You are on page 1of 14

J Am Soc Nephrol 11: 764 –777, 2000

HOMER W. SMITH AWARD LECTURE

Aquaporin Water Channels in Kidney


PETER AGRE
Departments of Biological Chemistry and Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland.

As a first year medical student, I attended nearly 200 lectures, ization of the molecular entities responsible for the function of
but I have clear recollections of only two. On the first day that biologic membranes has been a major goal of physiologists
autumn, Albert Lehninger canceled his planned lecture and during the past two decades. Using biochemical purification
spoke to us of a recent paper that he said would revolutionize and functional reconstitution techniques, multiple transporters
biomedical research—the discovery of reverse transcriptase. have been identified and characterized. In addition, expression
The following spring, James L. Gamble, Jr. lectured to us about cloning has led to the discovery of other transporters. Despite
renal physiology and the important contributions of a man this, the molecular identities of water transporters remained
named Homer Smith who had conducted research in a shed on unknown until a series of fortuitous events occurred in our
Mount Desert Island in Maine. At the time, the idea of a laboratory several years ago.
researcher at an idyllic site studying marine and tidal organ- The plasma membranes of all cells are known to exhibit
isms struck me as particularly appealing. Later that summer, I finite water permeability. Nevertheless, simple diffusion does
set out on a bicycle tour of Nova Scotia by way of Bar Harbor not account for all water movement through biologic mem-
where Dr. Gamble and his family had warmly invited me to branes, and specialized water channels were predicted to ex-
stop at their summer home. In addition to some delicious meals plain the high water permeability of certain tissues (reviewed
and a visit to multiple well-known sights in the area, Dr. in reference (2). In particular, the water permeability of red cell
Gamble drove me past the site of Homer Smith’s laboratory membranes is much higher than that observed for other cell
where 20 years later I was to spend time as a visiting scientist. types and artificial lipid bilayers. The activation energy of this
In many ways, the words of Smith were to have particular process (Ea ⬍ 5 kcal/mol) is equivalent to the diffusion of
meaning for me. water in solution (reviewed in reference (3), indicating that
water moves through aqueous pathways across these mem-
The history of renal physiology has erred, more often
branes. The observation of that HgCl2 and certain organomer-
than not, by attempts at oversimplification. The prob-
curials reversibly inhibit the water permeability predicted that
lems of water and salt excretion appear to be ex-
water channels are proteins with accessible sulfhydryl groups
tremely complex, and especially liable to this danger.
(reviewed in reference (4).
Homer W. Smith, 1937 Water permeability has been characterized in each segment
The Physiology of the Kidney (1) of the nephron (reviewed in reference (5). Renal proximal
tubules and descending thin limbs of Henle’s loop are known
Water is the major component of all living cells, so the
to have constitutively high water permeability allowing for the
ability to absorb and release water must be considered a fun-
reabsorption of the majority of the water in glomerular filtrate.
damental property of life. As recognized by Homer Smith, the
In contrast, the ascending thin limbs and thick limbs are
kidney is the champion organ with regard to fluid transport.
relatively impermeable to water but are highly permeable to
The epithelia lining the tubules in kidney are covered by
ions and other solutes. Renal collecting ducts are extremely
plasma membranes, which control the solute composition of
important in clinical water balance, since collecting duct water
the enclosed compartments by regulating the entry of ions,
permeability is regulated by vasopressin (antidiuretic hor-
small uncharged solutes, and water into and out of cells.
mone). Early studies of toad urinary bladder provided a model
Movement of these fluids from lumen to interstitium or in the
of vasopressin-regulated water permeability; stimulation of the
reverse direction is clearly related to the high degree of cellular
basolateral membrane of this epithelium with antidiuretic hor-
polarity in epithelial tissues whose cells have distinct apical
mone induces a redistribution of intracellular particles to the
and basolateral membranes. The identification and character-
apical membrane and increases the cellular water permeability
(6,7). Nevertheless, attempts to identify the membrane water
Received November 29, 1999. Accepted December 10, 1999. channels were unsuccessful due to the ubiquity of water, the
This lecture was updated and modified from an earlier review with permission presence of free sulfhydryls in many proteins, and relatively
(119). high diffusional permeability of most membrane bilayers (re-
Correspondence to Dr. Peter Agre, Department of Biological Chemistry, Johns viewed in reference (8).
Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205-
2185. Phone: 410-955-7049; Fax: 410-955-3149; E-mail: pagre@jhmi.edu
1046-6673/1104-0764 Aquaporin-1
Journal of the American Society of Nephrology A serendipitous observation accompanied by the wisdom of
Copyright © 2000 by the American Society of Nephrology a former mentor led to the recognition of the first molecular
J Am Soc Nephrol 11: 764 –777, 2000 Homer W. Smith Award Lecture 765

water channel. During the biochemical purification of the


32-kD core polypeptide of the red cell Rh blood group antigen,
a 28-kD polypeptide was noted and assumed to represent a
proteolytic fragment of Rh (9). Antiserum raised in rabbits
reacted exclusively with the 28-kD band, indicating that it is
unrelated to Rh (10). The 28-kD protein contained hydropho-
bic amino acids and exists in two forms: a nonglycosylated
28-kD polypeptide and a 40- to 60-kD N-glycosylated polypep-
tide. The protein proved easy to purify due to its limited
solubility in the detergent N-lauroylsarcosine. Biochemical
studies showed that the protein is a tetramer with intracellular
N- and C-termini and yielded the N-terminal amino acid se-
quence (11), which permitted cDNA cloning from an erythroid
library (12). The deduced amino acid sequence of the 28-kD
polypeptide was found to be related to a functionally undefined Figure 1. Water permeability of aquaporin-1 (AQP1) expressed in
Xenopus oocytes. When transferred to hypo-osmolar buffer for 2 min,
family of putative membrane channel proteins including major
control water-injected oocytes exhibit negligible swelling (left). Un-
intrinsic protein of lens (13). At the same time, radiation
der the same conditions, oocytes previously injected with AQP1
inactivation studies of water permeability by renal vesicles cRNA rapidly swell and explode (right). Reproduced and modified
yielded a target size of 30 kD (14). Abundance of the 28-kD with permission from Annu Rev Biochem (119).
polypeptide in highly water-permeable tissues, red cells, renal
proximal tubules, and descending thin limbs (10) led the late
John C. Parker (1935–1993) at the University of North Caro- similar to water channels in native membranes: reversible
lina at Chapel Hill to predict that it may be the sought-after inhibition by HgCl2 and low activation energy. Permeation by
water channel now known as aquaporin-1 (AQP1). Soon to other small solutes or even protons showed AQP1 to be water-
follow was the identification of multiple related proteins that selective (21). Thus, AQP1 is both necessary and sufficient to
comprise the aquaporin family of proteins and explain mem- explain the well-recognized membrane water permeability of
brane water permeability in diverse tissues and organisms the red cell. AQP1 functions as a constitutively active, water-
(8,15). selective pore. Although forskolin was once reported to induce
a cation current in AQP1-expressing oocytes (22), multiple
Measurement of Membrane Water Permeability other scientific groups have failed to reproduce this effect (23).
The measurement of water transport across cell membranes Recently, the permeation by CO2 has been evaluated, and rates
posed an experimental challenge. First proposed by Eric of pH change are higher in oocytes expressing AQP1 (24).
Windhager of Cornell Medical School, the Xenopus oocyte Because permeation of lipid bilayers by CO2, NO, and O2 may
expression system has proven to be a valuable method for be high, the potential physiologic relevance of AQP1 perme-
functional analysis of candidate water channel RNAs, since the ation by gases deserves further study (25), and the possibility
oocytes normally exhibit low membrane water permeability of yet undiscovered transport functions cannot be excluded.
(16,17). Fortunately, our colleague Bill Guggino at Johns Hop-
kins was very experienced in oocyte expression. When injected AQP1 Structure
with cRNA corresponding to the 28-kD polypeptide (18), the The late 1990s have proven to be a time of great progress in
oocytes became remarkably permeable to water (Pf approxi- the elucidation of membrane protein structures. Hydropathy
mately 200 ⫻ 10⫺4 cm/s) and exploded in hypotonic buffer. analysis of the deduced amino acid sequence of AQP1 suggests
Water-injected control oocytes swelled minimally (Figure 1). that the protein spans the lipid bilayer (12). Similar to the
Moreover, the AQP1 oocytes behaved like cells containing homolog major intrinsic protein from lens (now referred to as
native water channels: low activation energy, reversible inhi- AQP0), the deduced amino acid sequence of AQP1 is com-
bition by HgCl2, and no measurable increase in ion conduc- prised of two internal repeats (Figure 2), and each half contains
tance (18). The direction of water flow through AQP1 is the signature motif Asn-Pro-Ala (NPA) (26). Hydropathy anal-
determined by the orientation of the osmotic gradient (19). ysis predicts six bilayer-spanning domains, although connect-
The ability to isolate milligrams of AQP1 from human red ing loops B and E are also somewhat hydrophobic. Location of
cells permitted direct biophysical studies of the protein. Highly loop C between the two halves of the molecule is therefore
purified AQP1 protein from human red cells reconstituted with critical to the topology. In different recombinants, BamHI
pure phospholipid yielded proteoliposomes of approximately restriction sites were introduced into the cDNA for insertion of
100 nm in diameter (20). The change in volume of AQP1 a DNA encoding a 29-residue epitope. Expression in oocytes
proteoliposomes was compared to liposomes containing no confirmed that several of the recombinants remained func-
reconstituted protein by measuring quenching of internal car- tional, and using antibody binding and vectorial proteolysis,
boxyfluorescein. These measurements yielded the unit water loop C was shown to reside at the extracellular surface of the
permeability (Pf approximately 3 ⫻ 109 water molecules/ oocytes. Thus the N- and C-terminal halves of the molecule
subunit/s). The proteoliposomes exhibited water permeability exhibit a uniquely obverse symmetry with the two halves
766 Journal of the American Society of Nephrology J Am Soc Nephrol 11: 764 –777, 2000

