absorption o NH4 by rat medullary thick ascending
2003;179(4):325–330. +
1989;264(14):7784–7787.
ing duct: inner stripe. Am J Physiol. 1985;242:F521.
limb:
13. inhibition
Eldari D, Cheval by potassium.
L, Quentin Am J Physiol.
F, et al. 1988;255(1
Expression Pt of 2):F78–F87.
RhCG, a new puta- http://www.ncbi.nlm.nih.gov/pubmed/2542243
40. Koeppen BM. Electrophysiological identi cation of principal and in-
12.NH
tive Wall
3/NH SM.
4
Mechanisms
transporter, o NH
along +
the4 ratandnephron.
NH3 transport
J Am Soc during hypokalemia.
Nephrol. 2002;13: 35. Kudrycki
tercalated cells inKE,the Shull GE. outer
rabbit Primary structurecollecting
medullary o the rat kidney
duct. Pband ugers3 anion
Arch.
Acta Physiol Scand. 2003;179(4):325–330.
1999–2008. exchange
1987;409:138. protein deduced rom a cDNA. J Biol Chem. 1989;264(14): 8185–8192.
http://www.ncbi.nlm.nih.gov/pubmed/14656369
14. Verlander JW, Miller RT, Frank AE, et al. Localization of the ammonium 41. Alpern RJ. Mechanism of basolateral membrane H /OH–/HCO–3 transport
http://www.ncbi.nlm.nih.gov/pubmed/2722777
13. Eldariproteins,
transporter D, Cheval Rh L,B Glycoprotein
Quentin F, et and al. Expression o RhCG,ina the
Rh C Glycoprotein, newmouse
puta- in36.
the Schuster
rat proximal VL, convoluted
Bonsib SM,tubule. Jennings ML. Two types o electrogenic
A sodium-coupled collecting duct mito-J
process.
tive NHAm
kidney. 3
/NH +
transporter,
J Physiol
4
along the
Renal Physiol. rat nephron. J Am Soc Nephrol. 2002;13:
2003;284:F323–F337. chondria-rich (intercalated) cells: lectin and band 3 cytochemistry. Am J Physiol.
Gen Physiol. 1985;86(5):613–636.
1999–2008.
15. Han KH, Lee SY, Kim WY, et al. Expression of the ammonia transporter 1986;251(3
42. Boron Pt WF 1):C347–C355.
, Boulpaep EL. Intracellular pH regulation in the renal proximal
http://www.ncbi.nlm.nih.gov/pubmed/12138130
family members, Rh B Glycoprotein and Rh C Glycoprotein, in the developing 37. Wagner
tubule S, Vogen R,Na-H
of the salamander: Lietzke R, et al. JImmunochemical
exchange. characterization o
Gen Physiol. 1983;81(1):29–52.
rat14. Verlander
kidney. JW, Miller
Am J Physiol RenalRT,Physiol.
Frank AE, et al. Localization o the ammonium
2010;299:F187–F198. a 43.
bandSoleimani
3-like anion M, exchanger
Aronson PS. in collecting
Ionic mechanism duct o humanof Na:HCO kidney. Am J Physiol.
3 cotransport in
transporter
16. Biver S, proteins,
Belge H,RhBourgeois
B GlycoproteinS, et al.and Rh for
A role C Glycoprotein,
rhesus factor Rhcg in theinmouse
renal 1987;253(2
renal basolateral Pt 2):F213–F221.
membrane vesicles (BLMV). J Biol Chem. 1989;264(31):18302–
kidney. Am Jexcretion
ammonium Physiol Renal and Physiol. 2003;284:F323–F337.
male fertility. Nature. 2008;456:339–343. 38. Kudrycki KE, Shull GE. Rat kidney band 3 Cl–/HCO3– exchanger mRNA
18308.
15. Lee
17. HanHW, KH,Verlander
Lee SY, Kim WY, et JM,
JW, Bishop al. Expression
et al. Collecingo the ammoniactransporter
duct-speci Rh C Gly- is44.
transcribed
Yoshitomirom an alternative
K, Burckhardt BC, promoter.
Frömter E.Am J Physiol.sodium-bicarbonate
Rheogenic 1993;264(3 Pt 2):
amily members,
coprotein deletionRh B Glycoprotein
alters and Rh C Glycoprotein,
basal and acidosis-stimulated in the developing
renal ammonia excretion. F540–F547.
cotransport in the peritubular cell membrane of rat renal proximal tubule.
rat kidney.
Am J Physiol Am J Physiol
Renal Renal2009;296:F1364–F1375.
Physiol. Physiol. 2010;299:F187–F198. P39. Koeppen
ugers BM. Conductive properties o the rabbit outer medullary collect-
Arch. 1985;405(4):360–366.
16. Stone
18. Biver S, DK, Belge
Xie H,XS.Bourgeois S, et al. A role
Proton translocating or rhesus
ATPases: issuesactor Rhcg in renal
in structure and ing
45.duct:
DuBoseinnerTD stripe. Am J Physiol.
Jr, Pucacco 1985;242:F521.
LR, Lucci MS, et al. Micropuncture determination
ammonium
function. excretion
Kidney and male ertility. Nature. 2008;456:339–343.
Int. 1988;33(4):767–774. of40.
Ph, Koeppen
PCO2 and BM. total Electrophysiological
CO2 concentration in identif accessiblecation o principal
structures of the ratandrenal
in-
http://www.ncbi.nlm.nih.gov/pubmed/19020613
19. Codina J, Pressley TA, DuBose TD Jr. H ,K -ATPase functions as a Na - tercalated
cortex. J Clin cells in the
Invest. rabbit outer medullary collecting duct. Pf ugers Arch.
1979;64:476–482.
17. Lee HW,
dependent K (NH Verlander
4
)-ATPase JW, Bishop
in apical JM, et al. Collecing
membranes from duct-specif c Rh CJ Gly-
rat distal colon. Biol 1987;409:138.
46. DuBose TD Jr, Lucci MS, Hogg RJ, et al. Comparison of acidi ca-
coprotein
Chem. deletion alters basal and acidosis-stimulated renal ammonia excretion.
1999;274:19693–19698. 41. parameters
tion Alpern RJ. in Mechanism
super cialo and basolateral membraneofHthe
deep nephrons +
/OH –
rat./HCO
Am 3–Jtransport
Physiol.
Am20.J Physiol
CodinaRenal J, WallPhysiol.
SM, 2009;296:F1364–F1375.
DuBose TD Jr. Contrasting functional and regulatory in the rat proximal convoluted tubule. A sodium-coupled electrogenic process. J
1983;244(5):F497–F503.
pro18.lesStone
pro les DK,ofXie theXS.renal Proton
H ,Ktranslocating
-ATPases. Sem ATPases:
Nephrol. issues in structure and
1999;19:399–404. Gen
47.Physiol.
Chan 1985;86(5):613–636.
YL, Giebisch G. Relationship between sodium and bicarbonate
unction.
21. DuBose Kidney TDInt.Jr,1988;33(4):767–774.
Gitomer J, Codina J. H ,K -ATPase. Curr Opin Nephrol http://www.ncbi.nlm.nih.gov/pubmed/2999293
transport in the rat proximal convoluted tubule. Am J Physiol. 1981;240(3):
http://www.ncbi.nlm.nih.gov/pubmed/2838678
Hypertens. 1999;8:597–602. 42. Boron WF, Boulpaep EL. Intracellular pH regulation in the renal proximal
F222–F230.
19. DuBose
22. Codina J, TDPressley
Jr, Codina A, DuBose
J. H ,KTD Jr. H+,KCurr
-ATPase. +
-ATPase unctionsHypertens.
Opin Nephrol as a Na+- tubule o the PA,
48. Preisig salamander:
Ives HE, CragoeNa-H exchange.
EJ Jr, et al.J GenRolePhysiol.
of the Na 1983;81(1):29–52.
/H antiporter in rat
dependent K+(NH4+)-ATPase in apical membranes rom rat distal colon. J Biol
1996;5:411–416. http://www.ncbi.nlm.nih.gov/pubmed/6833996
proximal tubular bicarbonate absorption. J Clin Invest. 1987;80(4):970–978.
Chem.
23. Ono1999;274:19693–19698.
S, Guntupalli J, DuBose TD Jr. Role of H ,K -ATPase in pHi regulation 43. Soleimani M,
49. Schultheis PJ, Aronson
Clarke LL, PS. Meneton
Ionic mechanism o Na:HCO
P, et al. Renal cotransport
and intestinal
3 ab-
http://www.ncbi.nlm.nih.gov/pubmed/10391909
in inner medullary collecting duct cells in culture. Am J Physiol Renal Fluid Elec- in renal defects
sorptive basolateral membrane
in mice lackingvesicles
the NHE3 (BLMV). Na /HJ Biolexchanger.
Chem. 1989;264(31):
Nat Genet.
20. Codina
trolyte Physiol.J,1996;270(39):F1037–F1044.
Wall SM, DuBose TD Jr. Contrasting unctional and regulatory 18302–18308.
1998;19(3):282–285.
prof
24. les profSM,
Wall les oTruong
the renalAV,HDuBose,K -ATPases.
+ +
TD Jr.Sem H Nephrol.
,K -ATPase 1999;19:399–404.
mediates net acid http://www.ncbi.nlm.nih.gov/pubmed/2509453
50. Lucci MS, Tinker JP, Weiner IM, et al. Function of proximal tubule car-
21. DuBose
secretion in ratTD Jr, Gitomer
terminal innerJ, medullary
Codina J. H ,K -ATPase.
+ +
collecting duct. Curr
AmOpin Nephrol
J Physiol Hy-
Renal 44. Y
bonic oshitomi K,
anhydrase deBurckhardt
ned by selectiveBC, Frömter inhibition.E. Rheogenic
Am J Physiol sodium-bicarbonate
Renal Physiol.
pertens.
Fluid 1999;8:597–602.
Electrolyte Physiol. 1996;271(40):F1037–F1044. cotransport in the peritubular cell membrane o rat renal proximal tubule.
1983;245(4):F443–F449.
http://www.ncbi.nlm.nih.gov/pubmed/10541223
25. Brown D, Hirsch S, Gluck S. Localization of a proton-pumping ATPase in Pf51.
ugers Arch. 1985;405(4):360–366.
Weinman EJ, Cunningham R, Wade JB, et al. The role of NHERF-1 in the
rat22. DuBose
kidney. J Clin TDInvest.
Jr, Codina J. H+,K+-ATPase. Curr Opin Nephrol Hypertens.
1988;82(6):2114–2126. http://www.ncbi.nlm.nih.gov/pubmed/3936018
regulation of renal proximal tubule sodium-hydrogen exchanger 3 and sodium-
1996;5:411–416.
26. Brown D, Hirsch S, Gluck S. An H-ATPase in opposite plasma mem- 45. DuBose
dependent TD Jr, Pucacco
phosphate LR, Lucci
cotransporter 2a. JMS, et al. 2005;567(Pt
Physiol. Micropuncture determination
1):27–32.
http://www.ncbi.nlm.nih.gov/pubmed/8937809
brane domains in kidney epithelial cell subpopulations. Nature (London). o 52.
Ph, Romero
PCO2 and MFtotal CO2 MA,
, Hediger concentration
Boulpaep in EL,accessible structures
et al. Expression o theand
cloning rat char-
renal
23. Ono S, Guntupalli J, DuBose TD Jr. Role o H+,K+-ATPase in pHi regulation
1988;331(6157):622–624. cortex. J ClinofInvest.
acterization a renal 1979;64:476–482.
electrogenic Na /HCO3 cotransporter. Nature (London).

in27.
inner medullary
Manger TM, collecting
Koeppen BM. duct Characterization
cells in culture. Am J Physiol Renal
of acid-base Fluid Elec-
transporters in http://www.ncbi.nlm.nih.gov/pubmed/37258
1997;387(6631):409–413.
trolyte Physiol.
cultured outer 1996;270(39):F1037–F1044.
medullary collecting duct cells. Am J Physiol. 1992;263(6 Pt 2): 46. DuBose MF
53. Romero TD Jr, Lucci
, Fong P, Berger MS,UV Hogg
, et al. RJ,
Cloning et al.andComparison o acidif ca-
functional expression of
24. Wall SM, Truong AV, DuBose TD Jr. H+, K+-ATPase mediates net acid se-
F996–F1003. tion
rNBC,parameters
an electrogenic in superf
Na - HCO cial and– deep nephrons
3 cotransporter. o the rat.
Am J Physiol. Am J Physiol.
1998;274(2 Pt 2):
cretion in rat terminal
28. Wagner CA, Finberg inner KE,medullary
Bretoncollecting
S, et al. duct.
RenalAm J PhysiolHRenal
vacuolar Fluid
-ATPase. 1983;244(5):F497–F503.
F425–F432.
Electrolyte
Physiol Rev. Physiol. 1996;271(40):F1037–F1044.
2004;84(4):1263–1314. 47. Chan DW.
54. Good YL, Giebisch
Sodium-dependentG. Relationship bicarbonatebetween sodium by
absorption andcortical
bicarbonate
thick
25. Wang
29. BrownT,D,Malnic HirschG,S,Giebisch
Gluck S. G, Localization
Chan YL. Renal o a proton-pumping ATPase in
bicarbonate reabsorption transport
ascending in theofrat
limb rat proximal
kidney. Amconvoluted tubule. Am J Physiol. 1981;240(3):
J Physiol. 1985;248:F821.
rat the
in kidney.
rat. JIV Clin Invest. 1988;82(6):2114–2126.
. Bicarbonate transport mechanisms in the early and late distal F222–F230.
55. Aalkjaer C, Frische S, Leipziger J, et al. Sodium coupled bicarbonate trans-
http://www.ncbi.nlm.nih.gov/pubmed/2904451
tubule. J Clin Invest. 1993;91(6):2776–2784. 48. Preisig
porters in thePA, Ives an
kidney, HE,update.
CragoeActa EJ Jr,Physiol
et al. Role
Scand.o the Na+/H+ antiporter in rat
2004;181(4):505–512.
26. Amemiya
30. Brown D,M,Hirsch Lof ngS,J, Gluck
Lötscher S. M,
AnetH-ATPase
al. Expressionin opposite
of NHE-3plasma mem-
in the apical proximal
56. Krapf tubular bicarbonate
R. Basolateral absorption.
membrane J Clin Invest.
H/OH/HCO3 1987;80(4):970–978.
transport in the rat cortical
brane domains
membrane of rat in renalkidney
proximal epithelial
tubule celland subpopulations.
thick ascending limb. NatureKidney
(London).
Int. 49. ascending
thick Schultheis limb. PJ, Clarke
Evidence LL, Meneton P, et al. Renal
for an electrogenic and intestinal
Na/HCO3 absorptive
cotransporter in
1988;331(6157):622–624.
1995;48(4):1206–1215. de ects in
parallel with mice
a Na/Hlacking the NHE3
antiporter. J ClinNa +
/H exchanger.
Invest.+
Nat Genet. 1998;19(3):
1988;82(1):234–241.
http://www.ncbi.nlm.nih.gov/pubmed/2893294
31. Biemesderfer D, Pizzonia J, Abu-Alfa A, et al. NHE3: a Na /H exchanger 282–285.
57. Soleimani M. Functional and molecular properties of Na : HCO–3 cotrans-
27. Manger
isoform for renal TM, Koeppen
brush border.BM. AmCharacterization
J Physiol. 1993;265(5 o acid-base transporters in
Pt 2):F736–F742. 50. Lucci
porters (NBC). MS,Minerva
Tinker Urol JP, Weiner
Nefrolog.IM,2003;55(2):131–140.
et al. Function o proximal tubule car-
cultured
32. Orlowskiouter medullary
J, Kandasamy collectingRA, ductShullcells.GE.AmMolecular
J Physiol. 1992;263(6
cloning of Pt 2):
puta- bonic anhydrase
58. DuBose TD def ned by
Jr, Good DW,selective
Hamm inhibition. Am J Physiol
LL, et al. Ammonium Renal Physiol.
transport in the
F996–F1003.
tive members of the Na/H exchanger gene family. cDNA cloning, deduced 1983;245(4):F443–F449.
kidney: new physiological concepts and their clinical implications. J Am Soc
28. Wagner
amino CA, Finberg
acid sequence, and KE,mRNA Breton S, etexpression
tissue al. Renal vacuolar
of the rat H+-ATPase. Physiol
Na/H exchang- 51. Weinman
Nephrol. EJ, Cunningham R, Wade JB, et al. The role o NHERF-1 in the
1991;1(11):1193–1203.
Rev.NHE-1
er 2004;84(4):1263–1314.
and two structurally related proteins. J Biol Chem. 1992;267(13): regulation
59. Bailey o MA,
renal Giebisch
proximal G, tubule sodium-hydrogen
Abbiati exchanger 3 and
T, et al. NHE2-mediated sodium-
bicarbonate
http://www.ncbi.nlm.nih.gov/pubmed/15383652
9331–9339. dependent
reabsorption phosphate
in the distal cotransporter
tubule of NHE3 2a. J Physiol.
null mice. 2005;567(Pt
J Physiol.1):27–32.
2004;561(Pt 3):
29. Tse
33. Wang CM, T, Brant
MalnicSR, G, Walker
GiebischMS, G, Chan
et al. YL. Renaland
Cloning bicarbonate
sequencing reabsorption
of a rab- 52. Romero MF, Hediger MA, Boulpaep EL, et al. Expression cloning and char-
765–775.
in the
bit cDNArat.encoding
IV. Bicarbonate transport
an intestinal and mechanisms
kidney-speci inc the Na early and late distal
/H exchanger isoformtu- acterization
60. Chambrey o a R, renal electrogenic
Warnock Na+/HCORA,
DG, Podevin –
3
cotransporter. Nature (London).
et al. Immunolocalization of
bule. J ClinJ Biol
(NHE-3). Invest. 1993;91(6):2776–2784.
Chem. 1992;267(13):9340–9346. 1997;387(6631):409–413.
the Na /H exchanger isoform NHE2 in rat kidney. Am J Physiol. 1998;275(3
http://www.ncbi.nlm.nih.gov/pubmed/8390489
34. Brosius FC, III, Alper SL, Garcia AM, et al. The major kidney band 3 gene http://www.ncbi.nlm.nih.gov/pubmed/9163427
Pt 2):F379–F386.
30. Amemiya
transcript predicts M, Lo an famino-terminal
ng J, Lötscher M,truncated et al. Expression
band 3 opolypeptide.
NHE-3 in the api-
J Biol 53. Romero
61. Wang T, MF , FongM,P, Aronson
Hropot Berger UV , etetal.al.Cloning
PS, Role ofand NHEunctional
isoforms expression
in mediating o
cal membrane
Chem. o rat renal proximal tubule and thick ascending limb. Kidney Int.
1989;264(14):7784–7787. rNBC, an electrogenic
bicarbonate reabsorption Na along
+
- HCO –
the cotransporter. Am J Physiol. 1998;274(2 Pt 2):
3 nephron. Am J Physiol. 2001;281(6):F1117–
1995;48(4):1206–1215.
35. Kudrycki KE, Shull GE. Primary structure of the rat kidney band F425–F432.
F1122.
3http://www.ncbi.nlm.nih.gov/pubmed/8569082
anion exchange protein deduced from a cDNA. J Biol Chem. 1989;264(14): http://www.ncbi.nlm.nih.gov/pubmed/9486238
62. Koeppen BM, Helman SI. Acidi cation of luminal uid by the rabbit cortical
31. Biemesder er D, Pizzonia J, Abu-Al a A, et al. NHE3: a Na+/H+ exchanger
8185–8192. 54. Good
collecting DW.perfused
tubule Sodium-dependent
in vitro. Am bicarbonate absorption by cortical thick
J Physiol. 1982;242(5):F521–F531.
iso orm or renal brush border. Am J Physiol. 1993;265(5 Pt 2):F736–F742. ascending limb o rat kidney. Am J Physiol. 1985;248:F821.
228
22 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

55. Wingo
63. Aalkjaer CS,C,Madsen
Frische KM, S, Leipziger
SmolkaJ,A,etetal.al.Sodium H-K-ATPasecoupled bicarbonate trans-
immunoreactivity in 92. Akiba T, Rocco
80. Laghmani VK, Warnock
K, Sakamoto A, YanagisawaDG. Parallel
M, et al.adaptation
A consensus o the rabbitinrenal
sequence the
porters in
cortical andthe kidney,
outer an update.
medullary Acta Physiol
collecting Scand. 2004;181(4):505–512.
duct. Kidney Int. 1990;38(5):985–990. cortical sodium/proton
endothelin-B receptor secondantiporter and sodium/bicarbonate
intracellular loop is requiredcotransporter
for NHE3 activation in met-
http://www.ncbi.nlm.nih.gov/pubmed/15283764
64. Greenlee MM, Lynch IJ, Gumz ML, et al. The renal H,K-ATPases. Curr abolic
by acidosis and
endothelin-1. Amalkalosis.
J PhysiolJ Renal
Clin Invest.
Physiol. 1987;80:308.
2005;288(4):F732–F739.
56. Nephrol
Opin Krap R.Hypertens.
Basolateral2010;19(5):478–482.
membrane H/OH/HCO3 transport in the rat cortical http://www.ncbi.nlm.nih.gov/pubmed/3038953
93. Sartorius OW, Roemmelt JC, Pitts RF. The renal regulation of acid-base
thick ascending
65. Gumz ML,limb. Lynch Evidence
IJ, Greenleeor an MM, electrogenic
et al. The Na/HCO3
renal H cotransporter
-K -ATPases: in 81. Krap
balance in man; R. Mechanisms
the nature of o the
adaptation to chronic respiratory
renal compensations in ammonium acidosis in the
chloride
parallel withregulation,
physiology, a Na/H antiporter.
and structure. J Clin Invest. 1988;82(1):234–241.
Am J Physiol Renal Physiol. 2010;298(1): rabbit proximal
acidosis. tubule.1949;28(3):423–439.
J Clin Invest. J Clin Invest. 1989;83:890.
http://www.ncbi.nlm.nih.gov/pubmed/2839547
F12–F21. http://www.ncbi.nlm.nih.gov/pubmed/2537851
94. Sartorius OW, Roemmelt JC, Pitts RF. Changes in renal function in experi-
57. Gifford
66. Soleimani JD, M.Rome Functional
L, Galla and molecular properties
JH. H-K-ATPase o Na+: duct
in rat collecting HCOsegments.

3
cotrans- 82. Preisig
mental metabolic PA, acidosis
Alpern in RJ.theChronic
normalmetabolic
human subject. acidosisFedcauses an adaptation
Proc. 1948;7(1):108.
porters
Am (NBC).1992;262(4
J Physiol. Minerva Urol Pt Nefrolog.
2):F692–F695. 2003;55(2):131–140. in95.
theYapical
amaji membrane
Y, Moe OW, Na/H
Millerantiporter
RT, et al. andAcidbasolateral
activation membrane
of immediate Na(HCO early )
3 3
58. Wingo
67. DuBoseCS. TDActive
Jr, Good proton DW,secretion
Hamm and LL, potassium
et al. Ammonium absorption transport in the
in the rabbit symporter
genes in renal in the rat proximal
epithelial cells. Jconvoluted
Clin Invest.tubule. J Clin Invest. 1988;82:1445.
1994;94(3):1297–1303.
kidney:
outer new physiological
medullary collecting conceptsduct: functionaland their clinicalfor
evidence implications.
H/K-ATPase. J AmJ ClinSoc 83. Tsuganezawa
96. Talor Z, Yang H, WC, ShuS, Y
Sato eld
amajiJ, etY,al.etChronic
al. Rolehypercapnia
of c-SRC and enhances
ERK in Vmax acid-
Nephrol.
Invest. 1991;1(11):1193–1203.
1989;84:361. o Na-H activation
induced antiporter of o NHE3.
renal brush-border membranes. Am J Physiol. 1987;253(3
Kidney Int. 2002;62(1):41–50.
59. Silver
68. BaileyRB, MA,Frindt
Giebisch G, Abbiati identi
G. Functional T, et al.cation
NHE2-mediated
of H-K-ATPase bicarbonate
in interca- re- Pt97.
2):F394–F400.
Li S, Sato S, Yang X, et al. Pyk2 activation is integral to acid stimulation of
absorption
lated cells ofincortical
the distal tubuletubule.
collecting o NHE3 Am null mice.1993;262:F692.
J Physiol. J Physiol. 2004;561(Pt 3): 84. Amemiya M,exchanger
sodium/hydrogen Yamaji Y3. , Cano
J ClinA, et al.2004;114(12):1782–1789.
Invest. Acid incubation increase NHE-3
765–775.
69. Unwin RJ, Velázquez H, Giebisch G, et al. Active potassium absorption by mRNA abundance
98. Seuwen in OKP
K, Ludwig MG,cells.
Wolf Am RM.J Physiol.
Receptors 1995;269(1
for protons Pt or
1):C126–C133.
lipid messengers
http://www.ncbi.nlm.nih.gov/pubmed/15604231
the renal distal tubule. Am J Physiol 1992;262(3 Pt 2):F488–F493. or85.
both? Yang X, Amemiya
J Recept M, Peng Y
Signal Transduct , et 2006;26:599–610.
Res. al. Acid incubaion causes exocytic inser-
60. Wall
70. ChambreySM, Mehta R, Warnock
P, DuBose DG,TDPodevin
Jr. Dietary RA,K etrestriction
al. Immunolocalization
upregulates total o tion
99. oSun NHE3 X, Yin angOKP cells. Am
LV, Tiegs BC, JetPhysiol. 2000;279(2):C410–C419.
al. Deletion of the pH sensor GPR4 decreases
the Na
and +
Sch-/H28080-sensitive
+
exchanger iso orm NHE2 inabsorption
bicarbonate rat kidney.inAm ratJ tIMCD.
Physiol. 1998;275(3
Am J Physiol. Pt 86. acid
renal Guntupalli
excretion. J, DuBose
J Am Soc TDNephrol.
Jr. E ects o endothelin on rat renal proximal tu-
2010;21:1745–1755.
2):F379–F386.
1998;275:F543–F549. bule Na-P
100. Paunescu
i
co-transport
TG, Da Silvaand Na N, /H
+ +
Russo exchange.
LM, et al. AmAssociation
J Physiol Renal Fluid Electrolyte
of soluble adenylyl
61. Li
71. Wang T, Hropot
J, Codina M, Aronson
J, Petroske E, Werle PS,MJ, et al. Role
et al. The o carboxy
NHE isoterminus
orms in of medi-
the Physiol. with
cyclase 1994;266(35):F658–F666.
the V-ATPase in renal epithelial cells. Am J Physiol Renal Physiol.
ating bicarbonate
colonic H ,K -ATPase reabsorption
-subunitalong the nephron.
is required for stable Am -subunit
J Physiol. assembly
2001;281(6): and 87. Licht C, Laghmani K, Yanagisawa M, et al. An autocrine role or en-
2008;294(1):F130–F138.
F1117–F1122.
function. Kidney Int. 2004;65:1–10. dothelin-1
101. in the regulation
Pastor-Soler NM, Hallows o proximal
KR, Smolak tubule NHE3.
C, et Kidney pH-
al. Alkaline Int. 2004;65(4):
and cAMP-
62. Haggerty
72. Koeppen JG, BM,Agarwal
HelmanN, SI.Reilly
AcidiRF cation
, et al.oPharmacologically
luminal f uid by the rabbit Na/H
different corti- 1320–1326.
induced V-ATPase membrane accumulation is mediated by protein kinase A in
cal collecting
antiporters ontubule
the apical per andusedbasolateral
in vitro. Am J Physiol.
surfaces 1982;242(5):F521–F531.
of cultured porcine kidney cells http://www.ncbi.nlm.nih.gov/pubmed/15086471
epididymal clear cells. Am J Physiol Cell Physiol. 2008;294(2):C488–C494.
63. Wingo
(LLC-PK 1). ProcCS,Natl
Madsen AcadKM, Smolka
Sci USA. A, et al. H-K-ATPase immunoreactivity in
1988;85(18):6797–6801. 88. McKinney
102. Peng Y, Moe TD,OW,BurgChu MB.T,Bicarbonate
et al. ETB receptor
transportactivation
by rabbit leadscorticalto collecting
activation
cortical and outer
73. Preisig medullary
PA. Luminal owcollecting
rate regulates duct. Kidneytubule
proximal Int. 1990;38(5):985–990.
H-HCO3 transporters. and phosphorylation
tubules: effect of acid oand NHE3.
alkali Am loads J Physiol.
in vivo1999;276(4
on transportPtin1):C938–C945.
vitro. J Clin Invest.
http://www.ncbi.nlm.nih.gov/pubmed/7081439
Am J Physiol. 1992;262:F47. 89. Peng Y, Amemiya M, Yang T, et al. ETB receptor activation causes exo-
1977;60:766.
64. Alpern
74. Greenlee RJ,MM, Lynch IJ,
Chambers M.GumzCell pHML,inet the al. The renal H,K-ATPases.
rat proximal convoluted Curr Opin
tubule. cytic insertion
103. Schwartz oGJ,NHE3 Al-Awqatiin OKP Q. cells.
Role of Am J Physiol
hensin Renal Physiol.
in mediating 2001;280(1):
the adaptation of
Nephrol Hypertens.
Regulation by luminal 2010;19(5):478–482.
and peritubular pH and sodium concentration. J Clin F34–F42.
the cortical collecting duct to metabolic acidosis. Curr Opin Nephrol Hypertens.
65. Gumz
Invest. ML, Lynch IJ, Greenlee MM, et al. The renal H+-K+-ATPases:
1986;78(2):502–510. 90. Laghmani K, Preisig A, Moe OW, et al. Endothelin-1/endothelin-B recep-
2005;14:383.
physiology,
75. Aronson regulation,
PS, Nee and J, Suhm structure.
MA. Modi Am J Physiol
er role Renal Physiol.
of internal H 2010;298(1):
in activating tor-mediated
104. Bagnis C, increases
Marshansky in NHE3 activity
V, Breton S, in
et al.chronic metabolic
Remodeling theacidosis.
cellular pro J ClinleIn-
of
F12–F21.
the Na-H exchanger in renal microvillus membrane vesicles. Nature (London). vest. 2001;107(12):1563–1569.
collecting ducts by chronic carbonic anhydrase inhibition. Am J Physiol Renal
66. Gi ord JD, Rome L, Galla JH. H-K-ATPase in rat collecting duct segments.
1982;299:161. http://www.ncbi.nlm.nih.gov/pubmed/11413164
Physiol. 2001;280(3):F437–F448.
Am76.J Physiol.
Gluck S, 1992;262(4
Cannon C,PtAl-Awqati 2):F692–F695. Q. Exocytosis regulates urinary acidi ca- 91. Bastani
105. Laghmani K, Preisig
B, Purcell A, AlpernP, RJ.
H, Hemken The
et al. role o endothelin
Expression in proximal
and distribution of renaltu-
67.in Wingo
tion CS. Active
turtle bladder proton
by rapid secretion
insertion of H and potassium
pumps into the absorption in the rab-
luminal membrane. bule proton
vacuolar secretion and the adaptation
proton-translocating adenosine to a chronic metabolic
triphosphatase in responseacidosis. J Neph-
to chronic
bit outer
Proc Natl medullary
Acad Sci USA. collecting duct: unctional evidence or H/K-ATPase. J Clin
1982;79:4327. rol. 2002;15(Suppl
acid and alkali loads5):S75–S87.
in the rat. J Clin Invest. 1991;88(1):126–136.
Invest. 1989;84:361.
77. Schwartz GJ, Al-Awqati Q. Carbon dioxide causes exocytosis of vesicles 92. Satlin
106. Laghmani LM, K, Sakamoto
Schwartz GJ.A,Cellular
Yanagisawa M, et al.ofAHCO
remodeling consensus

3
-secretingsequence
cells in in
http://www.ncbi.nlm.nih.gov/pubmed/2544629
containing H pumps in isolated perfused proximal and collecting tubules. J Clin the endothelin-B
rabbit renal collecting receptor ductsecond intracellular
in response to an loopacidicis required
environment. or NHE3J Cellactiva-
Biol.
68. Silver
Invest. RB, Frindt G. Functional identi cation o H-K-ATPase in interca-
1985;75:1638. tion by endothelin-1. Am J Physiol Renal Physiol. 2005;288(4):F732–F739.
1989;109:1279.
lated
78. cells
Cogan o cortical
MG. Chronic collecting tubule. Am
hypercapnia J Physiol. proximal
stimulates 1993;262:F692.bicarbonate reab- 93. V
107. Sartorius
erlander OW, Roemmelt
JW, Madsen KM,JC, Pitts RF
Cannon JK,. The
et al.renal regulation
Activation o acid-base
of acid-secreting
69. Unwin
sorption in theRJ, rat.Velázquez
J Clin Invest. H, Giebisch
1984;74:1942.G, et al. Active potassium absorption by balance in man;
intercalated cells the naturecollecting
in rabbit o the renal ductcompensations
with ammonium in chloride
ammonium chloride
loading. Am
the
79.renal
KunaudistalRT,tubule.
Jr., Hart Am JI,JWalker
Physiol 1992;262(3
KA. Effect ofPt 2):F488–F493.
metabolic acidosis on proximal Jacidosis.
Physiol.J1994;266(4
Clin Invest. 1949;28(3):423–439.
Pt 2):F633–F645.
70. Wall
tubular totalSM,CO2Mehta P, DuBose
absorption. Am JTD Jr. Dietary
Physiol. K restriction upregulates total
+
1985;249:F62. 94. Schwartz
108. Sartorius OW, RoemmeltS,JC,
GJ, Tsuruoka Pitts RF. Changes
Vijayakumar S, et al.inAcid
renalincubation
unction inreverses
experi-
and
80. Sch-
Akiba 28080-sensitive
T, Rocco VK, Warnock bicarbonate DG.absorption in rat tIMCD.
Parallel adaptation of theAm J Physiol.
rabbit renal mental
the metabolic
polarity acidosis incell
of intercalated thetransporters,
normal human subject.
an effect Fed Proc.by1948;7(1):108.
mediated hensin. J Clin
1998;275:F543–F549.
cortical sodium/proton antiporter and sodium/bicarbonate cotransporter in http://www.ncbi.nlm.nih.gov/pubmed/18934503
Invest. 2002;109(1):89–99.
71. Li J,acidosis
metabolic Codinaand J, Petroske
alkalosis.E,J Werle MJ, et1987;80:308.
Clin Invest. al. The carboxy terminus o the 95. Soleimani
109. Yamaji Y, Moe OW, Miller
M, Bergman JA,RT, et al. Acid
Hosford MA, activation o immediate
et al. Potassium early
depletion
colonic
81. Krapf H ,KR.-ATPase
+ +
Mechanisms a -subunit is required
of adaptation or stable
to chronic b-subunitacidosis
respiratory assembly in and
the genes in renal
increases luminal epithelial
Na/H cells.
exchangeJ Clinand Invest. 1994;94(3):1297–1303.
basolateral Na :HCO3 :HCO3 cotrans-
unction.
rabbit Kidneytubule.
proximal Int. 2004;65:1–10.
J Clin Invest. 1989;83:890. http://www.ncbi.nlm.nih.gov/pubmed/8083371
port in rat renal cortex. J Clin Invest. 1990;86(4):1076–1083.
72. Preisig
82. Haggerty PA,JG, Agarwal
Alpern RJ. N, Reilly RF
Chronic , et al. Pharmacologically
metabolic acidosis causes an di erent Na/H
adaptation 96. Tannen
110. Tsuganezawa H, Sato S, Yamaji
RL. Relationship of renal Y, et al. Role production
ammonia o c-SRC and and ERK in acid-
potassium
antiporters
in the apicalonmembrane
the apicalNa/H and basolateral
antiportersur andaces o cultured
basolateral porcine Na(HCO
membrane kidney cells 3 )3 induced activation
homeostasis. Kidneyo Int.NHE3. Kidney Int. 2002;62(1):41–50.
1977;11(6):453–465.
(LLC-PK1).inProc
symporter theNatl Acad Sci USA.
rat proximal 1988;85(18):6797–6801.
convoluted tubule. J Clin Invest. 1988;82:1445. Adam WR, Koretsky AP, Weiner MW. 31P NMR in vivo measurement of
http://www.ncbi.nlm.nih.gov/pubmed/12081562
111.
73. Talor
83. Preisig Z, PA.
YangLuminal
WC, Shuf f oweldrate J, etregulates
al. Chronic proximal tubule enhances
hypercapnia H-HCO3 Vmax trans- 97. intracellular
renal Li S, Sato S, pH: Yangeffects
X, et al.of Pyk2
acidosisactivation is integral in
and K depletion to rats.
acid stimulation
Am J Physiol. o
porters.
of Na-H Am J Physiol.
antiporter of 1992;262:F47.
renal brush-border membranes. Am J Physiol. 1987;253(3 sodium/hydrogen exchanger 3. J Clin Invest. 2004;114(12):1782–1789.
1986;251:F904.
Pt74. Alpern RJ, Chambers M. Cell pH in the rat proximal convoluted tubule.
2):F394–F400. 98. Amemiya
112. Seuwen K,M,Ludwig Tabei MG, Wol RM.
K, Kusano E, etReceptors or protons
al. Incubation of OKP or lipid
cells messen-
in low-
Regulation
84. Amemiya by luminal
M, Yamaji and peritubular
Y, Cano A, pH et al.andAcidsodium concentration.
incubation increaseJ NHE-3
Clin In- Kgersmedia
or both? J ReceptNHE3
increases Signalactivity
Transduct Res.
after 2006;26:599–610.
early decrease in intracellular pH. Am J
vest. 1986;78(2):502–510.
mRNA abundance in OKP cells. Am J Physiol. 1995;269(1 Pt 1):C126–C133. 99. Sun
Physiol. X, Yang LVPt
1999;276(3 , Tiegs BC, et al. Deletion o the pH sensor GPR4 decreases
1):C711–C716.
http://www.ncbi.nlm.nih.gov/pubmed/3016029
85. Yang X, Amemiya M, Peng Y, et al. Acid incubaion causes exocytic insertion renal acid
113. Codina excretion. J AmTA,
J, Pressley Soc DuBose
Nephrol. TD 2010;21:1745–1755.
Jr. Effect of chronic hypokalemia on
of75.
NHE3 Aronson
in OKPPS, NeeAm
cells. J, JSuhm
Physiol. MA.2000;279(2):C410–C419.
Modi er role o internal H in activating H100.,K Paunescu TG, Da Silva
-ATPase expression in N, ratRusso
colon. LM, Amet Jal.Physiol
Association
Renalo Fluid soluble adenylyl
Electrolyte
the
86.Na-H exchanger
Guntupalli in renal
J, DuBose TDmicrovillus
Jr. Effects membrane
of endothelin vesicles.
on ratNature (London).
renal proximal cyclase with
Physiol. the V-ATPase in renal epithelial cells. Am J Physiol Renal Physiol.
1997;272(41):F22–F30.
1982;299:161.
tubule Na-Pi co-transport and Na /H exchange. Am J Physiol Renal Fluid 2008;294(1):F130–F138.
114. Codina J, Delmas-Mata JT, DuBose TD Jr. Expression of HK 2 protein is in-
http://www.ncbi.nlm.nih.gov/pubmed/7110335
Electrolyte Physiol. 1994;266(35):F658–F666. 101. Pastor-Soler
creased selectively in NM, renalHallows
medulla KR, bySmolak
chronicC, et al. Alkaline
hypokalemia. AmpH- and cAMP-
J Physiol Renal
76. Licht
87. GluckC,S,Laghmani
Cannon K, C,YAl-Awqati
anagisawa M, Q. et Exocytosis regulates
al. An autocrine roleurinary acidi ca-
for endothelin-1 induced 1998;275(44):F433–F440.
Physiol. V-ATPase membrane accumulation is mediated by protein kinase A in
tion
in theinregulation
turtle bladder by rapidtubule
of proximal insertion
NHE3. o H pumps
Kidney Int.into the luminal membrane.
2004;65(4):1320–1326. epididymal
115. Guntupalli clear cells. Am J Physiol
J, Onuigbo M, Wall CellSM,
Physiol. 2008;294(2):C488–C494.
et al. Adaptation to low K media
Proc
88. Natl
PengAcadY, MoeSci USA.
OW, 1982;79:4327.
Chu T, et al. ETB receptor activation leads to activation 102. McKinney
increases H,K-ATPase TD, Burg but MB.
not Bicarbonate
H,K-ATPase-mediated transport bypH rabbit corticalincollecting
i recovery OMCD1
http://www.ncbi.nlm.nih.gov/pubmed/6289300
and phosphorylation of NHE3. Am J Physiol. 1999;276(4 Pt 1):C938–C945. tubules:
cells. AmeJ Physiol.
ect o acid and alkaliPt
1997;273(2 loads in vivo on transport in vitro. J Clin Invest.
1):C558–C571.
77. Peng
89. Schwartz GJ, Al-Awqati
Y, Amemiya M, Yang Q. T,Carbon
et al. dioxide
ETB receptorcausesactivation
exocytosiscauses
o vesicles
exo- 1977;60:766.
116. DuBose TD, Jr., Good DW. Effects of chronic chloride depletion metabolic
containing
cytic insertionH pumps
of NHE3 in isolated
in OKP per cells.used
Am proximal
J Physiol and Renal collecting
Physiol. tubules.
2001;280(1): J Clin http://www.ncbi.nlm.nih.gov/pubmed/19497
alkalosis on proximal tubule transport and renal production of ammonium. Am
Invest. 1985;75:1638.
F34–F42. J103.
PhysiolSchwartz
Renal Fluid GJ, Al-Awqati
ElectrolyteQ.Physiol.
Role o 1995;269(38):F852–F861.
hensin in mediating the adaptation o
78. Laghmani
90. Cogan MG.K,Chronic Preisig hypercapnia
A, Moe OW, stimulates
et al. proximal bicarbonate reab-
Endothelin-1/endothelin-B the cortical
117. DuBose collecting
TD, Jr., duct to metabolic
Ca isch CR. Effect acidosis. Curr Opin
of selective Nephrol de
aldosterone Hypertens.
ciency
sorption in the rat. increases
receptor-mediated J Clin Invest. in 1984;74:1942.
NHE3 activity in chronic metabolic acidosis. 2005;14:383.
on acidi cation in nephron segments of the rat inner medulla. J Clin Invest.
Jhttp://www.ncbi.nlm.nih.gov/pubmed/6439738
Clin Invest. 2001;107(12):1563–1569. 104. Bagnis C, Marshansky V, Breton S, et al. Remodeling the cellular pro le
1988;82(5):1624–1632.
79. Laghmani
91. Kunau RT,K, Jr.,Preisig
Hart JI,A, Walker
Alpern KA.RJ.E The ect orolemetabolic acidosis in
of endothelin on proximal
proximal o collecting
118. DuBoseducts TD, by Jr.,chronic
Good carbonic
DW. Chronic anhydrase inhibition. impairs
hyperkalemia Am J Physiol ammoni-Renal
tubular proton
tubule total COsecretion
2
absorption. and Am the Jadaptation
Physiol. 1985;249:F62.
to a chronic metabolic acidosis. Physiol.
um 2001;280(3):F437–F448.
transport and accumulation in the inner medulla of the rat. J Clin Invest.
J Nephrol. 2002;15(Suppl 5):S75–S87. 1992;90(4):1443–1449.
CHAPTER 7 RENAL ACID–BASE TRANSPORT 2229
29

105. Al-Awqati
119. Bastani B, Q. Purcell H,ofHemken
Effect P, et al.
aldosterone onExpression
the coupling andbetween
distribution o renal
H transport 119. Al-Awqati
126. Alpern RJ, Q. E ectLL.
Hamm o Urinary
aldosterone
acidioncation.
the coupling between
In: DuBose TD, HHamm
transport
LL,
vacuolar
and proton-translocating
glucose adenosine
oxidation. J Clin Invest. triphosphatase in response to chronic
1977;60:1240. and
eds. glucose oxidation.
Acid-base J Clin Invest.
and Electrolyte 1977;60:1240.
Disorders: A Companion to Brenner and Rector’s
acid and
120. alkali
Stone DK,loads in the
Seldin DW, rat.Kokko
J Clin Invest. 1991;88(1):126–136.
JP, et al. Mineralocorticoid modulation of http://www.ncbi.nlm.nih.gov/pubmed/21197
The Kidney. Philadelphia: WB Saunders; 2002:23–40.
106. Satlin
rabbit medullaryLM, Schwartz
collectingGJ.duct Cellular
acidi remodeling o HCO3–-secreting cells
cation. A sodium-independent in
effect. 120. Stone DK,
127. DuBose TDSeldin DW,J,Kokko
Jr, Codina BurgesJPA,
, etet al.
al. Mineralocorticoid
Regulation of H -K modulation
-ATPase ex-o
Jrabbit renal collecting
Clin Invest. duct in response to an acidic environment. J Cell Biol.
1983;72(1):77–83. rabbit
pressionmedullary
in kidney.collecting duct 1995;269(4
Am J Physiol. acidi cation.PtA2):F500–F507.
sodium-independent e ect. J
1989;109:1279.
121. Eiam-ong S, Kurtzman NA, Sabatini S. Regulation of collecting tubule ade- Clin
128.Invest. 1983;72(1):77–83.
Wesson DE, Dolson GM. Endothelin-1 increases rat distal tubule acidi ca-
http://www.ncbi.nlm.nih.gov/pubmed/2549077
nosine triphosphates by aldosterone and potassium. J Clin Invest. 1993;91:2385. http://www.ncbi.nlm.nih.gov/pubmed/6874954
tion in vivo. Am J Physiol. 1997;273:F586–F594.
107. Hays
122. Verlander JW, Madsen KM,modulation
SR. Mineralcorticoid Cannon JK, of et al. Activation
apical o acid-secreting
and basolateral membrane 121. Eiam-ongDE.
129. Wesson S, Na/H
Kurtzman NA, Sabatini
exchange S. Regulation
and H-K-ATPase o collecting
increase tu-
distal tubule
intercalated cells in rabbit collecting duct with ammonium
H/OH/HCO3 transport processes in the rabbit inner stripe of outer medullary chloride loading. Am bule
acidi adenosine triphosphates
cation in chronic alkalosis.by aldosterone
Kidney and potassium. J Clin Invest.
Int. 1998;53:945–951.
J Physiol. 1994;266(4
collecting duct. J ClinPt 2):F633–F645.
Invest. 1992;90:180. 1993;91:2385.
130. Wesson DE. Combined K and Cl repletion corrects augmented H secretion
108. Schwartz GJ, Tsuruoka
123. Al-Awqati Q, Muller A, Steinmetz S, Vijayakumar S, et al.
PR. Transport Acid
of H incubation
against reverses
electrochemical 122. Hays
by distal SR. Mineralcorticoid
tubules modulation
in chronic alkalosis. o apical
Am J Physiol. and basolateral membrane
1994;266:F592–F603.
the polarity
gradients in oturtle
intercalated cell transporters,
urinary bladder. Am J Physiol.an e 1977;233:F502.
ect mediated by hensin. J Clin H/OH/HCO
131. Cogan3 MG, transport processes
Liu FY in the
. Metabolic rabbitininner
alkalosis stripe
the rat: o outer
evidence thatmedullary
reduced
Invest.DuBose
124. 2002;109(1):89–99.
TD. Metabolic alkalosis. In: Greenberg A, eds. Primer on Kidney collecting duct. J Clin Invest. 1992;90:180.
glomerular ltration rather than enhanced tubular bicarbonate reabsorption is
http://www.ncbi.nlm.nih.gov/pubmed/11781354
Diseases. Philadelphia: Elsevier Saunders; 2005:90–96. http://www.ncbi.nlm.nih.gov/pubmed/1321841
responsible for maintaining the alkalotic state. J Clin Invest. 1983;71:1141.
109. Galla
125. Soleimani M, Bergman
JH. Metabolic JA, HosIn:
alkalosis. ordDuBose
MA, et al. TD,Potassium
Hamm LL, depletion increas-
eds. Acid-Base 123. Al-Awqati
132. Alpern RJ, Q, Muller
Cogan MG,A, Steinmetz
Rector FCPR. Transport
Jr. Effects o H against electrochemi-
of extracellular uid volume
es luminal
and Na/HDisorders:
Electrolyte exchange and basolateral to
A Companion NaBrenner
+
:HCO3=:and HCO –
cotransport
Rector’s
3
in rat
The Kidney. cal gradients in turtle urinary bladder. Am J Physiol. 1977;233:F502.
and plasma bicarbonate concentration on proximal acidi cation in the rat. J Clin
renal cortex. JWB
Philadelphia: ClinSaunders;
Invest. 1990;86(4):1076–1083.
2002:109–128. 124.
Invest.DuBose TD. Metabolic alkalosis. In: Greenberg A, eds. Primer on Kidney
1983;71:736.
110. Tannen RL. Relationship o renal ammonia production and potassium ho- Diseases. Philadelphia: Elsevier Saunders; 2005:90–96.
meostasis. Kidney Int. 1977;11(6):453–465. 125. Galla JH. Metabolic alkalosis. In: DuBose TD, Hamm LL, eds. Acid-Base
http://www.ncbi.nlm.nih.gov/pubmed/17763 and Electrolyte Disorders: A Companion to Brenner and Rector’s The Kidney. Philadel-
111. Adam WR, Koretsky AP, Weiner MW 31P NMR in vivo measurement o phia: WB Saunders; 2002:109–128.
renal intracellular pH: e ects o acidosis and K depletion in rats. Am J Physiol. 126. Alpern RJ, Hamm LL. Urinary acidi cation. In: DuBose TD, Hamm LL,
1986;251:F904. eds. Acid-base and Electrolyte Disorders: A Companion to Brenner and Rector’s
112. Amemiya M, Tabei K, Kusano E, et al. Incubation o OKP cells in low- The Kidney. Philadelphia: WB Saunders; 2002:23–40.
K+ media increases NHE3 activity a ter early decrease in intracellular pH. Am J 127. DuBose TD Jr, Codina J, Burges A, et al. Regulation o H+-K+-ATPase ex-
Physiol. 1999;276(3 Pt 1):C711–C716. pression in kidney. Am J Physiol. 1995;269(4 Pt 2):F500–F507.
113. Codina J, Pressley TA, DuBose TD Jr. E ect o chronic hypokalemia on 128. Wesson DE, Dolson GM. Endothelin-1 increases rat distal tubule acidi ca-
H+,K+-ATPase expression in rat colon. Am J Physiol Renal Fluid Electrolyte Physiol. tion in vivo. Am J Physiol. 1997;273:F586–F594.
1997;272(41):F22–F30. 129. Wesson DE. Na/H exchange and H-K-ATPase increase distal tubule acidi-
114. Codina J, Delmas-Mata JT, DuBose TD Jr. Expression o HKa 2 protein is cation in chronic alkalosis. Kidney Int. 1998;53:945–951.
in creased selectively in renal medulla by chronic hypokalemia. Am J Physiol Renal http://www.ncbi.nlm.nih.gov/pubmed/9551402
Physiol. 1998;275(44):F433–F440. 130. Wesson DE. Combined K and Cl repletion corrects augmented H secretion
115. Guntupalli J, Onuigbo M, Wall SM, et al. Adaptation to low K+ media in- by distal tubules in chronic alkalosis. Am J Physiol. 1994;266:F592–F603.
creases H,K-ATPase but not H,K-ATPase-mediated pHi recovery in OMCD1 cells. 131. Cogan MG, Liu FY. Metabolic alkalosis in the rat: evidence that reduced
Am J Physiol. 1997;273(2 Pt 1):C558–C571. glomerular ltration rather than enhanced tubular bicarbonate reabsorption is
116. DuBose TD, Jr., Good DW E ects o chronic chloride depletion metabolic responsible or maintaining the alkalotic state. J Clin Invest. 1983;71:1141.
alkalosis on proximal tubule transport and renal production o ammonium. Am J 132. Alpern RJ, Cogan MG, Rector FC Jr. E ects o extracellular f uid volume
Physiol Renal Fluid Electrolyte Physiol. 1995;269(38):F852–F861. and plasma bicarbonate concentration on proximal acidi cation in the rat. J Clin
117. DuBose TD, Jr., Caf isch CR. E ect o selective aldosterone de ciency Invest. 1983;71:736.
on acidi cation in nephron segments o the rat inner medulla. J Clin Invest. http://www.ncbi.nlm.nih.gov/pubmed/6826733
1988;82(5):1624–1632.
http://www.ncbi.nlm.nih.gov/pubmed/3183058
118. DuBose TD, Jr., Good DW Chronic hyperkalemia impairs ammoni-
um transport and accumulation in the inner medulla o the rat. J Clin Invest.
1992;90(4):1443–1449.
C H A P T E R

8 Hormones and the Kidney


Sola Aoun Bahous • Maya Khairallah •
Joumana T. Chaiban • Kamal F. Badr

HORMONAL MODULATION OF Modulation of Tubule Water and


Electrolyte Transport
NEPHRON FUNCTION: AN OVERVIEW
Renal tubule cells express receptors for many circulat-
Modulation of Glomerular Filtration Rate ing and locally synthesized hormones. The effects of a
The major physiologic determinants of single nephron glo- particular hormone on water or electrolyte transport
merular ltration rate (GFR) are glomerular plasma ow are partly determined by the differential distribution of
(QA), glomerular transcapillary hydraulic pressure (PGC), its receptor on functionally specialized segments of the
and the ultra ltration coef cient (Kf).1 These variables are renal tubule. For example, arginine vasopressin (AVP)
determined, in part, by the contractile state of the affer- binds almost exclusively to principal cells in the col-
ent arteriole, efferent arteriole, and mesangial cells. Kf also lecting tubule (CT), where it influences the physiolog-
varies with alterations in the hydraulic permeability of the ic functions of this segment, primarily water and urea
capillary ltration barrier, which consists of endothelial absorption. 6 Parathyroid hormone (PTH), on the other
cells, visceral epithelial cells, and the glomerular basement hand, exerts its biologic actions on renal tubule segments
membrane. By binding to speci c receptors on cellular proximal to the collecting duct, where it modulates cal-
and structural components of the glomerulus, circulating cium, phosphate, magnesium, sodium, and bicarbonate
and locally produced hormones in uence one or more of transport.7 Table 8.1 summarizes the net effects of re-
the physiologic determinants of GFR. Figure 8.1 summa- nally relevant hormones on water and solute handling
rizes the effects of different hormones on pregomerular, in different tubule segments. Each hormone is discussed
glomerular, and postglomerular contractility. Because the in detail in the sections that follow. As in the glomeru-
same substance can act at different sites in the glomeru- lus, hormones may modulate their own actions by alter-
lar unit, the net effect on GFR will depend on whether its ing the production of counterregulatory hormones. For
actions are antagonistic or complementary. For example, example, AVP induces local synthesis of prostaglandin E
atrial natriuretic peptide (ANP) decreases afferent arteriolar (PGE), which opposes the effect of AVP on water perme-
resistance, whereas increasing efferent arteriolar resistance ability in the CT. 8
results in an augmented PGC and a rise in GFR.2 Under
certain conditions, ANP also increases Kf, which, in turn,
contributes to the enhancement of GFR.3 On the other
hand, angiotensin (Ang) II-mediated constriction of both ARGININE VASOPRESSIN
afferent and efferent arterioles results in opposite effects on In 1895, Oliver and Schafer were the rst to describe a sub-
glomerular plasma ow and PGC and, therefore, no change stance with potent vasopressor effects originating from the
in single nephron GFR.4 Regulation of glomerular hemo- posterior pituitary, hence the name, vasopressin.9 It was not
dynamics is further complicated by multiple interactions until later in the 20th century that the effects of this hormone
between hormones in the kidney. For example, infusion on modulation of water excretion by the kidney were dis-
of Ang II along with a cyclooxygenase inhibitor causes a covered. In fact, vasopressin, also called AVP or antidiuretic
signi cant decrease in single nephron GFR, suggesting that hormone (ADH), is a key hormone for osmoregulation and
endogenous prostaglandin production antagonizes the glo- maintenance of body homeostasis.10 AVP was also found
merular effects of Ang II.5 The net effects of renally relevant to exert effects on body temperature, glucose metabolism,
hormones on GFR are discussed in more detail later in this memory, social behavior, and the hypothalamic-pituitary-
chapter. adrenal axis.