Figure 2. Membrane topology of AQP1 subunit. The Asn-Pro-Ala


signature motifs are indicated (NPA). Polyhedrons enclose sequence-
related repeats in obverse orientation. Sites of N-linked glycan and
surface polymorphism are represented. Reproduced and modified with
permission from Annu Rev Biochem (119).

oriented at 180 degrees to each other (27) (Figure 2). Other


studies showed that Cys-189 is the site of mercurial inhibition Figure 3. Hourglass model for AQP1 membrane topology. (Top) Each
AQP1 subunit contains six bilayer-spanning domains comprised of
(28), and loops B and E were found to be functionally essential
two obversely symmetrical structures (TM1–3 ⫽ hemipore-1 and
for water permeability. Together, these studies led to the “hour- TM4 – 6 ⫽ hemipore-2). (Bottom) When NPA motifs in loops B and
glass” model (29), in which loops B and E overlap midway E are juxtaposed, they form a single aqueous channel spanning the
between the leaflets of the bilayer, creating a constitutively bilayer (the “hourglass”) flanked by the mercury-sensitive residue
open, narrow aqueous pathway (Figure 3). Although not un- (C189). Reproduced and modified with permission from J Biol Chem
derstood in detail, multiple studies indicate that the AQP1 is a (29).
tetramer comprised of functionally independent aqueous pores
(29,30). in a biologically active state (34). Microscopic examination of
AQP1 protein from human red cell membranes has provided negatively stained membranes showed that one side of the
increasingly detailed molecular insight into the protein’s struc- tetramer contains subunits widely separated around a central
ture. Shadowed freeze-fracture electron microscopic analyses stain-filled depression (the extracellular surface) or closely
of reconstituted AQP1 confirmed the tetrameric assembly of surrounding a central area (cytoplasmic surface) (35). Cryo-
AQP1 (21,31). Attenuated total reflection-FTIR (Fourier trans- electron microscopic studies have revealed the presence of
form infrared spectroscopy) of highly purified red cell AQP1 multiple bilayer-spanning domains (36 –38), and atomic force
reconstituted into membrane crystals has been directly com- microscopy further defined the orientation of the right-handed
pared to proteins with known structures, bacteriorhodopsin, helix bundle and extramembrane dimensions of AQP1 (39).
and the porin OmpF. These studies demonstrated that AQP1 Electron crystallography of cryopreserved specimens using a
lacks beta structure and contains multiple alpha helices tilted at liquid helium-cooled stage and tilts of up to 60 degrees re-
21 to 27 degrees (32). Using fluorescently labeled antibody vealed the presence of AQP1 tetramers with individual sub-
specific for the extracellular domain of loop A (Co antigen), units containing six bilayer-spanning alpha helices viewed at a
the lateral mobility of AQP1 in red cell membranes was shown resolution of 6 Å (40 – 42). These studies have revealed that the
to be regulated by passive steric hindrance from the membrane AQP1 subunit has a structure similar to the proposed hourglass
skeleton (33). (Figure 4 and cover photo). The next challenge is to merge
The structure of the AQP1 protein is being solved at increas- structural understandings derived from molecules with unam-
ing levels of resolution. Reconstitution of highly purified red biguous landmarks.
cell AQP1 into membranes under controlled conditions yields
membrane crystals with highly uniform lattices with p4221 Tissue Distribution of AQP1
symmetry. These resealed vesicles retain full water permeabil- In the initial report, AQP1 was noted to be abundant in the
ity, providing the opportunity to define the structure of AQP1 apical and basolateral membranes of renal proximal tubules
J Am Soc Nephrol 11: 764 –777, 2000 Homer W. Smith Award Lecture 767