23 0
CHAPTER 8 HORMONES AND THE KIDNEY 2231
31

FIGURE 8.1 Hormones regulating


glomerular, afferent, and efferent arte-
riolar contractility. Glucocorticoid action
may be pharmacologic and indirect. LTC4,
leukotriene C4; LTD4, leukotriene D4; LXA2,
leukotriene A2; TXA2, thromboxane A2; PGE2,
prostaglandin E2; PGI2, prostacyclin.

Structure and Synthesis of Physiology of Arginine Vasopressin


Arginine Vasopressin in the Kidneys
AVP is a 9-amino acid neuropeptide synthesized by mag- The most sensitive stimulus for AVP secretion into the blood-
nocellular neurons in the paraventricular and supraoptic stream is increased plasma osmolality. Osmosensitive neu-
nuclei of the hypothalamus.11 The AVP gene, located on rons that respond to changes in plasma osmotic pressure by
chromosome 20, consists of three exons that encode the varying their intracellular water content have been identi ed
signal peptide, AVP, neurophysin II (NPII), and a glyco- in the anterior hypothalamus, speci cally in the organum
peptide.10 After cleavage of the signal peptide within the vasculosum of the lamina terminalis (OVLT) and the sub-
ER, the resulting prohormone is folded and packaged into fornical organ (SFO).15 When stimulated by osmosensitive
secretory granules in neuronal bodies. These granules are neurons, the magnocellular neurons release the stored AVP
then transported along the axons to nerve terminals in the into the posterior pituitary (an area that lacks a blood–brain
posterior pituitary (neurohypophysis). 12 During axonal barrier) and AVP enters the general circulation. As little as
transport, further processing of the prohormone within 1% change in plasma osmolality leads to a change in AVP
secretory granules yields AVP, neurophysin II (NpII), and concentration that is suf cient to modify renal water excre-
a glycopeptide. Interestingly, mutations of NpII impair tion.16 AVP secretion is almost completely suppressed when
AVP secretion, suggesting that NpII assists in the pro- plasma osmolality decreases below an average of 280 mOsm
cessing or secretion of AVP.13 In addition to the posterior per kg of water in humans.17
pituitary, AVP synthesis has been detected in the pancreas, Secretion of AVP is also in uenced by alterations in
adrenal gland, ovary, testis, and regions of the brain.14 Its intravascular volume and blood pressure, sensed by barore-
physiologic function in these sites, however, remains to ceptors located in the heart, aortic arch, and carotid sinus.18
be clari ed. These signals are transferred through the vagal nerves to the
232
23 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

TA B L E
Hormonal Modulation of Tubular Transport
8.1
Hormones with Major Effects

Reabsorption Stimulatory Inhibitory

Proximal tubule Na Ang II, catecholamines, insulin Dopamine, PTH


Pi IGF-1 PTH
HCO3 Ang II PTH
Ca PTH

TALH Na AVP,a catecholamines PGE


Ca PTH, calcitonin, glucagon
Mg AVP, PTH, calcitonin, glucagon

DCT Pi PTH
Ca PTH

CCD H2O AVP PGE, bradykinin, ANP, -adrenergic agents


Na Aldosterone ANP, PGE, EGF

IMCD Urea AVP

SECRETION CCD K AVP, aldosterone 1 agonists


H Aldosterone

a
Unlikely in humans.
TALH, thick ascending limb of the loop of Henle; DCT, distal convoluted tubule; CCD, cortical collecting duct; IMCD, inner medullary collecting duct;
Ang II, angiotensin II; IGF, insulinlike growth factor; AVP, arginine vasopressin; PTH, parathyroid hormone; PGE, prostaglandin E; EGF, epidermal
growth factor.

nucleus solitarius in the brainstem, from which postsynap- (or C-terminal proarginine vasopressin, CT-proAVP) is the
tic pathways project to the magnocellular neurons. Whereas C-terminal part of AVP, which is secreted stoichiometrically
a 5% to 8% decrease in blood volume or systemic arterial with AVP in a manner similar to C-peptide and endogenous
pressure has little effect, further hemodynamic compromise insulin.23 CT-proAVP provides a reliable means for estimating
leads to a steep increase in circulating AVP levels. Signi - prevailing AVP levels in the circulation, thus facilitating the
cant reductions (10%–30%) in circulatory arterial volume study of AVP in human diseases. A recent cohort study in
or blood pressure can override osmoregulation and result in renal transplant patients suggests that high CT-proAVP
markedly increased AVP levels in the face of decreased plas- strongly correlates with a negative renal prognosis. In fact,
ma osmolality.19 Other less potent stimuli for AVP secretion in this study, the plasma concentrations of CT-proAVP pre-
include fever, emesis,20 and oropharyngeal osmoreceptors.21 dicted renal function loss over a 3.2-year follow-up.24
AVP circulates in the plasma nearly in an unbound AVP exerts its biologic actions through three speci c
form. Levels of circulating AVP depend on both the rate of cell-surface AVP receptors identi ed as V1R (also called
AVP release from the posterior pituitary and the rate of AVP V1aR), V2R, and V3R (also V1bR).25 These receptors belong
degradation. As discussed, the major factor controlling AVP to the G protein coupled receptor superfamily (Table 8.2).
release is plasma osmolality. The liver and the kidney both In the human kidney, mRNA for V1Rs predominates in cor-
contribute to the breakdown of AVP and the decline in AVP tical collecting ducts (CCD), gradually decreasing as the
levels when secretion ceases. In fact, the half-life of AVP collecting duct enters the medulla.26 In addition, whereas
in the circulation is 18 minutes due to rapid clearance by V1R mRNA is diffusely expressed in the CCD, it is restricted
hepatic and renal vasopressinases.22 Under physiologic con- to the intercalated cells in OMCD. V1Rs are responsible for
ditions, plasma vasopressin concentrations vary with serum mediating vascular smooth muscle cell vasoconstriction by
osmolarity between 0 to 5 pg per mL.22 activating G protein-dependent phospholipase C (PLC) and
It is noteworthy that AVP levels are dif cult to measure the downstream effectors, diacylglycerol (DAG), and inosi-
in plasma because of the instability of this peptide and tol 1,4,5-triphosphate (IP3). In turn, DAG stimulates protein
the low sensitivity of available AVP antibodies. Copeptin kinase C (PKC), whereas IP3 increases cytosolic Ca2 , thus
CHAPTER 8 HORMONES AND THE KIDNEY 2233
33

TA B L E

8.2 Vasopressin Receptor Types, Genetics, Location, and Main Physiologic Effects

No. of
Chromosome amino Main second
Receptor location acids Site of action messenger Main effects
V1 (V1a) 12 (q14–q15) 418 Vascular smooth PLC→IP3 Vasoconstriction, platelet
muscle, plate- DAG/calcium aggregation, glycoge-
lets, liver, testes, and PKC nolysis, stimulation of
brainstem, aldosterone and cortisol
adrenal glands synthesis
V2 X (q28) 371 Collecting duct cells Adenylate cyclase/ Water retention, stimulation
of kidney, inner cAMP of atrial natriuretic
medulla, heart, peptide, stimulation
pancreas of insulin synthesis,
coronary and pulmonary
artery vasodilation
V3 (V1b) 1 (q32) 553 Hypothalamus, PLC→IP3 Modulation of ACTH
anterior DAG/calcium synthesis; stimulation
pituitary gland and PKC of ACTH, GH, and
procaltin release
Oxytocin 3 (p25) 389 Uterus, breast, PLC→IP3 Myometrial contraction,
vascular DAG/calcium ductal myoepithelial
endothelium and PKC contraction, vasodilation

P2 Puriner- 11 (q13.5–14.1) Cardiac endothelium ATP Vasoconstriction, reduced


gic cardiac output

Reproduced from Favory R, Salgado DR, Vincent J-L. Investigational vasopressin receptor modulators in the pipeline. Expert Opin Investig Drugs.
2009;18:1119–1131.

initiating the second-messenger cascade responsible for the V2Rs, on the other hand, are heavily expressed in the med-
cellular actions of AVP.25 Stimulation of V1R, although not ullary TAL, macula densa (MD), connecting tubule, and corti-
directly involved in control of tubular water and electrolyte cal and medullary collecting duct, as well as weakly expressed
transport, increases sodium excretion because of the in u- in cortical thick ascending limb (TAL) and distal convoluted
ences on blood pressure, effective arterial circulating vol- tubule.35 These are the best characterized and studied vaso-
ume, glomerular ltration rate, and circulation in the vasa pressin receptors. By binding to V2R, AVP increases water re-
recta system.27,28 Additional biologic effects of AVP mediated absorption through multiple mechanisms.31 Activation of V2R
through V1Rs include platelet aggregation 29 and increased results in increased cyclic adenosine monophosphate (cAMP)
glycogenolysis and gluconeogenesis in the liver.30 levels and activation of PKA, which promotes insertion of wa-
The V3R (V1bR), also coupled to PLC signaling, is pres- ter channels into the luminal suface of the epithelial tubular
ent on neurons in the anterior pituitary (adenohypophysis) cells.36 This ultimately mediates the antidiuretic effect of AVP
and is thought to mediate AVP-induced corticotropin by allowing back diffusion of water down its concentration
secretion.25,31,32 They are also found elsewhere in the brain, gradient.37 In addition, V2Rs modulate sodium reabsorption
especially in the pyramidal neurons of the hippocampal through the epithelial Na channel (ENaC) across principal
CA2 eld, in which they mediate fundamental physiologic cells.38–41 This facilitates free water reabsorption by support-
actions such as memory and body temperature control ing the axial corticomedullary hyperosmotic gradient. It was
as well as social behavior.33 In addition, this receptor has also recently shown in rats that AVP modulates sodium re-
been localized to pancreatic islet cells, modulating insulin absorption even in the distal convoluted tubule by acting on
secretion.34 However, its presence and role at the level of the the thiazide-sensitive Na -Cl cotransporter (NCC).42 NCC is
medulla in rat kidney remain unclear. important in de ning sodium delivery to the collecting duct,
234
23 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

which is necessary for ENaC activity. Finally, V2Rs activate of antidiuretic hormone (SIADH). In addition to water
urea transporters, such as UTA1, in the distal nephron.43–45 metabolism, it has been recently suggested that AVP may
This increase in urea reabsorption and recycling maximizes play a role in the initiation and the progression of chronic
sodium reabsorption in the TAL by supporting the axial hy- kidney disease (CKD) and in the most prevalent form of he-
perosmotic gradient drawing water from the distal nephron.46 reditary renal disease, namely the adult polycystic kidney
The clinical importance of the V2R in water balance disorders disease (Fig. 8.2).59
is underlined by the current use of V2R antagonists in a clini- NDI is characterized by impaired AVP-induced water
cal setting (see later). reabsorption, resulting in polyuria and polydipsia.60 If water
It is noteworthy that AVP also binds to two nonspeci c intake is inappropriate, patients with NDI may fail to thrive,
receptors: oxytocin receptors and P2 purinoreceptors. Oxy- suffer from mental retardation, and die early. NDI can be
tocin receptors are found in the breast, ovary, uterus, and hy- acquired such as following lithium treatment, hypokalemia,
pothalamus. Since the af nity of this receptor for vasopressin hypercalcemia, or ureteral obstruction, all of which lead to
is relatively low, the clinical effects of this hormone are lim- downregulation of AQP2.61–64 NDI can also be inherited
ited under physiologic conditions.47 AVP may also act on P2 (congenital).65 In fact, two gene mutations have been linked
purinoreceptors in the heart, causing coronary vasoconstric- to congenital NDI: one in the AVPR2 gene encoding V2R
tion and contributing to the reduction in cardiac output.48 (X-linked NDI) or mutations in the AQP2 gene (autosomal
recessive or autosomal dominant NDI). More than 90% of
Water Channels patients with congenital NDI suffer from X-linked NDI. In
The discovery of the family of aquaporin water channels was both cases, patients cannot concentrate their urine despite
crucial to the understanding of the mechanism by which normal or elevated plasma concentrations of vasopressin,
AVP can increase water permeability in the kidney. The leading to a massive loss of water through the kidney. So far,
group of Peter Agre discovered the rst aquaporin in human NDI is managed by salt restriction combined with hydro-
erythrocytes.49 To date, seven different aquaporin (AQP) chlorothiazide diuretics to reduce urine output.66 However,
have been shown to be expressed in the human kidney and no cure is available so far.
to be involved in renal water reabsorption.50 Abnormal water handling of central origin includes
AQP2 is the vasopressin-sensitive water channel ex- SIADH in which AVP levels are abnormally elevated and not
pressed in the principal cells of the collecting duct, where it suppressed when plasma osmolality concentration falls below
shuttles between intracellular storage vesicles and the apical the osmotic threshold for physiologic AVP secretion.67 SIADH
membrane.51 Knocking out the AQP2 gene produces a severe is characterized by impaired water excretion in the absence
concentration defect in these mice, resulting in postnatal of renal insuf ciency, adrenal insuf ciency, or any recognized
death.52 It has now been shown to be involved in many clini- stimulus for AVP secretion. Renal water retention and extra-
cal disorders (see later). AQP1 is constitutively expressed on cellular uid expansion are compensated for by increased uri-
the basolateral and apical membrane of epithelial cells lin- nary Na excretion leading to life-threatening hyponatremia.
ing the proximal tubule and thin descending limb, as well The most common causes of the syndrome of inappropriate
as endothelial cells of the descending vasa recta. It not only AVP secretion are neoplasia, neurologic disorders, congestive
plays a role in water reabsorption from urine in these seg- heart failure, liver cirrhosis, preeclampsia, and drugs such
ments but is also critical for a functional countercurrent mul- as thiazide diuretics or selective serotonin reuptake inhibitor
tiplication system.53 AQP3 and AQP4 are expressed on the antidepressants.67 Four patterns of AVP dysregulation in pa-
basolateral membrane of the principal cells of the collecting tients with SIADH have been observed.68 The most common
duct, and they represent an exit pathway from these cells for pattern (in 40% of patients with SIADH) is the excessive
water entering through AQP2.54,55 Similar to AQP1, AQP7 is and unregulated release of AVP, which is unrelated to plasma
expressed at the apical membrane of proximal tubules (S3 seg- osmolality. In the second most common pattern ( 30% of
ment) and has been shown to mediate glycerol reabsorption patients), referred to as “reset osmostat,” AVP release contin-
in addition to water.56 AQP6 is found in intracellular vesicles ues to regulate water excretion at a lower plasma osmolal-
of acid-secreting -intercalated cells in the collecting duct.57 ity set-point. Although most tumors (e.g., lung carcinoma)
It is thought to be involved in urinary acid secretion. AQP11 manifest the rst type of SIADH, some also present with the
is localized to the endoplasmic reticulum (ER) in the proximal second type, thus the pattern of abnormal AVP secretion can-
tubule. Interestingly, knocking out the AQP11 gene in mice is not be utilized to predict the cause of SIADH. A third rare
fatal because of the onset of polycystic kidney disease.58 pattern is characterized by an inability to stop AVP secretion
at low plasma osmolalities, but the osmoregulation of AVP is
otherwise normal. This pattern may be due to dysfunction
Clinical Pathophysiologic Role of Arginine of inhibitory hypothalamic neurons, leading to persistent
Vasopressin in the Kidneys low-grade basal AVP secretion. In the fourth pattern, a rare
AVP is implicated in major clinical syndromes of alterations clinical picture of SIADH, the normal osmoregulation of AVP
in water metabolism, namely nephrogenic diabetes insipi- secretion is not altered ( 10% of patients) but AVP levels are
dus (NDI) and the syndrome of inappropriate secretion low or undetectable. It is thought that a nephrogenic SIADH
CHAPTER 8 HORMONES AND THE KIDNEY 2235
35

FIGURE 8.2 Vasopressin receptors: physiology and pathologic involvement in renal diseases. (Reproduced from Bolignano D, Zoccali
C. Vasopressin beyond water: Implications for renal diseases. Curr Opin Nephrol Hypertens. 2010;19(5):499–504.)

(NSIADH) may be responsible for this picture. In fact, in in urinary albumin excretion represents an early predictor of
some children, it appears to be due to an activating mutation glomerular damage in diabetes mellitus and a risk factor for
of V2R. In other patients, it may be due to abnormal control cardiovascular complications in hypertension. Studies show
of aquaporin-2 water channels in renal collecting tubules or that the Brattleboro rat, a model of central diabetes insipidus
production of an antidiuretic principle other than AVP.69 In a with complete lack of AVP, is protected from hyper ltration,
study with SIADH patients, treatment with V2R antagonists, albuminuria, and renal hypertrophy after streptozotocin-
commonly referred to as vaptans, increased serum Na con- induced diabetes mellitus.75 This suggests that AVP plays a
centration and decreased its excretion. Tolvaptan has been role in hyper ltration and glomerular damage induced by
recently approved in the United States and Europe for the diabetes. These observations are also of relevance to humans.
treatment of hyponatremia associated with SIADH, as well A marked increase in AVP plasma levels is well documented
as cirrhosis and congestive heart failure. Recently, a dual in diabetes mellitus.76 AVP through V1R induces contraction
vasopressin V1R and V2R antagonist, conivaptan, improves of cortical efferent, but not afferent, arterioles. Administration
hyponatremia in rats with SIADH, suggesting a therapeutic of a V1R selective antagonist to noninsulin-dependent diabetic
potential for conivaptan in the treatment of SIADH.70 patients modestly reduces albuminuria partly by decreasing
AVP has also been shown to modify vascular tone in renal intraglomerular capillary pressure.77 Thus, although V1Rs
microvessels. Short-term infusion of AVP does not alter either (but not V2Rs) are downregulated in diabetes mellitus, they
renal blood ow or the GFR.71 Alternatively, chronic AVP ad- could mediate part of the increase in albumin excretion. In-
ministration increases the intraglomerular capillary pressure terestingly, administering desmopressin, a selective V2R ago-
and GFR through tubuloglomerular feedback.72 In addition, nist, to healthy humans and patients with central diabetes
a sustained increase in water intake and the consequent AVP insipidus signi cantly increases urinary albumin excretion,
suppression reduce proteinuria and the severity of glomerular but this effect is absent in those with hereditary nephrogenic
and tubular damage in 5/6 nephrectomized rats.73,74 Thus, it diabetes insipidus secondary to V2R mutations.78 These nd-
seems likely that a chronic AVP-induced hyper ltration may ings suggest that the AVP-induced rise in albuminuria de-
alter the glomerular barrier and start a series of events leading pends on V2Rs. This is further con rmed by the observation
to enhanced protein loss and glomerulosclerosis. An increase that plasma copeptin levels correlate with microalbuminuria
236
23 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

in the PREVEND study.79 However, V2 receptors have not In conclusion, in recent years, AVP has been implicated
been found in glomeruli or proximal tubules, suggesting in- in the initiation and progression of many kidney diseases,
direct effects. Recent evidence suggests a strong interaction playing a role beyond water metabolism. Further studies
between AVP and the renin-angiotensin system (RAS).80,81 are required to determine whether AVP antagonists and/or
In fact, chronic RAS blockade by angiotensin-converting AVP suppression by a high water intake can be useful for
enzyme (ACE) inhibitors or angiotensin receptor blockers the treatment of nephropathies, from ADPKD to diabetic and
(ARBs) prevents the desmopressin-induced albuminuria, in- nondiabetic CKD.
dicating that RAS mediates the effects of AVP on glomerular
hemodynamics.82 Recent studies using V1R knockout mice THE RENIN-ANGIOTENSIN SYSTEM
show that AVP regulates body uid homeostasis and the GFR
by activating RAS through V1Rs in MD cells, and subsequent- Historical Review
ly the V2R-aquaporin 2 system.83 Simultaneous AVP and RAS The discovery of renin goes back to 1898, when the Finnish
blockade may represent a good therapeutic approach for de- physiologist Robert Tigerstedt and his student Per Gunnar
laying renal disease progression. Bergman found that extracts of rabbit renal cortex had a
Another interesting observation is that mesangial cells slowly developing and sustained pressor effect. Based on its
express V1Rs, which mediate AVP-induced cell contrac- origin, they named this substance renin.100 This effect was
tion.84 In addition, prolonged exposure to AVP promotes not observed with extracts of renal medulla and persisted
mitogenesis and proliferation of these cells, which ultimately despite removal of sympathetic activation. Subsequently, ef-
leads to an increased accumulation of extracellular matrix, a forts to verify these experiments were unsuccessful until the
pathologic feature found in various glomerular diseases.85,86 1930s when Harry Goldblatt and his colleagues demonstrat-
In fact, addition of AVP to cultured mesangial cells increases ed that clamping the renal artery in dogs produced chronic
in a dose-dependent manner the synthesis and release of ma- hypertension.101 This work converged with other subse-
trix proteins, such as type I and IV collagen, bronectin, and quent experiments performed by leading scientists such as
transforming growth factor .87 In addition, AVP inhibited Juan Fasciolo and Bernardo Houssay to suggest the presence
the synthesis of matrix metalloproteinase (MMP)-2, which of a vasoactive substance produced by the kidney, other than
degrades matrix proteins including type IV collagen, and renin.102,103 This substance was later isolated from the blood
stimulated endothelin (ET)-1 secretion from mesangial cells, and was named hypertensin. Based on their physiologic
another mitogenic factor.88 properties, renin and hypertensin were clearly two different
In the remnant kidney model in rat, a model of progres- compounds, but the relationship between them was not es-
sive CKD in humans, V1R antagonists (but not V2R) prevent tablished. Renin was later identi ed by an Argentine group
proteinuria and glomerulosclerosis in the initial phases of as a proteolytic enzyme that acts on a plasma constituent to
disease, but have limited effectiveness in established re- produce hypertensin as the nal product of the enzymatic
nal damage.89,90 Similar observations were reported in 5/6 reaction.104 Subsequent research led to the characterization
nephrectomized, salt-loaded spontaneously hypertensive of the components of the renin–angiotensin system and
rats (SHRs), in which the increase in urinary protein excre- Braun-Menéndez and Page gave the nal nomenclature of
tion and the progression of nephrosclerosis were attenuated the whole enzymatic system in 1958: the renin substrate was
with a V1R antagonist but not with a V2R antagonist.91 named angiotensinogen, hypertensin renamed angiotensin,
AVP has also been linked to autosomal dominant poly- and the enzymes that metabolize angiotensin were named
cystic disease (ADPKD), an inherited disorder characterized by angiotensinases.105,106
the development within renal tubules of innumerable cysts that
progressively expand to cause renal insuf ciency.92 It has been Components of the
shown that 3 -5 -cyclic adenosine monophosphate (cAMP) Renin-Angiotensin-Aldosterone System
stimulates tubule cell proliferation and transepithelial uid se-
cretion, both of which contribute to enlarge renal cysts.93 AVP Angiotensinogen
operates continuously in ADPKD patients to promote cAMP Angiotensinogen is a large molecular weight globulin pri-
production in the distal nephron and collecting ducts via V2Rs, marily formed by hepatic cells. It is constitutively secreted
thereby contributing to cyst enlargement and renal dysfunc- into the circulation by the liver, therefore plasma levels
tion.94 Studies in animal models of ADPKD provide compel- are generally stable.107 Other sources of angiotensinogen
ling evidence that blocking AVP’s actions dramatically improves have been identi ed and mRNA expression detected in tis-
disease progression.95,96 This prompted the initiation of an sues such as the kidney, heart, brain, vessels, placenta, and
international clinical trial97 testing the ef cacy of tolvaptan, a adrenal glands.108 It is currently accepted that intrarenal
V2R inhibitor, in the treatment of ADPKD.98 Alternatively, a re- angiotensinogen is formed and secreted locally for several
cent review discusses that the impact of simply increasing the reasons: rst, the molecular size of the molecule makes it
amount of solute-free water drunk evenly throughout the day unlikely for it to lter through the glomerular capillaries;
by patients with ADPKD on decreasing plasma AVP concentra- second, intrarenal angiotensinogen mRNA and protein
tions and mitigating the actions of cAMP on the renal cysts.99 have been identi ed in proximal tubule cells109; third,
CHAPTER 8 HORMONES AND THE KIDNEY 2237
37

concentrations of angiotensinogen in the proximal tubule of bradykinin and kallidin. Therefore, the functional activity of
anesthetized rats greatly exceeded the free angiotensin I and ACE results in enhanced vasoconstriction and reduced va-
II concentrations110; and fourth, human angiotensinogen sodilation. Differences between humans and various animal
was not detected in urine of normotensive rats infused with species regarding ACE localization in the kidney have been
the molecule.111 reported. Normal nonhypertensive human subjects show a
widespread expression of ACE on the brush border of tubule
Renin and Prorenin epithelial cells and less expression on glomerular vascular
Renin is produced and stored in granular juxtaglomerular endothelial cells, whereas the renal microvasculature of rats
cells, which are modi ed smooth muscle cells found in the show more preponderant ACE expression compared to epi-
media of afferent arterioles.112–114 Genomic analysis of the thelial cells. These ndings imply that the contribution of
renin gene identi ed a single locus in humans and rats des- circulating angiotensin I to the local formation of angioten-
ignated Ren-1, whereas mice have two renin genes, desig- sin II in the kidney may be minimal.118
nated Ren-1 and Ren-2.112 This duplicated renin gene in mice In the plasma, all conversion of Ang I to Ang II occurs
leads to production of substantial amounts of renin from by the activity of ACE with no species variation reported.
submandibular and submaxillary glands.115 Renin is synthe- However, non–ACE-dependent pathways exist at the tissue
sized in an inactive precursor form, preprorenin. Cleavage level and have species variation. In humans, tissue activity
of the signal peptide from the carboxyl terminal of prepro- of chymase can allow for the local formation of Ang II in the
renin results in prorenin, which is also biologically inactive. heart, arteries, and kidney. In rats and rabbits, tissue activity
Subsequent glycosylation and proteolytic cleavage leads to of chymase is associated with the local degradation (instead
formation of renin, a 37 to 40 kDa proteolytic enzyme. Both of formation) of Ang II. Therefore, one must carefully evalu-
circulating active renin and prorenin are released mostly ate experimental animal data when pharmacologic blockade
from the kidneys; however, other tissues also secrete these of the renin-angiotensin system is used.119
substances.116,117 Because prorenin is the major circulating
form, it is postulated that signi cant conversion of prorenin Angiotensin Peptides
to renin follows secretion. Prorenin-activating enzymes have
been localized to neutrophils, endothelial cells, and the kid- Ang II is the primary active product of the RAAS. It is an oc-
ney.112 In addition to juxtaglomerular cells, renin production tapeptide derived from Ang I after cleavage of the C-terminal
has also been detected in the submandibular gland, liver, dipeptide by the ACE. Most of the physiologic actions of the
brain, prostate, testis, ovary, spleen, pituitary, thymus, and RAAS on the vasculature and transport functions are me-
lung.112 Circulating renin, however, appears to be derived diated by Ang II action on angiotensin receptors, primarily
entirely from the kidney. type 1 (AT1) receptors. However, other angiotensin peptides
with reported biologic activity have been identi ed, such as
angiotensin III (Ang III) and IV (Ang IV), which are formed
Angiotensin I
from Ang II by the sequential removal of amino acids from
Angiotensin I is an inactive decapeptide formed upon cleav- the N-terminus by aminopeptidases (Fig. 8.3). They are
age of angiotensinogen by active renin in the circulation. predominantly seen in the kidney and brain, where amino-
Its rate of formation is highly determined by renin activity. peptidases A and N are prevalent.107,117 Ang III, also known
Angiotensin I is easily hydrolyzed to angiotensin II given the as angiotensin 2-8, is a heptapeptide with suggested role in
widespread availability of ACE on endothelial cells of many blood pressure maintenance in the brain. Ang IV (angioten-
vascular beds, including the lungs; the octapeptide angio- sin 3-8), on the other hand, is a hexapeptide that possibly
tensin II is therefore formed by cleavage of the C-terminal enhances Ang II signaling. The heptapeptide Ang (1-7) is
dipeptide of angiotensin I.107 currently considered one of the biologically active end prod-
ucts of the RAAS. It is formed from Ang I or Ang II in the
Angiotensin-converting Enzyme kidney and heart by the action of tissue peptidases at the C-
The ACE, also known as kininase II, is a membrane-bound terminus. Once formed, it is rapidly hydrolyzed by ACE; in
peptidase that catalyzes the conversion of angiotensin I conditions of ACE inhibition or AT1 receptor antagonism, its
(Ang I) to angiotensin II (Ang II), the primary active prod- concentration may increase severalfold. There are two major
uct of the renin-angiotensin-aldosterone system (RAAS). interactions between Ang (1-7) and bradykinin (BK): poten-
This enzyme is localized on the membrane of various cell tiation of BK by Ang (1-7) and mediation of vascular actions
types, mostly the endothelial cells. Other cell types include of Ang (1-7) by kinins. Both mechanisms are involved in
the epithelial cells of the kidney (e.g., the brush border of the cardioprotective effects of ACE inhibitors. At the kidney
the proximal tubule cells) and the neuroepithelial cells. The level, the proposed role of Ang (1-7) is natriuresis and diure-
ACE exists also in the plasma as a soluble circulating en- sis as opposed to Ang II, an effect blocked by AT1 receptor
zyme, but it is thought that the membrane-bound form is antagonists like losartan. In the vascular bed, it antagonizes
the physiologically active one.107 Other metabolic activities the vascular effect of Ang II by acting as a competitive an-
of ACE include the inactivation of the vasodilator peptides tagonist to AT1 receptors.120
238
23 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

expression in the human adult and is found in the kidney,


adrenal gland, heart, and brain. In the kidney, AT1 receptors
are found in the glomeruli, proximal tubule brush border
and basolateral membranes, thick ascending loop, proximal
convoluted tubule, renal vasculature, the proximal and dis-
tal nephron segments, and in both cortical and medullary
regions.125,126

Aldosterone
Aldosterone was identi ed by Simpson SA in 1953 and named
electrocortin.127 Later studies characterized more the nature
of this hormone and identi ed it as a mineralocorticoid syn-
thesized and secreted by the zona glomerulosa of the adrenal
cortex. The physiology of aldosterone action has been well es-
tablished after several breakthroughs in research experiences,
such as the identi cation of the mineralocorticoid receptor
(MR) as the principal aldosterone receptor, the characteriza-
tion of sites of aldosterone action in target tissues such as the
ENaC, and the demonstration of post-receptor processes in-
volved in physiologic responses to aldosterone.128–130 Aldo-
sterone secretion is determined by several stimuli, the most
important being Ang II and plasma potassium concentration.
The MR is the principal receptor for aldosterone, but other
target proteins can also bind aldosterone, such as the gluco-
corticoid receptor. Aldosterone has been implicated in the
pathophysiology of several cardiovascular and renal diseases
and will be discussed in details in other sections.129
FIGURE 8.3 Schematic representation of the renin-angiotensin
system showing plasma concentrations of some components as Physiologic Actions of Angiotensin II
measured in anesthetized rats. EP, endopeptidase; APA, amino-
Systemic Effects of Angiotensin II
peptidase A; APN, aminopeptidase N.
AT1 receptors have been shown to mediate many of the
functions of Ang II in the regulation of blood volume, cell
contraction, cell proliferation, aldosterone secretion, pres-
Angiotensin II Receptors sor and tachycardic responses, increased thirst, and hyper-
Circulating Ang II exerts its biologic effects by binding to tension secondary to renal artery stenosis. AT1 receptors
speci c receptors on the cell surface.112,121 At least four an- are positively coupled to phospholipase C and mitogen-
giotensin receptor subtypes have been identi ed. AT1 recep- activated protein kinases (PI3 and MAP) and negatively to
tors bind Ang II with higher af nity than Ang III and are adenylate cyclase.122,125,131 The predominant function of the
selectively blocked by the biphenylimidazole compound renin-angiotensin system is regulation of vascular tone and
losartan. AT2 receptors bind Ang II and III with similar af- renal salt excretion in response to changes in the volume of
nity and are selectively blocked by tetrahydroimidazopyri- extracellular uid or blood pressure. Ang II represents the
dines, such as PD123177.122 The type 3 angiotensin receptor effector limb of this hormonal system, acting on several or-
AT3 has no known function and the type 4 (AT4) is thought gans, including the vascular system, heart, adrenal glands,
to mediate the release of plasminogen activator inhibitor by central nervous system, and kidneys. Through direct ac-
Ang II, III, and IV. tion on smooth muscle cells, Ang II signi cantly increases
Megalin is an abundant membrane protein heavily in- arteriolar resistance in renal, mesenteric, dermal, coronary,
volved in receptor-mediated endocytosis. Megalin is a recep- and cerebral vascular beds.132 Skeletal muscle and pulmo-
tor for Ang II and Ang II internalization in some tissues is nary vessels, on the other hand, are not affected because of
megalin-dependent. Megalin may play a role in regulating Ang II-stimulated production of vasodilatory prostaglan-
proximal tubule Ang II levels.123 Ang (1-7) exert its vasodila- dins by endothelial and smooth muscle cells in these vas-
tor and natriuretic actions presumably through its binding to cular beds.133,134 Ang II exerts indirect pressor effects via the
a unique receptor, the Mas receptor.124 central and peripheral nervous systems. Its effects on the cen-
In rodents, two isoforms of AT1 receptors exist: AT1A tral nervous system include increased sympathetic discharge
(nephron) and AT1B (glomerulus), whereas in humans there and decreased vagal tone.135 Peripherally, Ang II augments
is only one AT1 isoform. The AT1 receptor has widespread the vasoconstrictive response to renal nerve stimulation in
CHAPTER 8 HORMONES AND THE KIDNEY 2239
39

dogs136 and its inhibition attenuates the pressor response to by endothelial-derived NO148 and constricts descending vasa
norepinephrine in humans.137 Experimental data suggest the recta (DVR) through Ca2 signaling in pericytes.149 The dis-
presence of a local renin–angiotensin system in the vascula- proportionate increase in postglomerular resistance results in
ture that contributes to the regulation of vascular tone.112 a marked increase in glomerular capillary hydrostatic pres-
A more recently recognized function of Ang II is its sure (PGC), ultra ltration pressure, and ltration fraction,
growth-promoting effects in smooth muscles of the vas- thus preserving GFR in the face of declining renal plasma
culature, heart, and the kidney. Ang II has been shown to ow (RPF). The selectivity of the vasoconstrictive action of
induce hypertrophy and mitogenesis in cultured vascular Ang II for the efferent arteriole results from stimulation of va-
smooth muscles.138,139 This effect is at least, in part, medi- sodilatory prostacyclin synthesis by the afferent arteriole and
ated through autocrine production of growth factors such not a preferential action of Ang II on the efferent arteriole.150
as platelet-derived growth factor (PDGF) and transforming In fact, Ang II increases both afferent and efferent resistance
growth factor- (TGF- ).140 Some studies suggest that the in the presence of a cyclooxygenase inhibitor.150 Under cer-
renin–angiotensin system contributes to neointimal for- tain pathophysiologic conditions, afferent arteriolar constric-
mation and restenosis after angioplasty.112 Ang II has been tion predominates, leading to a decrease in both RPF and
shown to have direct inotropic, chronotropic, mitogenic, GFR.151 Deep nephrons have higher postglomerular Ang II
and hypertrophic effects on isolated atria and ventricles.112 tone and also higher Ang II sensitivity than super cial neph-
Amelioration of hypertensive cardiomyopathy by ACE in- rons. The better preserved GFR in deep cortex during Ang II
hibitors suggests that the renin-angiotensin system plays a action may contribute to maintaining the renal-concentrating
role in cardiac hypertrophy.112 ability by providing NaCl for reabsorption by the ascending
The predominant physiologic role of the AT2 receptor is limb of the loop of Henle.152
to initiate vasodilation and natriuresis as a counterregulatory In addition to its vascular effects, Ang II induces me-
response to the vasoconstriction caused by activation of the sangial cell contraction, which leads to decreased Kf in
AT1 receptor.141 Other functions include inhibition of growth vivo.153,154 This effect, however, is attenuated by the con-
and hypertrophy, and stimulation of apoptosis.142 This has comitant production of prostaglandins by mesangial cells.155
been most clearly demonstrated in AT2 receptor knockout Ang II is one of the most potent sodium-retaining hor-
mice that have slightly elevated blood pressure in the basal mones in the body. Increased tubular sodium reabsorption is
state, but have an exaggerated increase in blood pressure in enhanced by both direct and indirect tubular effects of Ang II.
response to Ang II infusion compared to wild type mice.143,144 Physiologic concentrations of Ang II (10 12 to 10 10 M) stim-
The intracellular signaling pathways coupled to the AT2 ulate proximal tubule NaCl and NaHCO3 absorption at the
receptor are unclear. However, there is evidence that the AT2 proximal tubule.156 Indirectly, Ang II stimulates ion transport
receptor may be coupled to the production of a variety of renal in the proximal tubule by changing the peritubular milieu.
vasodilator substances, counterregulating the pressor effects Ang II can both decrease peritubular capillary hydrostatic
of Ang II via AT1 receptors.142 The most likely candidates are pressure and increase peritubular oncotic pressure, resulting
bradykinin and nitric oxide (NO)-stimulated cyclic guanosine in an increased driving force for ion reabsorption. Directly,
monophosphate (cGMP),145,146 but other candidates include Ang II can stimulate transport in the proximal tubule through
products of cyclooxygenase, such as PGE2 and PGI2.141 interaction with the AT1 receptors found on both the apical
and basolateral membranes of the tubule cells.157,158 It also
Renal Effects of Angiotensin II stimulates calcineurin phosphatase activity in proximal tubule
Ang II serves at least three important functions in the kidney: epithelial cells through a mechanism involving AT1 receptor-
regulation of blood ow and GFR, reduction of salt excretion mediated tyrosine phosphorylation of the PLC isoform, both
through direct and indirect actions on renal tubule cells, and linked to sodium transport in the proximal tubule.159 Spe-
growth modulation in renal cells expressing AT1 receptors. ci cally, Ang II stimulates the activity of apical membrane
In conditions of decreased renal blood ow, GFR is pre- Na–H antiporters and basolateral membrane Na–HCO3–CO2
served at nearly constant value over a wide range of perfusion cotransporters160 and the activity of Na–K-ATPase by chang-
pressures. This phenomenon is known as autoregulation of ing phosphorylation and conformation of Na–K-ATPase.161
GFR. At low levels of renal perfusion pressures, Ang II con- The net effect is increased proximal tubule reclamation of Na
tributes to this phenomenon.147 Micropuncture studies have and HCO3. Denervation of the proximal tubule results in at-
shown that Ang II exerts substantial effects on the renal mi- tenuation of the Ang II-stimulated NaCl, but not NaHCO3
crovasculature and hemodynamics; however, the individual absorption, suggesting that Ang II enhances proximal Na
contribution of the systemic and local renin-angiotensin sys- transport indirectly by increasing presynaptic catecholamine
tems to the overall regulation is still controversial. Ang II con- release.159 Of note is that supraphysiologic concentrations of
stricts both the afferent and efferent arterioles, reduces single Ang II (10 9 to 10 7 M) inhibit NaCl and water reabsorption
nephron glomerular ltration rate and plasma ow, increases in the proximal tubule and also inhibit Na–glucose cotrans-
the ltration fraction, and decreases glomerular ltration porter translocation by inactivation of PKA and decrease of
coef cient. Other studies have shown that Ang II infusion PI3-kinase activity mediated through the AT1 receptor.162,163
preferentially vasoconstricts efferent arterioles counteracted Ang II increases proximal tubule phosphate absorption by
240
24 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

direct stimulation of Na/Pi cotransport activity as a result of demonstrated in several animal hypertension models includ-
increase in the expression of brush-border membrane NaPi- ing renovascular hypertension.180,181 Clinical studies have
IIa protein level and that stimulation is most likely mediated also shown that local intrarenal Ang II formation was central
by posttranscriptional mechanisms.164 to the development of hypertension in humans.182
Intraluminal conversion of Ang I to Ang II can occur In addition to its effects on the maintenance of blood
in the cortical collecting duct, resulting in enhanced api- pressure, AT1 receptors may play a role in embryonic nephro-
cal sodium entry.165 The AT2 receptor regulates epithelial genesis. Blockage of the renin-angiotensin system with ACE
sodium channel (ENaC) abundance, consistent with a role inhibitors or AT1 inhibitors results in abnormal renal devel-
for Ang II in regulation of collecting duct function via AT1 opment that is characterized by both papillary and tubular
receptors.166,167 atrophy and by interstitial brosis and in ltration. In addi-
Ang II increases basolateral K-channel activity via the tion, knockout mice lacking both the AT1A and AT1B receptor
stimulation of AT1 receptors and the stimulatory effect of have similar renal abnormalities.183 Ang II also modulates
Ang II is mediated by a NO-dependent cGMP pathway.168 mesangial cell growth, and induces proximal tubular cell hy-
Other effects of Ang II on proximal tubule cells include pertrophy in humans, effectively inhibited by irbesartan, an
enhanced gluconeogenesis and ammonia production.169 Ang II receptor antagonist.184
The effects of Ang II on distal tubule transport of Na and K The AT2 receptor is expressed predominantly in fetal
are mediated through aldosterone release.170 In addition to tissues, but in almost all tissues there is postnatal down-
proximal tubule epithelial cells, vasa recta and outer med- regulation. AT2 receptor mRNA is expressed in the fetal and
ullary vascular bundles express high density of AT1 recep- neonatal rat kidney, but disappears after the neonatal period
tors.121 ACE inhibitors increase descending vasa recta blood and is not expressed in the normal adult. Although the AT2
ow, whereas Ang II infusion markedly decreases medullary receptor mRNA is not found in the adult kidney, both immu-
blood ow in rats.171 It is postulated that Ang II in uences nohistochemistry and Western blot analysis have detected
urinary dilution and concentration by modulating blood AT2 receptor protein in the glomeruli, cortical tubules, and
ow to the medulla. interstitial cells of the adult kidney.121,185 It was thought that
Ang II induces hypertrophy of proximal tubule epithe- AT2 receptors might play a role in the development of the
lial cells in vitro.172 It also exerts similar growth-promoting kidney and urinary tract given the high levels of expression
effects on mesangial cells.173 The signaling mechanism by in the fetus. However, although early studies of AT2 receptor
which Ang II exerts this effect is not precisely known, but knockout mice showed no gross morphologic abnormalities
p27Kip1 is required for Ang II-induced hypertrophy.174 of the kidney,143,144 a more recent study has demonstrated
Downstream potential targets of Ang II are the extracellular the presence of increased numbers of congenital anomalies
signal-regulated kinases 1 and 2 (ERK1/ERK2) and Ang II of the urinary tract.186 The AT2 receptor may be implicated
activates ERK1/ERK2 via the AT1 receptor.175 Studies have in some congenital abnormalities of the urinary tract or may
shown that connective tissue growth factor (CTGF) might be be involved in the pathophysiologic response to ureteral ob-
an important mediator of Ang II-induced renal hypertrophy, struction by protecting against the formation of interstitial
which suggests that inhibiting the production of CTGF might brosis.186,187 Ang II modulates the over-expression of AT2
be the new strategy in early prevention of renal brosis.176 receptors in renal ablation experiments through its own AT2
Ang II can stimulate human kidney broblast (KFB) prolif- receptor and functional expression of this effect may repre-
eration and enhance the expression of interleukin-6 in KFB. sent a counterregulatory mechanism to modulate the renal
These ndings suggest that Ang II might play a part in the damage induced by renal ablation.188
mechanisms for modulating tubulointerstitial changes and Ang II regulates renal parathyroid hormone-related pro-
inducing renal brosis.177 tein (PTHrP), a vasodilator and mitogenic agent upregulated
Previous in vivo studies in cardiomyopathic hamsters in kidney injury, and its type-I receptor (PTH1R) system via
suggested that the expression of vasopressin (AVP) V2 mRNA AT1 receptors.189
is upregulated by Ang II. Ang II caused a signi cant increase
in the AVP V2 mRNA in a dose-dependent manner medi- Local Effects of Ang II in Nonrenal Organs
ated by PKA, whereas PKC suppresses the expression of V2 Ang II exerts its actions on other organs, such as the adre-
mRNA in the inner medullary collecting duct (IMCD) of the nals and the brain. At the level of the adrenals, it stimulates
rat kidney.178 aldosterone synthesis and secretion.170 Acute angiotensin
The role of Ang II in the pathogenesis of hypertension administration stimulates the activity of 11 -hydroxysteroid
is complex. Several studies have demonstrated a crucial role dehydrogenase (HSD) type 2 in human kidneys and exerts
of the intrarenal renin-angiotensin system in the develop- a dual effect on the MR receptor (i.e., an indirect agonistic
ment of hypertension and kidney disease.117 Animal studies effect by increasing aldosterone availability and a direct or
have shown that blockade of the AT1 receptor resulted in indirect antagonistic effect by stimulation of renal 11 HSD
increased plasma Ang II levels while it limited renal Ang II type 2 activity).190
contents in response to Ang II infusion.179 This dissocia- At the central nervous system (CNS) level, it stimulates
tion between systemic and local Ang II regulation has been thirst and salt appetite191,192 and may increase secretion of
CHAPTER 8 HORMONES AND THE KIDNEY 2241
41

vasopressin and oxytocin from the posterior pituitary, and of postjunctional -adrenergic receptors increases renin re-
adrenocorticotropic hormone (ACTH), prolactin, and lu- lease. The role of -adrenergic receptors, on the other hand,
teinizing hormone from the anterior pituitary.193 is controversial.201 Ample evidence suggests that dopamine
stimulates renin secretion by direct activation of dopamine
A1 (DA1) receptors on juxtaglomerular cells.201,202
Regulation of the Systemic Several endocrine and paracrine hormones regulate re-
Renin-Angiotensin-Aldosterone System nin secretion by the kidney. ANP has been shown to inhibit
Ang II in the systemic circulation is produced primarily from renin release from isolated juxtaglomerular cells.203 Other in-
circulating angiotensinogen through the proteolytic action hibitory hormones include AVP, endothelin, and adenosine
of renin. Most of the circulating renin is derived from the (A1-receptor agonists).114,195 Regulation of renin secretion by
kidney, although other organs can secrete prorenin in the Ang II is probably the most physiologically relevant.169 Ang
circulation. Angiotensinogen is mostly formed and secreted II inhibits renin secretion and renin gene expression in a
in the circulation by liver cells, allowing for systemic for- negative feedback loop. Treatment of transgenic mice bear-
mation of angiotensin peptides, principally Ang II. The cir- ing the human renin gene with an ACE inhibitor increases
culating concentrations of angiotensinogen are more than renin expression in the kidney by ve- to tenfold.204 Simi-
1,000 times greater than those of Ang I and Ang II, therefore larly, ACE inhibition in rats augments renal renin mRNA ex-
changes in plasma renin activity is the major determinant of pression, an effect that is reversed by infusion of Ang II.205
Ang I formation (Fig. 8.3). Renin secretion is stimulated by The effects of Ang II are believed to be direct and not depen-
various dynamic parameters such as renal perfusion pres- dent on changes in renal hemodynamics or tubular trans-
sure, tubular uid sodium chloride concentration at the port. Arachidonic acid metabolites produced in the kidney
MD, sympathetic discharge to the kidney, and endocrine and also play an important role in renin secretion.195 Intrarenal
paracrine hormones and growth factors.107 infusion of arachidonic acid increases (and indomethacin
Stimulation of renin release by juxtaglomerular cells is decreases) plasma renin activity in rabbits.206 Several stud-
mediated by increased intracellular cAMP, whereas a rise in ies have since con rmed that prostaglandins of the I series
cytosolic free calcium is inhibitory.194 Renin release responds are potent stimulators of renin secretion.195,197 On the other
inversely to changes in renal perfusion pressure.114 Eleva- hand, lipoxygenase products of arachidonic acid metabolism
tion of intrarenal arterial pressure inhibits renin release and (12-HPETE, 15-HPETE, and 12-HETE) and cytochrome
induces a “pressure” natriuresis. At least two mechanisms P450-mediated epoxides (14,15-epoxyeicosatrienoic acid)
have been postulated. Increased afferent arteriolar wall ten- have been shown to inhibit renin release in renal cortical
sion secondary to increased renal perfusion elevates intra- slices.207,208
cellular calcium in juxtaglomerular cells and inhibits renin
secretion.114,194 Increased perfusion pressure also stimulates
NO production and release by endothelial cells. NO, in The Local Renin-Angiotensin System
turn, suppresses renin secretion.195,196 Conversely, decreased The renin-angiotensin system has been characterized as an
renal perfusion results in increased production of prosta- endocrine, paracrine, and autocrine system. Contribution
cyclin (prostaglandin I2), which enhances renin release.197 of systemically formed mediators to local control of dynam-
Mechanical strain leads to upregulation of the AT1 receptor ics within tissues is dif cult to delineate. Recent evidence
and increased Ang II production in conditionally immortal- suggests that local formation is a major determinant of Ang
ized podocytes. The resulting activation of a local tissue an- levels in organs and tissues. In the brain, for example, Ang
giotensin system leads to an increase in podocyte apoptosis, peptide levels are regulated in an autonomous manner.209
mainly in an AT1 receptor-mediated fashion.198 Local renin-angiotensin systems have been identi ed in sev-
Decreased NaCl delivery to MD cells stimulates renin se- eral organs, including the kidney, heart, vasculature, brain,
cretion, whereas increased urinary NaCl exerts an opposite and adrenals.210 Although most organs have elements of the
effect.180 Schlatter and coworkers199 demonstrate that chang- renin-angiotensin system, the adult kidney is unique in ex-
es in luminal Cl concentration alter the rate of Na -K -2Cl pressing all the components of the system.156,169
transport in MD cells.199 The precise mechanism by which Compared with plasma levels, the renal Ang II contents
variation in the activity of this transporter translates to a signal are much higher despite suppression of renin secretion and
that regulates renin release by adjacent juxtaglomerular gran- release.126 Renin is principally produced by juxtaglomerular
ular cells is not entirely clear. Postulated mediators include cells of the distal afferent arteriole, but has been shown to be
adenosine, which inhibits renin secretion via activation of A1 expressed in the proximal tubule cells,126 whereas its substrate,
receptors on juxtaglomerular cells, and alterations in inter- angiotensinogen, is expressed by proximal tubule cells. ACE
stitial osmolality, which may affect renin secretion directly.96 activity in the kidney has also been localized to the proximal
Experimental evidence also suggests that NO produced by tubule, with the highest concentration present on the brush
MD cells and endothelial cells regulates renin secretion.195,200 border. Several studies have provided evidence for production
The importance of renal sympathetic innervation in of Ang II by the kidney, mainly concentrated in the proximal
controlling renin secretion is well recognized.201 Stimulation tubules,126 suggesting that the intrarenal renin-angiotensin
242
24 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

system is, indeed, functional.156 Some investigators have pro- The role of the RAAS in end-stage renal disease (ESRD)
posed that this local system plays a role in proximal tubule is even more complicated and implicates many parameters
NaCl and HCO3 absorption, pathogenesis of essential hyper- related to residual renal function, presence of renal replace-
tension, and expression of the phenotype of autosomal domi- ment therapy and modality, and drug treatment of associated
nant polycystic kidney disease.156 Independent regulation conditions. In addition, renal transplantation adds a new
of renal Ang II production has not been de nitively demon- dimension to the complexity by introducing an immune
strated. Circulating Ang II stimulates renal angiotensinogen component through graft rejection and immunosuppressive
mRNA production and intact urine angiotensinogen suggests drugs. Despite the substantial decrease in renal blood ow
its presence along the whole nephron and that renin and ACE and function in patients with ESRD on dialysis, studies have
activity are available all through the nephron.126 shown that those with remaining kidneys have an activated
Endogenous Ang II in both peritubular blood and lu- intrarenal renin-angiotensin system.117 On the other hand,
minal uid is important for maximal expression of the stim- renal transplant diseases, especially rejection and cyclospo-
ulatory in uence of this peptide on proximal tubule uid rine-induced nephropathy, were associated with increased
uptake.211 Intraluminal conversion of Ang I to Ang II can activity of systemic and local RAAS in several studies.220,221
occur in the cortical collecting duct, resulting in enhanced In conclusion, the RAAS is one of the most powerful
apical sodium entry.165 pressure and volume regulatory systems in the body, involved
Renal degradation of Ang II is constitutively high, un- in physiologic responses and pathophysiologic processes at
affected by chronic levels of arterial blood pressure, and is both the systemic and local levels. Autonomous and inde-
independent of long-term changes in levels.212 pendent control of local renin-angiotensin systems in various
Low-density lipoproteins (LDLs) increase Ang II produc- organs and tissues may exist and contribute to disease pro-
tion by mesangial cells, which, in turn, results in increased gression. New areas of interest in the RAAS are continuously
O2 production, cell proliferation, and hypertrophy—these emerging, facilitating the understanding of mechanisms of
effects of Ang II are mediated by the AT1 receptor.213 diseases and development of speci c drug targets.

Established Clinical Pathophysiologic Role of


the Renin-Angiotensin-Aldosterone System ATRIAL NATRIURETIC PEPTIDE
in Humans Molecular and Biochemical Properties of
The RAAS has been implicated in the pathophysiology of Atrial Natriuretic Peptide
several diseases of the cardiovascular system and the kid- The cDNA for human ANP was isolated in 1984 and, shortly
ney, mostly hypertension and renal injury. The local renal afterward, the gene was localized to the short arm of chro-
renin-angiotensin system is activated in the renovascular mosome 1.222,223 The chromosomal gene consists of three
type of hypertension in the stenotic kidney and accounts for exons and two introns encoding for a mature mRNA tran-
most of the Ang II concentration in the renal tissue.214 In ad- script approximately 900 bases long.223
dition, it plays a crucial role in other forms of hypertension. Translation of human ANP mRNA results in a 151-amino
Administration of a renin inhibitor to normal subjects and acid preprohormone.224 Pro-ANP, a 126-residue molecule,
hypertensive patients showed sustained local renal vascular is formed after cleavage of the signal peptide sequence of
responses but time-dependent decreased drug activity at the prepro-ANP and represents the major storage form of the
systemic level.215 Several other studies demonstrated the im- hormone in atrial granules.225 The circulating, biologically
portance of the intrarenal renin-angiotensin system in the active form of ANP, often referred to as AN99–126 or ANP1-
pathophysiology of hypertension.216,217 28, is a peptide comprising the 28 carboxy-terminal amino
Ang II promotes brogenesis and oxidative stress in the acids of the parent molecule.224 The amino acid sequence of
kidney by stimulating brogenic mediators, altering renal he- ANP99–126 is highly conserved among mammalian species.224
modynamics especially facilitating glomerular hypertension, A disul de bond between cysteine residues 105 and 121
inducing tubulointerstitial hypoxia, and enhancing free radical gives ANP99–126 its ring structure, which is essential for bio-
formation. Studies have shown that the degree of mesangial logic activity.226
hypercellularity and expansion in IgA nephropathy correlated Pro-ANP (1–30), (31–67), and (68–98) are secreted
closely with glomerular expression of mRNAs for renin, ACE, from the heart and circulate in the plasma. Pro-ANP (1–30)
chymase, and AT1 and AT2 receptors.217,218 The role of the and (31–67) increase sodium and water excretion and bind-
intrarenal renin angiotensin system has been also established ing sites have been found in the proximal tubules and col-
in the pathogenesis of membranous nephropathy.219 In dia- lecting ducts. Pro-ANP (31–67) inhibits the Na -K pump
betic nephropathy, the renin-angiotensin system plays a cru- at the medullary collecting duct through a prostaglandin-
cial role in disease progression: the intrarenal generation of dependent mechanism and no effect on cGMP production.
Ang II is increased, despite suppression of the systemic RAAS. Pro-ANP (1–30) infusion in rats clearly increases urine out-
Details of the pathophysiologic mechanisms implicated in put, sodium, and potassium excretion, the mechanism of
diabetic nephropathy are beyond the scope of this chapter. which still needs to be elucidated.227
CHAPTER 8 HORMONES AND THE KIDNEY 2243
43

Secretion and Physiologic Regulation of Physiologic Actions of Atrial


Atrial Natriuretic Peptide 99–126 Natriuretic Peptide 99–126
Cardiac atria contain the highest concentrations of ANP Three subtypes of ANP receptors (NPRs) have been identi-
and serve as the major source of circulating hormone.224 ed.238–240 NPR-A and -B have intrinsic guanylate cyclase ac-
ANP99–126 secretion from cardiomyocytes occurs largely in tivity that catalyzes production of cGMP after ligand binding.
response to atrial stretch resulting from increased atrial cGMP then serves as an intracellular second messenger
transmural pressure.228 Physiologic stimuli for the release that mediates the biologic activities of ANP99–126.241 NPR-B
of ANP99–126, include acute salt and volume loading, su- appears to have 50-fold higher af nity for a related natri-
pine posture (head-down tilt), and head-out water immer- uretic factor originally puri ed from porcine brain, known
sion.228–230 An increased rate of atrial contraction has also as C-type natriuretic peptide (CNP).242 NPR-C, previously
been shown to stimulate ANP99–126 secretion.231,232 Ang known as the (clearance) receptor, is devoid of guanylate
II, vasopressin, epinephrine, and phenylephrine stimulate cyclase activity and, therefore, does not confer biologic ac-
ANP99–126 secretion from the heart largely because of their tivity and is thought to mediate clearance of circulating ANP
systemic vasopressor effects.233 On the other hand, gluco- along with metalloendoprotease (E.C.3.4.24.11)242 and of
corticoids and endothelin raise ANP99–126 levels possibly other related hormones, such as brain natriuretic peptide
by acting directly on atrial myocytes.234 Leptin decreases (BNP).243 Selective downregulation of NPR-C in the kidney
ANP secretion via an NO-mediated mechanism.235 Physi- in response to dietary salt supplementation may contribute
ologic and pathologic conditions in which elevated plas- to local elevation in ANP levels and may be functionally
ma levels of ANP99–126 have been detected are summarized signi cant in attenuating the development of salt-sensitive
in Table 8.3. hypertension.244 NPR density is decreased in diabetic rats
The local synthesis of natriuretic peptides is in- with signi cant increase in plasma ANP levels.245 ANP also
creased in the kidney and in the vasculature in obstructive antagonizes AVP-mediated increases in water permeability
uropathy.236 The activation of the renin-angiotensin system in IMCD cells.246
during low sodium intake antagonizes the biologic effect The major sites of action of ANP99–126 are the kidneys,
of ANP by interfering in the intracellular metabolism of adrenal glands, and vascular smooth muscle.247 Short-term
cGMP.237 administration of ANP99–126 in laboratory animals and in hu-
mans induces pronounced natriuresis, diuresis, alteration in
renal hemodynamics and tubular function, suppression of
renin release, inhibition of aldosterone secretion by the adre-
TA B L E
nal glands, and decreased vasomotor tone, resulting in tran-
13.2
8.3 Conditions Associated with sient drop in systemic blood pressure. From these actions it
Increased Levels of Circulating Atrial has been postulated that ANP plays an important physiolog-
Natriuretic Peptide ic role in protecting against extracellular volume overload.248

Physiologic Pathologic Renal Actions of Atrial


Natriuretic Peptide 99–126
Acute volume Congestive heart failure
expansion ANP-induced increase in the GFR is well established.249–251
ANP99–126 increases efferent arteriolar resistance, resulting in
Supine posture Atrial tachycardias increased PGC and ltration fraction.249 In addition, ANP99–126
relaxes mesangial cells in vitro, suggesting that it can increase
Head-out water Myocardial ischemia ltration area by cGMP generation in podocytes252 and Kf in
immersion vivo.253 Indeed, when baseline Kf is low, as in water-deprived
Mineralocorticoid Acute and chronic renal failure animals, ANP99–126 enhances GFR mainly by increasing Kf.253
escape If preexisting vascular constriction is present in isolated per-
fused kidney, ANP tends to vasodilate renal vessels and in-
Exercise Postobstructive diuresis crease renal blood ow (RBF).254 In whole animals, however,
ANP99–126 infusion causes either a decline or no change in
Neonatal period Nephrotic syndrome (subset) RBF.254 The effects of ANP99–126 on RBF are in uenced by its
Cirrhosis with ascites systemic actions on blood pressure and the renin-angiotensin
Severe hypertension system. ANP seems to increase NO production at both renal
Primary hyperaldosteronism and cardiac levels, further explaining its natriuretic and di-
Hypoxia uretic effects.255 Finally, ANP has been reported to induce
Other (SIADH myxedema) redistribution of blood ow from the cortex to the medulla
and to increase vasa recta ow, leading to dissipation of the
SIADH, syndrome of inappropriate antidiuretic hormone secretion. medullary solute gradient.256,257
244
24 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

ANP99–126 has both direct and indirect effects on tu- it is also secreted by cardiac ventricles and, to a lesser extent,
bular transport of Na, chloride, and water.258 In the proxi- from atria. The biologic effects of BNP infusion are simi-
mal tubule, ANP99–126 antagonizes Ang II-induced Na re- lar to those of ANP99–126. Unlike ANP, BNP secretion seems
absorption 259 and, along with endothelin-3, inhibits the to be constitutive and unrelated to myocyte stretch. The
sodium-glucose transporter260 and, like urodilatin, inhibits kidney-speci c degradation of ANP provides a mechanism
Na -ATPase activity.261 In the IMCD, it directly inhibits Na for preferential regulation of kidney function by BNP, inde-
transport by binding to ANP-R1 receptors and in uencing pendent of peripheral ANP concentration.270 In 1990, an-
amiloride-sensitive Na channels and the activity of apical other homologous peptide, CNP, was isolated from porcine
Na-K-2Cl cotransporters.262 Other mechanisms by which brain.271 CNP is produced in the brain, where it achieves
ANP99–126 induces natriuresis and diuresis include suppres- concentrations much higher than those of ANP and BNP. In
sion of renin and aldosterone release,263 inhibition of the contrast, circulating levels of CNP are lower. CNP lacks natri-
tubular actions of AVP,246 and dissipation of the medullary uretic, diuretic, and hypotensive effects and probably acts in a
solute gradient by impairing the increase in intracellular paracrine fashion in the CNS. The physiologic signi cance of
Ca2 concentration. ANP blocks both the stimulatory and BNP and CNP remains unclear.
inhibitory effects of AVP on Na -dependent pHi recovery.264
Clinical Use of Atrial Natriuretic Peptide and
Atrial Natriuretic Peptide Transgenic Mice Atrial Natriuretic Peptide Analogs in the
Transgenic mice over-express pro-ANP in hepatocytes. These Diagnosis and Treatment of Kidney Injury
transgenic animals have a hypotensive phenotype (20 to 30 and Congestive Heart Failure
mm Hg lower than control littermates) without compensato-
ry tachycardia. GFR remained normal despite hypotension. Measurement of circulating ANP and ANP analogs is now a
Moreover, signi cant diuresis or natriuresis during steady well established marker in assessment of volume overload
state was not detected. Contrary to observations made after and left ventricular dysfunction and for predicting mortal-
short-term infusion of ANP99–126, plasma renin activity also ity, in the presence or absence of renal dysfunction.272–276
did not change while aldosterone levels were elevated.265–267 A number of studies, reviewed recently,277 provide sup-
port for the use of ANP/ANP analogs in protection against
Physiologic Consequences of Interrupting and treatment of acute kidney injury (AKI).278–281 In these
studies, low (but not high)-dose ANP infusions reduced the
the Atrial Natriuretic Peptide Pathway need for renal replacement therapy and hospital and inten-
Disruption of the gene that encodes pro-ANP results in sive care unit length of stay, but did not appear to improve
knockout mice that lack expression of ANP. Homozygous mortality in the management of cardiac surgery–associated
ANP knockout mice fed a standard diet have both mildly AKI. Hyporesponsiveness to ANP in congestive heart failure
elevated blood pressure (average increase of 8 mm Hg) and may be due to reduced NPR-A expression.282 In dogs, maxi-
cardiac hypertrophy compared to wild type mice, suggesting mizing cGMP action with type V phosphodiesterase inhibi-
that ANP has a physiologic role in maintaining the normoten- tors augments the natriuretic responses to exogenous ANP.283
sive state. This hypertension appears to be sensitive to dietary Animal studies have shown the usefulness of encapsulated
salt intake as feeding the homozygous ANP knockout mice ANP gene transfected cells as a new tool for ANP gene deliv-
a diet with an intermediate salt content (2%) would further ery with possible implication for future therapy.284
increase blood pressure by an average of 20 mm Hg com-
pared to wild type mice.268 Knockout mice lacking ANP-R1
activity (known as GC-A null mice) have both elevated blood URODILATIN OR RENAL
pressure and cardiac hypertrophy, similar to pro-ANP knock- NATRIURETIC PEPTIDE
out mice, but the GC-A null mice have a salt-insensitive form Urodilatin is best described as a paracrine renal natriuretic
of hypertension.267 It is unclear why there should be a differ- peptide (RNP).285 It was rst isolated from human urine in
ence in the phenotype of these two types of knockout mice. 1988.286 Its amino acid sequence is identical to ANP99–126, ex-
It is possible that other guanylyl cyclase receptors, such as cept for an additional four amino acids at the amino terminal.
guanylyl cyclase C, can help regulate blood pressure in the Despite its high degree of homology to ANP99–126, speci c an-
face of changes in dietary salt intake and compensate for the tihuman RNP polyclonal antibody has been generated and
lack of GC-A receptors.268 ANP ( / ) mice have a blunted RNP levels can be measured by radioimmunoassay.287 To
pressure-natriuresis response and suppressed expression of date, RNP has not been detected in the circulation and the
local renal renin-angiotensin system.269 kidney is presumed to be its site of synthesis and action.285
RNP binds to ANP receptors in the kidney and stimu-
Atrial Natriuretic lates cGMP production.288 Its renal actions parallel those of
Peptide-Related Peptides 149,212 ANP99–126 and include more potent hyper ltration, diuresis,
BNP is a 32-amino acid peptide with structural homology to and natriuresis.288 RNP, like ANP99–126, inhibits sodium up-
ANP99–126. Although originally isolated from porcine brain,270 take by inner medullary duct cells by inhibiting entry of Na
CHAPTER 8 HORMONES AND THE KIDNEY 2245
45

through apical sodium channels.289 It appears that the na- limb of Henle’s loop and salivary and sweat glands. All MR-
triuretic effect of RNP is more potent than that of ANP, pos- expressing tissues also express GR. In the kidney, evidence
sibly because it is resistant to degradation by renal cortical exists for distribution of the GR receptors along the whole
metalloendopeptidase.290 Systemic infusion of RNP results parts of the nephron, except for the proximal tubule. No evi-
in effects similar to those of ANP99–126. dence exists for a glucocorticoid role in the colon.303–305
Several physiologic studies suggest that RNP functions MR has the same af nity for both aldosterone and glu-
as a paracrine hormone that regulates renal Na excretion. cocorticoids. Glucocorticoids concentration in plasma is
Drummer and coworkers291 demonstrated in the human that 100- to 1,000-fold higher than aldosterone and only 10%
urinary excretion of RNP, but not plasma ANP concentration, of it circulates as free, whereas all circulating aldosterone is
correlates with circadian variation in sodium excretion over free. MR selectivity exists to prevent complete occupancy of
a 9-day period. Moreover, acute infusion of normal saline in the MR receptor by glucocorticoids at physiologic concentra-
healthy subjects, balloon dilatation of left atrium, and water tions. This is mainly mediated by the colocalization of the
immersion induce a signi cant increase in urinary RNP.292,293 11 -hydroxysteroid dehydrogenase 2 (11 -HSD2) enzyme
in the distal nephron, along with MR. This enzyme trans-
forms glucocorticoids (cortisol in humans) into metabolites
GUANYLIN AND UROGUANYLIN (cortisone) that have weak af nity to MR.301,305–307 11 -HSD
Guanylin and uroguanylin are cGMP-regulating agonists has two main isoforms. 11 -HSD1 acts predominantly as a
isolated in 1994, respectively, from rat intestine and hu- reductase in vivo in many tissues, regenerating biologically
man/opossum urine that appear to have natriuretic proper- active glucocorticoids (mainly cortisol in human and cor-
ties.294,295 In humans and mice, earlier studies reported that ticosterone in rodents) from their inactive forms (cortisone
both genes for guanylin and uroguanylin are located close in human and 11-dehydrocorticosterone in rodents). 11 -
to each other on chromosomes 1 and 4, respectively, near HSD2, on the contrary, is a dehydrogenase that inactivates
the ANP A and B genes and probably arising from an ances- glucocorticoids.308 The major role of 11 -HSD2 is highlight-
tral uroguanylin/guanylinlike gene.296 Preproguanylin and ed in clinical situations, such as the syndrome of apparent
preprouroguanylin probably derived from a common pre- mineralocorticoid excess where it is inactivated or after in-
cursor gene, as they share approximately 35% homology.297 gestion of excessive amounts of licorice where glycyrrhetinic
Bioactive uroguanylin can be found in the urine at higher acid, a derivative of licorice, has been described to inhibit
concentrations than guanylin, suggesting that uroguany- 11 -HSD2.309,310 Both of these conditions lead to hypokale-
lin may be a hormonal link between the intestine and the mic hypertension with low renin and aldosterone levels. In
kidney.298 Uroguanylin, prouroguanylin, and proguanylin cirrhosis also, there is MR activation by cortisol explained by
peptides have been shown to circulate in the plasma of hu- a reduced activity of 11 -HSD2, which allows promiscuous
mans and other animals.299 activation of MR by the glucocorticoid cortisol as suggested
by Frey.311
GRs have approximately equal af nities for aldosterone
CORTICOSTEROIDS and endogenous glucocorticoids, but have the highest af ni-
Corticosteroids are steroid hormones synthesized by the ty for dexamethasone, a synthetic glucocorticoid.301 Because
adrenal cortex. On the basis of their physiologic functions, mineralocorticoids circulate at much lower concentrations
corticosteroids are traditionally divided into two groups— than glucocorticoids, signi cant binding of mineralocorti-
glucocorticoids (cortisone and cortisol) and mineralocorti- coids to GR does not occur under physiologic conditions.
coids (aldosterone)300—based on their potency in electrolyte
and metabolism regulation. Nongenomic Actions of Aldosterone
Corticosteroids bind to intracellular receptors: the min- The effects of aldosterone on its target cells have long been
eralocorticoid receptor (MR) and the glucocorticoid receptor considered to be mediated exclusively through the genomic
(GR). They are both part of the nuclear receptor family that pathway and were characterized by a 45-minute lag period;
also includes receptors for steroid and thyroid hormones, however, evidence has been provided for rapid effects of the
vitamin D3, and retinoic acids. These receptors translocate hormone that may involve nongenomic mechanisms.5–7,19,32
to the nucleus after ligand binding301 and regulate nuclear On the other hand, in a series of in vitro studies, aldoste-
gene transcription at speci c DNA response elements located rone was shown to have a half maximal effect on both rapid
in the ve regulatory regions near the promoters of speci c (15 minutes) and delayed (120 minutes) Na ux.4,32 The Na/H
target genes (Fig. 8.4).302 Both glucocorticoids and miner- exchanger has been identi ed as a target for nongenomic regu-
alocorticoids modulate renal function. Aldosterone-sensitive lation. Aldosterone rapidly increases Na/H exchanger activity
tissues include the distal parts of the nephron (distal tubule, in a variety of cells, including distal colon and renal epithe-
connecting tubule, and all along the collecting duct), the sur- lial cell lines,3 but rapidly inhibits apical NHE3 and HCO3
face epithelium of the distal colon (where it increases sodi- reabsorption in medullary thick ascending limb (MTAL) and
um absorption and potassium excretion), and other speci c has a dose-dependent biphasic effect on the Na/H exchanger
nuclear-binding sites for aldosterone in the thick ascending (low doses stimulate and high doses inhibit it).312
246
24 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 8.4 The intracellular concentrations of steroid molecules available for binding to glucocorticoid receptor (GR) or
mineralocorticoid receptor (MR) depend on the free extracellular concentrations available for diffusion into the cytoplasm and
the intracellular prereceptor control mechanism constituted by the 11 -hydroxysteroid dehydrogenase type 1 and 2 (11 -HSD1,
11 -HSD2) enzymes. Whereas 11 - HSD1 acts predominantly as a reductase and converts the 11-ketosteroid cortisone with virtually
no af nity for MR and GR into the 11 -hydroxyglucocorticoid cortisol with a high af nity for both GR29,58 and MR, 11 -HSD220
is exclusively an oxidase and inactivates cortisol into cortisone, which allows protection of MR-expressing cells from promiscuous
activation of MR by the glucocorticoid hormone cortisol. GRE, glucocorticoid-response element; MRE, mineralocorticoid-response
element. (Frey FJ, Odermatt A, Frey BM. Glucocorticoid-mediated mineralocorticoid receptor activation and hypertension. Curr Opin
Nephrol Hypertens. 2004;13:451–458.)

Aldosterone acts through nongenomic pathways to Rapid nongenomic aldosterone effects are characterized
regulate many different ion transport proteins and signaling by their rapid onset of action (within minutes) and an insen-
pathways in a variety of renal epithelial cells, such as proxi- sitivity to inhibitors of transcription (e.g., actinomycin D)
mal tubule cells derived from human renal cortex, MDCK- and of protein synthesis. Aldosterone also acts via rapid
C11 cells (a cell line that exhibits properties of collecting nongenomic effects in vivo in humans at the renal vascu-
duct intercalated cells), and principal cells isolated from lature. Antagonizing the endothelial NO synthase unmasks
rabbit cortical collecting duct and both M-1 and RCCD2 these effects. Therefore, rapid nongenomic aldosterone ef-
cortical collecting duct cell lines.6 Studies using isolated per- fects increase renal vascular resistance and thereby mediate
fused tubules also demonstrate that aldosterone, via nonge- arterial hypertension if endothelial dysfunction is present.314
nomic mechanisms, regulates the transepithelial transport Evidence suggests that there is a nonclassical membrane-
function of different nephron segments, such as proximal bound aldosterone receptor.315 These data come from kinet-
S3 segment,21 renal MTAL, type A intercalated cells of outer ic studies that demonstrate saturable, radiolabeled binding
medullary collecting ducts, and principal cells in the con- of aldosterone to cell surface membranes that have kinet-
necting tubule and inner medullary collecting ducts.6,313 In ics compatible with physiologic activity.316,317 In some cell
renal epithelial cells, an elevation in cytosolic Ca2 serves as lines, aldosterone can have very rapid physiologic effects
a second messenger for the nongenomic Na /H exchanger that are not blocked by inhibitors of cell transcription and
activation initiated by aldosterone.2 The existence of a non- translation. For instance, in human mononuclear leukocytes,
genomic action of aldosterone is in general attributed to con- aldosterone can stimulate release of IP3 or calcium within
ditions where this action is observed over a short period of 30 seconds of exposure.318,319 These membrane-bound re-
time (minutes) against the much longer time (hours, days) ceptors may explain some of the effects of aldosterone that
needed for a genomic action.313 occur prior to gene transcription, such as early stimulation of
CHAPTER 8 HORMONES AND THE KIDNEY 2247
47

sodium reabsorption320 or early stimulation of salt intake,321 model demonstrated a rapid induction of SGK-1 mRNA
possibly via phospholipase C/PKC signaling pathways. within 30 minutes of treatment.13,14 However, SGK-1 null
mice only show mild abnormalities in sodium homeosta-
Mineralocorticoid Actions in the Kidney sis, suggesting that other genomic targets are important for
overall regulation of sodium transport.15 Wong et al. dem-
Aldosterone originates primarily from the zona glomerulosa
onstrated that ET-1 is a direct aldosterone gene target in the
of the adrenal glands. Some recent studies have suggested
kidney and colon in Sprague dawly rats and may play an
that the heart, vasculature, and brain can also synthesize
important role in aldosterone-regulated ion homeostasis.323
aldosterone in response to local tissue injury, although ex-
Aldosterone also enhances Na reabsorption by increas-
traadrenal synthesis in humans is still debated.6–8,11,17,322 It is
ing Na–K-ATPase activity in basolateral membranes of prin-
the chief mineralocorticoid of the body. Its physiologic role
cipal cells in mammalian collecting duct and distal tubule.327
as a regulator of sodium and volume balance also allows al-
Studies in toad bladder and mammalian nephron suggest
dosterone the potential to play a pathologic role in the devel-
that aldosterone upregulates Na–K-ATPase activity by at
opment of hypertension in patients with renal disease.322 It
least three mechanisms: increased Na in ux due to open-
is also implicated in the pathophysiology of cardiac brosis
ing of amiloride-sensitive Na channels, induction of Na–K-
and cardiac hypertrophy in end-stage heart failure.3,4,323
ATPase subunit expression at the gene level, and induction
The major action of aldosterone in the kidney is regu-
of intracellular alkalosis, which occurs in tissues that contain
lation of Na, K, and H handling by the distal part of the
aldosterone-sensitive Na/H exchangers.331 Other hormones
nephron. Mineralocorticoid de ciency is associated with vol-
also modulate aldosterone’s action on Na transport; for ex-
ume depletion, hyperkalemia, and mild metabolic acidosis.
ample, ANP is inhibitory and vasopressin is stimulatory
Conversely, mineralocorticoid excess leads to Na retention,
(Table 8.3).332
hypokalemia, and metabolic alkalosis.
Hypersecretion of endogenous mineralocorticoids or the
Ang II, high serum K levels, and ACTH stimulate al-
administration of mineralocorticoids lead to transient sodium
dosterone secretion from the adrenal gland.324 ANP and do-
retention followed by a return to Na balance within a few
pamine, on the other hand, suppress aldosterone secretion.
days.333 The return to Na balance despite elevation of circu-
Dietary sodium also modulates aldosterone release through
lating mineralocorticoid levels is referred to as aldosterone
its effects on the renin-angiotensin system. In addition to
or mineralocorticoid “escape.” During mineralocorticoid
sodium and volume homeostasis, other triggers for aldoste-
escape, increased Na reabsorption by the distal tubule and
rone release have been cited, including hyperglycemia, adre-
collecting duct remains unchanged, but is offset by decreased
nocorticotropic hormone (ACTH), and, more importantly in
Na reabsorption in other nephron segments.332,334 The lat-
patients with CKD, angiotensin II and potassium.49
ter results from increased renal arterial pressure and elevated
plasma ANP levels, both of which suppress proximal tubule
Sodium Reabsorption transport of Na. Mineralocorticoid escape is also, in part, me-
One of the best-documented functions of aldosterone is its diated by decreased Na and water reabsorption in the loop
ability to increase Na reabsorption in the distal tubule and of Henle.334 Other factors, such as TGF- and interleukin-1
collecting duct.325–327 The rate-limiting step to sodium reab- (IL-1), may play a role in regulation of mineralocorticoid es-
sorption across tight epithelia is the permeability of the api- cape. These factors have been recently found to inhibit the
cal membrane of the transporting cell. Aldosterone increases action of aldosterone on the cells of the IMCD.335
apical Na permeability of tight epithelia, such as those found Ang II binds to Ang II type 1 (AT-1) receptors in the
in the mammalian distal tubule and descending colon, by kidney, which leads to glomerular hypertension, sclerosis,
increasing the activity of the amiloride-sensitive epithelial renal brosis, and cardiac remodeling, possibly through a
sodium channel (ENaC). ENaC is formed by three subunits TGF- -mediated pathway.30–32 ACE inhibitors provide reno-
(alpha, beta, and gamma) and, based on coexpression studies protection by inhibiting the conversion of Ang I to Ang II
in Xenopus oocytes, these subunits assemble into a complex and precluding the negative effects of AT-1 receptor activa-
heterooligomer forming the amiloride-sensitive pore.328–330 tion. Studies have shown that Ang II can be generated by
Other characterized aldosterone-induced targets in- ACE-independent pathways such as chymase in the heart,33
clude the serum and glucocorticoid-regulated kinase-1 which leads to “angiotensin escape.” Angiotensin escape is
(SGK-1), an important mediator of renal sodium homeosta- one of the factors frequently cited to explain aldosterone
sis; corticosteroid hormone-induced factor (CHIF), which breakthrough, which may be secondary to the genera-
regulates the activity of the sodium and potassium-depen- tion of non-ACE mediated angiotensin II.34 Potassium and
dent adenosine triphosphatase pump (Na-KATPase); the adipocyte-released factors may also contribute to the phe-
glucocorticoid-induced leucine zipper protein; a transcrip- nomenon of aldosterone breakthrough. Continual dietary
tion factor; and the G protein K-Ras2.10–12 SGK-1 is thought intake of potassium in the setting of reduced GFR may
to regulate Na ux by increasing ENaC activity at the api- promote elevated potassium levels that subsequently trig-
cal surface of epithelial cells. Aldosterone treatment of a rat ger aldosterone secretion, as seen in the rat remnant kidney
collecting duct cell line as well as an adrenalectomized rat model.28 ACE inhibitors and ARBs are also known to reduce
248
24 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

potassium excretion, so prolonged use of such agents may independent of Na ux.327,339 Physiologically, it is dif cult
enhance potassium retention and predispose a patient to to understand how one hormone can regulate the concen-
aldosterone breakthrough. Recent studies in rat models of tration of two different solutes that have varying levels of
metabolic syndrome with early nephropathy have shown en- dietary intake. Patch-clamp experiments have demonstrated
hanced aldosterone secretion due to adipocyte activity that that other nonaldosterone circulating factors exist that can
was not abolished by candesartan administration.35 These regulate K channel activity. Infusion of aldosterone by os-
results suggest that adipocyte-released factors outside of Ang motic minipump will increase the density of EnaC, but not
II may enhance aldosterone secretion and lead to increased of K channels.339 However, an increase in dietary K does
proteinuria and podocyte injury in rats.35 Thus, potassium, increase the K-channel density.340 Currently, it is unknown
angiotensin II, and adipocyte-released factors may all con- what other circulating factor controls K secretion.
tribute to the increase in aldosterone secretion in patients A large body of evidence suggests that SGK1 mediates,
on prolonged ACE inhibitors or ARB therapy. The exact at least in part, the effect of aldosterone on renal K secre-
de nition of aldosterone breakthrough has been a subject of tion.41,88,113,116 This notion is supported by studies per-
controversy, as there is no current consensus on its precise formed in SGK1 knockout mice demonstrating that the phe-
de nition. One of the common de nitions is a rise in plasma notype of SGK1 deletion is similar to MR knockout mice
aldosterone concentration, often past baseline values, fol- and displays impaired renal K secretion in response to high
lowing an initial decrease after the initiation of ACE inhibi- dietary K intake.41,341
tor or ARB therapy.322
Renal Acidi cation
Potassium Secretion The role of mineralocorticoids in regulation of renal acidi ca-
tion is supported by several clinical observations. Syndromes
Mineralocorticoids are the predominant hormonal in u-
of aldosterone de ciency are associated with metabolic acido-
ence on K secretion by principal cells of the collecting duct
sis because of reduced urinary acid excretion, whereas miner-
and connecting segment of the distal tubule.335,336 Although
alocorticoid excess results in metabolic alkalosis. Aldosterone
mineralocorticoids always increase K secretion by these
enhances urinary acidi cation through direct actions on epi-
nephron segments, this does not necessarily translate into a
thelial cells in the collecting duct and indirectly by in uenc-
kaliuresis because of the strong dependence of K excretion
ing various intrarenal and extrarenal factors.342 Aldosterone
on distal Na delivery and urinary ow rate.337 For example,
increases H secretion by type A intercalated cells in the col-
in conditions of decreased Na delivery and urinary ow to
lecting duct via two mechanisms: direct stimulation of the
the distal nephron, the kaliuretic effect of aldosterone is ei-
proton pump (H-translocating ATPase), and indirectly by
ther diminished or abolished. The mechanisms by which
stimulating Na in ux, which creates a lumen-negative poten-
aldosterone stimulates K secretion by principal cells overlap
tial difference.342 As with K, the overall effect of aldosterone
with those responsible for its Na-retaining action. The late
on renal acid excretion depends on Na delivery to the distal
distal convoluted and connecting tubules (CNTs) and corti-
part of the nephron. Reduction of Na transport in the col-
cal collecting duct (CCD) of the distal nephron mediate, in
lecting duct, because of either decreased Na delivery or in-
large part, the nal regulation of urinary K excretion.19 The
hibition of distal Na reabsorption by amiloride, signi cantly
traditional model by which K secretion is accomplished in
attenuates the effect of aldosterone on net H excretion.343
these segments can be summarized as follows. Na enters the
Aldosterone regulates the expression of aquaporin, AQP3,
CNT and principal cell from the urinary uid through the
with no effect on AQP1 and AQP2, in the collecting duct,
apical amiloride-sensitive ENaC and is then transported out
independently of Na intake.344
of the cell at the basolateral membrane in exchange for up-
take of K via the basolateral Na -K -ATPase. The high K
concentration within the cell and lumen-negative voltage, es- Effect on In ammation
tablished by electrogenic Na reabsorption, create a favorable Besides its classical effects on salt homeostasis in renal epi-
electrochemical gradient for K to diffuse into the urinary thelial cells, aldosterone promotes in ammation and bro-
space through apical K -selective channels. Thus vectorial K sis and modulates cell proliferation. As outlined by recent
secretion in these segments requires a favorable electrochemi- reports, mineralocorticoid also mediates in ammation
cal gradient and an apical permeability to K increases in and brosis through NF- B activation in liver, heart, and
extracellular K concentration directly stimulate aldosterone glomerular mesangial cells9–13 via a pathway involving the
production in zona glomerulosa cells of adrenal glands.6,59,338 aldosterone early-induced gene, serum, and glucocorticoid-
Aldosterone-induced Na in ux through the apical induced kinase 1 (SGK1).11,12,345
membrane leads to the generation of a lumen-negative po- Studies in vitro revealed that aldosterone could induce
tential difference that favors K secretion.326,327 In addition, proliferation of rat glomerular mesangial cells and promote
although mineralocorticoids do not increase the density of collagen gene expression and synthesis via activation of ex-
active K channels in the apical membrane, they increase tracellular signal-regulated kinase 1 and 2 (ERK1/2) mitogen-
the conductance of apical and basolateral K channels activated protein (MAP) kinase in renal broblast. Aldosterone
CHAPTER 8 HORMONES AND THE KIDNEY 2249
49

treatment of renal tubular epithelial cells increases calcium with higher levels of proteinuria in patients with elevated
in ow and intracellular cyclic adenosine monophosphate lev- urinary aldosterone levels.53 This suggests that aldosterone
els. The results suggest that aldosterone plays a pivotal role may work in concert with dietary salt to accelerate kidney
in tubulointerstitial brosis by promoting tubular epithelial– injury.322 Studies designed to delineate the factors respon-
mesenchymal transition and collagen synthesis in proximal sible for the renal injury associated with aldosterone have
tubular cells. The process is MR-dependent, and mediated by also been performed in humans. Sato and Saruta measured
ERK1/2 mitogen-activated protein kinase pathway.346 the urinary excretion of collagen type IV (a turnover product
Clinical trials have shown a potential role for MR of brosis) by patients with diabetes and found that those
blockers to further delay the development of end-stage re- on spironolactone showed a signi cant decrease in urinary
nal disease by completing renin–angiotensin blockade. MR collagen excretion, which suggests that collagen may be
blockade produces a signi cant antiproteinuric effect and one factor associated with aldosterone-mediated brosis.57
has minimal risk of causing hyperkalemia if the condition of Because aldosterone is a major regulator of sodium and vol-
the patient is closely monitored. As well as in the collecting ume balance, it could also, theoretically, mediate its negative
ducts, mineralocorticoid receptors are distributed through- effects by increasing blood pressure.322
out the body, including the proximal tubule, thick ascending
segment of the nephron, the heart, and the brain.14,20,21,31 Glucocorticoid Actions in the Kidney
Aldosterone is thought to mediate brosis by activating fac- Glucocorticoids appear to be important for the normal
tors such as reactive oxygen species; TGF- ; and increasing maintenance of GFR.347 In both adrenalectomized animals
collagen synthesis, the reduced form of nicotinamide ade- and in humans with adrenal insuf ciency, GFR is reduced
nine dinucleotide phosphate (NADPH) oxidase, and plas- compared to controls. Adrenalectomized rats given physi-
minogen activating factor I (PAI-1).7,20,22,23 The brotic effect ologic doses of glucocorticoids regain a normal GFR.348
of aldosterone is often enhanced by high dietary salt intake.24 Furthermore, short-term administration of pharmacologic
Fibrosis and kidney damage induced in uninephrectomized doses of glucocorticoids has been reported to increase inulin
rats placed on a high-salt diet and treated with angiotensin clearance in both normal animals and humans.349,350 Micro-
II were reduced by treatment with spironolactone and an puncture studies in normal rats indicate that glucocorticoids
aldosterone synthase inhibitor, FAD286.32,322 enhance GFR by increasing glomerular plasma ow.347,351
Quan et al. studied chronic kidney disease in rats The latter results from selective vasodilation of both afferent
subjected to subtotal nephrectomy.37 Rats with intact ad- and efferent renal arterioles.351 The mechanisms by which
renal glands were found to have increased proteinuria, glucocorticoids alter the glomerular microcirculation remain
renal histopathologic changes, and decreased inulin clear- obscure. Because amino acid infusion causes similar glomer-
ance when compared with rats subjected to adrenalectomy. ular hemodynamic changes, Baylis and colleagues suggest
Furthermore, the difference in the severity of kidney disease that glucocorticoids may increase GFR through their effects
between rats that had undergone adrenalectomy and those on catabolism of proteins to free amino acids.347
that had not was not abolished with corticosteroid admin- Glucocorticoid actions on the proximal tubule in-
istration, which suggested that some other factor from the clude enhancement of gluconeogenesis, ammoniagenesis,
adrenal gland was responsible for the renal defects.322 and Na reabsorption.352 Lag in ammonium excretion re-
Experiments conducted on saline drinking, stroke sulting in acid retention is well described in subjects in a
prone, spontaneously hypertensive (SHRSP) rats have shown glucocorticoid-depleted state.353 In adrenalectomized ani-
that inhibition of mineralocorticoid receptor activation may mals, glucocorticoid replacement restores proximal tubule
delay renal disease. Spironolactone, a mineralocorticoid re- ammoniagenesis and the ability of the kidney to respond
ceptor blocker, implanted into SHRSP rats results in a re- to the chronic phase of acidosis.354 Furthermore, in whole
duction in proteinuria. The expression of TGF- , NADPH animals, glucocorticoid excess accelerates renal base gen-
oxidase, and collagen I/IV was increased in the untreated eration, resulting in metabolic alkalosis.355 Glucocorticoids
diabetic rats, but was reduced in spironolactone-treated rats. regulate ammoniagenesis possibly through altering gluta-
Aldosterone was found to increase collagen expression in rat mine uptake and metabolism by proximal tubule cells.355
renal broblasts, which may contribute to brosis.20 Studies Glucocorticoids increase proximal tubule Na reabsorption
of humans have also shown that the deleterious effects of by at least two mechanisms: enhanced Na–K-ATPase activity
aldosterone are enhanced by high dietary salt intake, which and Na–H exchange.356,357 Several experiments suggest that
is typical of the Western diet. A study involving 2,700 par- glucocorticoids inhibit Na-dependent phosphate and sulfate
ticipants in the Framingham Offspring Study has shown that reabsorption in the proximal tubule.352 These observations
greater excretion of urinary sodium (a marker for dietary salt are supported by clinical reports of phosphaturia and lower
intake) correlates strongly with elevated urinary albumin- serum phosphate levels in patients with Cushing disease and
uria and weakly (albeit nonlinearly) with serum aldosterone subjects given high doses of glucocorticoids.358
levels.52 In a subsequent study conducted on patients with Both patients with Addison disease and adrenalecto-
resistant hypertension despite the use of three antihyper- mized animals have decreased urinary concentrating abili-
tensive medications, elevated dietary salt intake correlated ty,359 due in part to reduction in RBF, GFR, and hydroosmotic
250
25 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

permeability of the collecting tubule.360 In addition, adre- is one of the major long-term regulators of blood pressure
nal corticosteroids contribute to urinary concentration by (BP). This is achieved by the interplay of several hormonal,
stimulating Na, K, and HCO3 transport in the thick ascend- neural, and humoral factors that would regulate principally
ing limb of Henle’s loop.361 It is not entirely clear whether two parameters: sodium homeostasis and peripheral vascular
glucocorticoids, mineralocorticoids, or both mediate the ef- resistance.368 In the following section, we discuss the renal
fects of corticosteroids on renal-concentrating mechanisms. functions of catecholamines separately on their receptors.
Systemic excess of glucocorticoids is known to cause hy-
pertension. The effect of glucocorticoids may be in part me- Alpha-adrenergic Stimulation in the Kidney
diated by the suppression of endothelial nitric oxide synthase
Both 1- and 2-adrenergic receptors have been localized
(eNOS).2 Acute administration of glucocorticoids, however,
to vascular smooth muscle and tubule cells of the neph-
may have bene cial effects on the cardiovascular system in
ron. Norepinephrine is the major agonist at these levels and
part through nontranscriptional activation of eNOS.308
adrenergic stimulation causes renal arteriolar vasoconstric-
tion (increased afferent and efferent arteriolar resistance) by
CATECHOLAMINES activating mainly 1 receptors on vascular smooth muscle
cells.369 This 1-mediated renal vasoconstriction results in
Structure and Biosynthesis of decreased RBF and GFR. Recent evidence suggested that
Catecholamines norepinephrine increases afferent arteriolar sensitivity to an-
Norepinephrine (noradrenaline), epinephrine (adrenaline), giotensin II through activation of receptors and secondary
and dopamine are collectively called catecholamines. These increase in calcium sensitivity of mouse afferent arteriole.370
endogenous amines are derived from the amino acid tyrosine In the proximal convoluted tubule, where 1- and 2-
and the enzymes involved in their biosynthesis have been adrenergic receptors are expressed in high density, norepi-
identi ed, cloned, and characterized362 and include: tyrosine nephrine increases Na and water reabsorption, in part, by
hydroxylase, dopa decarboxylase, dopamine -hydroxylase, stimulation of Na -K -ATPase activity.371–373 This activ-
and phenylethanolamine-N-methyltransferase. The rst step ity is partially dependent on the cosecretion of neuropep-
in catecholamine synthesis involves the hydroxylation of ty- tide Y that acts to synergize the stimulatory -adrenergic
rosine by tyrosine hydroxylase, which is regarded as a rate- effects of norepinephrine and to antagonize the inhibitory
limiting step. This enzyme is activated by stimulation of -adrenergic effects. This is demonstrated by the fact that
sympathetic nerves or the adrenal medulla and is subject to norepinephrine alone does not affect Na -K -ATPase activ-
feedback inhibition by catechol compounds. Catecholamines ity in the proximal convoluted tubule unless neuropeptide
are synthesized in nerve terminals of the postsynaptic neurons Y or other -adrenergic inhibitors are present.374 In isolated
of the sympathetic nervous system and in the adrenal me- rat and rabbit proximal convoluted tubule cells, 1 and 2
dulla. Most of the steps occur in the cytoplasm whereas some agonists stimulate Na –H exchange, the overall effect of
take place in storage vesicles.363 Storage of catecholamines in which is enhanced Na and uid absorption.375,376
vesicles ensures their regulated release, protects them from In the loop of Henle, experimental evidence showed
enzymatic degradation, and prevents their leakage outside the that -adrenergic stimulation increases sodium and wa-
cell. Termination of action of norepinephrine and epineph- ter reabsorption at the thick ascending limb level through
377,378
rine includes reuptake by nerve terminals, reuptake by non- 1- and 2-mediated activation. In the collecting duct,
neuronal cells, and metabolic transformation. Major enzymes Krothapalli and Suki379 report that 2 agonists inhibit va-
involved in catecholamine metabolism include monoamine sopressin-stimulated water reabsorption by inhibiting ad-
oxidase (MAO) and catechol-O-methyl transferase (COMT). enylate cyclase activity.379 Other investigators, however,
The main source of epinephrine in the body derives from the challenge this observation.380
adrenal medulla whereas noepinephrine originates mostly
from nerve terminals.363 At the renal level, catecholamines Beta-adrenergic Stimulation in the Kidney
derive from renal efferent nerves (norepinephrine and, to a
-adrenergic receptors have been identi ed in the glomeru-
lesser extent, dopamine), from the circulation (epinephrine
lus, juxtaglomerular apparatus, thick ascending limb of loop
and norepinephrine), from the adrenal medulla (epineph-
of Henle, distal convoluted tubule, and collecting duct.380,381
rine), and from renal proximal tubule cells (dopamine) via 1,
1 stimulation enhances renin release from the juxtaglomer-
2, 1, and 2 receptors for epinephrine and norepinephrine
ular cells of the afferent arterioles. Otherwise, there are few
and D1- and D2-like receptors for dopamine.364–367
receptors in renal vessels. Although receptors have not been
localized to the proximal tubule, physiologic studies suggest
Physiology of Catecholamine that -adrenergic stimulation increases Na and uid trans-
Action in the Kidney port in this nephron segment independently of enhanced re-
Catecholamines play an important role in the regulation of nin secretion and angiotensin II production.380 In the thick
RBF, GFR, renin secretion, and tubular transport. Their ef- ascending limb, -adrenergic receptor activation stimulates
fects depend on site of action and receptor type. The kidney cAMP production and NaCl reabsorption.380 agonists also
CHAPTER 8 HORMONES AND THE KIDNEY 2251
51

increase Cl –HCO3 exchange and H -K -ATPase activity in and Kuchel388 point out that dopamine is the predominant
the collecting duct.371 The latter effect results in enhanced catecholamine in sh, in which salt excretion is a priority.
K reabsorption by type A intercalated cells (and an appar- On the other hand, norepinephrine predominates in terres-
ent decrease in K secretion).371 A potential mechanism of trial animals, in which salt retention is essential for survival.
action involves -adrenergic stimulation of cAMP produc- In addition to inhibition of Na -K -ATPase activity, other
tion and subsequent conversion to adenosine.381 studies suggest that dopamine suppresses Na –phosphate
cotransporter, antagonizes the stimulatory effect of Ang II
Dopamine and the Kidney on Na –H exchange in cortical brush-border membranes
Dopamine is the immediate metabolic precursor of norepi- mediated by cAMP and PKA,389,390 and stimulates renin
nephrine and epinephrine. Locally, dopamine is synthesized synthesis in cultured rat juxtaglomerular cells.391 Studies in
by proximal tubule cells via enzymatic decarboxylation humans have shown that dopamine does not induce natri-
of L-dopa by aromatic amino acid decarboxylase (AADC). uresis in Na -depleted subjects and that its natriuretic effect
L-dopa reaches the tubule cell after ltration and Na -cou- is more pronounced during conditions of volume expan-
pled reabsorption because renal proximal tubule cells lack sion.368,392 This demonstrates again that the natriuretic and
tyrosine hydroxylase. The contribution of presynthesized diuretic effects of D1-like receptors are dependent on sodium
and stored dopamine to local renal physiology is not yet balance.
clearly established.368,382 High salt intake increases AADC In the whole kidney, dopamine increases RBF and GFR
activity and dopamine synthesis in proximal tubule possibly through its D1 receptor-mediated vasodilatory effects.383,392
by enhancing Na -coupled uptake of L-dopa.383,384 Regu- Supraphysiologic concentrations of dopamine, however,
lation of intrarenal dopamine concentrations can occur at stimulate -adrenergic receptors, which lead to vasocon-
several levels: synthesis, storage, or degradation. Dopamine striction and decreased RBF. The natriuretic and vasodilating
is a substrate for both MAO and COMT. Synthesis and me- effects of dopamine have suggested a therapeutic role in
tabolism of dopamine differs between neural and nonneural patients with volume expansion, particularly when admin-
cells. Dopamine synthesized in the proximal tubule does istered in low doses that do not activate adrenergic recep-
not undergo further metabolism to norepinephrine and epi- tors. However, several studies have shown that dopamine
nephrine because of the lack of dopamine -hydroxylase385; has no major role as a therapeutic strategy in the prevention
instead, it diffuses to peritubular space and tubular lumen of further renal damage in acute kidney injury.393,394 More-
where it acts locally on its receptors. over, dysfunction of the renal dopamine system has been
Dopamine receptors are members of the G protein- postulated to contribute to the pathogenesis of systemic
coupled superfamily of heptahelical receptors. At least ve hypertension.368,383 Results from at least two studies suggest
receptors have been identi ed, subclassi ed into D1- and that defects in renal generation of dopamine are common in
D2-like subfamilies based on their molecular structure and patients with essential hypertension.395,396
pharmacology. Both of the cloned members of the D1-like The ability of the D1 receptor to induce both natriure-
receptor group (D1 and D5, also known as D1A and D1B in ro- sis and vasodilation makes D1 agonists, such as fenoldopam,
dents) are coupled with the stimulating G protein, Gs , and potential therapeutic agents for the treatment of both hy-
stimulate adenylyl cyclase. All three of the cloned D2-like pertensive urgencies and acute renal failure. In healthy nor-
receptors (D2, D3, and D4) are associated with the inhibitory motensive volunteers, fenoldopam has been shown to sig-
G protein, Gi/G0, and inhibit adenylyl cyclase. Both families ni cantly increase renal plasma ow while only minimally
of receptors are expressed in the kidney. D1 and probably D5 reducing systemic blood pressure.397 In people with hyper-
are localized in the smooth muscle layer of renal arterioles, tensive urgencies, fenoldopam has been shown to reduce
juxtaglomerular cells, proximal tubules, and cortical collect- systemic blood pressure by 23% while increasing natriuresis
ing duct. The D3 receptor is present in arterioles, glomeruli, by 200%, diuresis by 46%, and renal blood ow by 42%.398
proximal tubules, MTAL of loop of Henle, and the collecting Although the selective increase in renal plasma ow could be
duct. The D4 receptor is localized in the cortical collecting advantageous in the treatment of certain forms of acute renal
duct.386,387 failure, further studies must be done to de ne the speci c
Locally produced dopamine plays a central role in the utility of fenoldopam in this setting.
regulation of sodium excretion. Circulating dopamine con-
centrations are in the picomolar range, hence not suf ciently
high to activate dopamine receptors. The major signaling
THE RENAL KALLIKREIN-KININ
mechanism by which dopamine induces natriuresis is by in- SYSTEM
hibiting Na -K -ATPase in all the segments of the nephron In 1909, Abelous and Bardier reported for the rst time the
via D1-like receptors, whereas D2-like receptors stimulate hypotensive effect of human urine when injected into the
this enzyme. The end result of Na -K -ATPase inhibition is a bloodstream of dogs.399 Further studies by Werle and col-
dopamine-induced natriuresis. Several investigators suggest leagues from 1926 to 1939 attributed these effects to the
that the role of dopamine is to counterbalance the effects of kallikrein-kinin system (KKS) and described its basic com-
antinatriuretic factors in the kidney.383 Interestingly, Kuchel ponents: kallikreins, kinins, kininogens, and kininases. The
252
25 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

major actions of this system are mediated by bradykinin, a LK but is capable of cleaving HK, whereas plasma kallikrein
peptide hormone that exerts potent proin ammatory and cleaves HK exclusively.399 Kininogens are synthesized in the
vasodilatory effects. Interestingly, all components of the KKS liver and circulate in blood plasma at concentrations of 45 to
are also expressed in the kidney, especially in the distal con- 120 g per mL for LK and of 65 to 115 g per mL for HK,
voluted and connecting tubule as well as in the collecting but are also found in other body uids and organs such as
duct, and have been shown to regulate renal hemodynamic kidney.401
and tubular function. In addition, in the last few years, stud- Novel functions of kininogens have been recently dis-
ies have linked the KKS to different pathologic states includ- covered. Derivatives of HK have been shown to be involved
ing the diabetic nephropathy as reviewed here. in the regulation of endothelial cell proliferation, angiogen-
esis, and apoptosis.402 In addition, some seem to possess
Structure and Synthesis of Kinins potent and broad-spectrum microbicidal properties against
both gram-positive and gram-negative bacteria, and thus may
The KKS is a complex multienzyme system that can be divid-
represent an alternative to conventional antibiotic therapy.402
ed into two types: (1) a circulating KKS that belongs to the
coagulation system and (2) a tissue KKS that acts in a para-
crine or autocrine fashion. The KKS leads to the production Physiology of Kallikrein-Kinin System
of kinins, namely bradykinin and lys-bradykinin (kallidin), in the Kidneys
from kininogens through the action of kininogenase, namely Tissue kallikrein is secreted by many cells throughout the
kallikrein (Fig. 8.5).399 Two types of kallikreins have been body but some tissues produce particularly large quanti-
identi ed—a plasma and a tissue kallikrein—both of which ties such as the kidney, lung, intestine, brain, and glandular
are serine proteases that are encoded by different genes and tissues (salivary and sweat glands and pancreatic exocrine
differ in their distribution and regulation.399 Mice lacking gland). This enzyme is activated intracellularly from a pre-
tissue kallikrein show dramatically reduced levels of renal cursor, prokallikrein, to produce tissue kallikrein.403 The
kinin, suggesting that tissue kallikrein is the main system enzyme responsible for this conversion has not yet been
involved in the kidney,400 although little is known regard- identi ed.
ing the putative role of plasma kallikrein under pathologic Regulatory mechanisms of tissue kallikrein remain partly
conditions. Renal tissue kallikrein is synthesized in large unknown. Aprotinin, a polypeptide puri ed from the lung,
amounts by connecting tubule cells and is mainly secreted and kallistatin have been shown to inhibit the activity of re-
into the urinary uid and to a lesser extent to the peritubular nal and other tissue kallikreins.404 In addition, it has been
interstitium.399 shown that salt restriction stimulates the synthesis of renal
In humans, two types of kininogens have so far been de- kallikrein through an unclear mechanism, possibly implicat-
scribed: a high molecular weight (HK) form present in blood ing aldosterone.405 Interestingly, it was also recently shown
and a low molecular weight (LK) form present in various that potassium intake triggers an increase in renal kallikrein
tissues. It is generally accepted that tissue kallikrein prefers secretion through an aldosterone-independent mechanism