protein (56). Due to their linkage to human chromosome 7p14,


it was recognized that the Colton blood group antigen (Co)
may reside on the AQP1 protein. Co was shown to be an
Ala/Val polymorphism at the 45th residue located on the first
extracellular loop of red cell AQP1 (57) (Figure 2). The Co
null blood phenotype is exceedingly rare, and our laboratory
has studied Co null probands from three unrelated kindreds.
Each was born in a remote rural area and is believed to have
been the offspring of parental consanguinity. All three were
found to be homozygous for naturally occurring disruptions in
the AQP1 gene. Nonsense mutations were identified in two
probands: deletion of exon 1 and frameshift in exon 1. The
third proband is homozygous for a missense mutation: replace-
Figure 4. Stereoscopic view of AQP1 at 6 Å resolution established by
ment of proline by a leucine at the end of the first transmem-
cryoelectron microscopy. Six tilted, bilayer-spanning alpha helices
(numbered) in a single subunit surround the internal hourglass formed brane domain that produces an unstable polypeptide (56).
from loop B (green) and loop E (orange). Reproduced and modified These Co null individuals are all women who developed an-
with permission from Nature (40). ti-Co during pregnancy. Although the exceedingly rare blood
group phenotype makes them impossible to match for blood
transfusion, it was surprising that none of them exhibited any
and descending thin limbs of Henle’s loop (10). This was other obvious clinical phenotype, and it is uncertain whether
subsequently confirmed with polyclonal rabbit antiserum (43) this is explained by naturally occurring backup systems or the
and with affinity-purified Ig specific for the N- and C-terminal existence of compensating mutations. Evaluation of these Co
domains of AQP1 (44,45) (Figures 5 and 6). In all studies, null humans has been undertaken, and preliminary analyses
AQP1 is constitutively present in the apical plasma membranes revealed that they manifest a major concentration defect in the
(including the brush borders) and in basolateral membranes as proximal tubules but are not polyuric. When fluid-deprived for
well. A similar pattern of expression is found in the descending up to 24 h, the Co null individuals concentrate their urine to
thin limbs of Henle’s loop (Figure 7). AQP1 at the lumenal and less than half the normal level, and administration of exoge-
ablumenal surfaces of the tubule provides the transcellular nous vasopressin produced no additional effect (L. King and P.
pathway for water following small osmotic gradients at the Agre, unpublished observations).
apical and basolateral surfaces of the cell. Quantification of Mice homozygous for targeted AQP1 gene disruption have
AQP1 at these sites by dilutional immunoblotting and by been raised (58). The recent development of an Aqp1 gene
enzyme-linked immunosorbent assay verified that AQP1 alone knockout has revealed that renal water reabsorption in mice is
could serve as the conduit for the huge volumes of water very dependent on AQP1 protein, since the Aqp1 null animals
reabsorbed in the proximal nephron (44,46). Immunohisto- became hyperosmolar after fluid restriction. Detailed classical
chemical studies have also demonstrated AQP1 in renal vasa physiologic evaluation of the isolated proximal tubules from
recta (47). the Aqp1 null mice were undertaken to establish that trans-
AQP1 is also abundant in capillary endothelia in other membrane water permeability was reduced by 80%, and led to
organs. In lung, AQP1 is present in peribronchiolar capillary the observation of a compensatory reduction in glomerular
endothelium, where expression is induced by glucocorticoids filtration (59,60).
(48 –50) acting through the classic glucocorticoid response
elements in the proximal gene promoter (51). In brain, AQP1 Other Renal Aquaporins
has been defined in the apical membrane of choroid plexus The first functional definition of one member of a protein
epithelia, the site of cerebrospinal fluid secretion. AQP1 is also family often predicts the functions of the other members, and this
present at multiple locations in the eye: nonpigmented ciliary has certainly been the case for the aquaporins. Homology cloning
epithelium, lens epithelium, and corneal endothelium (48,52). has been undertaken by multiple laboratories, and 10 mammalian
Although not present in intestinal mucosa, AQP1 is rich in homologs have been reported, while preliminary evidence for
cholangiocytes, where it appears to be regulated by secretin additional homologs is emerging. Homology cloning of aquapor-
(53). Developmental expression of AQP1 has been found to be ins is most frequently undertaken using polymerase chain ampli-
complex in the rat model, and at least three patterns are fication with degenerate oligonucleotide primers corresponding to
recognized: (1) transient expression before birth (periosteum, the most highly conserved domains, the NPA sequences within
cardiac endothelium); (2) permanent expression beginning at loops B and E (61). All members of the family are believed to be
the time of birth (cornea, lung, kidney); (3) constitutive life- water channels, because all contain structural motifs similar to
long expression (choroid plexus) (50,54,55). AQP1, but each has features apparently needed for functional and
regulatory differences. The complete sequencing of the human
AQP1 Deficiency genome will almost certainly identify new aquaporin homologs.
AQP1 protein is not essential for survival in humans or Although an oversimplification, the 10 known mammalian aqua-
mice. Humans have been identified who totally lack the AQP1 porins have tentatively been broken into two subgroups: water-
768 Journal of the American Society of Nephrology J Am Soc Nephrol 11: 764 –777, 2000

Figure 5. Light and electron microscopic studies of AQP1 in rat proximal tubules. (a) Immunohistochemical analysis of S3 segment stained
with Meier reagent exhibits strong staining over apical brush border and basolateral membranes (arrows). (b) Tangentially sectioned S3 segment
stained with peroxidase-conjugated goat-anti-rabbit IgG shows no reaction over collecting duct (C). Bars, 20 ␮m. (c) Immunogold electron
microscopy of S3 segments in longitudinal section. (d) Cross section. Magnification, ⫻30,000. Reproduced and modified with permission from
J Cell Biol (44).

selective channels (Orthodox aquaporins) and channels permeated (18). Using the homology cloning approach, a cDNA was
by water, glycerol, and other small molecules (Aquaglyceropor- reported to encode a renal collecting duct water channel, AQP2
ins) (Figure 8). The Human Genome Organization has established (62). The combined efforts of multiple groups of investigators
an Aquaporin Nomenclature System for designation of novel have established major clinical significance for AQP2 (re-
family members (15), and updates are available on the Internet viewed in reference (63). AQP2 is expressed predominantly in
(http://www.gene.ucl.ac.uk/nomenclature). the principal cells of the renal collecting duct (62,64,65).
Regulation of AQP2 appears to be complex. The intracellular
AQP2, the Vasopressin-Regulated Water Channel trafficking of AQP2 is regulated by vasopressin through short-
Water permeability of renal collecting ducts is regulated by term exocytosis to the plasma membrane, whereas long-term
vasopressin. The failure to demonstrate AQP1 in collecting biosynthetic mechanisms are activated in response to chronic
ducts by immunohistochemistry (10) led to the prediction that thirsting.
other members of the aquaporin family must exist at this site Vasopressin has been known for more than two decades to
J Am Soc Nephrol 11: 764 –777, 2000 Homer W. Smith Award Lecture 769