FIGURE 8.5 Biosynthesis and


metabolism of kinins. CPM,
carboxypeptidase-M; ACE,
angiotensin I-converting enzyme;
NEP, neprilysin (endopepti-
dase 24.11); ECE, endothelin-
converting enzyme; red, active
peptides; blue, inactive pep-
tides. (Adapted from Kakoki M,
Smithies O. The kallikrein-kinin
system in health and in diseases
of the kidney. Kidney Int.
2009;75(10):1019–1030.)
(See Color Plate.)
CHAPTER 8 HORMONES AND THE KIDNEY 2253
53

involving membrane depolarization of kallikrein-secreting receptor (B1R) and B2 receptor (B2R) (Fig. 8.6). Although
cells in the renal connecting tubules, followed by enhanced B2R is ubiquitous and expressed throughout the kidney, B1R
calcium in ux.406,407 In addition to dietary sodium and is mainly expressed after induction by endotoxin, cytokines,
potassium intake, hereditary factors may determine tissue ischemia, and other noxious stimuli.411 Interestingly, treat-
kallikrein activity. In fact, a loss of function polymorphism ment with an in ammatory signal (lipopolysaccharide) in-
in the human tissue kallikrein gene (R53H) has been identi- duces the expression of B1R mRNA in all renal segments
ed with a frequency of 0.03 in Caucasians.408 Interestingly, except the outer medullary collecting ducts. B1R, once in-
these partially tissue kallikrein-de cient subjects develop a duced by in ammatory mediators and tissue damage, as-
form of arterial dysfunction characterized by remodeling of sumes some of the physiologic roles of B2Rs.413 Bradykinin
the brachial artery, which is not adapted to a chronic in- and kallidin are equipotent and both have higher af nity for
crease in wall shear stress.408 B2R. Interestingly, metabolites (des-Arg9-BK [DABK] and
Although of little relevance for the kidney, it is notewor- Lys-DABK) of bradykinin and kallidin, which result from
thy that plasma kallikrein is regulated through a different, the action of carboxypeptidases, have higher af nity for B1R.
more complex, mechanism. Plasma kallikrein is synthesized Both receptors activate similar intracellular signaling
and secreted by the liver as an inactive zymogen. It is acti- cascades involving phospholipase C activation and intracel-
vated by the intrinsic coagulation cascade whereby contact lular calcium mobilization. In addition, through calcium-
of plasma with negatively charged macromolecular surfaces dependent and -independent mechanisms, KKS increases
initiates a proteolytic cascade that ultimately converts prekal- NO and prostaglandin synthesis, both of which seem to me-
likrein to plasma kallikrein.409 diate some of the effects of kinins.410 Alternative mediators
The half-life of bradykinin in plasma is short ( 30 sec- of kinins also include endothelium-derived hyperpolarizing
onds), suggesting that its actions are regulated locally factor (EDHF), norepinephrine, substance P, cytokines, and
through its production and degradation within tissues. tissue plasminogen activator.410
In fact, both bradykinin and kallidin can be metabolized Experimental evidence suggests that kinins regulate re-
through two pathways.410 Kininase I (also called carboxy- nal blood ow and renal excretion of sodium and water.414,415
peptidase-N), as well as carboxypeptidase-M, remove the C- Studies on the role of the renal KKS, using congenitally ki-
terminal arginine from the kinins to generate their des-Arg ninogen-de cient Brown-Norway Katholiek rats and B2R
derivatives, which are agonists of B1R (see later). Kininase II knockout mice, amongst other models, revealed that this
(also known as ACE), neprilysin (endopeptidase 24.11), and system starts to induce natriuresis and diuresis when sodium
endothelin-converting enzyme cleave off the two C-terminal accumulates in the body as a result of excess sodium intake
amino acids (Phe and Arg) of the kinins, thereby inactivating or aldosterone release, for example, by angiotensin II. Thus,
them.411,412 It is noteworthy that kallidin can be converted it is hypothesized that the system works as a safety valve
into bradykinin by a plasma aminopeptidase. for sodium accumulation. In fact, mice lacking B2R and/or
Kinins exert their biologic effects by acting on two B1R develop salt-sensitive hypertension.416 Interestingly, hu-
types of G protein-coupled receptors called bradykinin B1 mans with essential hypertension have low levels of urinary

FIGURE 8.6 Bradykinin intracellular signaling


cascade. The thickness of arrows arising from
the kinins indicates the relative potency of
each peptide to elevate intracellular calcium
concentrations. PIP2, phosphatidylinositol-4,5-
bisphosphate; PI-PLC, phosphatidylinositol-
speci c phospholipase C; IP3, 1,4,5-inositol
triphosphate; ER, endoplasmic reticulum;
PL, phospholipids; PLA2, phospholipase A2.
(Reproduced from Kakoki M, Smithies O.
The kallikrein-kinin system in health
and in diseases of the kidney. Kidney Int.
2009;75(10):1019–1030.)
254
25 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

kallikrein.405 Conversely, hypertensive mice and rats over- or signaling pathway by which tissue kallikrein controls
expressing the human kallikrein develop hypotension.417 H -K -ATPase remains to be determined.
However, the renal KKS seems to be involved only in the In addition to sodium and potassium, the KKS seems
long-term regulation of blood pressure under conditions to regulate the metabolism of calcium, through a B2R/B1R-
of hypertensive insult such as high salt intake.418 Evidence independent mechanism. Picard et al. showed that knock-
suggests that bradykinin promotes natriuresis, in part, by ing out tissue kallikrein in mice leads to hypercalciuria of
decreasing sodium reabsorption in the distal part of the renal origin.427 In fact, the distribution of tissue kallikrein
renal nephron. Bradykinin has been shown to inhibit the overlaps that of the epithelial calcium channel (TRPV5) in
amiloride-sensitive component of conductive sodium uptake the distal nephron. Furthermore, kallikrein gene expression
in inner medullary collecting duct cells419 and to cause a re- was increased by a low-calcium diet.427 Interestingly, no such
versible inhibition of sodium reabsorption ex vivo in rat cor- abnormalities were observed in B2R knockout mice with or
tical collecting ducts.420 Furthermore, inhibition of B2Rs in without treatment with B1R antagonist. This suggests that
rats fed a normal salt diet decreases fractional sodium excre- the effect of kallikrein on calcium excretion is independent
tion with little effect on medullary and cortical perfusions or of kinin production.
on GFR.421 Recently, Zaika et al. con rmed that bradykinin Furthermore, products of the KKS may modulate cell
inhibits ENaC in the aldosterone-sensitive distal nephron.422 growth. Experimental evidence suggests that while under
It is noteworthy that during volume expansion, a condition certain conditions, in vitro bradykinin may inhibit growth
that requires further sodium reabsorption, at least one ad- of normal renal broblasts.428,429 It can also stimulate the
ditional mechanism is activated: kinins mediate the increase growth of broblasts, mesangial cells, and arterial smooth
in papillary blood ow, thereby indirectly inhibiting further muscle cells in other conditions.430 Over-expression in
tubular electrolyte reabsorption.421 hypertensive Dahl salt-sensitive (DSS) rats of the human
The renal KKS is believed to operate in concert with the kallikrein gene by adenoviral delivery results in reversal of
renin-angiotensin system to physiologically regulate the dis- the changes associated with hypertensive nephrosclerosis.430
tribution of RBF and the excretion of water and electrolytes. Chao et al. further con rmed that kinin exerts its renopro-
It has been con rmed that the effects of kinins antagonize the tective effects against salt-induced renal injury in the DSS
effects of RAAS on blood pressure and sodium handling. In rat model by inhibiting cellular apoptosis, in ammatory cell
fact, reduced KKS activity may contribute to the establish- recruitment, and brosis through suppression of oxidative
ment of a pathophysiologic state characterized by unopposed stress, TGF- expression, and MAPK activation, with no
hyperactivity of the renin-angiotensin system, resulting in salt effect on blood pressure.431 To a large degree, the bene ts of
retention.423 It has been shown that this reduced activity of kinins in that study were dependent on the elevated levels of
the KKS predominates over renin-angiotensin system over- renal NO.431 NO has been shown to play an important role
activity in all conditions of sodium balance in essential and in protection against hypertension and glomerulosclerosis
family-related hypertension.424 KKS and the renin-angiotensin in DSS rats.432 These renoprotective effects of NO may be
system are functionally coupled at the level of ACE. Inhibition attributed to a decrease in oxidative stress. In fact, NO can
of this enzyme not only blocks angiotensin II production but scavenge superoxide anions. It can also inhibit the assembly
also increases bradykinin production. It was shown that the of a functional NADH/NADPH oxidase, thereby attenuating
bene cial effects of ACE inhibitors were mainly due to NO the production of superoxide.433 In addition, given that oxi-
production following B2R activation.425 Interestingly, chronic dative stress can stimulate the expression of proin ammato-
treatment with ACE inhibitors induces B1R expression in vas- ry and pro brotic molecules, the increase in NO production
culature and in the kidney, which could mediate the renopro- triggered by kinins could not only decrease oxidative stress
tective effects of ACE inhibition.426 but consequently attenuate the in ammatory and brotic
Interestingly, the contribution of the KKS to renal responses.434 Tissue kallikrein has also been reported to at-
handling of potassium has been recently addressed by tenuate salt-induced renal damage by activation of B2R.435,436
Chambrey’s group.407 Usually, potassium is almost complete- The protective effect of the KKS against renal injury was also
ly reabsorbed by the end of the loop of Henle and active con rmed in B2R knockout mice.437 In addition, a human
secretion in the late distal nephron determines the fraction B2R gene polymorphism has been demonstrated in patients
that needs to be excreted in the urine. Thus, except in a with chronic renal failure, suggesting a role of this receptor
situation of potassium depletion, in which net potassium in the early development of this pathology.438
absorption also occurs in the collecting system, the latter is Furthermore, as mentioned previously, bradykinin,
generally a site of net potassium secretion. It was recently through its receptors, increases prostaglandin production
shown that cortical collecting ducts isolated from mice with in at least three ways. First, it promotes translocation into
tissue kallikrein gene disruption do not secrete potassium the cell membrane and phosphorylation of cytosolic phos-
but instead exhibit net transepithelial potassium absorption pholipase A2 through a Ca2 -dependent phosphorylation.
due to abnormal activation of H -K -ATPase.407 This sug- Second, it activates membrane-associated phospholipase A2
gests that potassium absorption is constantly under the neg- through a Ca2 -independent mechanism. Phospholipase
ative control of tissue kallikrein. However, the mechanism A2 frees arachidonic acid from membrane phospholipids.
CHAPTER 8 HORMONES AND THE KIDNEY 2255
55

This arachidonic acid gets converted to prostaglandins by prevented diabetic nephropathy in streptozotocin-induced
cyclooxygenase. The third mechanism by which bradykinin diabetic rats.450 Once again, NO seems to be mediating this
increases the production of prostaglandins is by inducing renoprotective role. In fact, L-arginine, a substrate of NO
cyclooxygenase-2.403,433 It has been shown that the prosta- synthase (NOS), reduces proteinuria associated with strepto-
glandins formed following stimulation of the bradykinin re- zotocin-induced diabetes.451 Conversely, an NOS inhibitor as
ceptors act through their speci c receptors to mediate some well as an endothelial NOS (eNOS) de ciency accelerates the
of the effects of kinins on vascular tone and on mitochondrial severity of diabetic nephropathy.452–455 Interestingly, cicap-
respiration. Of potential relevance to the kidney, Kopp and rost, a prostacyclin analog, also delays the pathogenesis of
Smith 439 demonstrated in a rat model of unilateral ureteral the diabetic nephropathy, suggesting that prostaglandins in
obstruction that indomethacin abolished BK-induced na- addition to NO mediate the bene cial effects of the KKS.456 It
triuresis and diuresis in the contralateral kidney.439 Further is noteworthy that some groups have reported that deletion
studies are required to determine the role of bradykinin- of B2R protects against the streptozotocin-induced diabetic
induced prostaglandins in the healthy and diseased kidney, nephropathy.457 In addition, it has been shown that glomeru-
especially because renal diseases present with several char- lar kinin receptors are induced by diabetes and this may con-
acteristics including inadequate ltration of proteins and in- tribute to the development of glomerular injury and to the
ammatory cell recruitment. development of microvascular complications of diabetes.458
Finally, recent evidence suggests that bradykinin par- Bradykinin also regulates the expression of connective tis-
ticipates in the regulation of neonatal glomerular function sue growth factor (CTGF), TGF- receptor II, and collagen
and acts as a growth regulator during renal development. I in mesangial cells, which provides a mechanistic pathway
An intact KKS is necessary for the normal functional de- through which B2R activation contributes to the develop-
velopment of the kidney.440 During nephrogenesis, tubular ment of diabetic nephropathy.459 In addition, Christopher et
and glomerular growth and differentiation, and acquisition al.460 demonstrated that glucose by itself regulates the expres-
of specialized functions, are coordinated in time and space sion of B2R in vascular smooth muscle cells. These contra-
with renal vasculogenesis, glomerulogenesis, and regional dictory results may be attributed to the differences between
hemodynamic changes. The end result ensures that tubu- strains of mice and/or the methods of induction of diabetes.
lar structure and function are tightly coordinated with glo- Furthermore, immune-mediated nephritis plays a major
merular ltration during normal kidney development. To role in the pathogenesis of systemic lupus erythematosus
achieve this delicate task of glomerulotubular balance, the (SLE) and Goodpasture syndrome, which is the experi-
developing kidney produces growth factors and vasoactive mental model of glomerulonephritis caused by antibodies
hormones that act in a paracrine manner to regulate nephro- against glomerular basement membrane (anti-GBM). Liu
vascular growth, differentiation, and physiologic functions. et al. recently showed that mouse strains with upregulated
One such paracrine system is the KKS, which generates bra- expression of renal and urinary kallikreins are less suscep-
dykinin. Bradykinin activates B2R to regulate RBF and salt tible to anti-GBM antibody-induced nephritis and lupus
and water excretion. Gene-targeting studies indicate that the nephritis.461 In addition, administration of kallikreins de-
fetal KKS plays an important role in the maintenance of ter- creased the anti-GBM antibody-induced nephritis, demon-
minal epithelial cell differentiation.441 In fact, gestational salt strating a link between KKS and immune-mediated renal
loading induces abnormal renal development in B2R knock- damage.461 These ndings corroborate the aforementioned
out mice.442 It was also recently shown that B1R is unlikely results that attribute a renoprotective role for KKS in nephri-
to play a role in early nephrogenesis but its enrichment in the tis following insults such as salt imbalance and diabetes.
maturing proximal tubule suggests a potential role for this re- The KKS has also been shown to protect against amino-
ceptor in terminal differentiation of the proximal nephron.443 glycoside-induced renal injury. Aminoglycoside antibiotics
have been used to treat infections by aerobic gram-negative
bacteria. However, they cause ototoxicity and nephrotoxic-
Clinical Pathophysiologic Role of the ity. Interestingly, administration of tissue kallikrein is protec-
Kallikrein-Kinin System in the Kidneys tive against gentamicin-induced renal injury in rats partly by
Diabetic nephropathy occurs in 30% of all patients with inhibiting the in ammatory response and apoptosis through
diabetes. ACE inhibition is protective in many models of dia- suppressing of oxidative stress.462 In addition, L-arginine
betic nephropathy, a protection partly mediated by the KKS. also prevents gentamicin-induced tubular damage in rats,
In fact, it has been shown in different diabetic models that whereas a NOS inhibitor aggravates the renal failure.463
the bene cial effects of ACE inhibitors were attenuated by Similarly to the diabetic nephropathy, endogenous prostacy-
a B2R antagonist.444–447 The same ndings were observed in clin production also prevented the apoptosis and oxidative
diabetic mice lacking B2R.448,449 Albuminuria, glomerular stress induced by gentamicin.464 Thus, the renoprotective
sclerosis, interstitial brosis, lipofuscin accumulation in prox- effects of tissue kallikrein against gentamicin toxicity seems
imal tubules, and lifespan shortening were enhanced in this to be once more mediated by NO and prostacyclin.
model. Corroborating these ndings, human tissue kallikrein Furthermore, KKS seems to play a protective role in
gene delivery ef ciently attenuated insulin resistance and ischemic acute renal failure (IARF). As previously discussed,
256
25 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

NO production induced by B2Rs is an important player in the luminal membrane. Oxygen availability in the renal medulla is
protective effects of ACE inhibitors.425 In addition, ARBs seem marginally adequate; consequently, increased Na -K -ATPase
to be less protective than ACE inhibitors against ischemia- activity may deplete ATP levels and lead to dephosphorylation
reperfusion injury,465 suggesting that ACE inhibitors may of adenine nucleotides to form adenosine. Reactive ischemia
partly mediate their bene cial effects by inhibiting the inacti- is a second example of the intracellular ATP pathway in the
vation of the kinins than by suppressing angiotensin II forma- kidney. A short period of ischemia in the kidney triggers a brief
tion. In addition, studies have reported that deleting B1R and/ increase in renal vascular resistance, a phenomenon known
or B2R aggravates renal damage following IARF; however, the as reactive ischemia. Renal ischemia activates the intracellular
double knockout had more detrimental effects than a de - ATP pathway of adenosine production and adenosine causes
ciency in B2R only, emphasizing that both B1R and B2R are reactive ischemia via activation of A1 receptors in the preglo-
necessary for protection in IARF.466 In addition to NO, older merular microvessels.478 Adenosine, produced intracellularly,
studies have shown that prostaglandins E1, E2, and I2 are can traverse cell membranes by facilitated diffusion and func-
also protective mediators of the KKS in IARF.467 Contrary to tion in a paracrine or autocrine fashion.
these ndings, results from exogenously administered brady- The extracellular ATP pathway is yet another mecha-
kinin on IARF are con icting and suggest that it aggravates nism of adenosine production in the kidney. Extracellular
IARF, although suppression of the endogenous KKS is also production of adenosine from AMP is possible because of
detrimental.468 Thus, it seems that the physiologic levels of the presence of ecto-5 -nucleotidase on the surface of many
bradykinin produced endogenously are important for the cell types. Release of adenine nucleotides into the extracel-
functional recovery after IARF, but the higher levels achieved lular compartment from renal sympathetic nerve terminals,
with exogenously administered bradykinin are detrimental. intrarenal platelets, renal endothelial cells, renal vascular
In support of the aforementioned results in animal mod- smooth muscle cells, and/or renal epithelial cells exposes
els, genetic studies in humans revealed that polymorphisms extracellular ATP to ecto-ATPases, ecto-ADPases, and ecto-
in several KKS-related genes are associated with higher inci- 5 -nucleotidases, and these enzymes metabolize adenine nu-
dence of renal problems. In fact, ACE polymorphism seems cleotides to adenosine.478 In the kidney, ecto-5 -nucleotidase
to in uence diabetic nephropathy469 and a polymorphism in activity is expressed on tubular luminal membranes, bro-
the human B2R gene has been correlated with a higher sus- blasts, and mesangial cells and is believed to be the major
ceptibility for nephropathy in diabetic patients.470 Both hu- source of renal adenosine.479
man SLE and spontaneous lupus nephritis were also recently When oxygen supply is adequate, enzymatic hydroly-
found to be associated with polymorphism in the kallikrein sis of S-adenosyl homocysteine (SAH) to L-homocysteine
gene family, particularly KLK1 and KLK3.471 Other studies and adenosine constitutes the major production pathway. It
have shown that the ACE D/D genotype is a risk factor for is the transmethylation pathway of adenosine production.
progression to chronic renal failure in patients with IgA Approximately one-third of the adenosine release to the
nephropathy.472,473 In addition, this ACE D/D genotype is also extracellular space by cardiomyocytes is through the trans-
a risk factor for patients with autosomal dominant polycystic methylation pathway, but the importance of this pathway in
kidney disease (ADPKD) to develop ESRD at an earlier age.474 the kidney is unclear.478
In fact, polymorphisms in most of the genes in the KKS have
been associated with the progression of chronic renal failure Adenosine Receptors
to ESRD, including ACE,475 B1R and B2R,476 and eNO.477
Purinoceptors or adenosine receptors, also called P1 re-
ceptors, are classic G protein-coupled receptors with four
known subtypes (A1, A2A, A2B, and A3 receptors).478,480 A1
ADENOSINE receptors (A1Rs) have a high af nity for adenosine, whereas
Adenosine, a purine nucleoside, is a paracrine hormone that the af nity of A2A receptors for adenosine is approximately
regulates cellular and physiologic functions in many tissues. threefold less compared with that of A1R. A2B and A3 recep-
Three major pathways produce adenosine: the intracellular tors are low-af nity adenosine receptors.478,480–482
pathway, the extracellular ATP pathway, and the transmeth- A1Rs are present in afferent arterioles, glomeruli includ-
ylation pathway. Intracellular generation of adenosine results ing mesangial cells, juxtaglomerular cells, vasa recta, as well
from the action of 5 -nucleotidase on adenosine monophos- as in various segments of the tubular and collecting duct
phate (AMP) during hypoxia (sequential dephosphorylation of system including proximal tubule, thin limbs of Henle, TAL,
intracellular ATP to adenosine). In cells that rapidly consume and collecting ducts.483 They evoke vasoconstriction by in-
ATP, enhanced utilization of ATP increases the intracellular pro- hibiting adenylate cyclase activity (via activation of Gi and
duction rate of adenosine. In the kidney there are two prime Go), thereby reducing cAMP generation in vascular smooth
examples of the intracellular ATP pathway. Increased delivery muscle and signaling.478 In renal epithelial cells and iso-
of Na to the thick ascending limb of loop of Henle stimulates lated rabbit afferent arterioles, A1R activation also appears
Na -K -ATPase activity in the basolateral membrane of to stimulate phospholipase C activity. A2A and A2B recep-
epithelial cells through the increased ux of Na across the tors produce vasodilation by stimulating adenylate cyclase
CHAPTER 8 HORMONES AND THE KIDNEY 2257
57

activity to increase cAMP generation; A2A signal by engaging Effect on Renal Blood Flow and
Gs, Golf, and p21ras and A2B receptors are also known to Glomerular Filtration Rate
stimulate phospholipase C via Gq. A3 receptors are thought
Infusion of adenosine into animals’ renal artery results in
to exert their physiologic effects through activation of cal-
transient reduction of RBF secondary to A1R mediated af-
cium signaling pathways (phospholipase via Gq families)
ferent arteriolar vasoconstriction, followed by a delayed A2R
and perhaps inhibition of cAMP accumulation (via Gi), but
mediated postglomerular vasodilation and return of RBF to
their role in regulating the renal microvascular and epithelial
normal.487–489 Selective A2R agonists increase RBF via vaso-
function has not been extensively examined.478,480
dilation of the medullary renal microcirculation. A1R impor-
P2 receptors were originally described as purinergic re-
tantly regulate renal function. Infusion of A1R agonists into
ceptors, re ecting the idea that they responded to ATP re-
the renal interstitium diminishes blood ow to both super -
leased as a neurotransmitter from peripheral sympathetic
cial and deep nephrons. In the outer cortex, A1R-induced va-
nerves. Currently P2 receptors are divided into two distinct
soconstriction is most likely caused by contraction of preglo-
families: P2X and P2Y.480
merular, rather than postglomerular, microvessels; however,
Distinct genes code for each receptor family and there are
in juxtaglomerular nephrons, A1R mediate vasoconstriction
marked differences in the structural and signal-transduction
by contracting preglomerular microvessels, efferent arterioles,
characteristics between the two families.
and outer medullary descending vasa recta. Ang II strongly
enhances A1R-induced preglomerular vasoconstriction. Con-
Renal Actions of Adenosine
traction of preglomerular microvascular smooth muscle cells
Adenosine regulates a wide array of physiologic functions, by A1R underlies the mediator role of adenosine in tubulo-
including cardiac rate and contractility, vascular smooth glomerular feedback (TGF) (Fig. 8.7), explains the ability of
muscle tone, neurotransmitter release, lipolysis, leukocyte adenosine to decrease GFR, and potentiates postjunctional
function, platelet function, and renal hemodynamics and vasoconstrictor responses to renal sympathetic neurotrans-
electrolyte transport.481,484,485 mission in the kidney.478 It is conceivable that A1R are pres-
Adenosine is produced in the kidney and acts in an au- ent in endothelial cells along the renal vasculature and that
tocrine or paracrine fashion.479,481 Both high-af nity A1R adenosine causes the release of NO and perhaps other endo-
and low-af nity A2R are widely distributed throughout the thelial vasodilators when administered from the vascular, but
renal vasculature and the nephron.481,486 The renal effects of not from the interstitial, aspect of the vessel. The resulting
adenosine are diverse and include alterations in RBF, GFR, A1R-induced constriction would, therefore, be blunted by
hormone production, neurotransmitter release, and tubular endothelial factors only when adenosine is given intravascu-
absorption (Table 8.4). larly. In a study in dogs, the administration of NOS inhibitors
caused a marked augmentation in the constrictor response
of RBF to bolus injections of adenosine, whereas the dilator
TA B L E
effect of the A2 agonist CGS-21680 was unaffected, indicating
8.4 Renal Actions of Adenosine that adenosine may cause NOS activation through an A1R-
mediated mechanism. It is now well recognized that the
majority of vasodilator agents act by binding to their receptors
Function Effect Receptor
on endothelial cells and by eliciting the generation and release
Renal blood ow Transient ↑ A1 of endothelial relaxing factors, most notably NO, endothe-
Delayed slight ↑ A2 lial hyperpolarizing factor, and prostaglandins. However, in
a number of studies, adenosine appears to augment NOS ac-
GFR ↓↓ A1 and A2 tivity and NO release through an A2R-mediated process, an
action that would enhance the dilator component rather than
Renin production ↓↓ A1 diminish the constrictor component of the adenosine actions.
↑ A2 Also adenosine has been shown to consistently stimulate
the production of NO in cultured endothelial cells, usually
Erythropoietin ↓ A1
through an A2AR-dependent mechanism.489
production ↑ A2 Adenosine induces a sustained decrease in GFR second-
ary to reduced PGC.490 Infused in humans, it results in an
Sodium excretion ↑↑ A2
insigni cant increase in RBF and a signi cant, moderate de-
↓ A1 crease in GFR.491,492 It is postulated that adenosine-mediated
reduction in GFR constitutes the underlying mechanism of
GMC production ↓ A2B
TGF.493,494 The hypothesis states that increased solute de-
and proliferation
livery to the MD stimulates sodium transport, resulting in
↓, decrease; ↑ , increase; GFR, glomerular ltration rate; GMC, glomerular ATP hydrolysis and generation of adenosine. Adenosine, in
mesangial cells. turn, completes the feedback loop by decreasing GFR and
258
25 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

FIGURE 8.7 Proposed mechanism of adenosine acting as


a mediator of the tubuloglomerular feedback. Numbers in
circles refer to the following sequence of events. 1, Increase in
concentration-dependent uptake of nitrogen (N), potassium
(K), and chlorine (Cl) via the furosemide-sensitive Na-K-2Cl
cotransporter (NKCC2); 2 and 3, transport-dependent, in-
tra- and/or extracellular generation of adenosine (ADO);
the extracellular generation involves ecto-5 -nucleotidase
(5 -NT); 4, extracellular ADO activates adenosine A1 receptors
triggering an increase in cytosolic Ca 2 in extraglomerular
mesangium cells (MC); 5, the intensive coupling between
extraglomerular MC, granular cells containing renin, and
smooth muscle cells of the afferent arteriole (VSMC) by gap
junctions allows propagation of the increased Ca 2 signal
resulting in afferent arteriolar vasoconstriction and inhibition
of renin release. Factors such as nitric oxide, arachidonic acid
breakdown products, or angiotensin (Ang) II modulate the de-
scribed cascade. NOS I, neuronal nitric oxide synthase; COX-2,
cyclooxygenase-2. See text for further explanations. (Adapted
from Vallon V, Muhlbauer B, Osswald H. Adenosine and kidney
function. Physiol Rev. 2006;86(3):901–940.)

normalizing solute delivery to the distal nephron. The hy- proximal tubule, which is a tubular segment with relatively
pothesis is supported by experiments demonstrating that high basal oxygen supply. In contrast, adenosine inhibits NaCl
A1R blockade inhibits TGF.495,496,497 reabsorption in medullary TAL and IMCD—that is, nephron
segments with relatively low oxygen delivery.483 Caffeine and
theophylline are nonselective adenosine antagonists, and re-
Effect on Sodium Reabsorption and Renal ports suggest high levels of each induced diuresis.500
Tubular Transport Adenosine acts as a mediator of the TGF, which establishes
In contrast to A1R, which directly stimulate Na reabsorption an inverse relationship between GFR and the NaCl concentra-
by increasing epithelial transport mechanisms, activation of tion at the MD. In the juxtaglomerular apparatus, extracellular
A2R in microvessels of juxtaglomerular nephrons and in the formation of adenosine by ecto-5 -nucleotidase contributes to
medullary microcirculation enhances medullary blood ow, the adenosine pool that mediates TGF-induced afferent arteri-
thus altering peritubular forces that modulate Na reabsorp- olar vasoconstriction. The TGF mechanism stablizes the NaCl
tion. The net result is an increase in Na excretion.478 Adenos- load to the distal nephron, which facilitates ne regulation of
ine-induced decrease in solute excretion, particularly sodium, body NaCl balance at these sites. Endogenous adenosine, the
has been generally attributed to the concomitant reduction in synthesis of which is increased by enhanced NaCl transport,
GFR and urine ow. The presence of A1R and A2R on renal limits via adenosine A1R activation the oxygen demand in rela-
epithelial cells, however, suggests that adenosine has direct tively hypoxic nephron segments by directly and differentially
effects on tubular transport.481 In rats, for example, infusing affecting reabsorption along the nephron.483
adenosine or adenosine receptor agonists in a dose that does Activation of basolateral P2 receptors in the collecting
not alter systemic blood pressure, RBF, or GFR still leads to so- duct inhibits vasopressin-stimulated water reabsorption,
dium and water retention.498,499 This effect was independent and stimulation of apical P2 receptors can affect solute trans-
of renal enervation, at least for A1-speci c agonists, and sug- port in both the proximal and the distal nephron: an in vivo
gested a direct tubular effect of adenosine on A1R to stimu- micropuncture study in the rat showed that stimulation of
late sodium reabsorption.499 A1R in the nephron are linked to apical P2Y1 receptors inhibits proximal tubular bicarbon-
Na transport in several segments. However, the natriuresis ate reabsorption through suppression of NHE3 activity,1 and
and diuresis associated with systemic inhibition of A1R are in vitro and in vivo evidence indicates that stimulation of
due primarily to reduced proximal tubule (PT) reabsorption. apical P2 receptors in the distal nephron inhibits amiloride-
Studies on kidney uid and electrolyte transport show that sensitive sodium reabsorption 2,3 and reduces the activity of
activation of A1R stimulates NaCl reabsorption in cortical apical K secretory channels.4,501
CHAPTER 8 HORMONES AND THE KIDNEY 2259
59

Effect on Renin example, selective antagonism of A1 receptors attenuates ne-


Adenosine suppresses renin release by the kidney.481 In phropathy caused by nephrotoxins such as cisplatin, genta-
sodium-depleted animals, renin release is inhibited by ma- micin, cephaloridine, glycerol, and radiocontrast media.478
neuvers that increase renal adenosine production,502,503 an
effect that results from direct action of adenosine on renin- A2R and Diabetic Nephropathy
producing cells.504 Inhibition of renin release is most likely Awad et al. showed that streptozotocin (STZ)-induced dia-
mediated by binding of adenosine to high-af nity A1R.505 betes in rodents leads to marked proteinuria and decreased
In this regard, A1R restrain renin release responses, a theory renal function that is attenuated with continuous subcutane-
known as the adenosine-brake hypothesis. A1R are coupled to ous administration of A2R agonists. Both nephrin and podo-
Gi and, therefore, inhibit adenylyl cyclase. Because stimu- cin mRNA levels were reduced after STZ-induced diabetes,
lation of renin release from juxtaglomerular cells by many an effect completely restored with A2R agonist treatment.511
stimuli involves activation of adenylyl cyclase, activation of They also demonstrated that chronic administration of se-
juxtaglomerular A1R attenuates renin release and antago- lective A2A agonists attenuates renal lesions and functional
nism of renal A1R increases renin release.478 In contrast, abnormalities characteristic of diabetic nephropathy. The
agonists selective for the low-af nity A2R stimulate renin renal tissue protective effect of the A2A agonist is believed
release, particularly when administered in high doses.481,506 to be mediated primarily by abrogating the in ammatory re-
This suggests that adenosine regulates renin release by sponse associated with diabetes. Chronic A2R activation in
exerting either an inhibitory or stimulatory effect, depending diabetic rats ameliorates histologic and functional changes in
on its local concentration. Studies employing acute block- kidneys induced by diabetes and causes reduced in amma-
ade or chronic de ciency of adenosine A1R receptors rather tion associated with diabetic nephropathy.511
indicate a modulating, tonic inhibition of the renin system
by adenosine. In addition, an MD-dependent source of ad- Ischemia and Reperfusion Injury
enosine and activation of adenosine A1R contribute to renin Activation of A1R in many organs, including the kidney, ini-
release inhibition under conditions of high NaCl concentra- tiates several cytoprotective kinase cascades including ERK
tions at the MD. An increase in intracellular cAMP is an im- MAPK, Akt, and PKC. Activation of cell surface A1R produc-
portant stimulator of renin release. Part of the cAMP could es cytoprotective effects against ischemia and reperfusion (IR)
be released by renin-secreting cells and is extracellularly injury in many organ systems including the heart, kidney,
converted to adenosine, which acts as a negative feedback and brain. Joo et al. demonstrated that transient A1R activa-
control or brake when renin secretion is stimulated.483 tion produces both acute and delayed protective effects in the
kidney including reduced renal cortical necrosis and apopto-
Effect on Renal Medullary Oxygenation sis after IR injury. Renal apoptosis is an important component
Renal medullary hypoxia is an obligatory part of the process of in the development of ARF after IR injury.512 A2R exert im-
urinary concentration. When O2 supply is further impaired, portant anti-in ammatory actions that may protect the kid-
however, medullary hypoxic injury can develop. Various neys from injury (Fig. 8.8). Their activation strongly inhibits
mechanisms act in concert to minimize medullary hypoxia. neutrophil endothelial cell interactions in vitro and, in vivo,
Adenosine-mediated actions like maintaining high proximal and markedly decreases the renal in ltration of neutrophils
reabsorption, inhibiting reabsorption in the medullary TAL, and attenuates renal dysfunction following IR injury.478
and reducing GFR by the TGF mechanism, when distal NaCl
concentrations increase, can be part of these mechanisms. Adenosine 5 -tetraphosphate
Furthermore, in the deep cortex and medulla, adenosine via Adenosine 5 -tetraphosphate (AP4) is the most potent va-
A2R activation causes vasodilation, which increases medul- soactive purinergic mediator identi ed to date, exerting the
lary blood ow and medullary oxygenation. Thus, adenosine vasoconstriction predominantly through P2X1 receptor acti-
through distinct actions on the vasculature and tubular trans- vation. The fact that AP4 is more resistant to degradation fur-
port system contributes to the stabilization of the O2 demand- ther favors potent and prolonged vasoconstrictive effects. The
to-supply ratio particularly in the renal medulla.483 Nishiyama vasoconstrictive in vitro effects of AP4 are paralleled by hy-
and associates480,507 demonstrated that hypoxia-induced renal pertensive effects in vivo. Therefore, it may be speculated that
vasoconstriction was associated with elevated interstitial ad- AP4 is a vasoconstrictor secreted by human endothelial cells,
enosine levels and could be blocked by adenosine A1R an- which also plays a role in the regulation of systemic blood
tagonists. Adenosine plays a role in balancing oxygen supply pressure under physiologic and pathologic conditions.513
and demand during renal hypoxia by regulating RBF, GFR,
renin secretion, and solute transport.493 Adenosine and Angiotensin II
Pathophysiologic conditions associated with increased Certain actions of adenosine, such as vasoconstriction of
renal production of adenosine include ARF, myoglobinuric the renal afferent arteriole, are either dependent on or sig-
ARF, and mercuric chloride-induced ARF.508–510 A1R may ni cantly enhanced by Ang II.481,514 Evidence suggests that a
also be involved in other drug-induced nephrotoxicity. For reduction in, or prevention of, Ang II formation and action
260
26 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

release.516 Postjunctionally, however, adenosine seems to en-


A1 receptors
hance sensitivity to NE.516 Because renal denervation does
not alter adenosine-induced changes in RBF and GFR, the
hemodynamic actions of adenosine in the kidney are most
likely independent of its effect on neurotransmitter release.481
Contraction of Inhibition of renin Alterations in
renal vascular release from transport in
smooth juxtaglomerular tubular Pancreatohepatorenal Extracellular
muscle cells cells epithelial cells
cAMP-Adenosine Pathway
In addition to an autocrine/paracrine role in the kidney, the
Mediation Enhanced extracellular cAMP-adenosine pathway may also function as
of TGF Decreased postjunctional
GFR response to NE
an endocrine system by which the pancreas and liver regulate
renal function. The pancreas releases glucagon directly into
the portal circulation in response to appropriate stimuli.
A2A receptors A2B receptors Glucagon in the portal circulation stimulates hepatic adenylyl
cyclase, which results in secretion of cAMP by hepatocytes
into the venous circulation. Liver-derived cAMP circulates to
the kidney, where it is ltered into the proximal tubule and
Relaxation of Inhibition of Inhibition of then metabolized to adenosine. Adenosine would then engage
renal vascular neutrophil mesangial and A1R in epithelial cells to enhance electrolyte transport. It is
smooth function vascular smooth
muscle cells
important to note that, in contrast to cAMP, which is stable
muscle cell growth
in blood, adenosine has a half-life in human blood of less
than 1 second. The liver has the capacity to release signi cant
Antiinflammatory amounts of cAMP into the hepatic vein.478
Increased medullary action
blood flow causing
The most recent studies strongly suggest that an auto-
decreased NaCl reabsorption crine/paracrine extracellular cAMP-adenosine pathway, in
fact, does not participate in the regulation of Na transport
by proximal epithelial cells. It appears that the pancreato-
FIGURE 8.8 Regulation of renal function by adenosine.
hepatorenal extracellular cAMP pathway is more important
TGF, tubuloglomerular feedback; GFR, glomerular ltration rate;
in modulating proximal tubular function. Although intra-
NE, norepinephrine. (From Bulut OP, Dipp S, El-Dahr S. Ontogeny
renal infusions of glucagons do not reduce Na excretion,
of bradykinin B1 receptors in the mouse kidney. Pediatr Res.
intraportal infusions of glucagon in sheep cause a marked
2009;66(5):519–523.)
antidiuresis. Although speculative, it is conceivable that
the pancreatohepatorenal cAMP-adenosine pathway par-
cause a marked attenuation of the vasoconstrictor response ticipates in physiologic adjustments of renal transport, as
of the intact kidney to adenosine. Conversely, an elevation well as in pathophysiologic processes. In normal mammals,
of ambient Ang II concentrations enhances the constrictor both hypoglycemia and exercise are powerful stimulants to
effect of A1R activation.489 glucagon release. Activation of the pancreatohepatorenal
cAMP-adenosine pathway by glucagon in response to hy-
Effects on Glomerular Mesangial Cells poglycemia might increase Na glucose symport in proxi-
mal tubules and, thus, increase the ef ciency of glucose
A2B receptors’ activation attenuates vascular smooth mus-
transport, an adaptive mechanism to combat hypoglycemia.
cle cell proliferation and collagen and protein synthesis
Activation of the pancreatohepatorenal cAMP-adenosine
(Fig. 8.8). Studies with a number of adenosine receptor
pathway by glucagon during exercise might enhance Na
agonists and antagonists and with antisense oligodeoxynu-
transport in the proximal tubules and thus increase the ef-
cleotides against the A2B receptor indicate that the growth
ciency of Na reabsorption, an adaptive mechanism to
inhibitory effects of adenosine on vascular smooth muscle
avoid volume depletion during sustained physical exertion.
cells are mediated by these receptors.478
The pancreatohepatorenal cAMP-adenosine pathway might
be overly activated in the metabolic syndrome. Although
Effect on Erythropoietin oral glucose normally strongly inhibits glucagon secretion
A1R stimulation inhibits erythropoietin (EPO) synthesis, by the pancreas, in animals and people with the metabolic
whereas that of A2R enhances EPO synthesis.515 syndrome, an oral glucose challenge markedly stimulates
pancreatic glucagon secretion by approximately 200%. If the
Effect on Norepinephrine pancreatohepatorenal cAMP-adenosine pathway exists, each
Activation of A1R on sympathetic neurons in the kid- time such a patient ingests a high carbohydrate meal the re-
ney causes presynaptic inhibition of norepinephrine (NE) nal tubules would be exposed to a wave of excess adenosine
CHAPTER 8 HORMONES AND THE KIDNEY 2261
61

production. Because adenosine causes increased reabsorp- that is reversed by PTH(1-34) and PTH(1-84). PTH(7-84)
tion of Na and vasoconstriction of the preglomerular mi- inhibits PTH(1-84)-induced bone resorption. Nakajima
crocirculation, this could contribute to the pathophysiology et al. studied the effect of PTH(7-84) on PTH(1-34)-induced
of hypertension in the metabolic syndrome. Importantly, production of 1,25(OH)2D3 in primary cultured murine
adenosine receptors also inhibit lipolysis in fat cells and may renal tubules. PTH(1-34) stimulated the conversion of 25-
reduce insulin sensitivity in skeletal muscle. However, at this OH D3 to 1,25(OH)2D3, and PTH(7-84) dose-dependently
time, both the physiologic and pathophysiologic roles of the inhibited this process. Real-time polymerase chain reaction
putative pancreatohepatorenal cAMP-adenosine pathway (PCR) revealed that PTH(1-34) increased the expression
are speculative.478 level of 1 -hydroxylase mRNA, whereas PTH(7-84) did not.
This may at least partly account for the decreased serum
PARATHYROID HORMONE AND level of 1,25(OH)2D3 in patients with severe primary hyper-
parathyroidism with renal failure.523
PARATHYROID HORMONE–RELATED Once secreted, PTH is rapidly cleared from plasma
PEPTIDE through uptake principally by the liver and kidney, where
In response to low levels of extracellular Ca, parathyroid PTH(1-84) is cleaved into amino- and carboxyl-terminal
glands secrete parathyroid hormone (PTH), an 84-amino acid fragments that are then cleared by the kidney. Hepatic
polypeptide hormone.517 PTH is initially synthesized as a 115- clearance of PTH involves rapid proteolysis by Kupffer
amino acid polypeptide, pre-pro-PTH, which is cleaved with- cells to N-terminal fragments and C-terminal fragments
in parathyroid cells at the N-terminal portion rst to pro-PTH (CPTH). CPTH fragments can exert direct effects on bone
(90 amino acids) and then to PTH (84 amino acids). PTH-re- cells via CPTH receptors. They are cleared predominantly by
lated peptide (PTHrP) was rst identi ed as a cause of humor- the kidney and accumulate disproportionately during renal
al hypercalcemia of malignancy and is secreted predominately failure.524 Intact PTH has a plasma half-life of 2 to 4 minutes.
as a 141-amino acid peptide. PTH and PTHrP have sequence In comparison, the C-terminal fragments, which are cleared
homology in the rst 13 amino acids (the amino terminus).518 principally by the kidney, have half-lives that are ve to ten
Increased circulating PTH or PTHrP leads to mobilization of times greater.
Ca from bone, enhancement of Ca reabsorption in the renal
tubule, and increased production of 1,25-dihydroxyvitamin
D3 (1,25[OH]2D3) from 25 hydroxyvitamin D3(25-OH D3) Parathyroid Hormone and Parathyroid
by proximal tubule cells through 1 hydroxylase stimula- Hormone–Related Peptide Receptors
tion. 1,25(OH)2D3, in turn, increases Ca absorption by the Two types of receptors exist. Type 1 PTH receptors (PTH1R)
intestine and possibly Ca reabsorption by the kidney. The bind PTH and PTHrP. Binding to PTH1R occurs in the 15-
combined actions of PTH and 1,25(OH)2D3 result in normal- to 34-amino acid region of both hormones (N-terminal
ization of the extracellular Ca concentration. In addition to sequence). It is interesting that these two peptides bind with
its regulatory actions on calcium balance, PTH can regulate almost equal af nity and yet do not share sequence homol-
phosphorus balance by inhibiting its reabsorption in the prox- ogy in this region. The PTH1R mediates the biologic activity
imal and distal tubules of the nephron. of PTH and PTHrP. PTH1R is heavily expressed in bone and
PTH synthesis and secretion by the parathyroid gland kidney, and is also present in other tissues such as breast,
is tightly regulated.517 Although a decreased extracellular skin, heart, blood vessels, pancreas, and other tissues. It ac-
Ca level stimulates PTH synthesis and secretion, increased tivates multiple cellular signaling pathways including cAMP,
extracellular levels of either Ca or 1,25(OH)2D3 are in- PLC pathway, PKC, and release of intracellular calcium stores.
hibitory. The extracellular phosphorus level regulates PTH Muller et al. showed also that activation of PTH1R engages
production directly at a posttranscriptional level519 and in- major apoptosis signaling pathways, namely in apoptosis
directly by altering circulating Ca and 1,25(OH)2D3 concen- of differentiating embryonic cells.525 Type 2 PTH receptors
trations. Increased serum phosphorus secondary to renal (PTH2R), which share 51% homology with PTH1R, can
insuf ciency, for example, decreases Ca concentration and bind PTH but they do not bind PTHrP with high af nity.
1,25(OH)2D3 production, leading to stimulation of PTH re- PTH2Rs are expressed in only a few tissues—their biologic
lease, but can itself increase PTH level without alterations in signi cance is unknown.518 Increasing evidence points to the
ionized calcium or serum 1,25(OH)2D3 levels.520,521 presence of novel PTH receptors (CPTHR) with speci city
Magnesium also regulates PTH secretion and its deple- for the carboxyl-terminal region of PTH. This portion of the
tion can decrease PTH secretion. Fibroblast growth factor 23 hormone was previously thought to be biologically inert but
(FGF23) decreases PTH mRNA and secretion. Ben-Dov et al. has now been shown to possess hypocalcemic activity. The
showed that FGF23 acts directly on the parathyroid through CPTHRs are present in various tissues but are most heavily
the MAPK pathway to decrease serum PTH.522 expressed in bone.
The biologic activity of PTH resides in its amino- Although PTH is a well-characterized endocrine regu-
terminus. There is increasing evidence that the C-terminal lator of mineral homeostasis, PTHrP is a key regulator of
fragment of PTH, PTH(7-84), exerts a hypocalcemic effect placental calcium transport in the fetus, and it appears to
262
26 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

be a physiologic modulator of smooth muscle tone. Current Renal Actions of Parathyroid Hormone–
concepts indicate that PTHrP is a developmental and/ Related Peptide
or growth-regulating factor, much more similar to other
In the kidney, PTHrP appears to modulate RPF and GFR,
known cytokines and growth factors than to PTH.526 Under
and induces proliferative effects on both glomerular mesan-
physiologic conditions, PTHrP levels are increased during
gial and tubuloepithelial cells. PTHrP is known to be upreg-
pregnancy and lactation.527 However, as described earlier, it
ulated in several experimental nephropathies such as acute
appears to play a predominately pathophysiologic role in the
renal failure, obstructive nephropathy, as well as diabetic
adult, causing hypercalcemia.
nephropathy. In transgenic mice models and in the former
condition, PTHrP appears to contribute to the progression of
Renal Actions of Parathyroid Hormone renal damage by increasing tubulointerstitial cell survival, in-
PTH receptors and PTH-sensitive adenylate cyclase have ammation, and renal brogenesis. In diabetic nephropathy,
been identi ed in glomeruli and basolateral membranes of PTHrP can promote renal hypertrophy and proteinuria. Ang
epithelial cells in the proximal tubule, thick ascending limb II, a critical factor in the progression of renal injury, appears
of Henle’s loop, and distal convoluted tubule (DCT).7,528 PTH to be, at least in part, responsible for endogenous PTHrP
has three major effects on the kidney: increased Ca reabsorp- upregulation in these pathophysiologic settings.526 PTHrP
tion, inhibition of phosphate reabsorption, and stimulation also participates in the hypertrophic signalling triggered by
of 1,25(OH)2D3 synthesis. Its other actions on the kidney high glucose on podocytes. In this condition, Ang II induces
include modulation of GFR, gluconeogenesis, magnesium the upregulation of PTHrP, which in turn might induce both
reabsorption, and acid-base handling. TGF- 1 and p27Kip1 expression and thereby promotes the
PTH decreases renal Ca excretion through multiple hypertrophy of podocytes.542
mechanisms. Ichikawa et al.529 demonstrated that PTH in- Conventional renal cell carcinoma (CRCC) originates
fusion in rats decreases GFR by reducing Kf. A decreased from the renal proximal tubular epithelium, a target tissue
GFR leads to decreased ltered load of Ca and, therefore, for PTHrP proliferation effects.543 It was shown that PTHrP,
Ca excretion. PTH also enhances tubular Ca reabsorption acting through its receptor PTH1R, is an essential growth
by stimulating active Ca transport in the thick ascending factor for CRCC in vitro and in vivo and a new target for
limb of Henle’s loop and distal tubule.530,531 The effect of the VHL gene products.544,545 It seems that PTHrP induces
PTH on Ca transport in the proximal tubule varies and tumor cell survival through inhibition of cell apoptosis.546
is probably related to Na and water reabsorption in this
nephron segment.
PTH causes phosphaturia primarily by inhibiting phos- VITAMIN D
phate transport in the proximal tubule, speci cally by inhib- Along with PTH, vitamin D plays a central role in calcium
iting sodium-phosphate (Na-P) cotransport.532 Two different and phosphate homeostasis. The active form of vitamin
renal Na-P cotransporters have been identi ed and have D, 1,25(OH)2D, is a steroid molecule synthesized from
been termed type 1 (Npt1) and type 2 (Npt2). Npt2 is a either vitamin D3 (cholecalciferol) or vitamin D2 (ergocal-
target for regulation by PTH and decreases Na-P transport by ciferol). These two forms of vitamin D differ only by the
endocytic retrieval and lysosomal degradation of the Npt2 side chain to the sterol skeleton. Vitamin D3 is present in
protein.533 This endocytic retrieval can occur either from the diet (animal sources, mainly sh oil) and is also syn-
proximal tubules exposed to apical or basolateral PTH and thesized by the skin from 7-dehydrocholesterol upon expo-
can signal via either the cAMP-PKA or the PLC-PKC path- sure to ultraviolet light. Vitamin D2 is available only from
way.534 Mice that are Npt2 null have profound phosphate dietary sources (plants, yeast, and fungi). Vitamins D3 and
wasting.535 Npt2-null mice are resistant to further phospha- D2 are biologically inactive. They require activation in the
turic effects from exogenous PTH.536 2-Adrenergic recep- liver and kidney. After binding to carrier proteins, in par-
tor stimulation blunts the phosphaturic response to PTH.537 ticular, vitamin D-binding protein (DBP), vitamin D is trans-
PTH-stimulated synthesis of 1,25(OH)2D3 in the proximal ported to the liver where it is enzymatically hydroxylated
tubule is discussed in the next section. to 25-hydroxyvitamin D (calcidiol, 25[OH]D) through the
Although no single hormone has been speci cally action of hepatic microsomal and mitochondrial cytochrome
shown to regulate Mg homeostasis, PTH appears to increase P450 vitamin D–25-hydroxylase. Subsequently, 25(OH)
Mg reabsorption in the kidney.538 PTH also plays a role in D, bound to DBP, is transported to the kidneys where it is
acid–base homeostasis by enhancing urinary acid excre- hydroxylated exclusively in the proximal tubule by the mito-
tion.539 Although PTH inhibits bicarbonate reabsorption in chondrial 25(OH)D–1 -hydroxylase.547 1 -Hydroxylation
the proximal tubule, it indirectly stimulates distal hydro- is the rate-limiting step in the formation of the most abun-
gen ion secretion and titratable acid excretion by increasing dant active metabolite 1,25(OH)2D3.548–550 This step is tight-
phosphate delivery to the distal nephron. PTH has also been ly regulated through multiple feedback mechanisms,549,550
shown to enhance proximal tubular gluconeogenesis540 and mainly PTH, calcium, phosphate, broblast growth factor
renin secretion by juxtaglomerular cells.541 23 (FGF23), and 1,25(OH)2D3 itself (Fig. 8.9). An increase
CHAPTER 8 HORMONES AND THE KIDNEY 2263
63

FIGURE 8.9 A: Calcium–parathyroid hormone (PTH)–vitamin D axis. The calcemic hormone PTH produced by the parathyroid gland
targets kidney to increase calcium absorption and 1,25(OH)2D3 production and bone to increase calcium ef ux. 1,25(OH)2D3 stimu-
lates calcium absorption from the gut, which, along with renal and bone calcium, restores serum calcium to normal. B: Fibroblast
growth factor 23 (FGF23)–bone–kidney axis. FGF23 produced by bone osteocytes has phosphaturic effects and suppresses
1,25(OH)2D3, thereby providing a means to lower serum phosphate in a PTH-independent manner. (From Stubbs J, Liu S, Quarles LD.
Role of broblast growth factor 23 in phosphate homeostasis and pathogenesis of disordered mineral metabolism in chronic kidney
disease. Semin Dial. 2007;20(4):302–308.)

in PTH levels, secondary to decreased serum Ca, stimu- of VDR functions as a transcription factor in the nucleus.
lates 1 -hydroxylase activity in the kidney. A low serum The VDR–1,25(OH)2D3 complex regulates transcription of
phosphorous or 1,25(OH)2D3 concentration also activates more than 60 genes by interacting with DNA sequences
1 -hydroxylase, whereas elevated 1,25(OH)2D3 levels and known as vitamin D response elements. Central to its role in
the phosphaturic FGF23 are inhibitory. Other less estab- Ca homeostasis, 1,25(OH)2D3 induces the transcription of
lished stimulators of 1 -hydroxylase activity include calci- genes coding for Ca-binding proteins.556 Vitamin D-depen-
tonin, growth hormone, insulin, insulin-like growth factor, dent Ca-binding proteins (CaBP-Ds) are found in high con-
estrogen, and prolactin.548–550 centrations in Ca-transporting tissues, such as the kidney,
In addition to the kidney, extrarenal sites of 1 -hydroxylase intestine, and placenta. CaBP-Ds bind calcium with high af-
activity have been identi ed. These include macrophages, ke- nity and are associated with Ca transport in these organs.
ratinocytes, hepatocytes, and human placenta, as well as skel- Another potential regulatory site for vitamin D signaling,
etal muscle cells551 and various bone cell preparations,552,553 other than the synthesis and degradation of 1,25(OH)2D3,
although the regulatory processes for this conversion are not is by regulation of VDR expression. Receptor regulation has
well understood. Extrarenal production of 1,25(OH)2D3 can been shown to be physiologically important, as 1,25(OH)2D3
lead to hypercalcemia in certain pathologic situations, as in signaling is dependent on both the number and occupancy
patients with active sarcoidosis or in patients with lymphoma. of VDRs on the cell surface membrane.557
The rst step in the inactivation and catabolism of vi- Aside from its biologic effects in the kidneys, calcitriol
tamin D is hydroxylation of 25(OH)D and 1,25(OH)2D by is transported by DBP to other vitamin D receptor VDR-
24-hydroxylase, which is present in the kidney, intestine, and positive target tissues (mainly bone, intestine, and para-
several other tissues that possess 1,25(OH)2D3 receptors.547,550 thyroid gland) to act in a genomic or nongenomic manner.
Importantly, kidney 24-hydroxylase and 1 -hydroxylase are Regulation of gene expression by calcitriol is mediated by
reciprocally regulated.547,550,554 1,25(OH)2D3 and hypophos- VDR and takes place within hours. By contrast, nongenomic
phatemia increase 24-hydroxylase activity whereas vitamin D responses of calcitriol are probably mediated by a speci c
de ciency and PTH (via CAMP) suppress it.554 membrane-bound VDR and occur within seconds to min-
25(OH)D has a long half-life (approximately 3 weeks) utes. Nongenomic effects of calcitriol include rapid changes
and is the best measure of vitamin D status. Calcitriol has a in phosphoinositide metabolism, increases in intracellular
short half-life (4 to 6 hours) and exists at circulating levels calcium levels, stimulation of intestinal calcium transport
1/1,000 of those of 25(OH)D. 1,25(OH)2D3 exerts its bio- and phosphate uxes, elevation in cyclic guanosine mono-
logic actions by binding to intracellular vitamin D receptors phosphate (cGMP) levels, and activation of protein kinase
(VDR), which in the unliganded form are located in the C.558 These activities have been found in many cells, includ-
cytosolic and nuclear compartments.555 Like other members ing keratinocytes, enterocytes, muscle cells, osteoblasts, and
of the steroid-thyroid family of receptors, the liganded form chondrocytes. VDR seems to be necessary for some of these
264
26 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

nongenomic transduction processes; however, another pro- worsening of hyperphosphatemia with more phosphate
tein named 1a,25-dihydroxy-membrane associated rapid re- released from bone than excreted in the kidney with di-
sponse steroid binding (MARRS) is also seemingly involved minished PTH sensitivity, ultimately leading to renal os-
in these rapid nongenomic actions.558 teodystrophy and possible calcium phosphate precipitation
in tissues and vessels with an associated increase in car-
Physiologic Actions of 1,25(OH)2 D3 diovascular events. The use of active vitamin D analogs in
1,25(OH)2D3 participates in a hormonal system that tightly an attempt to remedy secondary hyperparathyroidism may
regulates the extracellular Ca concentration.556 A decline in increase Ca while worsening hyperphosphatemia, therefore
serum Ca stimulates PTH release, which acts on the kidney increasing the Ca P product and rendering CaP precipi-
to increase production of 1,25(OH)2D3. 1,25(OH)2D3, in tation more likely. Phosphate binders can help decrease se-
turn, stimulates intestinal absorption of Ca and decreases rum phosphate levels.
its excretion by the kidneys. 1,25(OH)2D3 also acts on A new set of agents called calcimimetics have been ap-
bone to stimulate Ca mobilization.559 Normalization of se- proved in this setting (e.g., cinacalcet). They bind to CaSR in
rum Ca shuts off this cascade by suppressing 1 -hydrox- the parathyroids and increase its sensitivity to Ca, therefore
ylase activity in the kidney and release of PTH from the counteracting the excessive PTH secretion in a dose-
parathyroids. Increased 1,25(OH)2D3 also contributes to dependent manner and decreasing the Ca P product.567
turning off Ca-correcting mechanisms by inhibiting its own The presence of VDR on monocytes/macrophages and
production.560 In addition to its effects on Ca homeostasis, activated lymphocytes suggests that 1,25(OH)2D3 plays a
1,25(OH)2D3 also enhances phosphate absorption in the role in regulating the functions of these cells.568
intestine and kidney.
About 50% to 60% of ltered Ca is reabsorbed in the Renoprotective Effects of Vitamin D
proximal tubule. This process is Na-dependent, and the ma- It has been demonstrated that 1,25(OH)2D3 possesses reno-
jority of Ca is reabsorbed via a paracellular pathway.561,562 protective property against hyperglycemia-induced renal in-
Approximately 20% of ltered Ca is reabsorbed in Henle’s jury by suppressing the renal RAS. Evidence suggests that
loop, 10% to 15% in the distal tubule, and 5% in the col- 1,25(OH)2D3 is a negative endocrine regulator of renin bio-
lecting duct. Unlike the proximal tubule, Ca reabsorption synthesis and directly transrepresses renin gene transcrip-
in the distal tubule appears to be Na-independent.561 In ad- tion. 1,25(OH)2D3 suppresses renin biosynthesis in mice,
dition to VDR, distal tubule epithelial cells contain CaBP- and vitamin D de ciency stimulates renin production. VDR
Ds and ATP-dependent plasma membrane Ca pumps.563 knockout mice and those missing the 1 -hydroxylase en-
It has been postulated, therefore, that 1,25(OH)2D3 en- zyme develop hypertension and cardiac hypertrophy and
hances renal Ca reabsorption by direct action on the distal high renin levels.569 Also, diabetic mice lacking VDR devel-
tubule in a manner analogous to stimulation of Ca absorp- op more severe renal damage than wild type mice because
tion by intestinal cells.556 Several experimental studies sup- of more robust activation of the intrarenal RAS, including
port this hypothesis: Concentrations of CaBP-Ds and Ca more induction of renin and angiotensinogen.570 Deb et
transport rates in renal cells are increased by vitamin D, al. demonstrated that 1,25(OH)2D3 suppresses hyperglyce-
whereas vitamin D de ciency abolishes CaBP-D synthe- mia-induced angiotensinogen expression in the kidney by
sis and decreases Ca absorption.556 Experimental studies blocking NF- B activation of the angiotensinogen gene tran-
also suggest a role for 1,25(OH)2D3 in phosphate handling scription.571 Forman et al. showed that, among normoten-
by the kidney. In isolated perfused proximal tubule seg- sive individuals, lower 25(OH)D levels were associated with
ments, low concentrations of 1,25(OH)2D3 antagonize the higher circulating Ang II levels and a blunted renal plasma
phosphaturic action of PTH.563 In rats in which vitamin ow response to exogenous Ang II infusion, both ndings
D de ciency was induced, but the diet was manipulated consistent with activation of the RAS in the setting of lower
to maintain normocalcemia, normophosphatemia, and plasma 25(OH)D.572 Melamed et al. showed that low 25(OH)
normal PTH levels, 1,25(OH)2D3 stimulated tubular reab- D levels are associated with the development of ESRD.573
sorption of phosphate.564 It was thought that 1,25(OH)2D3 In animal studies, vitamin D and VDR agonists (VDRA) were
regulates phosphate reabsorption by direct modulation of shown to ameliorate glomerulosclerosis, glomerular hyper-
Na-P cotransport in renal tubule cells.565 However, data trophy and in ammation, podocyte hypertrophy, mesangial
on rats subjected to a low phosphate diet suggest that the proliferation, albuminuria, and interstitial brosis.574,575
Na-P cotransport in the kidney cannot be explained by the These effects may be independent of BP and PTH. In the kid-
1,25(OH)2D–VDR axis.566 ney, vitamin D may be important for maintaining podocyte
In patients with renal failure requiring dialysis, serum health, preventing epithelial-to-mesenchymal transforma-
phosphate levels increase as a result of the decreased ca- tion, and suppressing renin gene expression and in amma-
pacity of the kidney to excrete phosphate; the elevated se- tion. In human studies, CKD is associated with a very high
rum phosphate inhibits 1,25(OH)2D3 formation and leads prevalence of 25(OH)D de ciency. Emerging evidence in
to decreased serum Ca levels and to increased PTH secre- patients with CKD show that vitamin D can reduce protein-
tion (secondary hyperparathyroidism). PTH would lead to uria or albuminuria even in the presence of ACE inhibition.
CHAPTER 8 HORMONES AND THE KIDNEY 2265
65

In addition to reducing proteinuria, VDRA may reduce also FGF23 has been also implicated in the pathogenesis of
insulin resistance, BP, and in ammation and preserve podo- X-linked hypophosphatemic rickets/osteomalacia (XLH), tu-
cyte loss providing biologic plausibility to the notion that the mor-induced osteomalacia (TIO), and autosomal-dominant
use of VDRA may be associated with salubrious outcomes in hypophosphatemic rickets/osteomalacia (ADHR)—all enti-
patients with diabetic nephropathy.575 ties with common features, including hypophosphatemia
because of renal phosphate wasting and impaired mineral-
Fibroblast Growth Factor 23 ization of bone with normal serum Ca and PTH.579,580
FGF23, a 30-kDa protein primarily synthesized by osteo-
blasts and osteocytes, controls renal phosphate excretion
by regulating renal Na-dependent phosphate cotransport- EICOSANOIDS
ers (NaPi2a and NaPi2c). It is phosphaturic and decreases The eicosanoids are a group of locally acting hormones or
1 -hydroxylase levels by a VDR independent mechanism, autacoids that are derived from dietary polyunsaturated fatty
and induces 24-hydroxylase activity, therefore reducing acids. In humans, arachidonic acid, an essential fatty acid
1,25(OH)2D3 by a VDR-mediated mechanism.576 In vivo esteri ed into cellular membrane phospholipids, is the most
studies have shown that FGF23 is one of the most potent abundant and important precursor. After deesteri cation by
phosphatonins that induces renal phosphate wasting and re- phospholipases, free arachidonic acid may either rapidly re-
duction of 1,25(OH)2D3.577 Another unique characteristic of esterify into membrane lipids, avidly bind intracellular pro-
FGF23 is that this molecule derives from bone and exerts its teins, or undergo enzymatic oxygenation to yield the various
hormonal effects in the kidney despite the ubiquitous pres- biologically active molecules referred to as eicosanoids. The
ence of its receptors (FGFRs). type of product formed depends on the enzymes involved in
FGF23 directly acts on parathyroid glands and attenu- the oxygenation process (Fig. 8.10).581 Oxygenation of ara-
ates secretion of PTH in the presence of Klotho, an anti- chidonic acid by cyclooxygenase results in prostaglandin and
aging protein, as a cofactor. Klotho mutant mice display a thromboxane (TX) synthesis. Oxygenation by lipoxygenase
phenotype identical to that of FGF23 null mice, both of generates hydroxyeicosatetraenoic acids and leukotrienes.
which are characterized by premature aging–related phe- These two major enzymatic pathways are all expressed in the
notypes associated with hypercalcemia, hyperphosphate- kidney.582,583 The speci c nature of the products generated
mia, and paradoxically high 1,25(OH)2D levels. Klotho is varies with both cell type and initial stimulus for arachidonic
predominantly expressed in the distal tubule of the kidney. acid release. Eicosanoids have diverse biologic effects in the
Aside from its function as a cofactor for the stimulation of kidney, the signi cance of which will be discussed later.
FGF-23, it also colocalizes with epithelial Ca channel tran-
sient receptor potential vallinoid-5 (TRPV5). Mice overex- Cyclooxygenase Products
pressing the Klotho gene age slowly through a mechanism
that involves insulin and oxidant stress resistance.574 Klotho
Prostaglandins
is expressed in limited tissues such as the kidney, parathy- Prostoglandins (PGs) are a unique group of cyclic fatty acids
roid, and pituitary gland.577 with diverse biologic effects that are produced throughout
The identi cation of FGF23 and Klotho as a physi- the body. The kidney is a major site of PG production, me-
ologic regulator of phosphate and vitamin D metabolism tabolism, and action.584,585 PGs are important modulators
has considerably advanced the understanding of the min- of renal function in both physiologic and pathophysiologic
eral and bone disorder in CKD. It is now clear that FGF23 settings. The spectrum of their effects in the kidney encom-
plays a central role in the pathogenesis of altered mineral passes modulation of RBF, GFR, salt and water transport,
metabolism and secondary hyperparathyroidism in CKD and the release of renal hormones. It is within the setting of
patients.577 The primary systemic stimuli of FGF23 secre- compromised renal status that maintenance of renal function
tion are increased 1,25(OH)2D levels and increased dietary is most dependent on PGs. Under these circumstances, inhi-
phosphorus intake. In kidney failure, FGF23 levels increase bition of PG synthesis with nonsteroidal anti-in ammatory
early and steadily rise with progression of kidney disease, drugs (NSAIDs) is likely to impair renal function.586
likely as an appropriate physiologic adaptation to maintain
normal phosphorus balance by helping to augment urinary Structure and Synthesis of Prostaglandins Arachidonic
phosphate excretion in conjunction with increased PTH lev- acid (eicosatetraenoic acid) is the major substrate for the syn-
els and by decreasing gut phosphorus absorption through thesis of PGs in humans. The initial step is catalyzed by cy-
decreased 1,25(OH)2D. In the long term, this compensation clooxygenase (COX), a major therapeutic target of analgesic,
may become maladaptive by causing a progressive decline in antipyretic, and anti-in ammatory actions of NSAIDs. COX
1,25(OH)2D levels with attendant consequences such as sec- converts arachidonic acid to PGH2, which is subsequently me-
ondary hyperparathyroidism. Moreover, excess FGF23 levels tabolized by various PG synthases to more stable, biologically
have been independently linked with cardiovascular disease active, prostanoids, namely PGE2, prostacyclin (PGI2), PGF2 ,
and mortality, suggesting that chronically elevated FGF23 PGD2, and thromboxane A2 (TxA2) (Fig. 8.10). For a de-
levels may directly contribute to adverse CKD outcomes.578 tailed description of the biosynthetic pathways leading to the
266
26 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Membrane Phospholipids

90% recycled phospholipases


(deacylate arachidonic acid
from phospholipids)
oxidation products
epoxides and Arachidonic acid Hydroperoxy fatty acids
cytochrome P-450 lipoxygenases
diol derivatives
monooxygenases
cyclooxygenases 5-lipoxygenase 15-lipoxygenase
pathway pathway

Endoperoxides
PGG2 Leukotriene A4 15-HETE
PGH2
Isomerase synthetase
hydrolase synthase 12-LO 5-LO and
or reductase hydrolase
Prostaglandins Prostacyclin Thromboxanes LTB4 LTC4/D4/E4 Lipoxins
(E2,F2o,D2) (PGI2) (TxA2)

FIGURE 8.10 Renal eicosanoid synthesis. The oxygenated products of arachidonic-acid (eicosatetraenoic-acid) metabolism are
referred to as eicosanoids. These include lipoxygenase, cytochrome P450 monooxygenase, and cyclooxygenase products. The lipoxy-
genase pathway yields hydroxy fatty acids and leukotrienes. The cytochrome P450 monooxygenase pathway yields -oxidation prod-
ucts and diol derivatives. The cyclooxygenase pathway yields the prostanoids, which include the prostaglandins (PGE2, PGF2, PGD2,
and prostacyclin) and thromboxane. The most important prostaglandins are dienoic (i.e., possessing two double bonds outside the
ring structure)—hence the subscript 2.

different types of PGs and thromboxanes, collectively called donate tissue stores vary with dietary intake of essential fatty
prostanoids, the reader can refer to following reviews.587,588 acids and can be depleted when intake is de cient.602 Fish oil
The COX pathway is also the major pathway for arachi- diets (rich in omega-3 polyunsaturated fatty acids) will com-
donic acid metabolism in the kidney.582,583 In both animals pete for the arachidonate oxidation process and inhibit forma-
and humans, two separate COX enzymes have been identi- tion of active products.603 Table 8.5 lists several modulators of
ed that are encoded by two separate genes: COX-1 (589) the key steps involved in PG synthesis. Although under basal
and COX-2 (590). The human COX-1 enzyme is constitu- conditions both COX-1 and COX-2 contribute to prostanoid
tively present in the renal vasculature (arterial and arteriolar production in the kidney, some stimuli have been shown to
endothelial cells, mesangial cells),583,587,591–593 glomerular initiate a more selective response. Evidence indicates that
epithelial cells,594 renal interstitial cells,587,595 along most Ang II, through AT1 receptors, increases MD COX-2 expres-
segments of the tubule, although in markedly varying con- sion.604,605 In addition, conditions associated with activation
centrations,596–598 and in the medullary and papillary col- of the renin-angiotensin system, such as a low-salt diet or
lecting ducts in humans, monkeys, dogs, rabbits, and rats.587 diuretic administration, all signi cantly increase MD COX-2
The COX-2 enzyme, rst thought to be only inducible in expression.606,607 Recently, pressure was also shown to be an
in ammatory responses, was found to be constitutive in the important promoter of renal COX-2 expression in renal med-
kidney.599 Its expression is consistently focal and limited to ullary interstitial cells subjected to mechanical stress in vitro
the MD of the juxtaglomerular apparatus, epithelial cells of as well as in rats subjected to ureteral obstruction.608 In con-
the thick ascending limb, and papillary interstitial cells of trast to COX-2, cortical expression of COX-1 is independent
rats, rabbits, and dogs. In mice, COX-2 expression varied from Ang II but relies on hemodynamic changes.604 Several
with developmental stages, with high levels of expression in renal pathophysiologic states such as glomerulonephritis, in
the MD and thick ascending limbs of fetal kidneys and mini- addition to ureteral obstruction, are associated with increased
mal expression upon renal maturation, suggesting a puta- prostanoid production.609–611
tive role for COX-2 in nephrogenesis.600 In the adult human Prostanoids are rapidly degraded, which limits their ef-
kidney, expression of the COX-2 protein has been observed fects to the immediate vicinity of their site of synthesis and
in endothelial and smooth muscle cells of arteries and veins, accounts for their autocrine or paracrine function. They me-
and intraglomerularly in podocytes, but not in MD cells.587 diate diverse actions, in part related to their site of synthesis
It is noteworthy that the distribution of prostanoid synthases and the cells on which they act (Tables 8.6 and 8.7). Their
within the kidney remains incompletely characterized.601 principal physiologic role is mediation and/or modulation
of hormone action at these locations.583,587,592,612,613 Thus,
Physiology of Prostaglandins in the Kidneys The rate of cortical production by arterioles and glomeruli is related
PG production is dependent on the release of free arachidonic to regulation of RBF, GFR, and renin release. Other cortical
acid from tissue stores by phospholipase A2 (PLA2). Arachi- sites of PG production affect ammoniagenesis614 and calcium
CHAPTER 8 HORMONES AND THE KIDNEY 2267
67

TA B L E respectively interact with PGE2, PGF2 , PGI2, or TxA2


(Fig. 8.1). Multiple subtypes of each of these prostanoid re-
8.5 Modulators of Prostaglandin Synthesis a ceptors may exist, as in the case with the PGE2 receptor (EP
receptor), thus explaining the apparently contrasting effects
mediated by PGE2 on smooth muscle and collecting duct
Modulator Site of Action permeability to water.619 The differential sensitivity of tis-
Promoters sues to several structural PGE analogs has led to the identi-
cation of at least four distinct EP receptors: the two vaso-
Angiotensin II PLA2
dilator receptors, EP2 and EP4, and the two vasoconstrictor
AVP PLA2
receptors, EP1 and EP3.620 EP1 receptors signal mainly by
Bradykinin PLA2
IP3-mediated increased intracellular Ca2 .621–623 In contrast,
Norepinephrine PLA2
the vasodilator receptors EP2 and EP4, signal through in-
PAF PLA2
creased cAMP.624–626 EP3 receptors constrict smooth muscle,
Interleukin-1 PLA2 and COX
probably by inhibiting cAMP generation via a pertussis
TNF- PLA2
toxin-sensitive, Gi-coupled mechanism.627,628 In mesangial
PDGF COX
cells, the PGF2 receptor (FP receptor) seems to be coupled
EGF COX
to increased intracellular Ca2 . At higher concentrations,
Calcium PLA2
PGF2 also stimulates EP receptors.629,630 The TXA2 recep-
Diabetes PLA2
tor (TP receptor) appears to signal via phosphatidylinositol
Ischemia PLA2
hydrolysis, leading to increased intracellular Ca2 .631 There
Chronic AVP therapy COX
is pharmacologic evidence for existence of TP receptors in
Ureteral obstruction COX and TX synthase
the glomerulus.629 The PGI2 receptor (IP receptor) signals
Venous obstruction COX
via stimulation of cAMP generation.632,633 PGI2 has been
Glomerulonephritis COX
demonstrated to play an important vasodilator role in the
Nephrotic syndrome TX synthase
glomerular microvasculature, where the effects of PGI2 and
Inhibitors PGE2 to stimulate cAMP generation were distinct and ad-
ditive.634 Thus, because multiple PGs can be synthesized
Glucocorticolds PLA2 and COX
through the COX pathway and because these PGs can inter-
Potassium PLA2
act with different receptors, their pathophysiologic effects
Urea PLA2
are further diversi ed and depend on which prostanoid is
Mepacrine PLA2
produced and which receptor is available locally.
NSAIDs COX
a
Note that hormones are important physiologic modulators of prostaglan- Renal Hemodynamics There are some species differences in
din production. AVP, arginine vasopressin; PAF, platelet-activating factor; the renal actions of PGs, and this must be taken into account
TNF- , tumor necrosis factor- ; PDGF, platelet-derived growth factor; when extrapolating data from animals to humans. Although
EGF, epidermal growth factor; NSAIDs, nonsteroidal anti-in ammatory acute inhibition of PG synthesis does not change arterial
drugs; PLA, phospholipase A; COX, cyclooxygenase; TX, thromboxane.
pressure in normal circumstances, it does produce both an
increase in renal vascular resistance and a decrease in sodium
and water excretion.635–637 In general, PGE2 and PGI2 are va-
and phosphate transport.615 Medullary PG production is di- sodilators in most species, whereas TXA2, PGF2 , and PGE2
rected to regulating vasa recta blood ow, tubular sodium (in certain circumstances) are vasoconstrictors.584,638,639 The
and chloride transport, and the response of the collecting contribution of these vasoactive properties of COX products
duct to vasopressin. Inhibition of COX activity in the ab- to the regulation of renal vascular tone under normal physi-
sence of exogenous administration or endogenous release of ologic conditions is probably minimal.640–645
hormones such as Ang II, NE, or vasopressin has little effect In contrast, the local release of vasodilator PGs (PGE2
on renal functional parameters.616 Once their local release is and PGI2) in response to renal vasoconstrictors plays an
enhanced, COX products may themselves stimulate the local important role in maintaining RBF and GFR. There is com-
generation of other hormones. Under pathophysiologic con- pelling evidence indicating that mesangial cell synthesis
ditions, such as in ammatory injury, local release of pros- and release of PGE2 and PGI2 modulate the constrictor ac-
tanoids may mediate some of the functional derangements tions of Ang II, NE, and AVP.583,584,587,629,646 Activation of
that characterize these conditions.609–611 the renin-angiotensin and sympathetic nervous systems
Prostanoids act through speci c and distinct re- leading to enhanced release of angiotensin, catecholamines,
ceptors.617,618 These receptors are members of the G and AVP occurs in conditions, such as hemorrhage, volume
protein-coupled family of receptors. In the kidney, they depletion, general anesthesia, cirrhosis, and cardiac failure.
mainly include the E-prostanoid (EP), F-prostanoid (FP), While serving to maintain the systemic blood pressure,
I-prostanoid (IP), and T-prostanoid (TP) receptors, which these hormones constrict mesangial cells and glomerular
268
26 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

TA B L E

8.6 Renal Actions of Eicosanoids

Product Pathway
Action Cyclooxygenase P450 Epoxygenase Lipoxygenase
Vascular
Constriction TXA2 5,6-EET LTD4, LTC4
20-HETE LXA4, LXB4
Dilation PGE2, PGI2 5,6-EET LXA4, cyclooxygenase-dependent
Mesangial
Contraction TXA2, PGF2 LTD4, LTC4 LTC4
Relaxation PGE2, PGI2
Mitogenic PGF2 , TXA2
Antimitogenic PGI2, PGE2
Na transport
Inhibition PGE2 5,6-EET
Na-K-ATPase
Inhibition PGE2 11,12-DHT
Stimulation 19-HETE
Water transport
Inhibition PGE2 EETs, DHTs

TXA, thromboxane A; PGE, prostaglandin E; PGI, prostaglandin I; PGF, prostaglandin F; EET, epoxyeicosatrienoic acid; HETE, hydroxyeicosatetraenoic
acid; DHT, dihydrotestosterone; LTD, leukotriene D; LTC, leukotriene C; LXA, lipoxin A; LXB, lipoxin B. See text for references.

TA B L E

8.7 Renal Actions of the Different Prostanoids

Mediator Main Source Primary Effects


PGE2 Tubular epithelial cells Renal vasodilation
Interstitial cells Relaxation of mesangial cells
Modulation of glomerular capillary ultra ltration coef cient
Stimulates renin release from juxtaglomerular apparatus
Antagonizes hydroosmotic effect of ADH in collecting tubular
epithelial cells
Inhibits sodium chloride reabsorption
Mediates renal response to loop diuretics
PGI2 Vascular and glomerular Renal vasodilation
endothelial cells Relaxation of mesangial cells
Stimulates renin release from juxtaglomerular apparatus
PGF2 Mesangial cells Contracts smooth muscle
Glomeruli
TXA2 Glomeruli Contracts smooth muscle
Contracts mesangial cells

ADH, antidiuretic hormone; TXA, thromboxane A; PGE, prostaglandin E; PGI, prostaglandin I; PGF, prostaglandin F.
CHAPTER 8 HORMONES AND THE KIDNEY 2269
69

arterioles. Fortunately, their enhancement of renal PG release in the thick ascending limb probably involve inhibition of
locally opposes their constrictor effects. The vasodilatory ac- adenyl cyclase.667 Through Gi-coupled EP3 receptor ex-
tion of PGs on the afferent arteriole serves to maintain renal pressed in the thick ascending limb,668,669 PGE2 also blocks
perfusion, whereas their relaxant effects on mesangial cells the phosphaturic action of PTH in the proximal tubule.670
maintains the effective surface area for ltration.638 Inhibi- Recent evidence obtained from a randomized, placebo-con-
tion of PG generation in these circumstances is associated trolled, crossover study in healthy humans suggest that PGs
with a dramatic fall in RBF and GFR.645–648 Vasodilator PGs, may modulate sodium excretion through ENaC regulation
in particular PGI2, may also counteract the vasoconstrictor in the distal nephron.671 Natriuresis may also be regulated
responses to calcium in human subjects.638 In addition to by renal medullary COX-derived prostanoids acting in a
modulating the effects of vasoconstrictors, endogenous PGs paracrine manner. The prostanoid receptors regulating renal
mediate the actions of some vasodilator agents. These in- medullary blood ow through the descending vasa recta
clude a role for PGI2 in mediating the vasorelaxant actions of seem to be EP2, EP4, and/or IP receptors.672 The importance
dopamine650 and magnesium651 in humans. of EP2 and IP receptors has been con rmed to be associ-
Finally, PGs synthesized from the MD can trigger re- ated with salt-sensitive hypertension in animal models of
nin release,652 which leads to increased Ang II levels. Ang EP2 and IP receptor de ciency.673,674 Expression of EP3 in
II preferentially constricts the glomerular efferent arteriole, the renal medulla was shown to be induced by hypertonic-
thus increasing intraglomerular pressure and, ultimately, ity, suggesting a role for this receptor as well in natriuresis
maintaining GFR in volume-contracted conditions.653 This and protection of the renal medulla. EP3 activation leads to
response is further reinforced by the PGE2-induced afferent inhibition of Na -K -2Cl– transporter and aquaporin 2.675
vasodilation. In contrast, TXA2 exerts a negative effect on re- Sodium loading is associated with an increase in urinary
nin release.654 However, inhibition of COX activity reduces PG excretion, yet PGs are not important in the regulation
plasma renin activity, suggesting that the predominant in u- of sodium balance in normal euvolemic subjects. However,
ence of prostanoids is stimulatory. PG-mediated renin release in circumstances associated with sodium retention and com-
is independent of -adrenergic mechanisms.655 It has been promised renal function, PGs play a signi cant role. In fact,
recently reported that a decrease in extracellular tonicity leads high-salt diet increases renal medullary COX-2 and micro-
to renin release through a mechanism involving aquaporin somal prostaglandin E synthase-1 (mPGES-1) expression to
1–mediated water in ux in juxtaglomerular cells and PG- maintain blood pressure homeostasis.676–678 Inhibition of PG
dependent formation of cAMP and activation of PKA.645 synthesis or blocking their effects in such conditions is as-
sociated with sodium retention.647,679,680 Administration of
Solute Excretion Infusion of arachidonic acid or the COX furosemide is associated with increased PG excretion, which
products PGE2 or PGI2 directly into the renal artery results is, in part, mediated by a direct action on tubular cells.681,682
in natriuresis.656,657 Natriuresis is largely a direct tubular phe- Administration of NSAIDs diminishes the natriuretic action
nomenon originating in the distal nephron.657 PGE2 has mild of furosemide and other loop diuretics, suggesting a role for
or no effects on sodium transport in the proximal tubule and PGs in mediating the action of these agents. This cannot be
most segments of the ascending limb of Henle, with the ex- the sole mechanism, however, for the natriuretic response to
ception of the medullary thick ascending limb in some spe- diuretics outlasts the increase in PG excretion.
cies.658 This lack of effect is in keeping with both the low
rates of PG production and the low density of PG receptors in Water Excretion PGs, especially PGE2, affect water trans-
these nephron segments.597,598 Under normal circumstances, port in the collecting duct in many ways. PGs may indirectly
inhibition of COX does not result in alteration of sodium de- regulate water excretion by a reduction of the corticomedul-
livery out of the loop of Henle to the early distal tubule.659 lary osmotic gradient via inhibition of solute transport in
PGE2, however, has signi cant effects on sodium transport in the thick ascending limbs or by increase in renal medullary
the collecting duct, where it inhibits transepithelial sodium blood ow. In fact, during physiologic stress, PGE2 dilates
transport.656,658 In fact, in most mammalian species, the col- descending vasa recta, thereby buffering the constrictor ef-
lecting ducts are the major nephron segments responsible for fects of Ang II, AVP, and catecholamines, which is vital to pre-
PG synthesis597,598 and, along with the MTAL, express the vent hypoxic damage to renal medullary cells.683 Moreover,
majority of receptors for PGE2 in the kidney.660,661 PGs of the E series blunt the hydraulic conductivity response
There is evidence that PGE2 exerts its inhibitory effect of the collecting duct to AVP.684,685 In fact, in vivo infusions
on rabbit CCD sodium transport by at least two mechanisms. of arachidonic acid or PGE2 induce water diuresis, whereas
The rst involves inhibiting principal cell basolateral Na - inhibition of PG synthesis potentiates the urinary hyperos-
K -ATPase activity662–664 and the second by directly decreas- molality caused by AVP.686 However, in the absence of va-
ing the open probability of the apical amiloride-sensitive sopressin, basolateral PGE2 actually increases osmotic water
sodium channels.665,666 PGE2 utilizes multiple signal trans- reabsorption.687,688 These effects on water conductivity in the
duction pathways in the CCD. These include increase in in- collecting ducts have been explained by changes in cAMP ac-
tracellular Ca2 , activation of PKC, and modulation of cAMP cumulation. AVP mediates the increase in water conductivity
levels.584 The inhibitory effects of PGE2 on sodium transport in the collecting duct through increased cAMP generation.
270
27 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

Studies on the effect of PGE2 on cAMP metabolism in this output states. Studies in patients with congestive heart fail-
nephron segment demonstrated that PGE2 could both stimu- ure have con rmed that enhanced PG synthesis is crucial in
late basal cAMP generation and suppress AVP-stimulated protecting kidneys from the effects of elevated vasoconstric-
cAMP generation.689,690 The inhibitory effects of PGE2 on tor levels in these patients.706 Renal artery stenosis is another
AVP-stimulated cAMP generation and water conductivity in condition associated with increased renal PG secretion 707
the collecting duct are probably mediated through the EP3 that may locally act to enhance renal perfusion. Administra-
receptor, as discussed previously.675,690,691 In addition to af- tion of COX inhibitors in these settings with renal hypoper-
fecting water ow via modulation of cAMP levels, PGE2 has fusion is associated with adverse effects on RBF and GFR.708
been shown to inhibit AVP-induced water conductivity by With regard to intrinsic renal diseases, COX products
activation of PKC and elevation of intracellular calcium.619,692 have been implicated in modulating or mediating renal
Application of basolateral PGE2 probably increases water injury (or both). Single nephron GFRs increase signi -
absorption in the collecting duct by stimulating cAMP pro- cantly after renal ablation and are associated with increased
duction.687,688 The EP4 receptor, which is found on the epithe- glomerular synthesis and urinary excretion of prostanoids
lial cells of the ureter, bladder, and collecting duct, is coupled by the remaining nephron.709,710 In this model of renal ab-
to the Gs-stimulated cAMP signaling pathway.625,693 This sug- lation, increased expression of COX-2 has been described
gests that an EP4 receptor mediates cAMP-stimulated water and inhibition of COX-2 was associated with amelioration
absorption in the collecting duct. Recently, PGE2 was shown of the renal functional changes associated with renal abla-
to mediate lithium-induced polyuria by downregulating the tion.709,710 Similar results were observed with omega-3 fatty
expression of aquaporin 2 and the Na -K -2Cl– cotransporter acid 711 or ax oil712 supplementation after renal ablation.
(NKCC2) in the medulla but not in the renal cortex.694 Selective inhibition of TXA2 synthesis is associated with an
Another facet of the PGE2-AVP interaction is that AVP increase in GFR, lessening of proteinuria, and preservation
acutely stimulates endogenous PGE2 production by the collect- of renal histology.713,714 In addition, COX-2 inhibition was
ing duct. This effect has been demonstrated in rats695,696 and also found to slow progression in the rat model of polycystic
the current consensus is that it also occurs in humans.696,697 kidney disease.715 Dietary soy protein was recently shown to
It also suggests that PGE2 participates in a negative feedback decrease production of prostanoids and expression of COX
loop, whereby endogenous PGE2 production dampens the in a model of polycystic kidney disease and to be associated
action of AVP. In agreement with a functional role for the in- with reduced disease progression.716
crease in urinary PGE2 production seen during AVP infusions Enhanced TXA2 production has been implicated in the
are numerous observations of enhanced renal concentrating pathophysiology of the intense vasoconstriction that char-
ability in animals or humans pretreated with inhibitors of PG acterizes the obstructed kidney717,718 and in mediating the
production.698,699 Given that the concentration of AVP needed decrease in RBF and GFR that occurs in the early phase of
to stimulate PGE2 production is 10 to 100 times the concen- nephrotoxic serum nephritis.609,610,719,720 In patients with lu-
tration needed to maximally stimulate water conductivity, it pus nephritis, an inverse relation between TXA2 biosynthesis
is controversial whether AVP plays a physiologic role in PGE2 and GFR has been proposed.721,722 In this setting, renal func-
generation.700 At these concentrations, AVP has been shown tion improved after short-term therapy with a TX receptor
to acutely increase intracellular calcium and activate PKC, via antagonist, but not with aspirin.721,723 In addition, adminis-
activation of phospholipase C.700,701 tration of TXA2 synthesis inhibitors or receptor antagonists
has been associated with improved renal function in animals
Other Effects on Renal Function The tubuloglomerular with allograft rejection and cyclosporine toxicity.638 Although
feedback (TGF) response is modulated by multiple autacoids basal COX-2 levels are important for podocyte survival,
and hormones including ATP, Ang II, NO, and PGs, includ- overexpression of COX-2 in podocytes leads to increased
ing PGE2, PGI2, and TxA2.702,703 Under basal conditions, albuminuria and glomerular injury, partly through activa-
MD COX-2 directly modulates TGF response, particularly tion of the thromboxane receptor.724,725 Jia et al. also recently
through TXA2,702 with little contribution of COX-1, as well as showed that cisplatin-induced renal injury is mediated by
via inhibition of nNOS-dependent NO.704 When subjected to activation of the COX-2/mPGES-1 pathway, which may offer
low-dose chronic Ang II, the increase in TGF response is me- a new therapeutic target for management of the adverse effect
diated by the local release of vasoconstrictive COX-1 PGs.705 of cisplatin chemotherapy.726
These studies suggest that: (1) both COX isoforms regulate Ischemia/reperfusion injury has been shown to be
TGF but (2) their contribution differs in various settings. worsened by COX-2 inhibitors or in COX-2 knockout mice,
suggesting that PGs protect the kidney against acute renal
injury.727 Recent studies show that EP4 and EP2 receptor
Clinical Pathophysiologic Role of Prostaglandins agonists improve renal function and/or increase survival rate
in the Kidneys of a rat model of acute renal failure, supporting a protec-
PGs, through their vasodilator effects, play a salutary role tive role of EP2 and EP4 receptors in preventing the pro-
in maintaining RBF and GFR in several prerenal conditions gression of kidney failure.728 Lithium treatment was also
such as hemorrhage, septic shock, cirrhosis, and low cardiac shown to improve outcome of renal ischemia/reperfusion
CHAPTER 8 HORMONES AND THE KIDNEY 2271
71

injury through NO and/or COX pathways and represents a Finally, chronic inhibition of COX by regular use of
promising therapeutic approach to boost renal viability and NSAIDs leads to gastrointestinal toxicity and may increase
function after ischemia/reperfusion injury in the setting of the risk of chronic renal disease, especially in older patients
transplantation.729 However, other groups reported a bene - and patients with heart disease.756–758 Selective COX-2 in-
cial effect of COX-2 inhibitors that could be acting through hibitors have been developed and have been shown to
immune modulation.730,731 These opposite ndings could be spare gastric PG production. These nontraditional COX-2
partly explained by differences in the experimental models. selective anti-in ammatory agents might have represented
The role of COX products in mediating diabetic ne- a signi cant advance for the treatment of acute and chronic
phropathy remains controversial. Vasodilator PGs may in ammatory disorders759–761; however, the safety of their
contribute to the hyper ltration that occurs in early stages use has been placed into question after two large prospective
of diabetic nephropathy, whereas TXA2 may play a role in cohorts in elderly patients showed an increased risk of car-
the subsequent development of albuminuria and basement diovascular events in patients on selective COX-2 inhibitors
membrane changes.732,733 MD COX-2 expression is increased versus those on NSAIDs.762,763 Clinical studies have also
in models of hyper ltration, such as high-protein diet and demonstrated that selective COX-2 inhibitors can even re-
diabetes. COX-2 inhibition decreases hyper ltration and duce GFR and RBF in physiologically stressed volunteers or
proteinuria, and inhibits development of glomerular sclero- patients.764,765
sis in experimental diabetes.734,735 Interestingly, inhibition of In summary, prostanoids exert diverse and complex
COX-2 in patients with type 1 diabetes mellitus reduces, but functions in the kidney under physiologic and pathologic
does not correct, hyper ltration in subjects whose baseline conditions. Further understanding of pathways involving
GFR was 135 mL/min–1/1.73 m–2.736 However, in response and regulating COX, prostanoid synthases, and prostanoid
to COX-2 inhibition, women with type 1 diabetes mellitus receptors should provide targets for pharmacologic treat-
exhibited a signi cant renal hyper ltration response suggest- ments of renal disease.
ing that women have a greater dependence on vasodilatory
PGs than men.737 A role for decreased PGI2 synthesis in type
IV renal tubular acidosis associated with diabetes mellitus LIPOXYGENASE PRODUCTS
has also been suggested.738 The increased renal production Biosynthesis and Metabolism
of TXA2 and PGI2 in type 2 diabetes has also been suggested
Lipoxygenases (LOs) comprise a family of enzymes capable
as a role for these compounds in the pathogenesis of diabetic
of mediating selective lipid oxidation.766 Enzymatic lipoxy-
nephropathy.733,739,740 Recently, the lipocalin-type PGD2
genation of arachidonic acid leads to the generation of leu-
synthase (L-PGDS) knockout mouse was shown to develop
kotrienes (LTs), lipoxins (LXs), and hydroxyeicosatetraenoic
structural changes associated with diabetic nephropathy,
acids (HETEs). Formation of these compounds is initiated by
such as glomerular hypertrophy, brosis, and basement
5-, 12-, or 15-lipoxygenase, whereby a hydroperoxy group
membrane thickening.741 Urinary excretion of L-PGDS,
is introduced onto arachidonic acid at carbon-5, carbon-12,
found at elevated levels in type 2 diabetic patients, correlates
or carbon-15, respectively, to yield the corresponding 5-,
with the progression of diabetic nephropathy and re ects the
12-, or 15-hydroperoxytetraenoic acid (HPETE). HPETEs
underlying early increase in glomerular damage.742
are unstable compounds that are transformed into the corre-
Diminished vasodilator renal PG synthesis has also
sponding 5-, 12-, and 15-HETE, which, in turn, undergo en-
been implicated in the pathogenesis of the severe sodium
zymatic modi cation leading to the generation of the various
retention that occurs in patients with the hepatorenal syn-
LTs and LXs. The 5-lipoxygenase pathway is a major route of
drome.743 Pregnancy is associated with increased glomerular
arachidonic acid metabolism in the polymorphonuclear cells
synthesis and urinary excretion of PGE2, PGF2 , and PGI2.744
and macrophages leading to the formation of 5-HETE and
Augmented renal vasodilator PG production does not appear
LTs.767–769 5-Lipoxygenase requires activation by a cell mem-
to regulate GFR and RBF in normal pregnancy; however,
brane-bound protein called the 5-lipoxygenase-activating
diminished synthesis of PGI2 has been demonstrated in hu-
protein (FLAP).770 The 15-lipoxygenase enzyme catalyzes
man and animal models with pregnancy-induced hyperten-
the production of 15-HETE and initiates another major
sion.745–747 A bene cial effect of reducing TXA2 generation,
pathway of arachidonic acid metabolism in leukocytes. In
while preserving PGI2 synthesis, by low-dose (60 to 100 mg
activated neutrophils and macrophages, sequential lipoxy-
per day) aspirin therapy has been proposed in patients at
genation of arachidonic acid at carbons-15 and -5 yields tri-
risk for pregnancy-induced hypertension.748,749 In patients
hydroxy derivatives, the LXs.
with hypertension, COX inhibition by NSAIDs is associated
with increased salt retention and resistance to the diuretic
action of thiazides and furosemide.750,751 Short-term use of Renal Actions of Lipoxygenase Products
some NSAIDs was found to increase the mean arterial pres- In the rat, 5-LOX FLAP 12-LOX mRNA are present in the
sure of hypertensive patients.752,753 On the other hand, at- glomerulus as leukotriene B4 (LTB4) and leukotriene D4 (LTD4)
tempts to treat hypertension with PG analogs have generally receptors.771 15-LOX is localized to the distal nephron only.771
been disappointing.754,755 Products of lipoxygenase are classically proin ammatory;
272
27 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

however, lipoxins, 15-HPETE, and 15-HETE exhibit anti- the peptidyl LTs, by depressing Kf and GFR.773–779 Selective
in ammatory activity.674 blockade of the 5-lipoxygenase pathway, in the course of
The LTs are potent proin ammatory molecules. LTB4 glomerular injury, is associated with signi cant amelioration
has minimal spasmogenic properties, but is the most po- of the deterioration of renal hemodynamic and structural pa-
tent chemotactic substance yet described for polymorpho- rameters.788,789 In addition, dietary deprivation of essential
nuclear cells, and promotes their activation and adhesion fatty acids, which results in arachidonic acid and eicosanoid
to the endothelium.767 It has no signi cant effects on renal de ciency, confers protection against the histopathologic
hemodynamics in normal animals, but ampli es glomerular and the functional consequences of immune-initiated in-
in ammation and proteinuria in animals with glomerulo- jury in the glomerulus.790 In hemodialysis patients, 5-LOX
nephritic injury.773 The peptidyl LTs contract vascular, pul- activity and expression are signi cantly increased in periph-
monary, and gastrointestinal smooth muscle and increase eral blood monocytes, and can be markedly suppressed by
vascular permeability to macromolecules.767 LTC4 and LTD4 polyunsaturated fatty acid supplementation.1 In human glo-
exert potent effects on glomerular hemodynamics. In rats, merulonephritis, 5-lipoxygenase and 5-LO-activating pro-
systemic administration of LTC4 leads to reduction in RBF tein (FLAP) mRNA expression have been detected in kidney
and GFR.774 Similarly, infusion of either LTC4 or LTD4 in the biopsy specimens from patients with immunoglobulin A
isolated perfused kidney results in dramatic increase in re- (IgA) nephropathy and mesangial proliferative glomerulo-
nal vascular resistance and reduction in GFR.775 LTD4 medi- nephritis and were associated with a clinically worse renal
ates these effects by causing a signi cant increase in efferent status.791 Urinary LTE4 levels are also elevated in patients
arteriolar resistance, leading to a fall in glomerular plasma with active SLE.792 A pathophysiologic role for LTs has also
ow rate (QA), and a rise in glomerular capillary hydraulic been described in experimental acute allograft rejection,793
pressure (PGC). In addition, it markedly reduces the glomer- cyclosporine toxicity, and acute ureteral obstruction.787 LXA4
ular capillary ultra ltration coef cient (Kf) and, therefore, and 15-S-HETE are also generated during experimental glo-
its overall effect is to decrease single nephron GFR.776 LTC4 merular injury and may exert salutary effects on glomerular
and LTD4 contract mesangial cells777,778 and LTD4 stimulates function by antagonizing the proin ammatory actions of
neutrophil adhesion to these cells.779 In both rats and hu- LTs.784,794–797 In animals with experimental glomerulonephri-
mans, speci c mesangial cell LTD4 receptors have been iden- tis, 5-lipoxygenase inhibition results in marked reduction
ti ed. Intracellular signaling for LTD4 in these cells involves in proteinuria and preservation of GFR.786,798 The binding
receptor-activated phosphatidylinositol diphosphate (PIP2) of 5-lipoxygenase to FLAP is a prerequisite for subsequent
hydrolysis, release of inositol phosphates, and increased in- formation of leukotrienes from arachidonic acid.799 The use
tracellular calcium concentrations.780,781 of a FLAP antagonist has been shown to reduce proteinuria
LXA4 attenuates LTB4-induced neutrophil chemotaxis and restore glomerular size selectivity in human glomeru-
and inhibits natural killer cell cytotoxicity.767,769 The effects lonephritis.800 12/15-lipoxygenase (12/15-LO) expression
of LXA4 are mediated primarily by functional high-af nity is increased in high glucose (HG)-stimulated MC and in
LXA4 receptors.782 In rat glomerular mesangial cells, LXA4 experimental diabetic nephropathy.801–803
competes with LTD4 at a common receptor whereby LXA4
mediates partial agonist–antagonist effects.783 Different
LXs display distinct effects on renal hemodynamics.769,784,785 ENDOTHELIN
In rats, LXA4 causes a selective decrease in afferent arteriolar Endothelin (ET), originally isolated from porcine aortic en-
resistance, thereby increasing RBF, glomerular capillary pres- dothelial cells, is an extremely potent and long-lasting vaso-
sure, and GFR. The LXA4-induced increase in GFR, however, constrictor.804 Initially, ET’s effect on vascular tone regulation
is partially offset by its mild effect in decreasing K.783,785 The was the focus of research. Later, other actions—such as
vasodilator actions of LXA4 are mediated by prostaglandins. ET effects on cell growth and proliferation, ion transport,
eicosanoid synthesis, renin and ANP release, brosis, and
Role of Lipoxygenase Products in in ammation—emerged as important factors linking ET
to diseases.771,805,806 The kidney is an important site of ET
Kidney Disease production and expresses a high density of ET receptors.807
LTs are increasingly recognized as major mediators of glomeru- Endothelin may, therefore, act in an autocrine and paracrine
lar hemodynamic and structural deterioration during the early manner to in uence renal hemodynamics, tubular function,
phases of experimentally induced glomerulonephritis.784,786,787 and mesangial cell biology.
Mesangial cell (MC) proliferation is a central event in
the pathogenesis of glomerulonephritis. LTD4-induced pro-
liferation of mesangial cells is modulated by LXA4. Biochemistry, Synthesis, and
Increased glomerular generation of LTB4 and peptidyl- Receptor Biology
LTs has been demonstrated in several models of glomerular The term endothelin refers to a family of homologous
injury.784,786,787 LTB4 probably worsens glomerular injury 21-amino acid vasoconstrictor peptides found in three dis-
by augmenting leukocyte recruitment and activation and tinct isoforms: ET-1, ET-2, and ET-3. In humans, ET isoforms
CHAPTER 8 HORMONES AND THE KIDNEY 2273
73