pression of AQP2 protein has been found during the postan-


tidiuretic state known as “escape from vasopressin” (74).
Much clinical significance has been ascribed to AQP2, and
this protein may be involved in most, if not all, imbalances of
water metabolism. For example, impaired renal water reab-
sorption is associated with reduced levels of AQP2. Nephro-
genic diabetes insipidus (NDI) is a disorder that occurs when
vasopressin levels are elevated, but the kidney fails to respond
to the hormone. Mutations in genes encoding renal vasopressin
V2 receptors have been demonstrated in many patients with the
X-linked disorder (reviewed in reference (75). A family with
recessively inherited NDI and normal V2 receptors was found
to have mutations in the AQP2 gene causing loss of function
(76). Multiple other AQP2 mutations in aqueous pore domains
have been found in other patients with recessively inherited
NDI (77,78). One family with dominantly inherited NDI was
recently identified with a mutation in the C terminus of AQP2
(79). The products of the mutant and the normal AQP2 allele
have been shown to oligomerize, thereby restricting membrane
trafficking of both polypeptides. Individuals with NDI result-
ing from the dominant mutations experience a less severe form
of polyuria (Daniel Bichet, personal communication).
Secondary decreases in AQP2 expression probably occur
with relative frequency in medical clinics. Lithium is widely
prescribed for treatment of bipolar disorder, but polyuria is a
Figure 6. Diagram representing transcellular water permeation of potentially problematic side effect. While lithium-induced
proximal tubule cell via AQP1. polyuria may be managed by increasing fluid intake, this can
cause difficulties in elderly individuals or in patients deprived
of oral intake, such as in preparation for surgery. Lithium has
cause the redistribution of intracellular vesicles to the apical been found to cause a striking reduction (90%) in AQP2
surface of principal cells, a process referred to as the “mem- expression in rats (80). Other forms of polyuria have been also
brane shuttle mechanism” (66). Immunolocalization of AQP2 evaluated, and reduced AQP2 expression was also documented
in sections of rat kidney after treatment with vasopressin sup- after release of urinary obstruction, such as after prostate
ports the shuttle hypothesis (65,67,68). A much-cited study of surgery (81), in chronic hypokalemia-induced polyuria (82),
isolated rat renal collecting ducts showed that in the absence of and is suspected in nocturnal enuresis (83).
vasopressin, water permeability was low and AQP2 resided Secondary increases in AQP2 expression may also be com-
predominantly in intracellular vesicles (69). Administration of mon in clinical medicine and may cause problems with exces-
vasopressin caused the water permeability to rise fivefold and sive renal water retention. Patients with congestive heart fail-
was accompanied by a redistribution of AQP2 to the apical ure often succumb to refractory pulmonary edema, and
membrane. After removal of vasopressin, the water permeabil- increased expression of AQP2 has been documented in rat
ity declined and surface AQP2 was reinternalized (Figure 9). models of incompletely compensated congestive heart failure
The mechanism of vasopressin action is through a surface V2 (84,85). Increased expression of AQP2 may also explain fluid
receptor coupled to activation of adenylyl cyclase, inducing retention known to complicate pregnancy (86), cirrhosis, and
phosphorylation of the C terminus of the AQP2 protein (Figure syndrome of inappropriate antidiuretic hormone (87). AQP2
10). Cytoplasmic vesicles containing AQP2 are then targeted may be an effective target for pharmacologic intervention in
to the cell surface via the vesicle-targeting proteins syntaxin-4 some of these disorders.
and synaptobrevin-2. The molecular details of this pathway are
being evaluated and appear to be complex (70). The stoichi- Multifunctional AQP3
ometry of subunit phosphorylation and the possible presence of Using the homology cloning approach, three scientific
other transporters in the vesicle are being established (reviewed groups isolated cDNAs encoding AQP3, a member of the
in reference (63). The involvement of a heterotrimeric member aquaporin family with distinctive biophysical functions (88 –
of the Gi protein family is known to occur during membrane 90). Previously cloned aquaporins were only permeable to
trafficking in renal epithelial cells (71). The long-term mech- water, but AQP3 was noted to be genetically closer to the
anisms for regulation of AQP2 biosynthesis and removal are known Escherichia coli glycerol transport protein GlpF (Fig-
also being evaluated. Identification of a cAMP regulatory ure 8), and AQP3 is permeated by glycerol. AQP3 was also
element in the 5⬘ flanking DNA of AQP2 suggests that tran- reported to be slightly permeable to urea, but the biologic
scriptional regulation may be occurring (72,73). Reduced ex- significance of this is uncertain, since unrelated proteins spe-
770 Journal of the American Society of Nephrology J Am Soc Nephrol 11: 764 –777, 2000

Figure 7. Light and electron microscopic studies of AQP1 in rat descending thin limbs of Henle’s loop. (a) Immunohistochemical analysis of
outer medulla reveals labeling of apical and basolateral membranes of descending thin limbs (D) but not of ascending thin limbs (T) or
collecting ducts (C). (b) Higher magnification. Bars, 20 ␮m. (c) Immunogold electron microscopy of descending thin limb from short-looped
nephrons shows staining of cytoplasmic leaflets of apical membranes including microvilli (arrow) and basolateral membranes (BM). (d)
Long-looped nephron. Magnification, ⫻45,000. Reproduced and modified with permission from J Cell Biol (44).

cialized for urea transport have been identified (91). The struc- investigators (92), however, the existence of a single pore that
tural explanation for how AQP3 may permit transport of water permits the flow of water or glycerol has been reported by
and glycerol remains uncertain, and primary sequence differ- others (93). Although the significance is unclear, AQP3 ap-
ences in aquaglyceroporins include a motif (GLYY) in loop C pears to be gated shut at acid pH (94).
and an aspartate residue following the second NPA motif No humans have yet been identified with AQP3 deficiency,
(NPARD) rather than the motif (NPARS) found in orthodox and the clinical importance of this molecule remains to be
aquaporins. The possibility that the AQP3 has separate water- established. Because AQP3 is expressed at the basolateral
and glycerol-transporting domains has been proposed by some membranes of principal cells in the collecting duct (Figure 10),
J Am Soc Nephrol 11: 764 –777, 2000 Homer W. Smith Award Lecture 771

Figure 8. Phylogenetic comparisons of aquaporins. Mammalian aqua-


porins and Escherichia coli homologs. Water-selective “orthodox
aquaporins” and AqpZ (gray) and “aquaglyceroporins” and GlpF
(white). Reproduced and modified with permission from Annu Rev Figure 9. Immunogold electron microscopy of AQP2 in rat collecting
Biochem (119). duct principal cells before (top) or after (bottom) exposure to vaso-
pressin. Magnification, ⫻44,000. Reproduced and modified with per-
mission from Proc Natl Acad Sci USA (69).
a role in renal water reabsorption is expected (95–97). AQP3
appears to be regulated at a biosynthetic level similar to AQP2.
In addition to kidney, AQP3 is also expressed in lower levels AQP4 may facilitate rapid water flux needed to restore osmotic
at multiple other sites, including airways (95). AQP3 is strik- equilibrium. Of particular interest is the unexplained observa-
ingly abundant in nasopharyngeal epithelium, where it is sus- tion that AQP4 is reduced in fast twitch fibers in the MDX
pected to play a role in generation of mucosal secretions and mouse model of Duchennes muscular dystrophy (105).
allergic rhinitis (50,98). Based on their absence in tissues from Aqp4 null mice, it has
been proposed that AQP4 is the molecular basis of square
Brain AQP4 arrays within astroglial membranes (106). This has recently
Although primarily expressed in brain, AQP4 is also present been confirmed. Direct anti-AQP4 immunolabeling of freeze-
in the basolateral membranes of collecting duct principal cells fracture replicas from brain and spinal cord directly demon-
in the inner medulla (99). The protein presumably provides the strated AQP4 in the majority of square arrays (107). It has also
exit pore from these cells during vasopressin-dependent water recently been reported that protein kinase C activated by phor-
reabsorption. Disruption of the mouse Aqp4 gene has been bol diesters produces a 90% reduction in water permeability by
reported to result in a defect in renal concentration (100). AQP4 (108). Although this suggests that AQP4 may be ac-
AQP4 is the predominant aquaporin in brain (101,102). tively regulated, important control experiments are still needed.
Residing at the perivascular margin of astroglial cells, the Nevertheless, AQP4 may reversibly open and close the blood-
protein may provide an exit for excess brain water in severe brain barrier to the movement of water, a process that may be
clinical problems such as cerebral edema (103). AQP4 is of great significance in various clinical problems of brain
abundant in glial lamellae surrounding vasopressin secretory ischemia.
neurons (103) and in ependymal cells lining the cerebrospinal
fluid-filled cavities (95). In addition, AQP4 is expressed in AQP6, an Intracellular Ion Channel Permeated by
retina and optic nerve, where the protein resides in Müller cell Anions
endfeet adjacent to the vitreous body and vascular endothelium Another sequence-related protein initially referred to as
(104). WCH-3 (now designated AQP6) was identified in rat kidney
AQP4 is present in fast twitch skeletal muscle in rat (105). by homology cloning several years ago (109), but the protein
Fast twitch fibers accumulate high concentrations of lactate, so was not functionally defined. A highly related protein initially
772 Journal of the American Society of Nephrology J Am Soc Nephrol 11: 764 –777, 2000