are encoded by three separate genes located on chromo- veins, glomerular arterioles, and capillaries are a rich source
somes 6, 1, and 20, respectively.808,809 The initial ET pep- of ET.20 In the glomerulus, there is evidence for ET secre-
tide translation product is a large (approximately 200 amino tion by mesangial, endothelial, and epithelial cells.823 In the
acids) isopeptide-speci c prohormone named preproen- rest of the nephron, the internal medullary collecting duct
dothelin. Posttranslational processing of this prohormone (IMCD) has been demonstrated to be a major site of ET-1
to mature ET requires two steps. The rst involves its pro- and ET-3 production.823–825
teolytic cleavage by dibasic pair-speci c endopeptidases on Normally, blood vessels produce very little ET, and the
Lys-Arg and Arg-Arg pairs, which respectively ank the N- normal circulating level of ET is extremely low.24 Secretion of
and C-terminals of the preproendothelin molecule, to yield ET by endothelial cells is controlled at the level of transcrip-
an intermediate 38- or 39-amino acid proET polypeptide. tion and these cells do not store ET for future release.810,827
The subsequent step is accomplished by proteolytic cleavage Thus ET-1 probably acts primarily as a paracrine/autocrine
of proET between Trp 21 and Val22 by a putative endothelin- mediator and not as a circulating hormone.809 ET peptide se-
converting enzyme (ECE).810 All ET isopeptides have a hair- cretion is upregulated by various humoral mediators, such as
pin loop con guration structure imparted by two intrachain thrombin, BK, insulin, Ang II, AVP, endotoxin, IL-1, TGF- ,
disul de bonds bridging amino acid residues 1 through 15 and tumor necrosis factor (TNF).828 These mediators may
and 3 through 11, the reduction of which leads to a twofold be responsible for the increase in ET observed in various
loss of biologic activity.811 The two endothelin isoforms ET-2 pathophysiologic states. Hypoxia is also an important stimu-
and ET-3 differ from ET-1 in 2 and 6 amino acid residues, re- lus for ET production.829 In the kidney, increasing osmolar-
spectively. The three ET isoforms are highly homologous in ity serves as a stimulus for tubular production of ET.830 On
their amino acid sequences and tertiary structure to certain the other hand, NO, ANP, and prostacyclin exert inhibitory
scorpion and snake venoms, the sarafotoxins, which sug- in uences on ET synthesis and release.831 In summary, me-
gests common genetic evolutionary origins.812 Although all sangial cells’ ET-1 release is under complex regulation, with
isoforms of ET are potent vasoconstrictors, there are signi - vasoconstrictor, pro brotic, in ammatory, and proliferative
cant cell- and tissue-speci c differences in the secretion of, agents augmenting its release, whereas vasorelaxant agents
and biologic responses to, different isoforms.813,814 tend to inhibit its production.832
ECEs are metalloproteases that belong to the M13 group Endothelins exert their actions through two major re-
of proteins, a family including several proteins such as neu- ceptor subtypes known as ETA and ETB receptors. These be-
tral endopeptidases, X-converting enzyme, ECEs, and oth- long to the superfamily of G-protein coupled receptors and
ers.815 Three major ECE isoforms have been identi ed to have been identi ed in a variety of tissues.833 A third type of
date and named ECE-1, ECE-2, and ECE-3, of which ECE- ET receptors, ETC, has been cloned from Xenopus laevis.834
1 and ECE-2 are the most prominent.820 The ECEs differ Both ETA and ETB receptors have widespread distribution
from each other regarding cell/tissue distribution, localiza- and are abundantly expressed in the kidneys. The ETA re-
tion, pH of optimal activity, and substrate speci city.815 Four ceptor is believed to be involved in vasoconstrictive and
variants of ECE-1 have been reported in humans (ECE-1a, proliferative responses to ET-1 and binds endothelins with
ECE-1b, ECE-1c, and ECE-1d), and two ECE-2 variants the following af nity: ET-1 ET-2 ET-3.809 ETB receptor,
(ECE-2a and ECE-2b).809 Both ECE-1 and ECE-2 cleave on the other hand, is a nonisofrom selective receptor and
big ET-1 more ef ciently than either big ET-2 or big ET-3. recognizes all three ET isoforms with equal af nity. Its ac-
ECE-1 is expressed ubiquitously with highest expression tivation induces transient vasodilation. The recently cloned
in endothelium, lung, ovary, testis, and adrenal medulla, ETC receptor binds preferentially ET-3 and its stimulation
whereas ECE-2 is expressed in neural tissues. ECE-3, which causes NO release.823,833,834 Both ETA and ETB receptors are
selectively cleaves ET-3, has been found in the bovine iris.816 expressed on vascular smooth muscle.
ECE-1 was located at the cell surface and on intracellular The kidney expresses abundant mRNA transcripts for
vesicles.817 ECE-1 also hydrolyzes BK, substance P, and in- ETA and ETB receptors.823,833,835 Expression of ET recep-
sulin.809 Not all the enzymes responsible for the nal step tors is especially prominent in the renal artery, glomerular
of posttranslational processing of ET-1 have been identi ed. arterioles, endothelium, mesangium, vasa recta bundles,
Current evidence suggests that other proteases may be in- and collecting duct.771,823,833 Both receptor subtypes are
volved in the nal processing of ET, because mice lacking expressed in the glomerulus. Vascular smooth muscle cells
ECE-1 and ECE-2 can still produce mature ET-1.818 In ad- of the arcuate arteries and the renal medullary interstitial
dition, alternative, ECE-independent pathways have been cells display ETA receptors. Epithelial cells of the cortical,
suggested possibly involving tissue chymases and non-ECE inner medullary, and outer medullary collecting ducts have
metalloproteinases.819 Recently, several inhibitors speci c for ETB receptors.823
the ET-converting enzyme have been reported.820 ET receptor activation leads to diverse cellular respons-
Initial studies identi ed ET on the basis of its release es involving a chain of receptor-mediated ampli cation of
from large-vessel endothelial cells. Since then, ET immu- effectors. At the ETA receptor, these responses include the
noreactivity has been detected in the kidney, spleen, skel- phospholipase pathway (PLC) leading to the formation of
etal muscle, and lung.821 In the kidney, the arcuate arteries, inositol triphosphate and diacylglycerol and to the release of
274
27 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

stored calcium into the cytosol. This causes cellular contrac- water transport in the IMCD.805,844 In addition, a natriuretic
tion and vasoconstriction. ET-mediated effects persist after role for ET-1 is demonstrated in animals through ETB recep-
dissociation of ET from its receptor, probably because of per- tors engaging NO and leading to inhibition of chloride trans-
sistently high intracellular calcium or prolonged activation port in the MTAL of Henle’s loop.845 Furthermore, ET effect
of signaling pathways.815 Endothelial cells can express ETB on epithelial amiloride-sensitive sodium channel is dose-
receptors linked to formation of NO and prostacyclin and dependent including activation and inhibition through both
mediate endothelium-dependent vasorelaxation.836 In neph- ETB-mediated and non–ETB-mediated effects.846,847 The high
ron segments and other renal structures, ET mediates its ef- concentration of ET receptors in the human inner medul-
fects via a multiplicity of intracellular signal-transduction la835 and the recent observations suggesting ET production
pathways that involve phospholipase activation, tyrosine by human IMCD cells825 justify the assumption that a similar
phosphorylation of proteins, and elevation of intracellular physiologic role for intrarenal ET may also be operative in
free calcium.837 humans. ET also affects Na balance indirectly by stimulat-
ing release of ANP.848
Biologic Effects of Endothelin in the Kidney ET is well known to induce contraction of MCs in cul-
Endothelin effects on the kidney are broadly divided into ture and results of micropuncture experiments demonstrate
vascular and tubular. Endothelin is a potent renal vasocon- that ET-1 directly reduces the coef cient of ultra ltration.849
strictor, as much as 30-fold more potent in this regard than ET is also recognized as a growth factor with mitogenic ef-
Ang II.771,804,805,836,837 Indeed, the renal vasculature is more fects on MCs in culture, inducing changes in MC phenotype
sensitive than other vascular beds to the vasoconstrictive ef- and gene expression.771,805,833 Abundant evidence points
fects of ET-1.838 In the isolated perfused kidney, ET-1 admin- to a pivotal role for ET-1 in the biology and, particularly,
istration reduces GFR and causes a dose-dependent increase the pathology of the renal mesangium. The peptide is pro-
in renal vascular resistance. In whole animals, systemic ET duced by MC and can, in turn, act on MCs to elicit prolif-
infusion induces a decline in cortical blood ow, GFR, and eration, hypertrophy, contraction, and/or extracellular ma-
urine volume771,805; however, some studies have shown dif- trix accumulation. These effects are mediated in large part
ferential regulation of blood ow between the cortex and through activation of ETA and particularly involve PKC and
the medulla with exogenous ET-1 infusion, showing corti- MAPK. Excessive ET-1 production by, and action on, MCs is
cal vasoconstriction and NO-dependent medullary vaso- of pathogenic importance in glomerular damage in animal
dilation.839,840 The direct effects of ET-1 on preglomerular models of glomerulonephritis (GN), diabetes, and hyperten-
and postglomerular resistances are quantitatively similar at sion (Fig. 8.11).832
lower doses, so the glomerular transcapillary hydraulic pres- Studies with ET receptor antagonists demonstrated that,
sure and GFR are maintained.841 However, at higher doses, in the animal kidney, ET-1-induced reductions in total RBF are
a greater increase in preglomerular resistance occurs that, in mediated by ETA receptor, whereas selective medullary vasodi-
addition to a decrease in glomerular capillary ultra ltration lation is mediated by ETB receptor.850,851 In humans the situa-
coef cient (Kf), leads to a decline in GFR.841,842 Dihydro- tion is less clear. Antagonism studies have suggested that ET-1
pyridines inhibit ET-mediated vasoconstriction exclusively effects through ETA receptors may not be major contributors to
in the afferent arterioles.841 Renal hemodynamics may also the maintenance of renal vascular tone in health, whereas ET-1
be in uenced by indirect effects of ET, such as modulation of vasodilatory effects through ETB receptors are important.852
arachidonic acid metabolism and renin release.842 Local gen-
eration of prostaglandins (PGs), such as PGF2 , may medi- Endothelins in Kidney Disease
ate some of the vasoconstrictor effects of ET.843 ET-1 directly Endothelins have been implicated in a variety of diseases
inhibits renin release, but the net renin secretory response including cardiovascular (CV) and renal diseases, neopla-
in vivo varies with the ET-1 dose, as well as with the state of sia, wound healing, and others. ET-1 has been implicated
activation of intrarenal baroreceptors and the MD-mediated in the pathophysiology of congestive heart failure, showing
pathway.842 In humans, similar effects have been shown on cardiac pro-arrhythmic effects and promoting brogenesis
renal vasculature, with renal vasoconstriction and reduction and ventricular remodeling.853,854 Despite its central role in
in RBF and GFR, whereas no studies addressed the effect of CV disease pathogenesis, antagonism did not show any clear
ET-1 on intrarenal distribution of blood ow.838 bene cial effect.815
At the tubular level, ET-1 seems to play an important role The role of ET-1 in the pathogenesis of hypertension
in volume regulation. Despite compromise of RBF and GFR, has been evoked by several studies showing a cross-talk be-
infusion of nonpressor doses of ET in animals is associated tween all pressor and vasoconstrictor systems including ET,
with an increase in urinary ow and Na excretion.771,805 In the renin-angiotensin system, and the sympathetic nervous
addition, studies in the isolated perfused kidney have shown system.815 Collectively, study results suggest that dysregu-
that ET increases Na excretion despite a dramatic decline in lation of the endothelin system contributes to multisystem
GFR.805 These effects on Na and water balance are largely complications of hypertension.855–857
due to the ability of ET to reduce Na -K -ATPase activity At the renal level, various diseases were associated with
and reversibly inhibit AVP-stimulated cAMP generation and a dysfunctional endothelin system, including progression
CHAPTER 8 HORMONES AND THE KIDNEY 2275
75

ETRA ETRB

COX2
MAPK PGE2 Ca2+ (intracellular release)

CA2+ MMP-2 NO
c-fos, AP-1
ET-1 synthesis
cGMP
Extracellular
Proliferation matrix
accumulation

Contraction

FIGURE 8.11 Schematic representation of biologic effects mediated by endothelin receptor A (ETRA) and endothelin receptor
B (ETRB) in mesangial cells. The net effect of ETRA activation is mesangial cell contraction, extracellular matrix accumulation,
and proliferation. The net effect of activation of ETRB tends to be vasorelaxant as well as causing autostimulation of ET-1 production.
AP-1, activator protein-1; MMP-2, matrix metalloproteinase-2. (From Sorokin A, Kohan DE. Physiology and pathology of endothelin-1
in renal mesangium. Am J Physiol Renal Physiol. 2003;285:579, with permission.)

of chronic kidney disease, hypertension, and proteinuria. exacerbates proteinuria. Implicated mechanisms include in-
Declining renal function is associated with an increase in creased glomerular capillary hydrostatic pressure and per-
plasma ET levels.858 Patients undergoing hemodialysis have meability.838 Chronic selective ETA receptor blockade was
higher ET levels than do patients undergoing peritoneal associated with reduction of microalbuminuria in diabetic
dialysis or uremic patients not yet on renal replacement patients.62 Renal ischemia, on the other hand, is a potent
therapy.859 Erythropoietin administration, as well as acute stimulus for ET-1 production.63 ET-1 is upregulated after
volume contraction during hemodialysis, may contribute renal ischemia/reperfusion injury and may contribute to
to the elevation of ET-1 level in patients undergoing he- the injury by prolonging vasoconstriction. It is known that
modialysis.860 Although the signi cance of elevated plasma ETA-selective, but not ETB-selective, antagonist is protective
levels of a primarily autocrine or paracrine hormone (ET) against renal IR injury.64 ET-1 aggravates cell damage through
remains questionable at present, ET-secreting heman- the activation of Na /Ca exchanger suggesting that Ca
gioendotheliomas have been shown to produce marked may play a critical role in hypoxia/reoxygenation-induced
hypertension.861 renal tubular injury.65 Other renal diseases involving the
A pathophysiologic role for ET has been reported in ET system include: radiocontrast-induced renal injury,66,823
several conditions affecting the kidney.805,823,862 Urinary ET-1 cyclosporine-mediated nephrotoxicity,67 immune nephritis,
excretion increases in patients with several forms of chronic and the rat remnant kidney model.68 However, data support-
progressive glomerulopathies.862 MCs’ ET-1 production is ing a role for ET antagonists in the treatment of acute or
increased in rat models of immune complex GN, nephro- chronic kidney disease in humans are currently lacking.815
toxic serum nephritis, and mesangial proliferative GN. In In summary, ET is a potent vasoconstrictor peptide.
addition, MC ET-1 production is augmented in human SLE In the kidney, it reduces RBF and GFR, contracts mesangial
and urinary ET-1 excretion is proportional to the severity of cells, and may function as a paracrine–autocrine factor in
human mesangial proliferative GN.832 modulation of sodium and water balance. It is a potential
Although there is no direct evidence in vivo that MC mediator of growth and proliferative changes within the kid-
ET-1 production is increased in diabetic nephropathy (DN), ney. It is thought to play a pathophysiologic role in a number
there are in vitro data suggesting that MC ET-1 synthesis is of kidney diseases. ET receptor antagonists may prove to be
increased by hyperglycemia.832 Studies suggest that MC ET-1 bene cial in certain conditions.
production is enhanced in DN and that excessive ET-1 ac-
tion in the diabetic glomerulus can cause enhanced matrix
accumulation, proteinuria, and reduced GFR.832 NITRIC OXIDE
In animals subjected to surgical reduction of renal mass, NO is a paracrine mediator with a wide spectrum of physi-
ET-1 gene expression increases in parallel with proteinuria ologic actions, including the control of vascular tone, anti-
and glomerulosclerosis.863 Proteinuria is currently known to thrombotic actions, cell cycle regulation, neurotransmission,
be a renal and CV risk factor, and its reduction may con- signal transduction, and in ammation. NO is synthesized
fer CV protection.838 Upregulation of the renal ET system during conversion of its physiologic precursor L-arginine
276
27 6 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

(L-Arg) to L-citrulline.864,865 This reaction is catalyzed by a TGF-1 expression, was shown to be a major feature of re-
family of enzymes known as NO synthases (NOS).864,866–868 nal injury in rats with chronic inhibition of NO synthase.881
Three NOS isoforms (neuronal [nNOS, NOS1], inducible Both exogenous and endogenous NO have protective ef-
[iNOS, NOS2], and endothelial [eNOS, NOS3]) have been fects against ischemia/reperfusion-induced renal dysfunc-
identi ed in mammalian tissues. Neuronal nitric oxide syn- tion and tissue damage, probably through the suppression
thase (nNOS) was rst found in the neuronal cells of the of endothelin-1 overproduction in postischemic kidneys.882
brain, but is also constitutively expressed in the kidney and
may be of physiologic importance.867 iNOS is expressed Therapeutic Implications
upon activation of macrophages and other cells. The third Not only do ACE inhibitors decrease Ang II synthesis, they
isozyme is found in endothelial cells (eNOS) and, like nNOS, also prevent the degradation of bradykinin, which is an
is constitutive in nature.868,869 NO produced from endothe- important physiologic molecule involved in the release of
lial cells (eNOS-derived) is important as a tonic vasodilator NO. With ARBs, blocking the AT1 receptor favors the syn-
and inhibitor of platelet aggregation and adhesion. thesis of NO and stimulates factors that allow NO to be bio-
logically more active.881
Renal Action of Nitric Oxide nNOS is abundantly expressed in MD cells, and its ex-
All three NOS isoforms are expressed in the medulla and pression is stimulated in various high-renin states including
medullary NO production exceeds that in the cortex.864 salt restriction, administration of loop diuretics, or inhibition
eNOS is found predominantly in renal vasculature, whereas of the renin-angiotensin system—some of the same inter-
iNOS immunoreactivity has been shown in the preglomer- ventions that augment COX-2 expression. Direct evidence in
ular portion of the afferent arteriole871 and in glomerular support of MD NOS, presumably nNOS, acting as a positive
mesangial cells. Isolated rat proximal tubule and inner med- regulator of renin secretion came from studies in the in vitro
ullary collecting duct cells express iNOS following stimula- perfused juxtaglomerular apparatus (JGA). In this prepara-
tion with TNF- and IFN- .872 tion, administration of L-arginine-stimulated renin secretion
and NOS blockers almost completely abolished the stimula-
Studies at the Single Nephron Level tion of renin secretion by low levels of NaCl.883 Studies have
also demonstrated that tubuloglomerular feedback control
NO has been shown to participate in the regulation of renal
of afferent arteriolar resistance is in uenced by MD NO pro-
hemodynamics. The administration of a competitive inhibi-
duction, thereby enhancing autoregulation of RBF, GFR, and
tor of NO production, L-NMMA, to normal rats causes dra-
renin secretion.884
matic glomerular hemodynamic changes, including reduced
single nephron plasma ow, augmented afferent and efferent
arteriolar resistances, decreased ultra ltration coef cient, ERYTHROPOIETIN
and increased glomerular capillary pressure.873–876 Chronic Erythropoietin (EPO) is a 30.4-kDa glycoprotein hormone
oral supplementation with an L-arginine inhibitor in rats that acts on the bone marrow to stimulate red blood cell
caused proteinuria, increased glomerular capillary pressure, production.885 The kidneys produce 85% to 90% of circu-
and glomerular hemodynamic changes as described previ- lating erythropoietin in adults and the liver accounts for the
ously.875 These observations suggest that NO might be an remainder.885,886 The liver is the major source of erythropoi-
important regulator of glomerular capillary pressure and etin in the fetus.886 In situ hybridization studies performed
that its dysregulation might be involved in the development in anemic or hypoxic animals and erythropoietin-transgenic
of glomerular sclerosis through increases in glomerular cap- mice demonstrated that EPO is synthesized by peritubular
illary pressure. cells of the renal cortex, particularly at the corticomedullary
Dietary supplementation with L-arginine ameliorates junction.886–889 Other tissues were also found to produce
the progression of renal disease in rats with subtotal ne- EPO (peripheral endothelial cells, vascular smooth muscle
phrectomy,876 at least in part because of its inhibitory effects cells, neurons, astrocytes, microglia, and cardiomyocytes).
on the development of glomerular hypertension.877 EPO receptors (EPO-R), members of the cytokine receptor
superfamily, are localized in different parts of the kidney,
Role in Renal Injury as well as in the CNS, endothelial cells, solid tumors, liver,
Increasing evidence implicates a role for decreased NO and and uterus.890 In erythroid progenitor cells, EPO binds to
increased peroxynitrite production in the pathophysiol- EPO-R, leading to activation of the JAK2 and downstream
ogy of reactive oxygen species (ROS)-induced acute kidney signal transduction pathways including STAT5, PI3 kinase,
injury.878–880 Furthermore, Ang II promotes the production and MAPK.891 The main stimulus for EPO production
of O2 and Ang II synthesis is increased in the kidney when and secretion is decreased oxygen supply to renal tissue
there is a de ciency of NO. Once initiated, these events create which most commonly results from anemia or hypoxemia.
a cycle that continues to increase Ang II levels in the glomer- Decreased oxygen triggers a cascade of reactions mostly
ular circulation and to raise glomerular capillary pressure.881 mediated by the so-called hypoxia-inducible factors (HIF),
A locally activated renal RAS, in conjunction with increased which activate a wide set of genes involved in protecting the
CHAPTER 8 HORMONES AND THE KIDNEY 2277
77

kidney from hypoxia including EPO gene.892 This autocrine/ also explain the superiority of continuous peritoneal dialysis
paracrine secretion of EPO helps by decreasing apoptosis over hemodialysis in terms of decreasing blood urea nitro-
and cell necrosis in mesangial and tubular cells subjected to gen (BUN) and, at the same time, improving anemia.903
hypoxic (ischemia re ow injury) stress and toxic insult (cis-
platinum), suggesting a potential therapeutic use of EPO or
EPO analogs in tubular necrosis.893–895 Extensive evidence
INSULIN
indicates that EPO is a pleiotropic cytokine that confers Besides inhibiting gluconeogenesis in the proximal tubule,
broad tissue-protective properties as part of an innate re- insulin exerts a signi cant vasodilatory effect on the kidney,
sponse to stressors, promotes angiogenesis, and modulates thus increasing RPF; this effect appears to be mediated by
wound healing responses.896 It seems that SGK1 might PGs and partially counteracts the vasoconstrictor effect of
contribute to the mediation of EPO effects under ischemic Ang II.905 In contrast, insulin appears to enhance the contrac-
conditions.897 Conversely, when the kidney is hyperoxygen- tile effect of Ang II on mesangial cells.906 It also potentiates
ated, as occurs after red cell transfusion, EPO production is AVP action in terms of water reabsorption at the collecting
reduced. In addition to modulation by oxygen availability, ducts by increasing the gene transcription of aquaporin-2
EPO production is in uenced by several cytokines. IL-1 and (AQP2).907 In rats’ renal proximal tubule cells, insulin and
TNF- were shown to inhibit EPO mRNA levels and EPO dopamine receptors interact to regulate renal sodium trans-
formation in human hepatoma cell cultures and to reduce port.908 Studies using primarily cell culture have demonstrat-
EPO production in isolated perfused rat kidneys.898 Secre- ed that insulin can directly increase activity of the epithelial
tion of these cytokines by macrophages could contribute sodium channel, the sodium-phosphate cotransporter, the
to defective EPO production and anemia in infectious or sodium-hydrogen exchanger type III, and Na-K-ATPase.909
in ammatory diseases. Wang et al. showed that EPO has
antioxidative properties in organs affected by diabetes and C-peptide
may prevent incipient microvascular damage in the diabetic C-peptide, or connecting peptide, for a long time thought
retina.899 Schiffer et al. studied the effects of different EPO of as an inert by-product of the conversion of proinsulin to
molecules on podocyte signaling in vitro and on podocyte insulin, has revealed multiple biologic roles by partially re-
survival in an experimental model of diabetic kidney injury. versing or preventing complications of insulin-dependent
EPO activates pro-survival intracellular pathways in podo- diabetes.910 Physiologic concentrations of C-peptide in dia-
cytes in vitro, and ameliorates diabetes-induced podocyte betic rats activates pathways involved in cellular prolifera-
loss in vivo.900 tion, and limits or prevents the glomerular hypertrophy and
EPO has the potential to modulate oxygen delivery the mesangial matrix expansion seen in the posthyper ltra-
through regulation of endothelial NO production. In endo- tion phase of early diabetic nephropathy.911 In the kidney,
thelial cell cultures, although short-term exposure to EPO C-peptide reduces diabetes-induced glomerular hyper ltra-
decreases or leaves unchanged eNOS and endothelin-1 tion, albuminuria, and renal hypertrophy. C-peptide may in-
expression, the combination of EPO and hypoxia increases duce constriction of afferent arterioles in diabetic mice thus
EPO-R and eNOS expression, and nitric oxide (NO) and reducing GFR—one of the renoprotective mechanisms of
cGMP production, demonstrating a direct effect of EPO C-peptide in diabetes.912 Following in vivo administration of
on endothelial eNOS and NO production. EPO adminis- C-peptide to patients with type 1 diabetes (T1DM), micro-
tration for 14 days in healthy rats increased hematocrit as vascular blood ow to tissues and organs, including muscle,
well as eNOS expression and augmented NO-dependent skin, and kidney, is consistently augmented and likely re-
vasodilatation.901 lates to stimulatory effects on NO pathways.913 One month
In healthy volunteers, Ang II injection increases EPO treatment with C-peptide in T1DM patients results in a 6%
secretion in a dose-dependent manner. This effect is neu- decrease in GFR, and a 50% decrease in urinary albumin
tralized by a selective AT1 receptor blocker, inferring that excretion.914
the stimulation of EPO by Ang II is probably via AT1 recep- C-peptide reduces diabetes-induced hyper ltration
tors.902 It has been postulated that EPO has antinatriuretic via a net dilation of the efferent arteriole and inhibition
action, which could account for the worsening hypertension of tubular Na reabsorption, both potent regulators of the
observed in some patients receiving recombinant EPO. In glomerular net ltration pressure.915 In several studies, in-
one study in isolated Wistar rat kidneys, EPO decreased Na creased renal Na-K-ATPase activity has been linked to hy-
excretion, possibly by increasing Ang II production.904 per ltration and to the increase in oxygen consumption. Via
Cyanate, a compound that could spontaneously form inhibition of Na-K-ATPase, C-peptide may contribute to a
from urea, reacts with EPO leading to carbamylated EPO, normalization of the basal oxygen consumption of proximal
an EPO protein with reduced activity. In renal insuf ciency, tubules in diabetic animals. C-peptide may also in uence
cyanate can reach levels high enough to reduce the activity oxygenation through effects on NO release. In addition, C-
of both endogenous and exogenous EPO. This might explain peptide can improve diabetes-reduced erythrocyte deform-
one mechanism of suboptimal responses to recombinant ability. This has the potential to improve capillary blood
EPO in patients with ESRD on inadequate dialysis. It might ow, thus improving oxygen availability in the kidney, and
278
27 8 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

other affected tissues.914 In vivo, C-peptide supplementation such as erythropoietin, follicle-stimulating hormone, and
for 1 month improves body weight in STZ-induced diabet- luteinizing hormone.922,923
ic rats and decreases urinary sodium wasting.916 TGF- 1, Steroid hormones are derivatives of cholesterol and
overexpressed in renal cells in diabetes, has been suggested are typically eliminated by inactivating metabolic transfor-
as a major malefactor in the development of morphologic mations and excretion in urine or bile. Examples include
changes in diabetes by stimulating the mesenchymal for- testosterone, cortisol, and vitamin D. Their half-lives vary
mation of collagen IV, and eliciting brosis in renal tissues. considerably between a few minutes and a few hours. With
It has been reported that C-peptide has a protective effect the exception of vitamin D, the kidney plays only a minor
on early diabetic glomerular changes in response to TGF- role in the metabolsim of steroid hormones.920
1 in STZ-diabetic mice. In addition, an inhibitory effect Two major hormone groups are derivatives of amino
of C-peptide on TGF- 1-induced gene expression has been acids: thyroid hormones and catecholamines. These were
demonstrated in a mouse podocyte cell line. C-peptide is discussed in detail in previous sections of this chapter.
reported to effectively reverse TGF- 1-induced structural Thyroid hormones are inactivated primarily by intracellu-
changes in proximal tubular cells and to inhibit TGF- 1- lar deiodinases and catecholamines are rapidly degraded by
induced gene expression in a mouse podocyte cell line. The enzymes such as monoamine oxidase and catechol-O-methyl
mechanism by which this occurs is not fully understood. transferase.
It has also been reported that C-peptide, via activation of Fatty acid derivatives include mainly eicosanoids and
NF- B regulated survival genes, protects against TNF- – were discussed in other sections of this chapter. These hor-
mediated renal tubular injury.914 mones are rapidly inactivated by metabolism and have short
half-lives.
RENAL DEGRADATION OF HORMONES
Hormones fall in general into four structural groups: pep- ACKNOWLEDGMENTS
tides and proteins, steroid hormones, amino acid derivatives, The authors wish to thank Ms. Layla Carine Tannous for
and fatty acid derivatives. Peptide hormones such as insulin expert help in the preparation of the manuscript.
have generally a short half-life and are extracted by the kid-
ney, which, on average, removes between 16% and 40% of
the hormone entering the renal circulation.917 In exceptional REFERENCES
1. Maddox DA, Brenner BM. Glomerular ultra ltration. In: Brenner BM, Rec-
cases, the kinins for example, more than 90% is removed 1. Maddox DA, Brenner BM. Glomerular ultra ltration. In: Brenner BM,
tor FC Jr, eds. The Kidney, 4th ed. Philadelphia: Saunders; 1991.
during a single passage. The removal of biologically active Rector FC Jr, eds. The Kidney, 4th ed. Philadelphia: Saunders; 1991.
2. Dunn BR, Ichikawa I, P e er JM, et al. Renal and systemic hemodynamic
2. Dunn BR, Ichikawa I, Pfeffer JM, et al. Renal and systemic hemodynamic
and inactive peptides from the renal circulation occurs pre- e ects of
effects o synthetic
synthetic atrial
atrial natriuretic
natriuretic peptide
peptide in in the
the anesthetized
anesthetized rat.rat. Circ
Circ Res.
Res.
dominantly by glomerular ltration. 1986;59:237.
1986;59:237.
http://www.ncbi.nlm.nih.gov/pubmed/2945668
3. Fried TA, McCoy RN, Osgood RW, et al. Effect of atriopeptin II on deter-
The rate of ltration is in uenced by the size, shape, 3. Fried TA, McCoy ltration
RN, Osgood
minants of glomerular rate inRW,
the et
in al. E perfused
vitro ect o atriopeptin II on deter-
dog glomerulus. Am
and charge of the molecule. After ltration, peptide hor- minants o glomerular
J Physiol. 1986;250:F1119. ltration rate in the in vitro per used dog glomerulus. Am
mones are degraded in the proximal tubule via two mecha- J Physiol.
4. Myers 1986;250:F1119.
BD, Deen WM, Brenner BM. Effects of norepinephrine and angio-
4. Myers
tensin II on the BD, determinants
Deen WM, Brenner BM. E ects
of glomerular ultrao ltration
norepinephrine
and proximaland angio-uid
nisms. Larger peptides, such as insulin, require absorption tensin II on the determinants o glomerular ultra ltration and proximal f uid
reabsorption in the rat. Circ Res. 1975;37:101.
by epithelial cells and degradation in lysosomes or endo- reabsorption
5. Hura CE, in the rat. Circ
Kunau RT, Jr.Res.Angiotensin
1975;37:101. II-stimulated prostaglandin produc-
somes. Growth hormone, for example, is also absorbed http://www.ncbi.nlm.nih.gov/pubmed/1149180
tion by canine renal afferent arterioles. Am J Physiol. 1988;254:F734.
5. Harris
Hura CE, HW,Kunau RT,K,Jr.Zeidel
Angiotensin II-stimulated prostaglandin
of the produc-
by tubules where it undergoes peritubular degradation.918 tion
6.
by canine renal
Strange
a erent arterioles.
M. Current
Am J Physiol.
understanding
1988;254:F734.
cellular
biology and molecular structure of the anti-diuretic hormone-stimulated water
Smaller peptides, such as bradykinin, angiotensin, and 6. Harris
transport HW, Strange
pathway. K, Zeidel
J Clin Invest. M. Current understanding o the cellular
1991;88:1.
ANP, are degraded by hydrolysis on the brush border. Uri- biology and molecular
7. Dworkin structure
LD, Ichikawa o the anti-diuretic
I, Brenner BM. Hormonal hormone-stimulated
modulation of glomeru- water
transport
lar function. pathway. J Clin Invest.
Am J Physiol. 1991;88:1.
1983;244:F95.
nary hormone excretion accounts for less than 1% to 2% http://www.ncbi.nlm.nih.gov/pubmed/1683643
8. Breyer MD. Cellular signaling and mechanisms of vasopressin action.
of the ltered load.918 In most cases, the liver contributes In:7.Goldfarb
Dworkin S, LD, Ichikawa
Ziyadeh I, Brenner
FN, eds. BM. Hormonal
Contemporary Issues inmodulation
Nephrology:oHormones,
glomeru-
signi cantly to peptide hormone metabolism. A few hor- lar unction.
Autacoids, andAm
theJKidney,
Physiol. vol.
1983;244:F95.
23. New York: Churchill Livingstone; 1991.
8. Oliver
9. Breyer G, MD. Cellular
Schafer EA.signaling and mechanisms
On the physiological action o of
vasopressin
extracts ofaction.
pituitaryIn:
mones, however, undergo negligible hepatic extraction Gold arb
body and S,certain
Ziyadeh FN,glandular
other eds. Contemporary Issues in Nephrology:
organs: preliminary Hormones,
communication. Auta
J Physiol.
and the kidneys are their predominant site of degrada- coids, and the Kidney, vol. 23. New York: Churchill Livingstone; 1991.
1895;18(3):279.
tion.917 Examples of these hormones are calcitonin, the 9. Burbach
10. Oliver G,JP,Scha er EA.SM,
Luckman OnMurphy
the physiological
D, et al. Gene action o extracts
regulation in theomagnocel-
pituitary
bodyhypothalamo-neurohypophysial
lular and certain other glandular organs: preliminary
system. Physiol Rev.communication.
2001;81:1197– J Physiol.
1267.
amino-terminal fragment of PTH, and the C-peptide of
1895;18(3):279.
11. Majzoub JA. Vasopressin biosynthesis. In: Schrier RW, ed. Vasopressin. New
proinsulin. Complete removal of insulin is also dependent http://www.ncbi.nlm.nih.gov/pubmed/16992253
Y ork: Raven Press; 1985.
on intact renal function.919 Glomerular ltration accounts 10. Majzoub
12. Burbach JP , Luckman
JA. Vasopressin SM,biosynthesis.
Murphy D, etIn: al. Schrier
Gene regulation
RW, ed. V inasopressin.
the magnocel-
New
for approximately 60% of all the insulin removed by the lular
Y ork:hypothalamo-neurohypophysial
Raven Press; 1985. system. Physiol Rev. 2001;81:1197–1267.
http://www.ncbi.nlm.nih.gov/pubmed/11427695
13. Arima H, Oiso Y. Mechanisms underlying progressive polyuria in fa-
kidney and the remaining is extracted from the peritubu- 11. Majzoub
milial JA. Vasopressin
neurohypophysial biosynthesis.
diabetes insipidus. In: JSchrier RW, ed. Vasopressin.
Neuroendocrinol. 2010;22(7):New
lar circulation. Uptake and degradation occurs largely by York: Raven Press; 1985.
754–757.
receptor-mediated endocytosis of insulin on the basolateral 12. Grant
14. Majzoub FD,JA. Vasopressin
Reventos biosynthesis.
J, Kawabata S, et al.In: Schrier RW,
Transgenic ed. Vmodels
mouse asopressin. New
of vaso-
pressin
York: Ravenexpression. Hypertension. 1993;22:640.
Press; 1985.
membrane of tubule epithelial cells.920,921 The kidneys con- 15. Burbach
13. Arima H,JP,Oiso
Luckman SM, Murphy
Y. Mechanisms D, et al. Gene
underlying regulation
progressive in the magnocel-
polyuria in amilial
tribute signi cantly to disposal of glycoprotein hormones, lular hypothalamo-neurohypophysial
neurohypophysial diabetes insipidus.system. Physiol Rev. 2001;81(3):1197–1267.
J Neuroendocrinol. 2010;22(7): 754–757.
CHAPTER 8 HORMONES AND THE KIDNEY 2279
79

14. Bourque
16. Grant FD,CW. Reventos
CentralJ, mechanisms
Kawabata S, etofal. Transgenic mouse
osmosensation models oosmo-
and systemic vaso- 39.
45. CaiEcelbarger
Q, Nelson CA,SK,KimMcReynolds
GH, Terris J, MR,et et
al. al.
Vasopressin-mediated
Vasopressin increases regulation
expres-
pressin expression.
regulation. Hypertension.
Nat Rev Neurosci. 1993;22:640.
2008;9(7):519–531. osion
epithelial
of UT-A1, sodium
UT-A3, channel
and ER abundance
chaperone in GRP78
rat kidney. Amrenal
in the J Physiol Renal of
medulla Physiol.
mice
http://www.ncbi.nlm.nih.gov/pubmed/8406671
17. Robertson GL. Regulation of vasopressin secretion. In: Seldin DW, Giebisch G, 2000;279(1):F46.
with a urinary concentrating defect. Am J Physiol Renal Physiol. 2010;299(4):
15.The
eds. Burbach
Kidney:JP , Luckman
Physiology SM,
and Murphy D, et New
Pathophysiology. al. Gene
York:regulation in the
Raven Press; magnocel-
1992. 40. Blanchard A, Frank M, Wuerzner G, et al. Antinatriuretic e ect o vaso-
F712–F719.
lular
18. hypothalamo-neurohypophysial
Kakiya S, Arima H, Yokoi H, etsystem. al. EffectsPhysiol Rev. 2001;81(3):1197–1267.
of acute hypotensive stimuli on pressin in humans A,
46. Karakashian is amiloride
Timmer RT, sensitive,
Klein JD, thus ENaC
et al. dependent.
Cloning Clin J Am Soc
and characterization
16. Bourque
arginine CW Central
vasopressin mechanismsino the
gene transcription osmosensation
rat hypothalamus.and systemic osmo-
Am J Physiol. Nephrol.
of two new 2011;6(4):753–759.
mRNA isoforms of the rat renal urea transporter: UT-A3 and UT-A4.
regulation.Metab.
Endocrinol Nat Rev Neurosci. 2008;9(7):519–531.
2000;279(4):E886–892. http://www.ncbi.nlm.nih.gov/pubmed/21233458
J Am Soc Nephrol. 1999;10:230.
17. Gines
19. Robertson GL. Regulation
P, Abraham o vasopressin
WT, Schrier secretion. In:
RW. Vasopressin in Seldin DW, Giebisch
pathophysiological 41. Bugaj V, Pochynyuk
47. Manning M, Stoev S,O, Stockand
Chini JD.Peptide
B, et al. Activationandonon-peptide
the epithelial na+ chan-
agonists and
G, eds.Semin
states. The Kidney:
Nephrol. Physiology and Pathophysiology. New York: Raven Press; 1992.
1994;14:384. nel in the collecting
antagonists duct by vasopressin
for the vasopressin and oxytocin contributes to water
V1a, V1b, V2 andreabsorption.
OT receptors: Am J
18. Cunningham
20. Kakiya S, Arima ET,H,Sawchenko
Yokoi H, etPE. al. Re
E ects o acuteof
ex control hypotensive
magnocellular stimuli on
vaso- Physiol
ResearchRenal toolsPhysiol. 2009;297(5):F1411–1418.
and potential therapeutic agents. Prog Brain Res. 2008;170:473–512.
arginineand
pressin vasopressin gene transcription
oxytocin secretion. in the 1991;14:406.
Trends Neurosc. rat hypothalamus. Am J Physiol. 42. Pedersen NB, Ho
48. Zenteno-Savin meister MV,I,Rosenbaek
T, Sada-Ovalle Ceballos G,LL, et Effects
et al. al. Vasopressin
of arginine induces
vaso-
Endocrinol
21. KuramochiMetab. G, 2000;279(4):E886–892.
Kobayashi I. Regulation of the urine concentration mecha- phosphorylation
pressin in the heart o arethe mediated
thiazide-sensitive
by speci sodium chlorideendothelial
c intravascular cotransporter in the
receptors.
19. Gines
nisms P, Abraham WT,
by the oropharyngeal Schrier
afferent RW V
pathway inasopressin
man. Am JinNephrol.
pathophysiological
2000;20:42. distal convoluted2000;410(1):15–23.
Eur J Pharmacol. tubule. Kidney Int. 2010;78(2):160–169.
states. Semin Nephrol.
22. Vincent JL, Su F1994;14:384.
. Physiology and pathophysiology of the vasopressinergic http://www.ncbi.nlm.nih.gov/pubmed/20445498
49. Agre P. Homer W. Smith award lecture. Aquaporin water channels in kid-
http://www.ncbi.nlm.nih.gov/pubmed/7938953
system. Best Pract Res Clin Anaesthesiol. 2008;22(2):243–252. 43.J Am
ney. Sands SocJM. Mammalian
Nephrol. urea transporters. Annu Rev Physiol. 2003;65:543.
2000;11(4):764–777.
20. Morgenthaler
23. Cunningham NG. ET, Sawchenko
Copeptin: A PE.biomarker
Ref ex control o magnocellular
of cardiovascular vaso-
and renal 44. Sands Y,JM.
50. Noda Regulation
Sohara E, Ohta oE, renal urea transporters.
et al. Aquaporins in kidney J Am Soc Nephrol.
pathophysiology.
pressin and
function. oxytocin
Congest Heartsecretion. TrendsSuppl
Fail. 2010;16 Neurosc. 1991;14:406.
1:S37–44. 1999;10:635.
Nature Reviews.Nephrology, 2010; 6(3):168–178.
21. Meijer
24. Kuramochi G, Kobayashi
E, Bakker SJ, de Jong I. Regulation
PE, et al.oCopeptin,
the urineaconcentration
surrogate marker mecha-of 45. Cai Q, Nelson
51. Nedvetsky SK, McReynolds
PI, Tabor V, Tamma MR, G, etetal.al.Reciprocal
Vasopressinregulation
increasesof expres sion
aquapo-
nisms by theisoropharyngeal
vasopressin, associated with a erent pathway
accelerated renalin man. Am Jdecline
function Nephrol.in 2000;20:42.
renal trans- orin-2
UT-A1, UT-A3, and
abundance and ER chaperonebyGRP78
degradation protein in kinase
the renal A medulla
and p38–MAP o micekinase.with a
http://www.ncbi.nlm.nih.gov/pubmed/10644867
plant recipients. Transplantation. 2009;88(4):561–567. urinary concentrating de ect. Am J Physiol Renal Physiol. 2010;299(4): F712–F719.
JASN. 2010;21(10):1645–1656.
22. Favory
25. VincentR,JL,Salgado
Su F. Physiology
DR, Vincent and JL.pathophysiology
Investigationalo vasopressin
the vasopressinergic
receptor 46. Karakashian
52. Rojek A, Timmer
A, Fuchtbauer EM,RT,KwonKleinTH, JD, etet al. Severe
Cloningurinaryand characterization
concentrating
system. Best in
modulators Pract
the Res Clin Anaesthesiol.
pipeline. Expert Opin2008;22(2):243–252.
Investig Drugs. 2009;18(8):1119–1131. odefect
two in new mRNA
renal iso orms
collecting o the rat renal
duct-selective AQP2 ureaconditional-knockout
transporter: UT-A3 and mice.UT-A4.
Proc
http://www.ncbi.nlm.nih.gov/pubmed/18683471
26. Carmosino M, Brooks HL, Cai Q, et al. Axial heterogeneity of vasopressin- JNatl
Am Acad
Soc Nephrol.
Sci U S A. 1999;10:230.
2006;103(15):6037–6042.
23. Morgenthaler
receptor subtypes along NG.theCopeptin:
human and A mouse
biomarker o cardiovascular
collecting duct. Am J Physiol and Renal
renal http://www.ncbi.nlm.nih.gov/pubmed/10215321
53. Ma T, Yang B, Gillespie A, et al. Severely impaired urinary concentrat-
unction.2007;292(1):F351–360.
Physiol. Congest Heart Fail. 2010;16 Suppl 1:S37–44. 47.ability
ing Manning M, Stoev mice
in transgenic S, Chini B, etaquaporin-1
lacking al. Peptide and non-peptide
water channels. agonists
J Biol Chem.and
24. Aoyagi
27. Meijer E, T, Bakker
Izumi YSJ, de Jong PE,
, Hiroyama M, et al. Copeptin,
Vasopressina regulates
surrogate the marker o
renin- antagonists or the vasopressin and oxytocin V1a, V1b, V2 and OT receptors: Re-
1998;273(8):4296–4299.
vasopressin, is associatedsystem
angiotensin-aldosterone with accelerated renal unction
via V1a receptors in maculadecline
densain renal
cells. trans-
Am J search
54. Ma tools and potential
T, Song Y, Yang B, therapeutic agents. Prog
et al. Nephrogenic Braininsipidus
diabetes Res. 2008;170:473–512.
in mice lacking
plant recipients.
Physiol Renal Physiol.Transplantation. 2009;88(4):561–567.
2008;295(1):F100–107. http://www.ncbi.nlm.nih.gov/pubmed/18655903
aquaporin-3 water channels. Proc Natl Acad Sci U S A. 2000;97(8):4386–4391.
http://www.ncbi.nlm.nih.gov/pubmed/19696640
28. Carmosino M, Brooks HL, Cai Q, et al. Axial heterogeneity of vasopressin- 48.
55. V Zenteno-Savin
an Hoek AN, Ma T, Sada-Ovalle
T, Yang B, etI,al. Ceballos
Aquaporin-4G, et al.
is E ects o arginine
expressed vaso-
in basolateral
25. Favory
receptor subtypes R, Salgado
along theDR, Vincent
human JL. Investigational
and mouse collecting duct. vasopressin
Am J Physiol receptor
Renal pressin
membranes in the ofheart
proximalare mediated
tubule S3by speci c in
segments intravascular
mouse kidney. endothelial
Am J Physiolreceptors.
Renal
modulators
Physiol. in the pipeline. Expert Opin Investig Drugs. 2009;18(8):1119–1131.
2007;292(1):F351–360. Eur J Pharmacol.
Physiol. 2000;410(1):15–23.
2000;278(2):F310–F316.
26. Hasan
29. Carmosino KN, M, Brooks
Shoji HL, CaiS,Q,etetal.
M, Tsutaya al.Study
Axial heterogeneity
of V1a vasopressin o vasopressin-
receptor http://www.ncbi.nlm.nih.gov/pubmed/11134652
56. Sohara E, Rai T, Sasaki S, et al. Physiological roles of AQP7 in the kid-
receptor
gene subtypes
single nucleotide alongpolymorphisms
the human and in mouse collecting
platelet duct. Am
vasopressin J Physiol RenalJ
responsiveness. 49. Lessons
ney: Agre P. Homer from W. AQP7Smithknockout
award lecture. mice.Aquaporin
Biochimicawater channels inActa,
Et Biophysica kid-
Physiol.
Clin Lab2007;292(1):F351–360.
Anal. 2006;20(3):87–92. ney. J Am Soc Nephrol. 2000;11(4):764–777.
2006;1758(8):1106–1110.
27. Aoyagi
30. Aoyagi T, T, Birumachi
Izumi Y, Hiroyama
J, Hiroyama M, M, et al. Vasopressin
et al. Alterationregulates
of glucose thehomeo-
renin- 50.
57. Y Noda
asui M, Y, Sohara
Kwon E,TH, Ohta E, et al.MA,
Knepper Aquaporins in kidney pathophysiology.
et al. Aquaporin-6: An intracellu-
angiotensin-aldosterone
stasis in V1a vasopressinsystem via V1acient
receptor-de receptors
mice. in macula densa
Endocrinology. cells. Am J
2007;148(5): Nature
lar vesicleReviews.Nephrology,
water channel protein 2010;in 6(3):168–178.
renal epithelia. Proc Natl Acad Sci U S A.
Physiol Renal Physiol. 2008;295(1):F100–107.
2075–2084. http://www.ncbi.nlm.nih.gov/pubmed/20101255
1999;96(10):5808–5813.
28. Inoue
31. Carmosino M, Brooks HL,
T, Nonoguchi Cai Q, etK.al. Physiological
H, Tomita Axial heterogeneityeffectso of vasopressin-
vasopres- 51. Nedvetsky
58. Okada PI, Tabor
S, Misaka V, Tamma
T, Tanaka Y, G, et al.
et al. Reciprocal regulation
Aquaporin-11 knockoutomice aquapo-and
receptor
sin and subtypes along thepeptide
atrial natriuretic human and in themouse collecting
collecting duct.
duct. Am J Physiol Renal
Cardiovascular Res. rin-2 abundance
polycystic kidney and diseasedegradation
animals share by protein
a common kinase A and p38–MAP
mechanism kinase.
of cyst formation.
Physiol. 2007;292(1):F351–360.
2001;51(3):470. JASN.
FASEB2010;21(10):1645–1656.
J. 2008;22(10):3672–3684.
29. Jard
32. Hasan KN, Shoji
S, Gaillard RC,M, Tsutaya
Guillon G, S,
et et
al. al. Study o antagonists
Vasopressin V1a vasopressinallow receptor
demon- http://www.ncbi.nlm.nih.gov/pubmed/20724536
59. Knepper MA, Star RA. Vasopressin: Friend or foe? Nature Med.
gene single
stration of a nucleotide
novel type polymorphisms
of vasopressin receptorin platelet vasopressin
in the responsiveness.
rat adenohypophysis. MolJ 52. Rojek A, Fuchtbauer EM, Kwon TH, et al. Severe urinary concentrating
2008;14(1):14–16.
Clin Lab Anal.
Pharmacol. 2006;20(3):87–92.
1986;30:171. de60.ect Boone
in renal M,collecting
Deen PM.duct-selective AQP2 conditional-knockout
Congenital nephrogenic diabetes insipidus:mice. What Proccan
30. Caldwell
33. Aoyagi T, HK, Birumachi
WersingerJ, Hiroyama
SR, Young M, WSet al.3rd.
Alteration
The roleo ofglucose homeo-
the vasopres- Natl Acadfrom
we learn Sci Umouse
S A. 2006;103(15):6037–6042.
models? Exp Physiol. 2009;94(2):186–190.
stasis1binreceptor
sin V1a vasopressin
in aggressionreceptor-de
and other cientsocial
mice.behaviours.
Endocrinology. Prog2007;148(5):
Brain Res. 53.
61. KimMa T,GH, YangChoiB, NW,
Gillespie
Jung A, JY,etet al.
al. Severely impaired urinarynephrogenic
Treating lithium-induced concentrat-
2075–2084.
2008;170:65–72. ing ability
diabetes in transgenic
insipidus with amiceCOX-2 lacking aquaporin-1
inhibitor improveswater polyuria channels. J Biol Chem.
via upregulation of
http://www.ncbi.nlm.nih.gov/pubmed/17303660
34. Lee B, Yang C, Chen TH, et al. Effect of AVP and oxytocin on insulin release: 1998;273(8):4296–4299.
AQP2 and NKCC2. Am J Physiol Renal Physiol. 2008;294(4):F702–709.
31. Inoue of
involvement T, V1b
Nonoguchi
receptors.H,AmTomita
J Physiol.K. 1995;269(6
Physiological e ects o 1100.
Pt 1):E1095– vasopres- 54. Ma T, Song
62. Amlal Y, YangCM,
H, Krane B, etChen
al. Nephrogenic
Q, et al. Early diabetes insipidus
polyuria andinurinary
mice lackingcon-
sin
35.andMutigatrial natriuretic
K, Paliege peptide
A, Kahl in Vasopressin
T, et al. the collecting duct. Cardiovascular
V2 receptor expression along Res. aquaporin-3
centrating defect water channels.
in potassium Proc Natl Acad Sci U S
deprivation. Am A. 2000;97(8):4386–4391.
J Physiol Renal Physiol.
2001;51(3):470.
rat, mouse, and human renal epithelia with focus on TAL. Am J Physiol Renal http://www.ncbi.nlm.nih.gov/pubmed/10737773
2000;279(4):F655– 663.
http://www.ncbi.nlm.nih.gov/pubmed/11476737
Physiol. 2007;293(4):F1166–1177. 55. Van Hoek
63. Earm AN, Ma T, YBM,
JH, Christensen ang Frokiaer
B, et al. Aquaporin-4 is expressed
J, et al. Decreased in basolateral
aquaporin-2 expres-
32. Goligorsky
36. Jard S, Gaillard MS. RC,
CellGuillon
biologyG, ofetsignal
al. Vasopressin
transduction: antagonists
an overviewallowofdemon-
mem- membranes
sion and apical o proximal tubule S3 delivery
plasma membrane segmentsininkidney mousecollecting
kidney. Am J Physiol
ducts Renal
of polyuric
stration
brane o a novel
receptors, type o vasopressin
G proteins, and secondreceptor
messengers. in theIn:ratGoldfarb
adenohypophysis.
S, Ziyadeh FN, Mol Physiol. 2000;278(2):F310–F316.
hypercalcemic rats. J Am Soc Nephrol. 1998;9(12):2181–2193.
Pharmacol.
eds. 1986;30:171.
Contemporary Issues in Nephrology: Hormones, Autacoids, and the Kidney, vol 56. Sohara AM,
64. Jensen E, RaiBae T, EH,
Sasaki S, et al.
Fenton RA,Physiological roles o IIAQP7
et al. Angiotensin in the
regulates V2kid-
re-
http://www.ncbi.nlm.nih.gov/pubmed/3016500
23. New York: Churchill Livingstone; 1991. ney:
ceptorLessons
and pAQP2 rom during
AQP7 ureteral
knockoutobstruction.
mice. Biochimica
Am J Physiol Et Biophysica
Renal Physiol.Acta,
33. Knepper
37. CaldwellMA, HK,InoueWersinger SR, Young
T. Regulation WS 3rd. Thewater
of aquaporin-2 role channel
o the vasopres sin
traf cking 2006;1758(8):1106–1110.
2009;296(1):F127–134.
1b vasopressin.
by receptor inCurr aggression
Opin Cell and other social behaviours. Prog Brain Res.
Biol. 1997;9:560–564. http://www.ncbi.nlm.nih.gov/pubmed/16860289
65. Bichet DG. Nephrogenic diabetes insipidus. Adv Chronic Kidney Dis.
2008;170:65–72.
38. Carmichael MC, Kumar R. Molecular biology of vasopressin receptors. 57. Yasui M, Kwon TH, Knepper MA, et al. Aquaporin-6: An intracellu-
2006;13(2):96–104.
http://www.ncbi.nlm.nih.gov/pubmed/18655872
Semin Nephrol. 1994;14:341. lar66.vesicle water channel
Boussemart T, Nsotaprotein in renal epithelia.
J, Martin-Coignard D, et Proc Natl Acad Scidiabetes
al. Nephrogenic U S A.
34. Ecelbarger
39. Lee B, YangCA, C, Chen
Kim GH,TH, et al. EJ,ect
Terris o AVP
et al. and oxytocin on insulin
Vasopressin-mediated release:
regulation of 1999;96(10):5808–5813.
insipidus: Treat with caution. Pediatr Nephrol. 2009;24(9):1761–1763.
involvement
epithelial o V1bchannel
sodium receptors. Am J Physiol.
abundance in rat1995;269(6
kidney. AmPtJ1):E1095–
Physiol Renal 1100.Physiol. 58.
67. RaiOkada S, Misaka T, Tanaka
A, Whaley-Connell Y, et al. Aquaporin-11
A, McFarlane knockout mice
S, et al. Hyponatremia, and
arginine
35. Mutig K, Paliege A, Kahl T, et al. Vasopressin V2 receptor expression along
2000;279(1):F46. polycystic
vasopressinkidney disease animals
dysregulation, share a common
and vasopressin receptor mechanism
antagonism. o cyst
Am ormation.
J Nephrol.
rat,
40.mouse,
Blanchard and A, human
Frankrenal epithelia with
M, Wuerzner G, et ocus on TAL. Am Jeffect
al. Antinatriuretic Physiol Renal
of vaso- FASEB J. 2008;22(10):3672–3684.
2006;26(6):579–589.
Physiol. in
pressin 2007;293(4):F1166–1177.
humans is amiloride sensitive, thus ENaC dependent. Clin J Am Soc 59. Knepper MJ,
68. Hannon MA, Star RA.CJ.Vasopressin:
Thompson The syndrome Friend or oe? Nature
of inappropriate Med.
antidiuretic
36. Goligorsky
Nephrol. MS. Cell biology o signal transduction: an overview o mem-
2011;6(4):753–759. 2008;14(1):14–16.
hormone: Prevalence, causes and consequences. Eur J Endocrinol. 2010;162 Sup-
brane receptors,
41. Bugaj G proteins,
V, Pochynyuk O,and secondJD.
Stockand messengers.
ActivationIn: Gold
of the arb S, Ziyadeh
epithelial na chan- FN, http://www.ncbi.nlm.nih.gov/pubmed/18180711
pl 1:S5–12.
eds.inContemporary
nel the collectingIssues duct inbyNephrology:
vasopressin Hormones,
contributesAutacoids, and the Kidney,Am
to water reabsorption. volJ 60. Boone M, Deen
69. Robertson PM. Congenital
GL. Regulation nephrogenic
of arginine vasopressin diabetes
in theinsipidus:
syndromeWhat can
of inap-
23. New
Physiol York:
Renal Churchill
Physiol. Livingstone; 1991.
2009;297(5):F1411–1418. we learn rom
propriate mouse models?
antidiuresis. Am J Med. Exp2006;119(7
Physiol. 2009;94(2):186–190.
Suppl 1):S36–42.
37. Pedersen
42. Knepper MA, NB, Inoue T Regulation
Hofmeister o aquaporin-2
MV, Rosenbaek LL, et water channel trainduces
al. Vasopressin cking http://www.ncbi.nlm.nih.gov/pubmed/18790812
70. Wada K, Matsukawa U, Fujimori A, et al. A novel vasopressin dual V1A/V2
by vasopressin. Curr
phosphorylation of theOpin Cell Biol. 1997;9:560–564.
thiazide-sensitive sodium chloride cotransporter in the 61. Kim
receptor GH, Choiconivaptan
antagonist, NW, Jung JY , et al. Treatingimproves
hydrochloride, lithium-induced
hyponatremia nephrogenic
in rats
http://www.ncbi.nlm.nih.gov/pubmed/9261056
distal convoluted tubule. Kidney Int. 2010;78(2):160–169. diabetes
with syndromeinsipidus with a COX-2 secretion
of inappropriate inhibitor of improves polyuria
antidiuretic via upregulation
hormone (SIADH). Bio o
38. Sands
43. Carmichael MC, Kumar
JM. Mammalian R.transporters.
urea Molecular biology Annu Rev o Physiol.
vasopressin receptors.
2003;65:543. AQP2
Pharmand Bull.NKCC2. Am J Physiol Renal Physiol. 2008;294(4):F702–709.
2007;30(1):91–95.
Semin Nephrol.JM.
44. Sands 1994;14:341.
Regulation of renal urea transporters. J Am Soc Nephrol. 62. AmlalM,
71. Gellai H, Silverstein
Krane CM,JH, Chen Q, etJC,
Hwang al.etEarly
al. Inpolyuria
uence of and urinary con
vasopressin oncentrating
renal he-
http://www.ncbi.nlm.nih.gov/pubmed/7938949
1999;10:635. de ect in potassium
modynamics deprivation.
in conscious Am J Physiol
brattleboro rats. AmRenal Physiol.
J Physiol. 2000;279(4):F655–
1984;246(6 Pt 2):F819–827. 663.
280
28 0 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