Figure 11. Immunogold electron microscopy of AQP6 and H⫹-


ATPase in alpha-intercalated cell of rat collecting duct. Anti-AQP6
(small particles, arrowheads) is found over intracellular vesicles,
whereas anti-H⫹-ATPase (large particles, arrows) is also found at the
apical plasma membrane. Magnification, ⫻83,000. Reproduced and
modified with permission from Nature (112).
Figure 10. Diagram representing vasopressin-regulated transcellular
water permeation of apical membrane of collecting duct principal cell
via AQP2 and constitutive water permeation of basolateral membrane
via AQP3. Ion replacement techniques showed that the AQP6 currents
were anion-selective. Moreover, AQP6 contains a unique pos-
itively charged residue lysine that is predicted to reside at the
referred to as hKID was identified in human kidney, and the cytoplasmic mouth of the pore (Figure 13). Replacement of
chromosomal locus at human 12q13 coincided with that of this residue with a negative charge K72E produced a loss of ion
several other aquaporins. AQP6 was also reported to have selectivity.
extremely low inherent water permeability and was further Together these studies have documented highly unusual
inhibited by HgCl2 (110). The apparent lack of functional behavior for an aquaporin and suggest that some members of
information reduced the scientific interest in this protein. this protein family may participate in acid secretion or other
Recently, there has been renewed interest in AQP6. This processes in addition to simple transmembrane water move-
aquaporin has been shown to reside in intracellular vesicles at ments. These studies of AQP6 suggest that the functional
three sites in rat kidney: within foot processes of glomerular repertoire of aquaporins may be much broader than previously
podocytes, in subapical vesicles of proximal tubules, and in thought. The intracellular vesicles in alpha-intercalated cells
alpha-intercalated cells of collecting duct (111). Exploring the are believed to permit renal acid secretion by generating HCl.
hypothesis that AQP6 may have a low basal activity which Much work has firmly established the role of the H⫹-ATPase
may be activated, functional studies of AQP6 were recently (reviewed in references (113) and (114). To maintain electro-
undertaken in Xenopus laevis oocytes using protein kinase A neutrality, a pathway for anion secretion must exist, and the
and protein kinase C to phosphorylate the protein; however, no ClC-5 has been located at this site (115). However, this protein
increase in water permeability was noted. Using the known is known to become inactive at low pH (116). Thus, the role of
aquaporin inhibitor HgCl2, AQP6 unexpectedly became acti- AQP6 in alpha-intercalated cells may be to function as a
vated and was accompanied by a large electrical current (112). chloride channel during later stages of acid secretion.
AQP6 was observed to colocalize with H⫹-ATPase in alpha-
intercalated cells of the collecting duct (Figure 11), suggesting Perspectives
that low pH may be the natural activator of the protein. Expo- Organs including kidney, lung, eye, and brain have tissues
sure to low pH produced slight increases in water permeability, with complex expression patterns. Some of these tissues have
and the membrane currents were notably activated below pH multiple different aquaporins expressed even within individual
5.5 but were rapidly reduced after return to pH 7.5 (Figure 12). cells. Precise subcellular locations are being elucidated by light
J Am Soc Nephrol 11: 764 –777, 2000 Homer W. Smith Award Lecture 773

Figure 12. Electrophysiologic analyses of Xenopus laevis oocytes expressing AQP6 and water-injected control oocytes. (a) Osmotic water
permeability at pH 4 and 7.5. (b) Representative currents of AQP6 or water-injected oocytes at indicated pH values. (c) Representative
current-voltage plots of AQP6 oocytes at pH 7.5 (E), after 1 min at pH 4.0 (F), or 1 min after return to pH 7.5 (Œ). (Inset) Continuous current
measurement. (d) Conductances measured at indicated pH values. Reproduced and modified with permission from Nature (112).
774 Journal of the American Society of Nephrology J Am Soc Nephrol 11: 764 –777, 2000

regulation may involve members of this protein family. Plant


geneticists are already utilizing aquaporin genes in attempts to
develop pest- or drought-resistant organisms. Thus, the cellular
and molecular biology of the aquaporins is far from under-
stood.
In summary, there are no small problems. Problems
that appear small are large problems that are not
understood. Nature is a harmonious mechanism
where all parts, including those appearing to play a
secondary role, cooperate in the functional whole.
No one can predict their importance in the future. All
natural arrangements, however capricious they may
seem, have a function.
Santiago Ramón y Cajal, 1897
Advice for a Young Investigator (118)
Figure 13. Hourglass model for AQP6 showing conserved Asn-Pro-
Ala motifs (NPA), functionally important cysteines (C), and selectiv- Acknowledgments
ity-determining lysine (K). Reproduced with permission from Nature
This work was supported by grants from National Institutes of
(112).
Health, the Cystic Fibrosis Foundation, and the Human Frontier
Science Program. Credit for our studies should be shared with our
colleagues and the members of their groups: William B. Guggino,
and electron microscopy, and generally it is found that indi- Søren Nielsen, Mark Knepper, Landon King, Andreas Engel, Yoshi-
vidual aquaporin homologs are expressed at unique intracellu- nori Fujiyoshi, Ole Petter Ottersen, Mark Zeidel, Carel van Os, Peter
lar sites. Because of its major importance in transport, this Deen, Bruce Baum, Nick LaRusso, Erhard Bremer, Suresh Ambud-
kar, Erik Ilsø Christensen, John Rash, Carolyn Bondy, David Anstee,
review has focused on aquaporins in the mammalian kidney,
John Moulds, Bill Bishai, Peter Maloney, Narla Mohandas, Tom
since this organ has been studied most extensively and multiple Pallone, and Thomas Zeuthen. Several junior scientists have been
aquaporins have been documented. It is generally believed that particularly helpful: Thomas Walz, Mahmood Amiry-Moghaddam,
renal aquaporins function together to provide transcellular wa- Erlend Nagelhus, Tae-Hwan Kwon, Akihiro Hazama, Kaoru Mit-
ter flow. In principal cells of collecting duct, AQP2 traffics to suoka, Teruhisa Hirai, Miriam Echevarria, and Bernard Heymann.
the apical membrane where water enters from the lumen (Fig- Former members of my own laboratory contributed much to these
ure 9), while AQP3 and AQP4 reside at the basolateral mem- studies: Andy Asimos, Brad Denker, Barb Smith, Greg Preston, Jin
branes in the outer medulla and in the inner medulla, respec- Sup Jung, Surabhi Raina, Giuseppe Calamita, Mingqi Lu, Mélanie
tively, where they appear to provide the outflow pathways to Bonhivers, Matt Hall, Doug Lee, John Mathai, Ruben Baumgarten,
the interstitium. Because the precise roles of AQP6 and other Sabine Mulders, Chulso Moon, Christy Turtzo, Kushal Bhakta, and
Zev Waldman. Our present laboratory group is continuing the aqua-
uncharacterized aquaporins are still unclear, it is fair to say that
porin story: John Neely, Mario Borgnia, Jennifer Carbrey, Masato
we still do not fully understand the importance of aquaporins in
Yasui, Jason Hoffert, Virginia Leitch, and David Kozono. Generous
kidney or other organs. long-term support and guidance have been provided by John C.
The list of 10 recognized mammalian aquaporins is still Parker, Anita Aperia, Arvid Maunsbach, Joseph Handler, and Dan
growing, and aquaporins are being identified in invertebrates, Lane.
microbes, and plants. In particular, plants are likely to use
aquaporins for numerous physiologic processes involving hy- References
draulics. Accordingly, the list of recognized plant aquaporins is 1. Smith HW: The Physiology of the Kidney, New York, Oxford
already three times the length of the mammalian list. Sequenc- University Press, 1937, p 233
ing the entire genomes from microbes led to recognition of 2. Finkelstein A: Water Movement Through Lipid Bilayers, Pores,
microbial aquaporin homologs, and the much-awaited se- and Plasma Membranes: Theory and Reality, New York, John
quence of the human genome will certainly contain genes for Wiley & Sons, 1987
still unrecognized members of the aquaporin family. It will be 3. Solomon AK: Characterization of biological membranes by
a major challenge to define the ontogeny and the sites of equivalent pores [Abstract]. J Gen Physiol Suppl 51: 335S, 1968
expression. At the same time, the recent functional surprises 4. Macey RI: Transport of water and urea in red blood cells. Am J
Physiol 246: C195–C203, 1984
found with AQP3 and AQP6 indicate that aquaporins may not
5. Knepper MA, Wade JB, Terris J: Renal aquaporins [Review].
simply serve as water transporters. It is also likely that cotrans-
Kidney Int 49: 1712–1717, 1996
port proteins that are genetically unrelated to aquaporins may 6. Bourguet J, Chevalier J, Hugon JS: Alterations in membrane-
also transport water along with the specific solutes (117). The associated particle distribution during antidiuretic challenge in
already recognized primary and secondary roles of aquaporins frog urinary bladder epithelium. Biophys J 16: 627– 639, 1976
in human pathophysiology suggest that other clinical problems 7. Brown D, Orci L: Vasopressin stimulates formation of coated
such as cataracts, glaucoma, brain edema, or body temperature pits in rat kidney collecting ducts. Nature 302: 253–255, 1983
J Am Soc Nephrol 11: 764 –777, 2000 Homer W. Smith Award Lecture 775

8. Agre P, Preston GM, Smith BL: Aquaporin CHIP: The arche- tants by vectorial proteolysis. J Biol Chem 269: 1668 –1673,
typal molecular water channel [Review]. Am J Physiol 265: 1994
F463–F476, 1993 28. Preston GM, Jung JS, Guggino WB: The mercury-sensitive
9. Agre P, Saboori AM, Asimos A: Purification and partial char- residue at cysteine 189 in the CHIP28 water channel. J Biol
acterization of the Mr 30,000 integral membrane protein associ- Chem 268: 17–20, 1993
ated with the erythrocyte Rh(D) antigen. J Biol Chem 262: 29. Jung JS, Preston GM, Smith BL: Molecular structure of the water
17497–17503, 1987 channel through aquaporin CHIP: The hourglass model. J Biol
10. Denker BM, Smith BL, Kuhajda FP: Identification, purification, Chem 269: 14648 –14654, 1994
and partial characterization of a novel Mr 28,000 integral mem- 30. Shi LB, Skach WR, Verkman AS: Functional independence of
brane protein from erythrocytes and renal tubules. J Biol Chem monomeric CHIP28 water channels revealed by expression of
263: 15634 –15642, 1988 wild-type mutant heterodimers. J Biol Chem 269: 10417–10422,
11. Smith BL, Agre P: Erythrocyte Mr 28,000 transmembrane pro- 1994
tein exists as a multisubunit oligomer similar to channel proteins. 31. Verbavatz JM, Brown D, Sabolic I: Tetrameric assembly of
J Biol Chem 266: 6407– 6415, 1991 CHIP28 water channels in liposomes and cell membranes: A
12. Preston GM, Agre P: Isolation of the cDNA for erythrocyte freeze-fracture study. J Cell Biol 123: 605– 618, 1993
integral membrane protein of 28 kilodaltons: Member of an 32. Cabiaux V, Oberg KA, Pancoska P: Secondary structures com-
ancient channel family. Proc Natl Acad Sci USA 88: 11110 – parison of aquaporin-1 and bacteriorhodopsin: A Fourier trans-
11114, 1991 form infrared spectroscopy study of two-dimensional membrane
13. Gorin MB, Yancey SB, Cline J: The major intrinsic protein crystals. Biophys J 73: 406 – 417, 1997
(MIP) of the bovine lens fiber membrane: Characterization and 33. Cho MR, Knowles DW, Smith BL: Membrane dynamics of the
structure based on cDNA cloning. Cell 39: 49 –59, 1984 water transport protein AQP1 in intact human red cells. Biophys
14. van Hoek AN, Hom ML, Luthjens LH: Functional unit of 30 kDa J 76: 1136 –1144, 1999
for proximal tubule water channels as revealed by radiation 34. Walz T, Smith BL, Zeidel ML: Biologically active two-dimen-
inactivation. J Biol Chem 266: 16633–16635, 1991 sional crystals of aquaporin CHIP. J Biol Chem 269: 1583–1586,
15. Agre P: Molecular physiology of water transport. Aquaporin 1994
35. Walz T, Smith BL, Agre P: The three-dimensional structure of
nomenclature workshop: Mammalian aquaporins. Biol Cell 89:
human erythrocyte aquaporin CHIP. EMBO J 13: 2985–2993,
255–257, 1997
1994
16. Fischbarg J, Kuang KY, Vera JC: Glucose transporters serve as
36. Walz T, Typke D, Smith BL: Projection map of aquaporin-1
water channels. Proc Natl Acad Sci USA 87: 3244 –3247, 1990
determined by electron crystallography [Letter]. Nat Struct Biol
17. Zhang RB, Logee KA, Verkman AS: Expression of mRNA
2: 730 –732, 1995
coding for kidney and red cell water channels in Xenopus oo-
37. Jap BK, Li H: Structure of the osmo-regulated H2O channel,
cytes. J Biol Chem 265: 15375–15378, 1990
AQP-CHIP, in projection at 3.5 Å resolution. J Mol Biol 251:
18. Preston GM, Carroll TP, Guggino WB: Appearance of water
413– 420, 1995
channels in Xenopus oocytes expressing red cell CHIP28 protein.
38. Mitra AK, van Hoek AN, Wiener MC: The CHIP28 water
Science 256: 385–387, 1992
channel visualized in ice by electron crystallography [Letter].
19. Meinild A, Klaerke DA, Zeuthen T: Bidirectional water fluxes
Nat Struct Biol 2: 726 –729, 1995
and specificity for small hydrophilic molecules in aquaporins 39. Walz T, Tittmann P, Fuchs KH: Surface topographies at sub-
0 –5. J Biol Chem 273: 32446 –32451, 1998 nanometer-resolution reveal asymmetry and sidedness of aqua-
20. Zeidel ML, Ambudkar SV, Smith BL: Reconstitution of func- porin-1. J Mol Biol 264: 907–918, 1996
tional water channels in liposomes containing purified red cell 40. Walz T, Hirai T, Murata K: The three-dimensional structure of
CHIP28 protein. Biochemistry 31: 7436 –7440, 1992 aquaporin-1. Nature 387: 624 – 627, 1997
21. Zeidel ML, Nielsen S, Smith BL: Ultrastructure, pharmacologic 41. Li H, Lee S, Jap BK: Molecular design of aquaporin-1 water
inhibition, and transport selectivity of aquaporin channel-form- channel as revealed by electron crystallography [Letter]. Nat
ing integral protein in proteoliposomes. Biochemistry 33: 1606 – Struct Biol 4: 263–265, 1997
1615, 1994 42. Cheng A, van Hoek AN, Yeager M: Three-dimensional organi-
22. Yool AJ, Stamer WD, Regan JW: Forskolin stimulation of water zation of a human water channel. Nature 387: 627– 630, 1997
and cation permeability in aquaporin 1 water channels [see 43. Sabolic I, Valenti G, Verbavatz JM: Localization of the CHIP28
Comments]. Science 273: 1216 –1218, 1996 water channel in rat kidney. Am J Physiol 263: C1225–C1233,
23. Agre P, Lee MD, Devidas S: Aquaporins and ion conductance 1992
[Letter; Comment]. Science 275: 1490 –1492, 1997 44. Nielsen S, Smith BL, Christensen EI: CHIP28 water channels are
24. Nakhoul NL, Davis BA, Romero MF: Effect of expressing the localized in constitutively water-permeable segments of the
water channel aquaporin-1 on the CO2 permeability of Xenopus nephron. J Cell Biol 120: 371–383, 1993
oocytes. Am J Physiol 274: C543–C548, 1998 45. Maunsbach AB, Marples D, Chin E: Aquaporin-1 water channel
25. Reuss L: Focus on “Effect of expressing the water channel expression in human kidney [Published erratum appears in J Am
aquaporin-1 on the CO2 permeability of Xenopus oocytes.” Am J Soc Nephrol 8: 358 –360, 1997]. J Am Soc Nephrol 8: 1–14, 1997
Physiol 274: C297–C298, 1998 46. Maeda Y, Smith BL, Agre P: Quantification of aquaporin-CHIP
26. Pao GM, Wu LF, Johnson KD: Evolution of the MIP family of water channel protein in microdissected renal tubules by fluo-
integral membrane transport proteins. Mol Microbiol 5: 33–37, rescence-based ELISA. J Clin Invest 95: 422– 428, 1995
1991 47. Pallone TL, Kishore BK, Nielsen S: Evidence that aquaporin-1
27. Preston GM, Jung JS, Guggino WB: Membrane topology of mediates NaCl-induced water flux across descending vasa recta.
aquaporin CHIP: Analysis of functional epitope-scanning mu- Am J Physiol 272: F587–F596, 1997
776 Journal of the American Society of Nephrology J Am Soc Nephrol 11: 764 –777, 2000