63. Bouby
72. Earm JH, N,Christensen
Ahloulay M, BM,Nsegbe
FrokiaerE, J, etetal.al.
Decreased
Vasopressin aquaporin-2
increases expres-
glo- 86. Tahara
polycystic A, Tsukada
kidney diseaseJ, Tomura
and its Youtcomes)
, et al. Vasopressin
3–4 study.inducesAm human
J Kidney mesan-
Dis.
sion and apical
merular ltration plasma
rate inmembrane
conscious delivery in kidney
rats through its collecting
antidiuretic ducts o polyuric
action. JASN. gial cell growth via
2011;57(5):692– 699. induction o vascular endothelial growth actor secretion.
hypercalcemic rats. J Am Soc Nephrol. 1998;9(12):2181–2193.
1996;7(6):842–851. Neuropeptides.
99. Torres VE, 2011;45(2):105–111.
Bankir L, Grantham JJ. A case for water in the treatment of
http://www.ncbi.nlm.nih.gov/pubmed/9848772
73. Bouby N, Bachmann S, Bichet D, et al. Effect of water intake on the pro- 87. Tahara
polycystic A, Tsukada
kidney disease. J,Clin
Tomura Y, etNephrol.
J Am Soc al. Vasopressin stimulates the produc-
2009;4(6):1140–1150.
64. Jensen
gression AM, Bae
of chronic renalEH,failure
Fenton RA, 5/6
in the et al. Angiotensin IIrat.
nephrectomized regulates V2 re-
Am J Physiol. tion oTigerstedt
100. extracellular matrix byPG.
R, Bergman cultured
Niere rat undmesangial
kreislauf.cells. Clin Exp
Skandinav Pharmacol
Arch Physiol.
ceptor and Pt
1990;258(4 pAQP2 during ureteral obstruction. Am J Physiol Renal Physiol.
2):F973–979. Physiol. 2008;35(5–6):586–593.
1898;8:223–271.
2009;296(1):F127–134.
74. Torres VE. Vasopressin in chronic kidney disease: An elephant in the 88. Goldblatt
101. Tahara A, H, Tsukada
LynchJ,J,Tomura
Hanzal YRF , et
, etal.al.
Vasopressin
Studies onstimulates
experimentalthe produc-
hyper-
65. Bichet
room? Kidney Int.DG.2009;76(9):925–928.
Nephrogenic diabetes insipidus. Adv Chronic Kidney Dis. tion o extracellular
tension. I. The productionmatrix of bypersistent
cultured rat mesangial
elevation cells. Clin
of systolic blood Exppressure
Pharmacolby
2006;13(2):96–104.
75. Bardoux P, Martin H, Ahloulay M, et al. Vasopressin contributes to Physiol.of2008;35(5–6):586–593.
means renal ischemia. J Exp Med. 1934;59:347–380.
66. Boussemart
hyper T, Nsota J, Martin-Coignard
ltration, albuminuria, and renal hypertrophy D, et al. Nephrogenic
in diabetes diabetes
melli- http://www.ncbi.nlm.nih.gov/pubmed/18177476
102. Houssay BA, Taquini AC. Acción vasoconstrictora de la sangre venosa del
insipidus:
tus: Study Treat with caution. Pediatr
in vasopressin-de Nephrol. 2009;24(9):1761–1763.
cient brattleboro rats. Proc Natl Acad Sci U S A. 89. isquemiado.
riñón Windt WA, Tahara Rev SocA, ArgKluppel AC, et al. Early, but not late therapy with
Biol. 1937;13:284–287.
67. Rai A, Whaley-Connell
1999;96(18):10397– 10402. A, McFarlane S, et al. Hyponatremia, arginine a vasopressin
103. FascioloV1a-antagonist
JC. Hipertensión ameliorates the development
arterial nefrógena: estudioo experimental.
renal damageTesis a ter
vasopressin
76. Kamoidysregulation,
K, Ishibashi M,and vasopressin
Yamaji T. Thirstreceptor
and plasma antagonism.
levels ofAm J Nephrol.
vasopressin, 5/6 nephrectomy.
Doct Med Ferrari. JRAAS. 2006;7(4):217–224.
Hnos, Buenos Aires; 1939.
2006;26(6):579–589.
angiotensin II and atrial natriuretic peptide in patients with non-insulin-depen- http://www.ncbi.nlm.nih.gov/pubmed/17347933
104. Braun-Menéndez E, Fasciolo JC, Leloir LF, et al. Hipertensión Arterial
http://www.ncbi.nlm.nih.gov/pubmed/17170524
dent diabetes mellitus. Diabetes Res Clin Pract. 1991;11(3):195–202. 90. Torres
Nefrógena. VE. Vasopressin
Buenos Aires, Argentina: in chronic
El Ateneo; kidney1943.disease: An elephant in the
68. YHannon
77. amada K, MJ,Nakano
Thompson CJ. The syndrome
H, Nishimura M, et al.o Effect inappropriate
of AVP.V1– antidiuretic
receptor room?Braun-Menéndez
105. Kidney Int. 2009;76(9):925–928.
E, Page IH. Suggested revision of nomenclature: angio-
hormone: Prevalence,
antagonist on urinary causes and consequences.
albumin excretion and Eur Jrenal
Endocrinol. 2010;162 Sup-
hemodynamics in 91. Otsuka
tensin. Science.F,1958;127:242.
Ogura T, Yamauchi T, et al. E ects o OPC-21268, a vasopres-
pl 1:S5–12.
NIDDM nephropathy: Role of AVP.V1–receptor. J Diabetes Complications. sin V1–receptor
106. Basso N, Terragnoantagonist, on expression
NA. History about the o discovery
growth actors rom glomeruli
of the renin- angiotensin in
69. Robertson329.
1995;9(4):326– GL. Regulation o arginine vasopressin in the syndrome o inap- spontaneously
system. hypertensive
Hypertension. rats. Regul Pept. 1997;72(2–3):87–95.
2001;38:1246–1249.
propriate
78. Bardouxantidiuresis.
P, BichetAm DG, J Med.
Martin 2006;119(7 Suppl 1):S36–42.
H, et al. Vasopressin increases urinary albu- 92. Atlas
107. TorresSA. VE,TheBankir L, Grantham JJ.aldosterone
renin-angiotensin A case or system:
water in pathophysiologi-
the treatment o
70.excretion
min Wada K,inMatsukawa U, Fujimori
rats and humans: A, et al. Aof
Involvement novel vasopressin
V2 receptors anddual
theV1A/V2
renin- polycystic
cal role andkidney disease. CJASN.
pharmacologic 2009;4(6):1140–1150.
inhibition. J Manag Care Pharm. 2007;13(8)(Suppl
receptor antagonist,
angiotensin conivaptan
system. Nephrol hydrochloride,
Dial Transplant. improves hyponatremia in rats
2003;18(3):497–506. http://www.ncbi.nlm.nih.gov/pubmed/19443627
S-b):S9–S20.
with
79. syndrome o inappropriate
Meijer E, Bakker SJ, Halbesma secretion
N, etoal.antidiuretic
Copeptin, hormone
a surrogate (SIADH).
marker Bio of 93. Morgan
108. Gattone L, VHBroughton
2nd, Wang PF,X,Kalsheker
Harris PC, N.etAngiotensinogen:
al. Inhibition o renal cysticbiol-
molecular dis-
Pharm Bull. 2007;30(1):91–95.
vasopressin, is associated with microalbuminuria in a large population cohort. ease biochemistry
ogy, development and and physiology.
progressionInt byJaBiochem
vasopressin V2 receptor
Cell Biol. antagonist. Nat
1996;28:1211–1222.
71. Gellai
Kidney M, Silverstein JH, Hwang JC, et al. Inf uence o vasopressin on re-
Int. 2010;77(1):29–36. Med. 2003;9(10):1323–1326.
109. Lantelme P, Rohrwasser A, Vincent M, et al. Signi cance of urinary angio-
nal
80.he-modynamics
Wang W, Li C, in conscious
Summer S, etbrattleboro
al. Interaction rats.between
Am J Physiol. 1984;246(6
vasopressin and an- Pt http://www.ncbi.nlm.nih.gov/pubmed/14502283
tensinogen in essential hypertension as a function of plasma renin and aldoste-
2):F819–827.
giotensin II in vivo and in vitro: Effect on aquaporins and urine concentration. 94. status.
rone Gattone VH 2nd, Wang
J Hypertens.. X, Harris PC, et al. Inhibition o renal cystic dis-
2005;23:785–792.
Am72.J Physiol
BoubyRenal N, Physiol.
Ahloulay2010;299(3):F577–584.
M, Nsegbe E, et al. Vasopressin increases glo- ease development
110. Navar LG, Nishiyamaand progressionA. Why byarea vasopressin
angiotensinV2 receptor antagonist.
concentrations so highNat in
merular
81. Schrier ltration rate in conscious
RW. Interactions between ratsangiotensin
through itsII antidiuretic
and arginineaction. JASN.
vasopressin Med.kidney?
the 2003;9(10):1323–1326.
Curr Opin Nephrol Hypertens. 2004;13:107–115.
1996;7(6):842–851.
in water homeostasis. Kidney Int. 2009;76(2):137–139. 95. Kobori
111. GattoneH,VH 2nd, Wang
Nishiyama X, Harris PC, et al.
A, Harrison-Bernard LM,Inhibition o renal
et al. Urinary cystic dis-
angiotensino-
http://www.ncbi.nlm.nih.gov/pubmed/8793792
82. Bardoux P, Bichet DG, Martin H, et al. Vasopressin increases urinary albu- easeasdevelopment
gen an indicator and progression
of intrarenal by a vasopressin
angiotensin status in V2 receptor antagonist.
hypertension. Nat
Hypertension.
73.excretion
min Bouby N, in Bachmann S, BichetInvolvement
rats and humans: D, et al. E of ectV2o receptors
water intake andon thethe pro-
renin- Med. 2003;9(10):1323–1326.
2003;41:42–49.
gression o system.
angiotensin chronic Nephrol
renal ailure in the 5/62003;18(3):497–506.
Dial Transplant. nephrectomized rat. Am J Physiol. 96. Castrop
112. Torres VE, Wang X,K,Qian
H, Höcherl KurtzQ, A, etet al. Physiology
E ective treatment
of kidneyorenin. an ortholo-
Physiol
1990;258(4
83. AoyagiPtT,2):F973–979.
Izumi Y, Hiroyama M, et al. Vasopressin regulates the renin- gous2010;90(2):607–673.
Rev. model o autosomal dominant polycystic kidney disease. Nat Med.
74. Torres VE. Vasopressin
angiotensin-aldosterone systeminviachronic kidney in
V1a receptors disease:
macula Andensa
elephant
cells.inAm theJ 2004;10(4):363–364.
113. Peti-Peterdi J, Harris RC. Macula densa sensing and signalling mechanisms
room? Kidney
Physiol Int. 2009;76(9):925–928.
Renal Physiol. 2008;295(1):F100–107. http://www.ncbi.nlm.nih.gov/pubmed/14991049
of renin release. J Am Soc Nephrol. 2010;21(7):1093–1096.
http://www.ncbi.nlm.nih.gov/pubmed/19829311
84. Bolignano D, Zoccali C. Vasopressin beyond water: Implications for renal 97. Lopez
114. TEMPO ML,NCT00428948.
Gomez RA. The Amrenin
J Kidney Dis. 2011;57(5):692–699.
phenotype: roles and regulation in the
75. Bardoux
diseases. Curr Opin P, Martin
Nephrol H, Hypertens.
Ahloulay M, et al. Vasopressin contributes to hyper-
2010;19(5):499–504. http://www.ncbi.nlm.nih.gov/pubmed/21333426
kidney. Curr Opin Nephrol Hypertens. 2010;19(4):366–371.
ltration,
85. albuminuria,
Tahara A, Tsukadaand renal hypertrophy
J, Tomura in diabetesregulates
Y, et al. Vasopressin melli-tus:rat Study in vas-
mesangial 98. Catanzaro
115. Torres VE,DF Meijer E, Bae
, Mullins KT, et BJ.
JJ, Morris al. The
Rationale and design
biosynthetic pathwayo the TEMPO
of renin in
opressin-de
cell growth bycient brattleboro
inducing autocrinerats.secretion
Proc NatlofAcad Sci Uendothelial
vascular S A. 1999;96(18):10397–
growth factor. (tolvaptan
mouse e cacy andgland.
submandibular sa etyJin management
Biol o autosomal dominant polycys-
Chem. 1983;258:7364–7368.
10402.
JPS. 2011;61(2):115–122. tic kidney
116. Mercure disease and its outcomes)
C, Lacombe MJ, Khazaie3–4 K, etstudy. Am J Kidney
al. Cathepsin B is Dis. 2011;57(5):
not the process-
http://www.ncbi.nlm.nih.gov/pubmed/10468619
86. Tahara A, Tsukada J, Tomura Y, et al. Vasopressin induces human mesan- 692–699.
ing enzyme for mouse prorenin. Am J Physiol. 2010;298(5): R1212–R1216.
76.cell
gial Kamoi
growth K, Ishibashi
via induction M, Yamaji T. Thirst
of vascular and plasma
endothelial levelsfactor
growth o vasopressin,
secretion. http://www.ncbi.nlm.nih.gov/pubmed/21333426
117. Kobori H, Nangaku M, Navar LG, et al. The intrarenal renin-angiotensin
angiotensin II 2011;45(2):105–111.
Neuropeptides. and atrial natriuretic peptide in patients with non-insulin-depen- 99. Torres
system: VE, BankirtoL,the
from physiology Grantham
pathobiology JJ. Aof case or water and
hypertension in the treatment
kidney disease. o
dent
87. diabetes
Tahara A,mellitus.
TsukadaDiabetes
J, Tomura ResYClin Pract.
, et al. 1991;11(3):195–202.
Vasopressin stimulates the production polycystic Rev.
Pharmacol kidney disease. Clin J Am Soc Nephrol. 2009;4(6):1140–1150.
2007;59:251–287.
http://www.ncbi.nlm.nih.gov/pubmed/1828024
of extracellular matrix by cultured rat mesangial cells. Clin Exp Pharmacol Physiol. 100. Danser
118. Tigerstedt AH,R, Bergman
Admiraal PJ,PG.
Derkx Niere
FH,und et al.kreislau . Skandinav
Angiotensin Arch Physiol.
I-to-II conversion in
77. Yamada K, Nakano H, Nishimura M, et al. E ect o AVP.V1– receptor antag-
2008;35(5–6):586–593. 1898;8:223–271.
the human renal vascular bed. J Hypertens.. 1998;16:2051–2056.
onist
88. on urinary
Tahara albuminJ,excretion
A, Tsukada Tomura Y and
, et renal hemodynamics
al. Vasopressin in NIDDM
stimulates nephrop-
the production 101. Hollenberg
119. Goldblatt H, NK. Lynch J, Hanzal
Implications RF, et al.
of species Studies for
difference on clinical
experimental hyper-
investigation.
athy:
of Role o AVP
extracellular .V1–receptor.
matrix by cultured J Diabetes Complications.
rat mesangial cells. Clin1995;9(4):326–329.
Exp Pharmacol Physiol. tension.on
Studies I. the
Therenin–angiotensin
production o persistent system. elevation
Hypertension.o systolic blood pressure by
2000;35:150.
78. Bardoux P, Bichet DG, Martin H, et al. Vasopressin increases urinary albu-
2008;35(5–6):586–593. meansAugustos
120. o renal ischemia.
P, Tomagnini J ExpPK, Med. 1934;59:347–380.
Pesquero JB, et al. Interactions between angio-
min
89. excretion
Windt WA, in rats
TaharaandA,humans:
KluppelInvolvement
AC, et al. Early, o V2 butreceptors and the with
not late therapy renin-a http://www.ncbi.nlm.nih.gov/pubmed/19870251
tensin-(1–7):kinins, and angiotensin II in kidney and blood vessels. Hypertension.
angiotensin V1a-antagonist
vasopressin system. Nephrol ameliorates
Dial Transplant. 2003;18(3):497–506.
the development of renal damage after 102. Houssay
2001;38(pt BA, Taquini AC. Acción vasoconstrictora de la sangre venosa del
2):660.
http://www.ncbi.nlm.nih.gov/pubmed/12584270
5/6 nephrectomy. JRAAS. 2006;7(4):217–224. riñón Griendling
121. isquemiado.KK, Rev Soc Arg Biol.RW.
Alexander 1937;13:284–287.
The angiotensin (AT1) receptor. Semin
79. Torres
90. Meijer VE. E, Bakker SJ, Halbesma
Vasopressin in chronic N, etkidney
al. Copeptin,
disease:a An surrogate
elephant marker
in the o 103. Fasciolo
Nephrol. JC. Hipertensión arterial ne rógena: estudio experimental. Tesis
1993;13:558.
vasopressin,
room? KidneyisInt. associated with microalbuminuria in a large population cohort.
2009;76(9):925–928. Doct Med
122. Ferrari.
Bernstein KE,Hnos, BuenosRW.
Alexander Aires; 1939.
Counterpoint: molecular analysis of the an-
Kidney Int. 2010;77(1):29–36.
91. Otsuka F, Ogura T, Yamauchi T, et al. Effects of OPC-21268, a vasopres- 104. Braun-Menéndez
giotensin II receptor. Endocr E, Fasciolo JC, Leloir LF, et al. Hipertensión Arterial Nefró
Rev. 1992;13:381.
http://www.ncbi.nlm.nih.gov/pubmed/19847155
sin V1–receptor antagonist, on expression of growth factors from glomeruli in gena. Buenos
123. Aires, Argentina:
Gonzalez-Villalobos El Ateneo;
R, Klassen RB. 1943.
Megalin binds and internalizes angio-
80. Wang W,hypertensive
spontaneously Li C, Summer rats.S, Regul
et al. Pept.
Interaction between vasopressin and an-
1997;72(2–3):87–95. 105. Braun-Menéndez
tensin II. Am J Physiol Renal E, Page IH. 2005;288(2):F420.
Physiol. Suggested revision o nomenclature: angio-
giotensin
92. Torres II inVE, vivo and in
Bankir L, vitro:
GranthamE ectJJ. onAaquaporins
case for water and urine
in theconcentration.
treatment of tensin.Schefe
124. Science. JH,1958;127:242.
Neumann C, Goebel M, et al. Prorenin engages the (pro)re-
Am J Physiol
polycystic Renaldisease.
kidney Physiol. CJASN.
2010;299(3):F577–584.
2009;4(6):1140–1150. http://www.ncbi.nlm.nih.gov/pubmed/17750687
nin receptor like renin and both ligand activities are unopposed by aliskiren.
81. Gattone
93. Schrier RW. Interactions
VH 2nd, Wang X,betweenHarris PC, angiotensin II and arginine
et al. Inhibition of renalvasopressin
cystic dis- J106. Basso N,
Hypertens.. Terragno NA. History about the discovery o the renin- angioten-
2008;26(9):1787–1794.
in water
ease homeostasis.
development and Kidney Int. 2009;76(2):137–139.
progression by a vasopressin V2 receptor antagonist. Nat sin system.
125. Allen Hypertension. 2001;38:1246–1249.
AM, Zhuo J, Mendelsohn FA. Localization and function of angioten-
82. 2003;9(10):1323–1326.
Med. Bardoux P, Bichet DG, Martin H, et al. Vasopressin increases urinary albu- http://www.ncbi.nlm.nih.gov/pubmed/11751697
sin AT1 receptors. Am J Hypertens.. 2000;13:31s.
min
94. excretion
Gattone VH in rats
2nd,andWanghumans: Involvement
X, Harris PC, et al.oInhibition
V2 receptors and cystic
of renal the renin-
dis- 107. Navar
126. Atlas SA.GL, The renin-angiotensin
Harrison-Bernard aldosteroneA,system:
LM, Nishiyama pathophysiologi-cal
et al. Regulation of intrare-
angiotensin
ease developmentsystem. andNephrol Dial Transplant.
progression 2003;18(3):497–506.
by a vasopressin V2 receptor antagonist. Nat roleangiotensin
nal and pharmacologic inhibition.Hypertension.
II in hypertension. J Manag Care Pharm. 2007;13(8)(Suppl
2002;39(part 2):316. S-
83. 2003;9(10):1323–1326.
Med. Aoyagi T, Izumi Y, Hiroyama M, et al. Vasopressin regulates the renin- b):S9–S20.
127. Simpson SA, Tait JF, Wettstein A, et al. Isolation from the adrenals of a
angiotensin-aldosterone
95. Gattone VH 2nd, Wang systemX,via V1aPC,
Harris receptors in maculaofdensa
et al. Inhibition renal cells.
cysticAm dis-J 108. crystalline
new Morgan L,hormone Broughton withPFespecially
, Kalshekerhigh N. Angiotensinogen:
effectiveness on mineralmolecular biol-
metabo-
Physiol
ease Renal Physiol.
development and2008;295(1):F100–107.
progression by a vasopressin V2 receptor antagonist. Nat ogy, biochemistry
lism. and physiology. Int J Biochem Cell Biol. 1996;28:1211–1222.
Experientia. 1953;9:333–335.
84. 2003;9(10):1323–1326.
Med. Bolignano D, Zoccali C. Vasopressin beyond water: Implications or renal http://www.ncbi.nlm.nih.gov/pubmed/9022280
128. Thomas W, Harvey BJ. Mechanisms underlying rapid aldosterone effects in
diseases.
96. TorresCurr VE,Opin
Wang Nephrol
X, Qian Hypertens. 2010;19(5):499–504.
Q, et al. Effective treatment of an orthologous model 109.kidney.
the LantelmeAnnuP,Rev Rohrwasser A, Vincent M, et al. Signi cance o urinary angio-
Physiol. 2011;73:335–357.
http://www.ncbi.nlm.nih.gov/pubmed/20689424
of autosomal dominant polycystic kidney disease. Nat Med. 2004;10(4):363–364. tensinogen
129. Chen SY in ,essential
Bhargavahypertension
A, Mastroberardino as a unction o plasmasodium
L, et al. Epithelial renin and aldoste-
channel reg-
85. TEMPO
97. Tahara A, Tsukada J, Tomura
NCT00428948. Y, et al.Dis.
Am J Kidney Vasopressin regulates rat mesangial
2011;57(5):692–699. rone status.
ulated J Hypertens.. 2005;23:785–792.
by aldosterone-induced protein sgk. Proc Natl Acad Sci. 1999;96:2514–2519.
cell
98.growth
TorresbyVE, inducing
Meijerautocrine
E, Bae KT, secretion
et al. oRationale
vascular and endothelial
design growth
of the TEM-actor. http://www.ncbi.nlm.nih.gov/pubmed/15775783
130. Ho K, Nichols CG, Lederer WJ, et al. Cloning and expression of an in-
JPS. (tolvaptan
PO 2011;61(2):115–122.
ef cacy and safety in management of autosomal dominant 110. Navar
wardly LG, ATP-regulated
rectifying Nishiyama A. potassium
Why are angiotensin
channel. Nature.concentrations so high in
1993;362:31–38.
http://www.ncbi.nlm.nih.gov/pubmed/21229342 the kidney? Curr Opin Nephrol Hypertens. 2004;13:107–115.
CHAPTER 8 HORMONES AND THE KIDNEY 2281
81

111. Alpert
131. KoboriMA, H, Nishiyama
Govindarajan A, Harrison-Bernard
G, Del Rosario ML, LM,etet al.
al. The
Urinary
roleangiotensino-
of the renin- 137.
159. Lea Smith DHG,
JP, Jin SG, Neutel
RobertsJM, BR,Weber MA. E ectsIIostimulates
et al. Angiotensin angiotensin II on pressor
calcineurin activ-
gen as an indicator
angiotensin system oinintrarenal angiotensin status
the pathophysiology, in hypertension.
prevention, and treatment Hypertension.
of renal responses
ity in proximalto norepinephrine
tubule epithelia in humans.
through Life AT-1Sci. 1991;48:2413. tyrosine phos-
receptor-mediated
2003;41:42–49.
impairment in patients with the cardiometabolic syndrome or its components. 138. Liang of
phorylation L, the
TamPLC-gamma1
CW, Pozsgaiisoform. G, et al.J Protection
Am Soc Nephrol.o angiotensin II-induced
2002;13(7):1750.
Jhttp://www.ncbi.nlm.nih.gov/pubmed/12511528
Cardiometab Syndr. 2009;4(1):57–62. vascular
160. Turban hypertrophy
S, Beutler in vascular
KT, Morris smooth
RG, muscle-targeted
et al. Long-termreceptor activity-mod-
regulation of proxi-
112. Stegbauer
132. Castrop H,J, Höcherl CoffmanK,TM. Kurtz
NewA,insights
et al. Physiology o kidney
into angiotensin renin.actions:
receptor Physiol imal
yingtubule
proteinacid-base
2 transgenic mice. Hypertension.
transporter abundance2009;54(6):1254–1261.
by angiotensin II. Kidney Int.
Rev. 2010;90(2):607–673.
from blood pressure to aging. Curr Opin Nephrol Hypertens. 2011;20(1):84–88. 139. Stou er GA, Owens GK. Angiotensin II-induced mitogenesis o
2006;70(4):660–668.
http://www.ncbi.nlm.nih.gov/pubmed/20393195
133. De Beer VJ, de Graaff HJ, Hoekstra M, et al. Integrated control of pulmo- spontane
161. Yingst ouslyDR,hypertensive
Massey KJ, rat-derived
Rossi NF, et cultured smoothIImuscle
al. Angiotensin directlycells is de-
stimulates
113. vascular
nary Peti-Peterdi tone J,byHarris RC. Macula
endothelin densa sensing
and angiotensin II inand signalling
exercising mechanisms
swine depends pendent
activity and on autocrine
alters the production
phosphorylation o trans orming growth
of Na-K-ATPase in actor-beta.
rat proximalCirc Res.
tubule
o renin
on gender. release. J Am Soc
Am J Physiol Nephrol.
Heart Circ 2010;21(7):1093–1096.
Physiol. 2010;298(6):H1976–1985. 1992;60:820.
with a rapid time course. Am J Physiol Renal Physiol. 2004;287(4):F713.
http://www.ncbi.nlm.nih.gov/pubmed/20360309
134. Hassid A, Williams C. Vasoconstrictor-evoked prostaglandin synthesis in 140.
162. Han Gibbons GH, Pratt
HJ, Park SH, Lee RE,YJ.Dzau VJ. Vascular
Signaling cascade smooth
of ANG muscle cell hypertrophy
II-induced inhibition
114. Lopez
cultured ML, smooth
vascular Gomez RA. TheAm
muscle. renin phenotype:
J Physiol. roles and regulation in the
1983;245:C278. vs. hyperplasia:
of alpha-MG autocrine
uptake in renaltransproximal
orming growth actor-b1
tubule cells. Am Jexpression
Physiol Renal determines
Physiol.
kidney.Diz
135. Curr
DL,OpinArnoldNephrolAC,Hypertens.
Nautiyal M, 2010;19(4):366–371.
et al. Angiotensin peptides and central growth response to angiotensin II. J Clin Invest. 1992;90:456.
2004;286(4):F634.
http://www.ncbi.nlm.nih.gov/pubmed/20502328
autonomic regulation. Curr Opin Pharmacol. 2011;11(2):131–137. http://www.ncbi.nlm.nih.gov/pubmed/1644917
163. Lee YJ, Lee YJ, Han HJ. Regulatory mechanisms of Na( )/glucose cotrans-
115. Wong
136. Catanzaro DF, Mullins
PC, Hart JJ, Morris BJ.
SD, Timmermans The biosynthetic
P. Effect of angiotensin pathway o reninon
II antagonism in 141.
portersSteckelings
in renal proximalUM, Rompe tubuleFcells.
, Kaschina
KidneyE,Int. et Suppl.
al. The evolving story o the
2007;106:S27–S35.
mouse renal
canine submandibular
sympathetic gland.
nerveJ Biol Chem.Hypertension.
function. 1983;258:7364–7368. 1991;17:1127. RAAS
164. Xu in hypertension,
L, Dixit MP, Chen diabetes R, and
et al.CV disease:
Effects moving romIImacrovascular
of angiotensin on NaPi-IIa co- to
116. Smith
137. Mercure C, Lacombe
DHG, Neutel JM, MJ, Weber
KhazaieMA. K, et al. Cathepsin
Effects B is notIIthe
of angiotensin on process-
pressor microvascular
transporter expressiontargets. Fundam lin Pharmacol.
and activity in rat renal 2009;23(6):693–703.
cortex. Biochim Biophys Acta.
ing enzymeto or
responses mouse prorenin.
norepinephrine Am J Physiol.
in humans. 2010;298(5):
Life Sci. 1991;48:2413. R1212–R1216. http://www.ncbi.nlm.nih.gov/pubmed/19817870
2004;1667(2):114.
117. Liang
138. KoboriL,H,Tam Nangaku M, Navar
CW, Pozsgai G, et LG,al.etProtection
al. The intrarenal renin-angiotensin
of angiotensin II-induced 142.
165. PechArimaV,S,Kim Ito YH,
S. Angiotensin
Weinstein AM, II type 2 receptors
et al. Angiotensin in the kidney: evidence
II increases chloride
system: rom
vascular physiologyintovascular
hypertrophy the pathobiology
smooth omuscle-targeted
hypertension and kidneyactivity-
receptor disease. or endothelial-cell
absorption in the cortical mediated renalduct
collecting vasodilatation.
in mice through Nephrol Dial Transplant.
a pendrin-dependent
Pharmacol Rev.
modifying protein2007;59:251–287.
2 transgenic mice. Hypertension. 2009;54(6):1254–1261. 2000;15:448.
mechanism. Am J Physiol Renal Physiol. 2007;292(3):F914–920.
http://www.ncbi.nlm.nih.gov/pubmed/17878513
139. Stouffer GA, Owens GK. Angiotensin II-induced mitogenesis of spontane- http://www.ncbi.nlm.nih.gov/pubmed/10727534
166. Beutler KT, Masilamani S, Turban S, et al. Long-term regulation of ENaC
118. hypertensive
ously Danser AH, Admiraal rat-derivedPJ, Derkx
cultured FH,smooth
et al. Angiotensin
muscle cells I-to-II conversionon
is dependent in 143. Hein in
expression L, kidney
Barsh GS, Pratt RE, et II.
by angiotensin al.Hypertension.
Behavioral and cardiovascular e ects o
2003;41(5):1143.
the humanproduction
autocrine renal vascular bed. J Hypertens..
of transforming growth1998;16:2051–2056.
factor-beta. Circ Res. 1992;60:820. disrupting the angiotensin
167. Peti-Peterdi J, Warnock II type-2
DG, receptor
Bell PD. in mice. Nature.
Angiotensin 1995;377:744.
II directly stimulates
http://www.ncbi.nlm.nih.gov/pubmed/9886896
140. Gibbons GH, Pratt RE, Dzau VJ. Vascular smooth muscle cell hypertrophy http://www.ncbi.nlm.nih.gov/pubmed/7477266
ENaC activity in the cortical collecting duct via AT (1) receptors. J Am Soc
119.
vs. Hollenberg
hyperplasia: NK. Implications
autocrine transforming o species
growth di factor-b1
erence orexpression
clinical investigation.
determines 144.
Nephrol. Ichiki T, Labosky PA, Shiota C, et al. E ects on blood pressure and ex-
2002;13(5):1131.
Studies response
growth on the renin–angiotensin
to angiotensin II.system. Hypertension.
J Clin Invest. 1992;90:456.2000;35:150. ploratory
168. Wei behavior
Y, Wang W. o Angiotensin
mice lackingII angiotensin II type-2 Kreceptor.
stimulates basolateral channelsNature.
in rat
http://www.ncbi.nlm.nih.gov/pubmed/10642291
141. Steckelings UM, Rompe F, Kaschina E, et al. The evolving story of the 1995;377:748.
cortical collecting ducts. Am J Physiol Renal Physiol. 2003;284(1):F175.
120. Augustos
RAAS P, Tomagnini
in hypertension, diabetesPK,andPesquero JB, et moving
CV disease: al. Interactions between angio-
from macrovascular to http://www.ncbi.nlm.nih.gov/pubmed/7477267
169. Zhuo JL. Intracrine renin and angiotensin II: a novel role in cardiovascular
tensin-(1–7):kinins,
microvascular targets.and angiotensin
Fundam II in kidney
lin Pharmacol. and blood vessels. Hypertension.
2009;23(6):693–703. 145.
and renalSiragy HM. regulation.
cellular The potential role o the
J Hypertens.. angiotensin subtype 2 receptor in
2006;24(6):1017–1020.
2001;38(pt
142. Arima2):660.S, Ito S. Angiotensin II type 2 receptors in the kidney: evidence cardiovascular
170. Badzinskaprotection. B, Sadowski Current Hypertens
J. Opposed Rep.
effects of2009;11(4):260–262.
prostaglandin E2 on perfusion
121.endothelial-cell
for Griendling KK,mediated Alexander RW.vasodilatation.
renal The angiotensin (AT1) Dial
Nephrol receptor. Semin
Transplant. http://www.ncbi.nlm.nih.gov/pubmed/19602326
of rat renal cortex and medulla: interactions with the renin-angiotensin system.
Nephrol. 1993;13:558.
2000;15:448. 146. Siragy2008;93(12):1292–1302.
Exp Physiol. HM, Ja a AA, Margolis HS, et al. Renin-angiotensin system modu-
http://www.ncbi.nlm.nih.gov/pubmed/8278689
143. Hein L, Barsh GS, Pratt RE, et al. Behavioral and cardiovascular effects of lates
171. renal
Madsen bradykinin
K, Marcussenproduction. Am J Physiol.
N, Pedersen M, et al.1996;271:R1090.
Angiotensin II promotes devel-
122. Bernstein
disrupting KE, Alexander
the angiotensin RW. receptor
II type-2 Counterpoint: in mice. molecular analysis o the an-
Nature. 1995;377:744. 147.
opment Seeliger
of the E, Wronski
renal T, Ladwig M,
microcirculation et al. The
through AT1renin-angiotensin
receptors. J Am Soc system and
Nephrol.
giotensin
144. IchikiII receptor.
T, Labosky Endocr
PA, Rev.
Shiota1992;13:381.
C, et al. Effects on blood pressure and ex- the third mechanism o renal blood f ow autoregulation. Am J Physiol Renal Physi
2010;21(3):448–459.
http://www.ncbi.nlm.nih.gov/pubmed/1618167
ploratory behavior of mice lacking angiotensin II type-2 receptor. Nature. ol.
172.2009;296(6):F1334–F1345.
Wolf G, Neilson EG. Angiotensin II induces cellular hypertrophy in cul-
123. Gonzalez-Villalobos R, Klassen RB. Megalin binds and internalizes angio-
1995;377:748. 148. Patzak A,
tured murine Kleinmann
proximal tubularF, Lai EYAm
cells. , etJal. Nitric1990;259:F768.
Physiol. oxide counteracts angioten-
tensinSiragy
145. II. Am HM.J PhysiolTheRenal Physiol.
potential role2005;288(2):F420.
of the angiotensin subtype 2 receptor in sin
173.II Chen
in ducedJ, Chencontraction
JK, Neilson in e EG, erentet arterioles
al. Role ofinEGF mice. Acta Physiol
receptor Scand.
activation in
124. Sche e JH,
cardiovascular NeumannCurrent
protection. C, Goebel M, et al.
Hypertens Rep. Prorenin engages the (pro)re-nin
2009;11(4):260–262. 2004;181(4):439.
angiotensin II-induced renal epithelial cell hypertrophy. J Am Soc Nephrol.
receptor
146. like HM,
Siragy reninJaffa
and AA,
bothMargolis
ligand activities
HS, et al. areRenin-angiotensin
unopposed by aliskiren. systemJmodu-
Hyper- http://www.ncbi.nlm.nih.gov/pubmed/15283756
2006;17(6):1615–1623.
tens.. renal
lates 2008;26(9):1787–1794.
bradykinin production. Am J Physiol. 1996;271:R1090. 149.
174. WolfZhangG,Z,Jablonski
Rhinehart K, Kwon W,
K, Schroeder R, etetal.al.Angiotensin
ANG II signaling II-inducedin vasa recta
hypertro-
125. Seeliger
147. Allen AM, E, Zhuo
Wronski J, Mendelsohn
T, Ladwig M, FA.etLocalization and unction osystem
al. The renin-angiotensin angioten-
and pericytes
phy of proximalby PKCtubular and reactive oxygenp27Kip1.
cells requires species. Am KidneyJ Physiol Heart Circ Physiol.
Int. 2003;64(1):71.
sin AT1
the thirdreceptors.
mechanism AmofJrenal
Hypertens..
blood 2000;13:31s.
ow autoregulation. Am J Physiol Renal Physi- 2004;287(2):H773.
175. Gorin Y, Ricono JM, Wagner B, et al. Angiotensin II-induced ERK1/ERK2
http://www.ncbi.nlm.nih.gov/pubmed/10678286
ol. 2009;296(6):F1334–F1345. 150. Huraand
activation CE,protein
Kunau synthesis
RT, Jr. Angiotensin II-stimulated
are redox-dependent in prostaglandin
glomerular mesangialproduc-
126. Navar A,
148. Patzak GL,Kleinmann
Harrison-Bernard
F, Lai EY, LM,et al.Nishiyama
Nitric oxideA,counteracts
et al. Regulation o intrare-
angiotensin II in- tion
cells.by canineJ.renal
Biochem a erent arterioles.
2004;381(Pt 1):231. Am J Physiol. 1988;254:F734.
nal angiotensin
duced contraction II in
in efferent
hypertension.
arterioles Hypertension.
in mice. Acta2002;39(part
Physiol Scand. 2):316.
2004;181(4):439. 151.
176. Liu Velez BC,JC.SunTheJ,importance
Chen Q, etoal.the Roleintrarenal renin-angiotensin
of connective tissue growth system.
factorNatin
127. Zhang
149. Simpson Z, SA, Tait JF, Wettstein
Rhinehart K, KwonA,W,et et al. al.
Isolation
ANG IIrom the adrenals
signaling o arecta
in vasa new Clin Pract Nephrol.
mediating hypertrophy 2009;5(2):89–100.
of human proximal tubular cells induced by angiotensin
crystallinebyhormone
pericytes PKC andwith especially
reactive oxygen high e ectiveness
species. on mineral
Am J Physiol Heart Circ metabo lism.
Physiol. http://www.ncbi.nlm.nih.gov/pubmed/19065132
II. Am J Nephrol. 2003;23(6):429.
Experientia. 1953;9:333–335.
2004;287(2):H773. 152.
177. Guo ChiltonDF, L, Loutzenhiser
Chenier I, LavoieK,JL, Morales E, et al. Inward
et al. Development recti er K(+) and
of hypertension currents
kid-
http://www.ncbi.nlm.nih.gov/pubmed/13107656
150. Hura CE, Kunau RT, Jr. Angiotensin II-stimulated prostaglandin produc- and kir2.1 expression
ney hypertrophy in renal amice
in transgenic erent and e erent arterioles.
overexpressing ARAP1 gene J Am
in Soc
the Nephrol.
kidney.
128.byThomas
tion canine renalW, Harvey BJ. arterioles.
afferent Mechanisms Amunderlying rapid aldosterone e ects in
J Physiol. 1988;254:F734. 2008;19(1):69–76.
Hypertension. 2006;48:453–459.
the kidney.
151. Velez AnnuJC. The Revimportance
Physiol. 2011;73:335–357.
of the intrarenal renin-angiotensin system. Nat 153.
178. WongDu J, DingNL, M, Tsui Sours-Brothers
JK. Angiotensin S, etIIal.upregulates
Mediation othe angiotensin
expressionII-induced
of vaso-
http://www.ncbi.nlm.nih.gov/pubmed/20809792
Clin Pract Nephrol. 2009;5(2):89–100. Ca 2+
pressinsignaling
V2 mRNA by polycystin
in the inner 2 in glomerular
medullary mesangial
collecting ductcells.
of the Amrat.
J Physiol Renal
Metabolism.
129.Chen
152. ChiltonSY, Bhargava
L, LoutzenhiserA, Mastroberardino
K, Morales E,L,etetal. al.Inward
Epithelial sodium
recti er K(channel reg-
) currents Physiol. 2008;294(4):F909–F918.
2003;52(3):290.
ulated
and by aldosterone-induced
kir2.1 expression in renalprotein afferentsgk. andProc Natl Acad
efferent Sci. 1999;96:2514–2519.
arterioles. J Am Soc Nephrol. 154. Roos MH,
179. Kobori Eringa EC, A.
H, Nishiyama vanEffects
Rodijnen WF, et on
of tempol al. renal
Preglomerular and postglo-
angiotensinogen pro-
http://www.ncbi.nlm.nih.gov/pubmed/10051674
2008;19(1):69–76. merular
duction basalin Dahl diameter changesrats.
salt-sensitive and Biochem
reactivityBiophys
to angiotensin
Res Commun. II in obese
2004;315: rats.
130. Du
153. Ho J,K,DingNichols CG, Lederer WJ,
M, Sours-Brothers S, etetal.al. Cloning of
Mediation and expressionII-induced
angiotensin o an in- Diabetes
746–750. Obes Meta. 2008;10(10):898–905.
2
wardly recti ying
Ca signaling byATP-regulated
polycystin 2 inpotassium
glomerular channel.
mesangial Nature.
cells.1993;362:31–38.
Am J Physiol Renal 155.
180. V Foidart
anegas V JB, Mahieu A,
, Ferrebuz P. Quiroz
GlomerularY, et al. mesangial cell contractility
Hypertension. in vitro
in Page (cellophane-
131. Alpert
Physiol. MA, Govindarajan G, Del Rosario ML, et al. The role o the renin-
2008;294(4):F909–F918. is controlled
wrapped) kidney by isandue angiotensin–prostaglandin
to interstitial nephritis. Kidney balance. Mol Cell Endocrinol.
Int. 2005;68:1161–1170.
angiotensin
154. Roos MH, systemEringain the
EC,pathophysiology,
van Rodijnen WFprevention, and treatment
, et al. Preglomerular o renal
and postglo- 1986;47:163.
181. Prieto-Carrasquero MC, Botros F, Kobori H, et al. Angiotensin II regulates
impairment
merular basal in diameter
patients with the cardiometabolic
changes and reactivity tosyndrome angiotensin or itsII components.
in obese rats.J http://www.ncbi.nlm.nih.gov/pubmed/3091426
distal nephron renin gene expression in Goldblatt hypertensive rats indepen-
Cardiometab
Diabetes ObesSyndr.
Meta.2009;4(1):57–62.
2008;10(10):898–905. 156.
dentlyShalamanova
from high blood L, Wilkinson MC, McArdle
pressure (Abstract). F, et al. Characterisation
Hypertension. 2005;46:871. o the
132. Foidart
155. Stegbauer JB,J, Mahieu
Co manP.TM. New insights
Glomerular into angiotensin
mesangial receptor in
cell contractility actions:
vitro expression
182. NavaroLG. theThe renin-angiotensin system in primary
intrarenal renin-angiotensin system andin immortalised
hypertension. human Kidney
isrom blood pressure
controlled by an toangiotensin–prostaglandin
aging. Curr Opin Nephrol Hypertens. balance. Mol 2011;20(1):84–88.
Cell Endocrinol. renal proximal tubular cells. Nephron Exp Nephrol. 2010;116(3):e53–61.
Int. 2004;65:1522–1532.
http://www.ncbi.nlm.nih.gov/pubmed/21076298
1986;47:163. 157. Navar LG,
183. Abadir PM.Prieto The MC, frail Satou R, et al. Intrarenal
renin-angiotensin system. angiotensin
Clin Geriatr II and its
Med.
133. Shalamanova
156. De Beer VJ, deL,Graa HJ, Hoekstra
Wilkinson MC, McArdle M, et al.F, etIntegrated control o pulmo-
al. Characterisation of the contribution
2011;27(1):53–65. to the genesis o chronic hypertension. Curr Opin Pharmacol.
nary vascular
expression tonerenin-angiotensin
of the by endothelin and angiotensin
system in primary II in and
exercising swine depends
immortalised human 2011;11(2):180–186.
184. Matavelli LC, Huang J, Siragi HM. Angiotensin AT2 receptor stimulation
on gender.
renal proximal Am tubular
J Physiolcells.
HeartNephron
Circ Physiol. 2010;298(6):H1976–1985.
Exp Nephrol. 2010;116(3):e53–61. http://www.ncbi.nlm.nih.gov/pubmed/21339086
inhibits early renal in ammation in renovascular hypertension. Hypertension.
134. Navar
157. HassidLG, A, Williams
Prieto MC, C. Vasoconstrictor-evoked
Satou R, et al. Intrarenal prostaglandin
angiotensinsynthesis
II and itsin 158. Liu FY, Cogan MG. Angiotensin II stimulation o hydrogen ion secretion in
2011;57(2):308–313.
cultured vascular
contribution to thesmooth muscle.
genesis Am J Physiol.
of chronic 1983;245:C278.
hypertension. Curr Opin Pharmacol. the
185.ratOzono
early proximal
R, Wang tubule:Z, Moore modes
AF, etoal.action, mechanisms,
Expression and kinetics.
of the subtype-2 J Clin
angioten-
135. Diz DL, Arnold AC, Nautiyal M, et al. Angiotensin peptides and central
2011;11(2):180–186. Invest. 1988;82:601.
sin II (AT2) receptor protein in rat kidney. Hypertension. 1997;30:1238.
autonomic
158. Liu FY regulation.
, Cogan MG. Curr Opin Pharmacol.
Angiotensin 2011;11(2):131–137.
II stimulation of hydrogen ion secretion in http://www.ncbi.nlm.nih.gov/pubmed/2841357
186. Siomou E, Bouba I, Kollios KD, et al. Angiotensin II type 2 receptor gene
http://www.ncbi.nlm.nih.gov/pubmed/21367658
the rat early proximal tubule: modes of action, mechanisms, and kinetics. J Clin 159. Lea JP, JininSG,
polymorphism Roberts children
Caucasian BR, et al. with
Angiotensin II stimulates
a wide spectrum calcineurin
of congenital activ-
anoma-
136. Wong
Invest. PC, Hart SD, Timmermans P. E ect o angiotensin II antagonism on
1988;82:601. ity
liesinofproximal
the kidney tubule epithelia
and urinary through
tract. Pediatr AT-1
Res.receptor-mediated
2007;62(1):83–87. tyrosine phos-
canine renal sympathetic nerve unction. Hypertension. 1991;17:1127. phorylation o the PLC-gamma1 iso orm. J Am Soc Nephrol. 2002;13(7):1750.
282
28 2 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