48. Nielsen S, Smith BL, Christensen EI: Distribution of the aqua- of a collecting duct water channel, aquaporin, in hydrated and
porin CHIP in secretory and resorptive epithelia and capillary dehydrated rats. Exp Nephrol 3: 193–201, 1995
endothelia. Proc Natl Acad Sci USA 90: 7275–7279, 1993 69. Nielsen S, Chou CL, Marples D: Vasopressin increases water
49. King LS, Nielsen S, Agre P: Aquaporin-1 water channel protein permeability of kidney collecting duct by inducing translocation
in lung: Ontogeny, steroid-induced expression, and distribution of aquaporin-CD water channels to plasma membrane. Proc Natl
in rat. J Clin Invest 97: 2183–2191, 1996 Acad Sci USA 92: 1013–1017, 1995
50. King LS, Nielsen S, Agre P: Aquaporins in complex tissues. I. 70. Katsura T, Gustafson CE, Ausiello DA: Protein kinase A phos-
Developmental patterns in respiratory and glandular tissues of phorylation is involved in regulated exocytosis of aquaporin-2 in
rat. Am J Physiol 273: C1541–C1548, 1997 transfected LLC-PK1 cells. Am J Physiol 272: F817–F822, 1997
51. Moon C, King LS, Agre P: AQP1 expression in erythroleukemia 71. Valenti G, Procino G, Liebenhoff U: A heterotrimeric G protein
cells: Genetic regulation of glucocorticoid and chemical induc- of the Gi family is required for cAMP-triggered trafficking of
tion. Am J Physiol 273: C1562–C1570, 1997 aquaporin 2 in kidney epithelial cells. J Biol Chem 273: 22627–
52. Hasegawa H, Lian SC, Finkbeiner WE: Extrarenal tissue distri- 22634, 1998
bution of CHIP28 water channels by in situ hybridization and 72. Hozawa S, Holtzman EJ, Ausiello DA: cAMP motifs regulating
antibody staining. Am J Physiol 266: C893–C903, 1994 transcription in the aquaporin 2 gene. Am J Physiol 270: C1695–
53. Roberts SK, Yano M, Ueno Y: Cholangiocytes express the C1702, 1996
aquaporin CHIP and transport water via a channel-mediated 73. Matsumura Y, Uchida S, Rai T: Transcriptional regulation of
mechanism. Proc Natl Acad Sci USA 91: 13009 –13013, 1994 aquaporin-2 water channel gene by cAMP. J Am Soc Nephrol 8:
54. Bondy C, Chin E, Smith BL: Developmental gene expression 861– 867, 1997
and tissue distribution of the CHIP28 water-channel protein. 74. Ecelbarger CA, Nielsen S, Olson BR: Role of renal aquaporins in
Proc Natl Acad Sci USA 90: 4500 – 4504, 1993 escape from vasopressin-induced antidiuresis in rat. J Clin Invest
55. Smith BL, Baumgarten R, Nielsen S: Concurrent expression of 99: 1852–1863, 1997
erythroid and renal aquaporin CHIP and appearance of water 75. Fujiwara TM, Morgan K, Bichet DG: Molecular biology of
channel activity in perinatal rats [see Comments]. J Clin Invest diabetes insipidus [Review]. Annu Rev Med 46: 331–343, 1995
92: 2035–2041, 1993 76. Deen PM, Verdijk MA, Knoers NV: Requirement of human
56. Preston GM, Smith BL, Zeidel ML: Mutations in aquaporin-1 in renal water channel aquaporin-2 for vasopressin-dependent con-
phenotypically normal humans without functional CHIP water centration of urine. Science 264: 92–95, 1994
channels. Science 265: 1585–1587, 1994 77. van Lieburg AF, Verdijk MA, Knoers VV: Patients with auto-
57. Smith BL, Preston GM, Spring FA: Human red cell aquaporin somal nephrogenic diabetes insipidus homozygous for mutations
CHIP. I. Molecular characterization of ABH and Colton blood in the aquaporin 2 water-channel gene. Am J Hum Genet 55:
group antigens. J Clin Invest 94: 1043–1049, 1994 648 – 652, 1994
58. Ma T, Yang B, Gillespie A: Severely impaired urinary concen- 78. Deen PM, Croes H, van Aubel RA: Water channels encoded by
trating ability in transgenic mice lacking aquaporin-1 water chan- mutant aquaporin-2 genes in nephrogenic diabetes insipidus are
nels. J Biol Chem 273: 4296 – 4299, 1998 impaired in their cellular routing. J Clin Invest 95: 2291–2296,
59. Chou CL, Knepper MA, van Hoek AN: Reduced water perme- 1995
ability and altered ultrastructure in thin descending limb of Henle 79. Mulders SM, Bichet DG, Rijss JP: An aquaporin-2 water channel
in aquapoirn-1 null mice. J Clin Invest 103: 491– 496, 1999 mutant which causes autosomal dominant nephrogenic diabetes
60. Schnermann J, Chou CL, Ma T: Defective proximal tubular fluid insipidus is retained in the Golgi complex. J Clin Invest 102:
reabsorption in transgenic aquaporin-1 null mice. Proc Natl Acad 57– 66, 1998
Sci USA 95: 9660 –9664, 1998 80. Marples D, Christensen S, Christensen EI: Lithium-induced
61. Agre P, Mathai JC, Smith BL: Functional analyses of aquaporin downregulation of aquaporin-2 water channel expression in rat
water channel proteins. Methods Enzymol 294: 551–572, 1998 kidney medulla. J Clin Invest 95: 1838 –1845, 1995
62. Fushimi K, Uchida S, Hara Y: Cloning and expression of apical 81. Frøkiær J, Marples D, Knepper MA: Bilateral ureteral obstruc-
membrane water channel of rat kidney collecting tubule. Nature tion downregulates expression of vasopressin-sensitive AQP-2
361: 549 –552, 1993 water channel in rat kidney. Am J Physiol 270: F657–F668, 1996
63. Knepper MA, Inoue T: Regulation of aquaporin-2 water channel 82. Marples D, Frøkiær J, Dorup J: Hypokalemia-induced down-
trafficking by vasopressin [Review]. Curr Opin Cell Biol 9: regulation of aquaporin-2 water channel expression in rat kidney
560 –564, 1997 medulla and cortex. J Clin Invest 97: 1960 –1968, 1996
64. Nielsen S, DiGiovanni SR, Christensen EI: Cellular and subcel- 83. Frøkiær J, Nielsen S: Do aquaporins have a role in nocturnal
lular immunolocalization of vasopressin-regulated water channel enuresis? [Review] Scand J Urol Nephrol Suppl 183: 31–32,
in rat kidney. Proc Natl Acad Sci USA 90: 11663–11667, 1993 1997
65. Sabolic I, Katsura T, Verbavatz JM: The AQP2 water channel: 84. Xu DL, Martin PY, Ohara M: Upregulation of aquaporin-2 water
Effect of vasopressin treatment, microtubule disruption, and dis- channel expression in chronic heart failure rat. J Clin Invest 99:
tribution in neonatal rats [Published erratum appears in J Membr 1500 –1505, 1997
Biol 145: 107–108, 1995]. J Membr Biol 143: 165–175, 1995 85. Nielsen S, Terris J, Andersen D: Congestive heart failure in rats
66. Wade JB, Stetson DL, Lewis SA: ADH action: Evidence for a is associated with increased expression and targeting of aqua-
membrane shuttle mechanism. Ann NY Acad Sci 372: 106 –117, porin-2 water channel in collecting duct. Proc Natl Acad Sci USA
1981 94: 5450 –5455, 1997
67. DiGiovanni SR, Nielsen S, Christensen EI: Regulation of col- 86. Ohara M, Martin PY, Xu DL: Upregulation of aquaporin 2 water
lecting duct water channel expression by vasopressin in Brattle- channel expression in pregnant rats. J Clin Invest 101: 1076 –
boro rat. Proc Natl Acad Sci USA 91: 8984 – 8988, 1994 1083, 1998
68. Yamamoto T, Sasaki S, Fushimi K: Localization and expression 87. Fujita N, Ishikawa SE, Sasaki S: Role of water channel AQP-CD
J Am Soc Nephrol 11: 764 –777, 2000 Homer W. Smith Award Lecture 777