160. Ma
187. Turban S, Beutler
J, Nishimura H, KT,
FogoMorris
A, et al.RG, et al. Long-term
Accelerated regulation
brosis and collagenodeposi-proxi- 184. Miyake-Ogawa
218. Matavelli LC, Huang J, SiragiM,
C, Miyazaki HM. AbeAngiotensin AT2 receptorc stimulation
K, et al. Tissue-speci expression
mal develop
tion tubule in acid-base
the renaltransporter
interstitiumabundance
of angiotensin by type
angiotensin
2 receptor II. null
Kidney Int.
mutant inhibits
of early renal inf
renin-angiotensin ammation
system in renovascular
components hypertension.Am
in IgA nephropathy. Hypertension.
J Nephrol.
2006;70(4):660–668.
mice during early ureteral obstruction. Kidney Int. 1998;53:937. 2011;57(2):308–313.
2005;25:1–12.
http://www.ncbi.nlm.nih.gov/pubmed/16807546
188. Vazquez E, Coronel I, Bautista R, et al. Angiotensin II-dependent induc- http://www.ncbi.nlm.nih.gov/pubmed/21189405
219. Mezzano SA, Aros CA, Droguett A, et al. Renal angiotensin II up-regulation
161.ofYAT(2)
tion ingst DR, receptorMassey KJ, Rossi
expression NF,renal
after et al.ablation.
Angiotensin
Am J II directly
Physiol Renalstimulates
Physiol. 185.myo
and Ozono R, Wang
broblast Z, Moore
activation in AF , et al.membranous
human Expression o nephropathy.
the subtype-2Kidney angioten-
Int.
activity and alters the phosphorylation o Na-K-ATPase in rat proximal tubule
2005;288(1):F207. sin II (AT2) receptor
2003;Suppl 64:S39–S45. protein in rat kidney. Hypertension. 1997;30:1238.
with aLorenzo
189. rapid time course. Am JM,
O, Ruiz-Ortega Physiol
EsbritRenal
P, et Physiol. 2004;287(4):F713.
al. Angiotensin II increases parathy- 186. Kengne-Wafo
220. Siomou E, Bouba I, KolliosL,KD,
S, Massella et al. AngiotensinF, IIettype
Diomedi-Camassei 2 receptor
al. Risk factorsgene
for
162.hormone-related
roid Han HJ, Park SH, Lee YJ.
protein Signaling
(PTHrP) and cascade
the typeo1 PTH/PTHrP
ANG II-induced receptorinhibition
in the polymorphism
cyclosporine in Caucasian children
A nephrotoxicity in childrenwith with
a wide spectrum o congenital
steroid-dependent nephriticanoma-
syn-
o alpha-MG
kidney. J Am Soc uptake
Nephrol.in renal proximal tubule cells. Am J Physiol Renal Physiol.
2002;13(6):1595. lies o the
drome. Clinkidney
J Am Soc andNephrol.
urinary 2009;4(9):1409–1416.
tract. Pediatr Res. 2007;62(1):83–87.
2004;286(4):F634.
190. Kerstens MN, van der Kleij FG, Boonstra AH, et al. Angiotensin admin- 187. Oka
221. Ma J,K, Nishimura
MoriyamaH,T,Fogo A, et al.S,Accelerated
Takahara et al. Increased brosis and collagen
expression deposi-
of renin in
163. Leestimulates
istration YJ, Lee YJ, renalHan HJ. 11 Regulatory mechanisms
beta-hydroxysteroid o Na(+)/glucose
dehydrogenase cotrans-
activity in tion develop
chronic in the
allograft renal interstitium
nephropathy. Transplanto angiotensin type 2 receptor null mutant
Proc. 2005;37:2131–2134.
porters in
healthy men.renal proximal
Kidney tubule cells. Kidney Int. Suppl. 2007;106:S27–S35.
Int. 2004;65(6):2065. mice during
222. Oikawaearly ureteral
S, Imai M, obstruction.
Ueno A, etKidney Int. 1998;53:937.
al. Cloning and sequence analysis of
164. Zimmerman
191. Xu L, Dixit MP MC. , Chen R, et al.II Eand
Angiotensin ectsangiotensin
o angiotensin II on NaPi-IIa
1–7 redox signalingco-in http://www.ncbi.nlm.nih.gov/pubmed/9551401
cDNA encoding: a precursor human atrial natriuretic For polypeptide. Nature.
transporter
the expression
central nervous system.andCurractivity
OpininPharmacol.
rat renal 2011;11(2):138–143.
cortex. Biochim Biophys Acta. 188. Vazquez E, Coronel I, Bautista R, et al. Angiotensin II-dependent induc-
1984;309:724.
2004;1667(2):114.
192. Thornton SN. Thirst and hydration: physiology and consequences of dys- tion oNemer
223. AT(2) M, receptor expression
Chamberland M,aSirois
ter renal
D, etablation.
al. GeneAm J PhysiolofRenal
structure human Physiol.
car-
http://www.ncbi.nlm.nih.gov/pubmed/15581846
function. Physiol Behav. 2010;100(1):15–21. 2005;288(1):F207.
diac hormone precursor, pronatriodilatin. Nature. 1984;312:654.
165. Unger
193. Pech V , Kim
T, Badoer YH,E,Weinstein
Ganten D,AM, et al.etBrain
al. Angiotensin
angiotensin:IIpathways
increasesand chloride
phar- 189. Y
224. Lorenzo
andle TG. O, Minisymposium:
Ruiz-Ortega M, Esbrit P, et al. Angiotensin
the natriuretic peptide hormonesII increases parathy-
biochemis-
absorptionCirculation.
macology. in the cortical collecting duct
1988;75(Suppl 1):1.in mice through a pendrin-dependent roidofhormone-related
try natriuretic peptides. protein J Int(PTHrP) and the type 1 PTH/PTHrP receptor in the
Med. 1994;235:561.
mechanism
194. HannerAm F, JChambrey
Physiol Renal R, Physiol.
Bourgeois 2007;292(3):F914–920.
S, et al. Increased renal renin content kidney.Bloch
225. J AmKD, Soc Scott
Nephrol. JA, 2002;13(6):1595.
Zisfein JB, et al. Biosynthesis and secretion of proatrial
166.
in miceBeutler
lacking KT,the Masilamani
Na /H S, exchanger
Turban S, et al. Long-term
NHE2. regulation
Am J Physiol Renal oPhysiol.
ENaC http://www.ncbi.nlm.nih.gov/pubmed/12039989
natriuretic factor by cultured rat cardiocytes. Science. 1985;230:1168.
expression in kidney by angiotensin II. Hypertension. 2003;41(5):1143.
2008;294(4):F937–944. 190. Misono
226. KerstensK,MN, Fukumi van H, derGrammer
Kleij FG,RT, Boonstra
et al. Rat AH, et al.
atrial Angiotensin
natriuretic factor:admin-
com-
http://www.ncbi.nlm.nih.gov/pubmed/12682079
195. Bie P, Damkjaer M. Renin secretion and total body sodium: pathways of istration
plete amino stimulates
acid sequencerenal and 11 beta-hydroxysteroid
disul de linkage essential dehydrogenase
for biological activity in
activity.
167. Peti-Peterdi
integrative control. J,Clin Warnock DG, Bell
Exp Pharmacol PD. Angiotensin
Physiol. II directly stimulates
2010;37(2):e34–42. healthy men.
Biochem Biophys Kidney Int. 2004;65(6):2065.
Res Commun. 1984;119:524.
ENaCKurtz
196. activity A, in the cortical
Kaissling collecting
B, Busse duct
R, et al. via AT (1)cells
Endothelial receptors.
modulate J Am Socsecre-
renin Neph- http://www.ncbi.nlm.nih.gov/pubmed/15149319
227. Dietz JR, Scott DY, Landon CS, et al. Evidence supporting a physiological
rol. 2002;13(5):1131.
tion from isolated mouse juxtaglomerular cells. J Clin Inves. 1991;88:1147. 191.forZimmerman
role proANP-(1–30) MC. in Angiotensin
the regulation II andof angiotensin
renal excretion. 1–7 Am redox signaling
J Physiol in
Regul
168. Schweda
197. Wei Y, Wang F, KurtzW A. Angiotensin
RegulationIIofstimulates
renin release basolateral
by local K andchannels
systemicinfac-rat the central
Integr Compnervous
Physiol. system. Curr Opin Pharmacol. 2011;11(2):138–143.
2001;280(5):R1510.
cortical
tors. Revcollecting
Physiol Biochem ducts. Pharmacol.
Am J Physiol2009; RenalFebPhysiol.
17. 2003;284(1):F175. 192. Espiner
228. Thornton EA.SN. Thirst and hydration:
Mini-symposium: physiology
the natriuretic and consequences
peptide hormones. Physiol-o dys-
169. Durvasula
198. Zhuo JL. Intracrine
RV, Petermann renin AT,
andHiromura
angiotensin K, II: a novel
et al. role inofcardiovascular
Activation a local tissue unction.
ogy Physiol Behav.
of natriuretic peptides.2010;100(1):15–21.
J Int Med. 1994;235:427.
and renal cellular
angiotensin systemregulation.
in podocytes J Hypertens..
by mechanical 2006;24(6):1017–1020.
strain. Kidney Int. 2004;65(1):30. http://www.ncbi.nlm.nih.gov/pubmed/20211637
229. Shenker Y, Sider RS, Osta n EA, et al. Plasma levels of immunoreactive
http://www.ncbi.nlm.nih.gov/pubmed/16685198
199. Schlatter E, Salomansson M, Persson AE, et al. Macula densa cells sense 193. natriuretic
atrial Unger T, Badoer factor E,inGanten
healthyD,subjects
et al. Brainandangiotensin:
in patients pathwayswith edema. and Jphar-
Clin
170. Badzinska
luminal B, Sadowski via
NaCl concentration J. Opposed
furosemide e ects sensitive Na 2Cl–K E2
o prostaglandin on per u-
cotransport. macology.
Invest. Circulation. 1988;75(Suppl 1):1.
1985;76:1684.
Psion o rat
ugers Arch.renal cortex and medulla: interactions with the renin-angiotensin sys-
1989;414:286. 194. Hodsman
230. Hanner F, GP Chambrey
, Phillips R,PA,Bourgeois
Ogawa K, S, et al. Increased renal renin
Atrial natriuretic factorcon-
in
tem. Exp
200. Yao Physiol.
J, Oite 2008;93(12):1292–1302.
T, Kitamura M. Gap junctional intercellular communication in tent in mice
normal man:lacking
effects the Na+/H+
of tilt, exchanger
posture, exercise, NHE2.and Am J Physiol Renal
hemorrhage. Physiol.
J Hypertens.
http://www.ncbi.nlm.nih.gov/pubmed/18586855
the juxtaglomerular apparatus. Am J Physiol Renal Physiol. 2009;296(5):F939–946. 2008;294(4):F937–944.
1986;4(Suppl):S503.
171. Koppa
201. MadsenUC, K, Dibona
Marcussen GF. N, Pedersen
Neural M, et al.
regulation Angiotensin
of renin II promotes
secretion. devel-
Semin Nephrol. 195. Schiebinger
231. Bie P, Damkjaer M. Renin
RJ, Linden secretion
J. Effect and total
of atrial body sodium:
contraction frequency pathways
on atrialo
opment o the renal microcirculation through AT1 receptors. J Am Soc Nephrol.
1993;13:543. integrative peptide
natriuretic control. secretion.
Clin Exp Pharmacol
Am J Physiol. Physiol. 2010;37(2):e34–42.
1986;251:H1095.
2010;21(3):448–459.
202. Zeng C, Zhang M, Asico LD, et al. The dopaminergic system in hyperten- 196. Nishimura
232. Kurtz A, Kaissling
K, Ban B, T, Busse
Saito R, Y, etetal.al.Endothelial
Atrial pacing cellsstimulates
modulatesecretion
renin secre-of
http://www.ncbi.nlm.nih.gov/pubmed/20056745
sion. Clin Sci. 2007;112(12):583–597. tion rom
atrial isolatedpolypeptide
natriuretic mouse juxtaglomerular
without elevation cells. JofClin Inves.
atrial 1991;88:1147.
pressure in awake dogs
172. Charloux
203. Wol G, Neilson EG. Angiotensin
A, Brandenberger G, PiquardII induces
F, et al.cellular hypertrophy
Dysregulation in cul-
of pulsatility http://www.ncbi.nlm.nih.gov/pubmed/1717509
with experimental complete atrioventricular block. Circ Res. 1990;66:115.
tured
in murine
aging proximal
IV. Pulsatile tubular cells.
signalling Am J Physiol. 1990;259:F768.
and cardiovascular aging: functions and regula- 197. Manning
233. Schweda FPT, , Kurtz A. Regulation
Schwartz D, Katsube o renin NC, release
et al.byVasopressin-stimulated
local and systemic ac-
173.ofChen
tion J, Chen
natriuretic JK, Neilson
peptide signaling. EG,Ageing
et al.ResRole Rev.o 2008;7(3):151–163.
EGF receptor activation in tors. RevofPhysiol
release Biochem endocrine
atriopeptin: Pharmacol. antagonists
2009; Feb 17. in uid homeostasis. Science.
angiotensin
204. Sigmund II-induced
CD, Jonesrenal epithelial
CA, Kane CM, etcell hypertrophy.
al. Regulated tissue-J Amand Soc Nephrol.
cell-speci c 198. Durvasula RV, Petermann AT, Hiromura K, et al. Activation o a local tissue
1985;229:395.
2006;17(6):1615–1623.
expression of the human renin gene in transgenic mice. Circ Res. 1992;70:1070. angiotensin
234. Staschsystem
JP, Hirthin podocytes
C, Kazda by mechanical
S, et al. Endothelin strain.stimulates
Kidney Int.release
2004;65(1):30.
of atrial
174. Johns
205. Wol G, DW, Jablonski
Peach MJ, K, Gomez
Schroeder RA,R,etetal.al.Angiotensin
AngiotensinII II-induced
regulates renin hypertro-
gene http://www.ncbi.nlm.nih.gov/pubmed/14675034
natriuretic peptides in vitro and in vivo. Life Sci. 1989;45:869.
phy o proximal
expression. Am J tubular
Physiol. cells requires p27Kip1. Kidney Int. 2003;64(1):71.
1990;259:F882. 199. Y
235. Schlatter
uan K, YE, u J,Salomansson
Shah A, et al. M, Leptin
PerssonreducesAE, et plasma
al. Macula ANP densa
levelcells sense
via nitric
175. Larsson
206. Gorin Y,C, Ricono
Weber JM,P, Wagner
AnggardB,E.et Arachidonic
al. Angiotensin acidII-induced
increasesERK1/ERK2
and indo- luminal NaCl concentration
oxide-dependent mechanism. Am viaJ Physiol
urosemide sensitive
Regul Integr CompNa +
2Cl–K2010;298(4):
Physiol. +
cotransport.
activation decreases
methacin and protein synthesis
plasma reninare redox-dependent
activity in the rabbit.inEur glomerular
J Pharmacol. mesangial
1974; Pf ugers Arch. 1989;414:286.
R1007–1016.
cells. Biochem J. 2004;381(Pt 1):231.
28:391. http://www.ncbi.nlm.nih.gov/pubmed/2780213
236. Kim SW, Li Y, Kim S, et al. Local renal and vascular natriuretic peptide
176. Antonipallai
207. Liu BC, SunI,J,Nadler Chen Q, JL, et al. Role
Robin EC, oet connective tissue growth
al. The inhibitory role of 12– actor
andin 200. Yin
system ao obstructive
J, Oite T, Kitamura
uropathicM.rats. GapUrol junctional intercellular communication in
Res. 2002;30(2):97.
mediating hypertrophy
15–lipoxygenase products o human
on reninproximal tubular cells induced
release. Hypertension. 1987;10:61. by angiotensin the juxtaglomerular
237. Kalinowski L, apparatus. Am J PhysiolM,
Szczepanska-Konkel Renal Physiol. 2009;296(5):F939–946.
Jankowski M, et al. Studies on po-
II. AmHeinrich
208. J Nephrol.WL, 2003;23(6):429.
Falck JR, Campbell WB. Inhibition of release by 14, 201. Koppa
tential UC, Dibona
involvement of proteinGF. Neural
kinase regulation
C in glomerular o renin secretion. Semin
insensitivity to atrialNephrol.
natri-
177. Guo DF, Chenier I,acid
15–epoxyeicosatrienoic Lavoie JL, et
in renal al. Development
cortical slices. Am JoPhysiol.
hypertension and kid-
1990;258:E269. 1993;13:543.
uretic factor on low sodium intake. Med Sci Monit. 2001;7(4):628.
ney hypertrophy
209. Bader M. Tissue in transgenic mice overexpressing ARAP1
renin-angiotensin-aldosterone systems: gene in theforkidney.
Targets phar- http://www.ncbi.nlm.nih.gov/pubmed/8278687
238. Takayanagi R, Snajdar RM, Imada T, et al. Puri cation and characterization
Hypertension.therapy.
macological 2006;48:453–459.
Annu Rev Pharmacol Toxicol. 2010;50:439–465. 202.
of two Zeng
typesC, of Zhang M, Asico LD,
atrial natriuretic factor et receptors
al. The dopaminergic
from bovine adrenal system in hyperten-
cortex: gua-
http://www.ncbi.nlm.nih.gov/pubmed/16801480
210. Fyhrquist F, Saijonmaa O. Renin-angiotensin system revisited. J Intern Med. sion. Clin
nylate Sci. 2007;112(12):583–597.
cyclase-linked and cyclase-free receptors. Biochem Biophys Res Commun.
178. Wong NL, Tsui JK. Angiotensin II upregulates the expression o vaso-
2008;264(3):224–236. http://www.ncbi.nlm.nih.gov/pubmed/17492945
1987;144:244.
pressinHiranyachattada
211. V2 mRNA in the inner medullary
S, Harris PJ. Regulation collecting
of renal duct o the rat.
proximal uidMetabolism.
uptake by 203. Leitman
239. CharlouxDC, A, Brandenberger
Andresen JW, Catalano G, PiquardRM, F, etetal.al.Dysregulation
Atrial natriuretic o pulsatility
peptide
2003;52(3):290.
luminal and peritubular angiotensin II. J Renin Angiotensin Aldosterone System. in aging IV
binding, Pulsatile signalling
cross-linking and cardiovascular
and stimulation of cyclic GMP aging: unctions and
accumulation andregulation
particu-
http://www.ncbi.nlm.nih.gov/pubmed/12647265
2004;5(2):89. o natriuretic
late guanylatepeptidecyclase signaling.
activity in Ageing
cultured Rescells.
Rev. 2008;7(3):151–163.
J Biol Chem. 1988;263:3720.
179. Jackson
212. Kobori EK, H, Herzer
Nishiyama A. E extraction
WA. Renal ects o tempol on renalII. angiotensinogen
of angiotensin J Pharmacol Exp http://www.ncbi.nlm.nih.gov/pubmed/18243818
240. Forssmann W, Meyer M, Forssmann K. The renal urodilatin system: clini-
production
Ther. in Dahl salt-sensitive rats. Biochem Biophys Res Commun. 2004;315:
2003;307(3):1001. 204.
cal Sigmund CD,
implications. Jones CA,
Cardiovasc Res.Kane CM, et al. Regulated tissue- and cell-speci c
2001;51(3):450.
746–750.
213. Park SY, Song CY, Kim BC, et al. Angiotensin II mediates LDL- expression
241. Zhao oD,the human
Pandey KN,reninNavar gene
LG.inANP-mediated
transgenic mice. Circ Res.
inhibition of1992;70:1070.
distal nephron
http://www.ncbi.nlm.nih.gov/pubmed/14975764
induced superoxide generation in mesangial cells. Am J Physiol Renal Physiol. 205. Johns
fractional DW, Peach
sodium MJ, Gomez
reabsorption RA, et al.and
in wild-type Angiotensin II regulates natriuretic
mice overexpressing renin gene
180. Vanegas V, Ferrebuz A, Quiroz Y, et al. Hypertension. in Page (cellophane-
2003;285(5):F909. expression.
peptide Am J Physiol.
receptor. Am J Physiol 1990;259:F882.
Renal Physiol. 2010;298(1):F103–108.
wrapped)
214. Raizadakidney is due toB,interstitial
V, Skipper Luo W, Grif nephritis. Kidney Int. and
th J. Intracardiac 2005;68:1161–1170.
intrarenal renin- 206. Koller
242. Larsson KJ,C,Lowe
Weber DG, P, Anggard
Bennett GL, E. Arachidonic
et al. Selective acidactivation
increases of and indometh-
B natriuretic
http://www.ncbi.nlm.nih.gov/pubmed/16105047
angiotensin systems: mechanisms of cardiovascular and renal effects. J Investig acin decreases
peptide receptor plasma reninnatriuretic
by C-type activity in the rabbit.
peptide. Eur J Pharmacol.
Science. 1991;252:120. 1974; 28:391.
181. 2007;57(7):341–359.
Med. Prieto-Carrasquero MC, Botros F, Kobori H, et al. Angiotensin II regulates 207. Maack
243. Antonipallai
T, SuzukiI, Nadler JL, Robin
M, Almeida FA, et EC, al.etPhysiological
al. The inhibitory role of role
silento receptors
12– and
distal Fisher
215. nephron ND, renin
Allan geneDR,expression
Gaboury in CL,Goldblatt
Hollenberg hypertensive
NK. Intrarenal rats indepen-
angio- 15–lipoxygenase
of atrial natriuretic products on renin1987;238:675.
factor. Science. release. Hypertension. 1987;10:61.
dently IIrom
tensin high blood
formation pressure (Abstract).
in humans: evidence from Hypertension. 2005;46:871.
renin inhibition. Hypertension. http://www.ncbi.nlm.nih.gov/pubmed/3298043
244. Sun JZ, Chen SJ, Majid-Hasan E, et al. Dietary salt supplementation
182. Navar LG. The intrarenal renin-angiotensin system in hypertension. Kid-
1995;25:935–939. 208. Heinrich
selectively WL, FalckNPR-C
downregulates JR, Campbell
receptorWB. Inhibition
expression o release
in kidney by 14, 15–
independently
ney Int.
216. 2004;65:1522–1532.
Perlstein TS, Gumieniak O, Hopkins PN, et al. Uric acid and the state of the epoxyeicosatrienoic
of ANP. Am J Physiolacid RenalinPhysiol.
renal cortical slices. Am J Physiol. 1990;258:E269.
2002;282(2):F220.
183. Abadir
intrarenal PM. The rail
renin-angiotensin systemrenin-angiotensin
in humans. Kidney system. Clin Geriatr Med.
Int. 2004;66:1465–1470. 209. Obineche
245. Bader M. Tissue renin-angiotensin-aldosterone
EN, Adeghate E, Chandranath IS, etsystems: Targets in
al. Alterations or phar-
atrial
2011;27(1):53–65.
217. Siragi HM, Carey RM. Role of the intrarenal renin-angiotensin-aldosterone macologicalpeptide
natriuretic therapy.and Annu its Rev Pharmacol
receptors Toxicol. 2010;50:439–465.
in streptozotocin-induced diabetic rat kid-
http://www.ncbi.nlm.nih.gov/pubmed/21093722
system in chronic kidney disease. Am J Nephrol. 2010;31(6):541–550. http://www.ncbi.nlm.nih.gov/pubmed/20055710
neys. Mol Cell Biochem. 2004;261(1–2):3.
CHAPTER 8 HORMONES AND THE KIDNEY 2283
83

210. Klokkers
246. FyhrquistJ,F, Saijonmaa
Langehanenberg O. Renin-angiotensin
P, Kemper B, et system revisited.
al. Atrial J Intern pep-
natriuretic Med. 234. Stasch S,
273. Satyan JP,Light
HirthRPC, Kazda S,
, Agarwal R. et al. Endothelin
Relationships stimulatespro-B-natriuretic
of N-terminal release o atrial
2008;264(3):224–236.
tide and nitric oxide signaling antagonizes vasopressin-mediated water per- natriuretic
peptide andpeptides in vitro and
cardiac troponin T toinleft
vivo. Life Sci. mass
ventricular 1989;45:869.
and function and mortality
http://www.ncbi.nlm.nih.gov/pubmed/18793332
meability in inner medullary collecting duct cells. Am J Physiol Renal Physiol. http://www.ncbi.nlm.nih.gov/pubmed/2552240
in asymptomatic hemodialysis patients. Am J Kidney Dis. 2007;50(6):1009–1019.
211. Hiranyachattada S, Harris PJ. Regulation o renal proximal f uid uptake
2009;297(3):F693–703. 235. Yuan K,MH.
274. Rosner Yu J,Measuring
Shah A, et risk al. Leptin reducesrenal
in end-stage plasma ANP level
disease: via nitric pro
is N-terminal ox-
by luminal
247. Brenner andBM,peritubular
Ballermanangiotensin
BJ, Gunning II. ME,
J Renin Angiotensin
et al. Aldosterone
Diverse biological System.
actions of ide-dependent
brain natriureticmechanism.
peptide a useful Am J Physiol
marker?Regul KidneyIntegr
Int.Comp Physiol. 2010;298(4):
2007;71(6):481–483.
2004;5(2):89.
atrial natriuretic peptide. Physiol Rev. 1990;70:665. R1007–1016.
275. Madsen LH, Ladefoged S, Corell P, et al. N-terminal pro brain natriuretic
http://www.ncbi.nlm.nih.gov/pubmed/15295721
248. Nicholls MG. Mini-symposium: the natriuretic peptide hormones. Edito- 236.
peptide Kim SW, Li
predicts Y, Kim in
mortality S, patients
et al. Local withrenal and vascular
end-stage natriuretic
renal disease peptide
in hemodialy-
212.andJackson
rial EK,review.
historical HerzerJ IntWA. Med.Renal extraction o angiotensin II. J Pharmacol
1994;235:507. system in obstructive
sis. Kidney uropathic rats. Urol Res. 2002;30(2):97.
Int. 2007; 71(6):548–554.
Exp Ther.
249. Fried 2003;307(3):1001.
TA, McCoy RN, Osgood RW, et al. Effect of atriopeptin II on deter- 237. Kalinowski
276. Wang AY, Lam L, Szczepanska-Konkel
CW, Yu CM, et alN-terminal M, Jankowski M, etnatriuretic
pro-brain al. Studiespeptide:
on po-
213. Park
minants SY, Song CY
of glomerular , Kimrate
ltration BC,in ettheal. Angiotensin
in vitro perfusedIIdog mediates
glomerulus.LDL-in-
Am tential involvement
an independent risko predictor
protein kinase C in glomerular
of cardiovascular insensitivity
congestion, to atrial
mortality, andnatri-
ad-
Jduced
Physiol.superoxide
1986;250:F1119.generation in mesangial cells. Am J Physiol Renal Physiol. uretic actor on low sodium
verse cardiovascular outcomes intake. Med Sciperitoneal
in chronic Monit. 2001;7(4):628.
dialysis patients. J Am Soc
2003;285(5):F909.
250. Cogan MG. Atrial natriuretic factor can increase solute excretion primarily 238.
Nephrol.Takayanagi R, Snajdar RM, Imada T, et al. Puri cation and characterization
2007;18(1):321–330.
214.
by Raizada
raising V, Skipper
glomerular B, Luorate.
ltration W, AmGri J Physiol.
th J. Intracardiac
1986;250:F710.and intrarenal renin- o277.
twoNigwekar
types o atrial SU, natriuretic
Navaneethan actor
SD,receptors
Parikh CR, rometbovine adrenal
al. Atrial cortex: pep-
natriuretic gua-
angiotensin
251. Yukimura systems:
T, Ito mechanisms
K, Takenaga T, o etcardiovascular
al. Renal effects and renal e ects.
of synthetic human J Investig
atrial nylate
tide forcyclase-linked
preventing andand cyclase-
treating acutereekidney
receptors.
injury.Biochem
Cochrane Biophys Res Commun.
Database Syst Rev.
Med. 2007;57(7):341–359.
natriuretic polypeptide in anesthetized dogs. Eur J Pharmacol. 1984;103:363. 1987;144:244.
2009;(4):CD006028.
215. Golos
252. FisherM,ND, LewkoAllan DR, E,
B, Bryl Gaboury
et al. EffectCL,ofHollenberg
angiotensinNK. II onIntrarenal
ANP-dependentangio- http://www.ncbi.nlm.nih.gov/pubmed/2883969
278. Shimada M, Ejaz AA, Beaver TM. Role of natriuretic peptides in cardiovas-
tensin II cyclase
guanylyl ormation in humans:
activity in cultured evidence
mouse and romratrenin inhibition.
podocytes. KidneyHypertension.
Blood Press 239. LeitmanExpert
cular surgery. DC, Andresen
Rev CardiovascJW, Catalano RM, et al. Atrial natriuretic peptide
Ther. 2009;7(5):515–519.
1995;25:935–939.
Res. 2002;25(5):296. binding, cross-linking
279. Nigwekar SU, Hix and JK.stimulation
The role o of
cyclic GMP accumulation
natriuretic and particu-
peptide administration
http://www.ncbi.nlm.nih.gov/pubmed/7737730
253. Singhal PC, Decandido S, Satriano JA, et al. Atrial natriuretic peptide late guanylate cyclase
in cardiovascular activity in cultured
surgery-associated cells.dysfunction:
renal J Biol Chem. a1988;263:3720.
systematic review
216. nitroprusside
and Perlstein TS,causesGumieniak O, Hopkins
relaxation of culturedPN, etmesangial
al. Uric acid and
cells. AmtheJ Physiol.
state o http://www.ncbi.nlm.nih.gov/pubmed/2894373
and meta-analysis of randomized controlled trials. J Cardiothorac Vasc Anesth.
the intrarenal renin-angiotensin system in humans. Kidney Int. 2004;66:1465–
1989;257:C86. 240. Forssmann W, Meyer M, Forssmann K. The renal urodilatin system: clini-
2009;23(2):151–160.
1470. Camargo MJF, Kleinert HD, Atlas SA, et al. Ca-dependent hemodynam-
254. cal
280.implications.
Nigwekar SU, Cardiovasc
NavaneethanRes. 2001;51(3):450.
SD, Parikh CR, et al. Atrial natriuretic peptide
http://www.ncbi.nlm.nih.gov/pubmed/15458439
ic and natriuretic effects of atrial extract in isolated rat kidney. Am J Physiol. http://www.ncbi.nlm.nih.gov/pubmed/11476735
for management of acute kidney injury: a systematic review and meta-analysis.
217. Siragi HM, Carey RM. Role o the intrarenal renin-angiotensin-aldosterone
1984;246:F447. 241. Zhao
Clin J Am SocD,Nephrol.
Pandey 2009;4(2):261–272.
KN, Navar LG. ANP-mediated inhibition o distal nephron
systemDe
255. inlos
chronic
Angeleskidney
Costadisease. Am J Nephrol.
M, Elesgaray R, et al.2010;31(6):541–550.
Atrial natriuretic peptide in u- ractional
281. Murraysodium reabsorption
P. Brain natriureticinpeptide
wild-type and mice
therapy overexpressing
to prevent acute kidney natriuretic
injury
218. onMiyake-Ogawa
ence nitric oxide systemC, Miyazaki
in kidney M,and Abe K, etRegul
heart. al. Tissue-speci c expression
Peptide. 2004;118(3):151. peptide receptor.
after cardiac surgery.Am Am J Physiol Renal
J Kidney Dis.Physiol. 2010;298(1):F103–108.
2008;51(1):5–9.
o renin-angiotensin
256. Takezawa K, Cowley systemAW components
Jr, SkeltoninM,IgA et nephropathy.
al. AtriopeptinAm III Jalters
Nephrol.
re- 242. Koller PM,
282. Bryan KJ, Lowe
Xu X,DG, Dickey Bennett
DM, GL,et al.etRenal
al. Selective activation o Btonatriuretic
hyporesponsiveness atrial na-
2005;25:1–12.
nal medullary hemodynamics and the pressure–diuresis response in rats. Am J peptide
triuretic receptor
peptide in bycongestive
C-type natriuretic
heart failurepeptide.
resultsScience. 1991;252:120.
from reduced atrial natriuretic
219. Mezzano
Physiol. SA, Aros CA, Droguett A, et al. Renal angiotensin II up-regulation
1987;252:F992. http://www.ncbi.nlm.nih.gov/pubmed/1672777
peptide receptor concentrations. Am J Physiol Renal Physiol. 2007;292(5):F1636–
and myo
257. Van de broblast
Stolpeactivation
A, JamisoninRL. human membranous
Micropuncture studynephropathy.
of the effect Kidney
of ANP Int.on 243.
1644. Maack T, Suzuki M, Almeida FA, et al. Physiological role o silent receptors
2003;Suppl
the papillary64:S39–S45.
collecting duct in the rat. Am J Physiol. 1988;254:F477. o283.
atrial natriuretic
Chen HH, Huntley actor. Science. 1987;238:675.
BK, Schirger JA, et al. Maximizing the renal cyclic
220. Bailly
258. Kengne-Wa
C. Effecto S,ofMassella L, Diomedi-Camassei
luminal atrial natriuretic peptide F, on
et al. Risk actors
chloride reabsorp- or http://www.ncbi.nlm.nih.gov/pubmed/2823385
3 -5 -guanosine monophosphate system with type V phosphodiesterase in-
cyclosporine
tion in mouseAcortical
nephrotoxicity in children
thick ascending limb:with steroid-dependent
inhibition by endothelin.nephritic
J Amsyn-
Soc 244.
hibitionSunand JZ,exogenous
Chen SJ, Majid-Hasan
natriuretic peptide:E, et al.aDietary salt supplementation
novel strategy to improve renal se-
drome. Clin
Nephrol. J Am Soc Nephrol. 2009;4(9):1409–1416.
2000;11(10):1791. lectively
function downregulates
in experimental NPR-C
overtreceptor expression
heart failure. J Am in Sockidney
Nephrol. independently
2006;17(10): o
221. Harris
259. Oka K,PJ,Moriyama
Thomas D, T, Takahara
Morgan TO. S, etAtrial
al. Increased
natriuretic expression o reninan-
peptide inhibits in ANP. Am J Physiol Renal Physiol. 2002;282(2):F220.
2742–2747.
chronic allogra t nephropathy.
giotensin-stimulated proximal Transplant
tubular sodium Proc. 2005;37:2131–2134.
and water reabsorption. Nature 245. Obineche
284. Chen LG, WangEN, Adeghate
ZR, Wan E, CM, Chandranath IS, et al.
et al. Encapsulated Alterations
transgene cellsinattenu-
atrial
http://www.ncbi.nlm.nih.gov/pubmed/15964360
(London). 1987;326:697. natriuretic peptidecardiac
ate hypertension, and itshypertrophy
receptors in and streptozotocin-induced
enhance renal function diabetic rat kid-
in Goldblatt
222. Majowicz
260. Oikawa S,MP Imai M, Ueno
, Gonzalez BoscA,LVet al. Cloning
, Albertoni and sequence
Borghese MF, et al.analysis
Atrial na- o neys. Mol Cellrats.
hypertensive Biochem.
J Gene2004;261(1–2):3.
Med. 2004;6(7):786.
cDNA encoding:
triuretic peptide and a precursor
endothelin-3humantarget atrialrenal
natriuretic For polypeptide.
sodium-glucose Nature.
cotransporter. 246. KlokkersJPJ,
285. Valentin Langehanenberg
, Humphreys P, Kemper aB,paracrine
MH. Urodilatin: et al. Atrial
renal natriuretic
natriuretic pep-
1984;309:724.
Peptides 2003;24(12):1971. tide
tide. and
Seminnitric
Nephrol.oxide signaling antagonizes vasopressin-mediated water per-
1993;13:61.
http://www.ncbi.nlm.nih.gov/pubmed/6203042
261. Caruso-Neves C, Vives D, Dantas C, et al. Ouabain-insensitive Na -ATPase meability in inner medullary
286. Schulz-Knappe P, Forssmann collecting duct cells.
K, Herbst AmIsolation
F, et al. J Physioland Renal Physiol.
structural
223.
of Nemertubules
proximal M, Chamberland
is an effector M, for
Sirois D, et al. and
urodilatin Geneatrial
structure o human
natriuretic car-
peptide. 2009;297(3):F693–703.
analysis of “urodilatin,” a new peptide of the cardiodilatin-(ANP)-family, extract-
diac hormone
Biochim Biophysprecursor, pronatriodilatin. Nature. 1984;312:654.
Acta. 2004;1660(1–2):93. 247.
ed from Brenner
humanBM, Ballerman
urine. BJ, Gunning
Klin Wochenshr. ME, et al. Diverse biological actions o
1988;66:752.
http://www.ncbi.nlm.nih.gov/pubmed/6095118
262. Ziedel ML. Medullary collecting duct sodium transport. Am J Physiol. atrial
287. natriuretic
Drummer C, peptide.
Fiedler Physiol
F, Bub A,Rev.et al.1990;70:665.
Development and application of a urodilatin
224. Yandle TG. Minisymposium: the natriuretic peptide hormones biochemis-
1993;265:F159. http://www.ncbi.nlm.nih.gov/pubmed/2141944
(CDD/ANP-95–126)-speci c radioimmunoassay. P ugers Arch. 1993;423:372.
try o Metzler
263. natriureticCH,peptides.
Ramsay DJ. J IntPhysiological
Med. 1994;235:561. doses of atrial peptide inhibit angio- 248. NichollsSK,
287a.Dubois MG. Mini-symposium:
Kishimoto I, Lillis TO, theetnatriuretic
al. A genetic peptide
model hormones.
de nes the Edito-
im-
225. Bloch
tensin KD, Scott
II-stimulated JA, Zis einsecretion.
aldosterone JB, et al. Biosynthesis and secretion o proatrial
Am Physiol. 1989;256:R1155. rial and historical
portance of the atrial review. J Int Med.peptide
natriuretic 1994;235:507.
receptor (guanylyl cyclase-A) in the
natriuretic
264. actor by cultured
Oliveira-Souza rat cardiocytes.
M, Mello-Aires M. EffectScience. 1985;230:1168.
of arginine vasopressin and ANP http://www.ncbi.nlm.nih.gov/pubmed/8207358
regulation of kidney function. Proc Natl Acad Sci U S A. 2000;97(8):4369.
2
226.
on Misono K,pH
intracellular Fukumi H, Grammer
and cytosolic RT, et ]al.regulation
free [Ca Rat atrial innatriuretic
MDCK cells.actor:Kidney
com- 249. Fried TA, McCoy
288. Hidebrandt DA, MizeueRN, OsgoodHL, BrandsRW, etMW, al. Eetectal.oComparison
atriopeptin of II on
the deter-
renal
plete2001;60(5):1800.
Int. amino acid sequence and disul de linkage essential or biological activity. minants
actions ofo urodilatin
glomerular andltration rate in thepeptide.
atrial natriuretic in vitro Am per Jused dog1992;262:R395.
Physiol. glomerulus. Am
Biochem
265. Koh Biophys
GY, KlugRes Commun.
MG, Field1984;119:524.
LJ. Atrial natriuretic factor and transgenic mice. J289.
Physiol. 1986;250:F1119.
Gunning ME, Otuechere G, Zeidel ML. Mechanism of urodilatin (ANP
http://www.ncbi.nlm.nih.gov/pubmed/6538787
Hypertension. 1993;22:634. 250.
95–126; Cogan
URO) MG. Atrial natriuretic
inhibition actor caninincrease
of Na transport solutemedullary
rabbit inner excretion collecting
primarily
227. Field
266. Dietz LJ.
JR, Transgenic
Scott DY, Landon mice inCS, et al. Evidence
cardiovascular supporting
physiology. a physiological
Annu Rev Physiol. by
ductraising
cells. glomerular
J Am Soc Nephrol. ltration rate. Am J Physiol. 1986;250:F710.
1991;2:402.
role or proANP-(1–30) in the regulation o renal excretion. Am J Physiol Regul
1993;55:97. 251. Yukimura T,M,
290. Gagelmann ItoHock
K, Takenaga T, et al.WG.
D, Forssman Renal e ects o (CDD/ANP-95–126)
Urodilatin synthetic human atrial is
Integr Steinhelper
267. Comp Physiol. ME,2001;280(5):R1510.
Cochrane KL, Field LJ. Hypotension in transgenic mice natriuretic polypeptide
not biologically inactivated in anesthetized
by a peptidase dogs.
from EurdogJ Pharmacol.
kidney cortex 1984;103:363.
membranes in
228. Espiner
expressing EA.
atrial Mini-symposium:
natriuretic factor fusion the genes.
natriuretic peptide hormones.
Hypertension. 1990;16:301. Physiol- http://www.ncbi.nlm.nih.gov/pubmed/6237925
contrast to atrial natriuretic peptide/cardiodilatin (a-ANP/CDD-99–126). FEBS
ogy o John
268. natriuretic
SWM, peptides.
Krege JH,J Int Med.PM,
Oliver 1994;235:427.
et al. Genetic decreases in atrial natri- 252. Golos M, Lewko B, Bryl E, et al. E ect o angiotensin II on ANP-dependent
Lett. 1988;233:249.
229. peptide
uretic Shenkerand Y, Sider RS, Ostahypertension.
salt-sensitive n EA, et al. Science.
Plasma 1995;267:679.
levels o immunoreactive guanylyl
291. Rieggercyclase GAJ,activity
ElsnerinD, cultured
Forssmann,mouseetand rat podocytes.
al. Effects Kidney Blood
of ANP-(95–126) Press
in dogs
atrial Lopez
267. natriuretic actor in healthy
MJ, Kishimoto I, Machsubjects
V, et al. and in patients
Salt-resistant with edema.
hypertension in Jmice
Clin Res.
before2002;25(5):296.
and after induction of heart failure. Am J Physiol. 1990;259:H1643.
Invest. 1985;76:1684.
lacking the guanylyl cyclase-A receptor for atrial natriuretic peptide. Nature http://www.ncbi.nlm.nih.gov/pubmed/12435875
291a.Drummer C, Fiedler F, Konig A, et al. Urodilatin, a kidney-derived na-
(http://www.ncbi.nlm.nih.gov/pubmed/2932471
London). 1995;378:65. 253. Singhal
triuretic factor,PC, Decandido
is excreted withS,a Satriano
circadian JA, et al.and
rhythm Atrial natriureticbypeptide
is stimulated saline
230. Sudoh
268. Hodsman GP, Phillips
T, Kanagawa PA, OgawaN,K,etetal.al.A Atrial
K, Minamino natriureticpeptide
new natriuretic actor inin and nitroprusside
infusion in man. J Am causes relaxation
Soc Nephrol. o cultured mesangial cells. Am J Physiol.
1991;1:1109.
normal brain.
porcine man: Nature
e ects (London).
o tilt, posture,
1988;332:78. exercise, and hemorrhage. J Hypertens. 1989;257:C86.
292. Drummer C, Heer M, Baisch F, et al. Diuresis and natriuresis following iso-
1986;4(Suppl):S503.
269. Altered regulation of renal interstitial hydrostatic pressure and the renal 254. Camargo
tonic saline MJF, in
infusion Kleinert
healthyHD, young Atlas SA, et al.before,
volunteers Ca-dependent
during, and hemodynam-
after HDT.
231. Schiebinger system
renin-angiotensin RJ, Lindenin theJ.absence
E ect oofatrial atrial contraction requency
natriuretic peptide. on atrial
J Hypertens. ic and
Acta natriuretic
Physiol Scand. e1992;144(S604):101.
ects o atrial extract in isolated rat kidney. Am J Physiol.
natriuretic peptide secretion. Am J Physiol. 1986;251:H1095.
2008;26(2):303–311. 1984;246:F447.
293. Goetz KL. Renal natriuretic peptide (urodilatin?) and atriopeptin: evolving
232. Kishimoto
270. NishimuraI,K,Hamra Ban T,FK, Saito Y, et DL.
Garbers al. Atrial
Apparentpacing stimulates
B-type secretion
natriuretic peptide o 255. De Am
concepts. los JAngeles Costa M, Elesgaray R, et al. Atrial natriuretic peptide
Physiol. 1991;261:F921.
atrial natriuretic
selectivity in the polypeptide
kidney due to without elevation
differential o atrialCan
processing. pressure in awake
J Physiol dogs
Pharmacol. inf
294.u ence
Hamra onRK, nitric
Forte oxide
LR, Eber system
SL, et al.in Uroguanylin:
kidney andstructure heart. andRegul Peptide.
activity of a
with experimental complete atrioventricular block. Circ Res. 1990;66:115.
2001;79(8):715. 2004;118(3):151.
second endogenous peptide that stimulates intestinal guanylate cyclase. Proc Natl
http://www.ncbi.nlm.nih.gov/pubmed/2136811
271. Sudoh I, Minamino N, Kanagawa K, et al. C-type natriuretic peptide (NP): 256.
Acad SciTakezawa K, Cowley AW Jr, Skelton M, et al. Atriopeptin III alters re-
U S A. 1993;90:10464.
a233.
new Manning
member ofPT, Schwartzpeptide
natriuretic D, Katsube NC, etedal.in Vasopressin-stimulated
family identi porcine brain. Biochem nal
295.medullary
Kita T, Smithhemodynamics
CE, Fok KF and
, et the pressure–diuresisofresponse
al. Characterization in rats. Am J
human uroguanylin:
release Res
Biophys o Commun.
atriopeptin: endocrine antagonists in f uid homeostasis. Science.
1990;168:863. Physiol.
member1987;252:F992.
of the guanylin peptide family. Am J Physiol. 1994;266:F342.
1985;229:395.
272. Use of cardiac biomarkers in end-stage renal disease. J Am Soc Nephrol. 257. Van deLRStolpe
296. Forte A, Jamisonand
Jr. Uroguanylin RL.guanylin
Micropuncturepeptides: study o the e ectand
pharmacology o ANP on
experi-
http://www.ncbi.nlm.nih.gov/pubmed/2990050
2008;19(9):1643–1652. the papillary
mental collecting
therapeutics. duct in the
Pharmacol rat.2004;104(2):137.
Ther. Am J Physiol. 1988;254:F477.
284
28 4 SECTION I STRUCTURAL AND FUNCTIONAL CORRELATIONS IN THE KIDNEY

258. Schulz
297. Bailly C. S, E ect o luminal
Chrisman atrial natriuretic
TD, Garbers DL. Cloning peptide on chloride
and expression reabsorp-
of guanylin: 325. Nigwekar
279. Garty SU, Hix JK.
H. Regulation of NaThepermeability
role o natriuretic peptide Semin
by aldosterone. administration
Nephrol.
tionexistence
its in mouse in cortical
various thick ascending
mammalian limb:
tissues. inhibition
J Biol by endothelin. J Am Soc
Chem. 1992;267:16019. in cardiovascular surgery-associated renal dys unction: a systematic review
1992;12:24.
Nephrol.
298. 2000;11(10):1791.
Wiegand RC, Kato J, Currie MG. Rat guanylin cDNA: characterization of and meta-analysis
326. Palmer LG, Frindt o randomized
G. Regulation controlled
of apicaltrials. J Cardiothorac
membrane Na and KV asc Anesth.
channels in
259.precursor
the Harris PJ, Thomas
of an D, Morgan
endogenous activator TO.ofAtrial natriuretic
intestinal guanylatepeptide
cyclase.inhibits an-
Biochem 2009;23(2):151–160.
rat renal collecting tubules by aldosterone. Semi Nephrol. 1992;12:37.
giotensin-stimulated
Biophys proximal tubular sodium and water reabsorption. Nature
Res Commun. 1992;185:812. http://www.ncbi.nlm.nih.gov/pubmed/19167908
327. O’Neil RG. Aldosterone regulation of sodium and potassium transport in
(London).
299. Forte 1987;326:697.
LR, Fan X, Hamra K. Salt and water homeostasis: uroguany- 280. Nigwekar
cortical collectingSU, Navaneethan
duct. Semin Nephrol.SD, Parikh CR, et al. Atrial natriuretic peptide
1990;10:365.
http://www.ncbi.nlm.nih.gov/pubmed/2951600
lin is a circulating peptide hormone with natriuretic activity. Am J Kidney Dis. or management
328. Lingueglia E, o V acute
oilleykidney injury: aR,systematic
N, Waldmann review and
et al. Expression meta-analysis.
cloning of an epi-
260. Majowicz MP, Gonzalez Bosc LV, Albertoni Borghese MF, et al. Atrial na-
1996;28:296. Clin J Am
thelial Soc Nephrol. 2009;4(2):261–272.
amiloride-sensitive Na channel. FEBS Lett. 1993;318:95.
triuretic
300. peptide
Haynes RC and endothelin-3 target hormone:
Jr. Adrenocorticotropic renal sodium-glucose cotransporter.
adrenocorticosteroids and http://www.ncbi.nlm.nih.gov/pubmed/19073785
329. Canessa CM, Schild L, Buell G, et al. Amiloride-sensitive epithelial Na
Peptides
their 2003;24(12):1971.
synthetic analogues; inhibitors of the synthesis and action of adrenal corti- 281. Murray
channel is madeP. up Brain natriuretic
of three peptide
homologous therapyNature
subunits. to prevent acute1994;367:463.
(London). kidney injury
http://www.ncbi.nlm.nih.gov/pubmed/15127950
cal hormones. In: Goodman Gilman A, Rall TW, Nief AS, et al, eds. The pharma- a ter cardiac
330. Canessasurgery. Am J KidneyJD,
CM, Horisberger Dis.Schild
2008;51(1):5–9.
L, et al. Expression cloning of the
261. Caruso-Neves
cological C, VivesNew
basis of therapeutics. D, Dantas C, et al. Ouabain-insensitive
York: Pergamon; 1990:1431. Na+-ATPase http://www.ncbi.nlm.nih.gov/pubmed/18155527
epithelial sodium channel. Kidney Int. 1995;48:950.
o proximal
301. Palevsky tubules
P, Szerlipis an
HM,e Cox
ector M. or urodilatin
Steroid and mechanisms
hormones: atrial natriuretic of cellpeptide.
signal- 282. Marver
331. Bryan PM, D. Xu X, DickeyofDM,
Regulation et al. Renal by
Na,K-ATPase hyporesponsiveness
aldosterone. Semin to atrial na-
Nephrol.
Biochim
ing. Biophys Acta.
In: Goldfarb 2004;1660(1–2):93.
S, Ziyadeh FN, eds. Contemporary issues in nephrology: hormones, triuretic peptide in congestive heart ailure results rom reduced atrial natriuretic
1992;l12:56.
http://www.ncbi.nlm.nih.gov/pubmed/14757224
autacoids, and the kidney, vol. 23. New York: Churchill Livingstone; 1991. peptide
332. receptor concentrations.
Gonzalez-Campoy JM, RomeroAm J Physiol
JC, Knox Renal FG.Physiol.
Escape 2007;292(5):F1636–
from the sodium-
262. Farman
302. Ziedel ML. Medullary
N. Steroid collecting
receptors: duct sodium
distribution alongtransport.
the nephron. Am J Physiol.
Semin 1644. effects of mineralocorticoids: role of ANF and intrarenal hormone sys-
retaining
1993;265:F159.
Nephrol. 1992;12:12. 283. Kidney
tems. Chen HH, Huntley BK, Schirger JA, et al. Maximizing the renal cyclic
Int. 1989;35:767.
263. Wong
303. Metzler CH, Ramsay
S, Brennan FE, YDJ.
oung Physiological
MJ, Fuller PJ,doses o atrial
Cole TJ. peptide
A direct effect of inhibit angio-
aldosterone 3’-5’-guanosine
333. August JL, monophosphate
Nelson DH, Thorn system with typeofVnormal
GW. Response phosphodiesterase
subjects to large in-
tensin
on II-stimulated
endothelin-1 gene aldosterone
expression insecretion. Am Physiol.
vivo. Endocrinology. 1989;256:R1155.
2007;148(4):1511–1517. hibition and
amounts exogenous Jnatriuretic
of aldosterone. Clin Invest.peptide: a novel strategy to improve renal
1958;37:1549.
264. Funder
304. Oliveira-Souza M, Mello-Aires
JW. Corticosteroid M. E and
receptors ect orenal
arginine vasopressin anddehy-
11b-hydroxysteroid ANP unction
334. Haas inJA,
experimental overt heartforailure.
Knox FG. Mechanisms escapeJ from
Am Soc Nephrol. 2006;17(10):
the salt-retaining effects of
on intracellular
drogenase pHSemin
activity. and cytosolic
Nephrol. ree [Ca ] regulation in MDCK cells. Kidney Int.
1990;10:311.2+
2742–2747.
mineralocorticoids: role of deep nephrons. Semin Nephrol. 1990;10:380.
2001;60(5):1800.
305. Farman N, Rafestin-Oblin ME. Multiple aspects of mineralocorticoid selec- 284. Stokes
335. Chen LG, JB. Wang ZR, Wan
Physiologic CM, ettoal.the
resistance Encapsulated transgene cells
action of aldosterone. attenu-
Kidney Int.
265. Am
tivity. KohJ GY , Klug
Physiol MG,Physiol.
Renal Field LJ. Atrial natriuretic actor and transgenic mice.
2001;280(2):F181. ate hypertension, cardiac hypertrophy and enhance renal unction in Goldblatt
2000;57:1319.
Hypertension.
306. Funder JW, 1993;22:634.
Pearce PT, Smith R, et al. Mineralocorticoid action: target tissue hypertensive
336. Field MJ, rats. J Gene Med.
Giebisch GH. 2004;6(7):786.
Hormonal control of renal potassium excretion.
http://www.ncbi.nlm.nih.gov/pubmed/8406670
speci city is enzyme, not receptor, mediated. Science. 1988;242:583. http://www.ncbi.nlm.nih.gov/pubmed/15241786
Kidney Int. 1990;27:379.
266. Kenouch
307. Field LJ. Transgenic
S, Alfaidy N, mice in cardiovascular
Bonvalet physiology.ofAnnu
JP, et al. Expression Rev Physiol.
11 beta-OHSD 285. Field
337. ValentinMJ,JPStanton
, Humphreys MH. Urodilatin:
BA, Giebisch a paracrine
GH. Differential acuterenal natriuretic
effects pep-

You might also like