in water retention in SIADH and cirrhotic rats. Am J Physiol 269: membrane domains for water transport in glial cells: High-
F926 –F931, 1995 resolution immunogold cytochemistry of aquaporin-4 in rat
88. Ishibashi K, Sasaki S, Fushimi K: Molecular cloning and expres- brain. J Neurosci 17: 171–180, 1997
sion of a member of the aquaporin family with permeability to 104. Nagelhus EA, Veruki ML, Torp R: Aquaporin-4 water channel
glycerol and urea in addition to water expressed at the basolateral protein in the rat retina and optic nerve: Polarized expression in
membrane of kidney collecting duct cells [see Comments]. Proc Muller cells and fibrous astrocytes. J Neurosci 18: 2506 –2519,
Natl Acad Sci USA 91: 6269 – 6273, 1994 1998
89. Echevarria M, Windhager EE, Tate SS: Cloning and expression 105. Frigeri A, Nicchia GP, Verbavatz JM: Expression of aquaporin-4
of AQP3, a water channel from the medullary collecting duct of in fast-twitch fibers of mammalian skeletal muscle. J Clin Invest
rat kidney. Proc Natl Acad Sci USA 91: 10997–11001, 1994 102: 695–703, 1998
90. Ma T, Frigeri A, Hasegawa H: Cloning of a water channel 106. Verbavatz JM, Ma T, Gobin R: Absence of orthogonal arrays in
homolog expressed in brain meningeal cells and kidney collect- kidney, brain and muscle from transgenic knockout mice lacking
ing duct that functions as a stilbene-sensitive glycerol trans- water channel aquaporin-4. J Cell Sci 110: 2855–2860, 1997
porter. J Biol Chem 269: 21845–21849, 1994 107. Rash JE, Yasumura T, Hudson CS: Direct immunogold labeling
91. You G, Smith CP, Kanai Y: Cloning and characterization of the of aquaporin-4 in square arrays of astrocyte and ependymocyte
vasopressin-regulated urea transporter. Nature 365: 844 – 847, plasma membranes in rat brain and spinal cord. Proc Natl Acad
1993 Sci USA 95: 11981–11986, 1998
92. Echevarria M, Windhager EE, Frindt G: Selectivity of the renal 108. Han Z, Wax MB, Patil RV: Regulation of aquaporin-4 water
collecting duct water channel aquaporin-3. J Biol Chem 271: channels by phorbol ester-dependent protein phosphorylation.
25079 –25082, 1996 J Biol Chem 273: 6001– 6004, 1998
93. Kuwahara M, Gu Y, Ishibashi K: Mercury-sensitive residues and
109. Ma T, Frigeri A, Skach W: Cloning of a novel rat kidney cDNA
pore site in AQP3 water channel. Biochemistry 36: 13973–
homologous to CHIP28 and WCH-CD water channels. Biochem
13978, 1997
Biophys Res Commun 197: 654 – 659, 1993
94. Zeuthen T, Klaerke DA: Transport of water and glycerol in
110. Ma T, Yang B, Kuo WL: cDNA cloning and gene structure of a
aquaporin-3 is gated by H⫹. J Biol Chem 274: 21631–21636,
novel water channel expressed exclusively in human kidney:
1999
Evidence for a gene cluster of aquaporins at chromosome locus
95. Frigeri A, Gropper MA, Umenishi F: Localization of MIWC and
12q13. Genomics 35: 543–550, 1996
GLIP water channel homologs in neuromuscular, epithelial and
111. Yasui M, Kwon TH, Knepper M: Aquaporin-6: An intracellular
glandular tissues. J Cell Sci 108: 2993–3002, 1995
vesicle water channel protein in renal epithelia. Proc Natl Acad
96. Ecelbarger CA, Terris J, Frindt G: Aquaporin-3 water channel
Sci USA 96: 5808 –5813, 1999
localization and regulation in rat kidney. Am J Physiol 269:
F663–F672, 1995 112. Yasui M, Hazama A, Kwon TH: Rapid gating and anion perme-
97. Ishibashi K, Sasaki S, Fushimi K: Immunolocalization and effect ability of an intracellular aquaporin. Nature 402: 184 –187, 1999
of dehydration on AQP3, a basolateral water channel of kidney 113. Schwartz GJ, Al-Awqati QF: Regulation of transepithelial H⫹
collecting ducts. Am J Physiol 272: F235–F241, 1997 transport by exocytosis and endocytosis. Annu Rev Physiol 48:
98. Nielsen S, King LS, Christensen BM: Aquaporins in complex 153–161, 1986
tissues. II. Subcellular distribution in respiratory and glandular 114. Gluck S, Iyori M, Holliday S: Distal urinary acidification from
tissues of rat. Am J Physiol 273: C1549 –C1561, 1997 Homer Smith to the present. Kidney Int 49: 1660 –1664, 1996
99. Terris J, Ecelbarger CA, Marples D: Distribution of aquaporin-4 115. Günther W, Luchow A, Cluzeaud F: ClC-5, the chloride channel
water channel expression within rat kidney. Am J Physiol 269: mutated in Dent’s disease, colocalizes with the proton pump in
F775–F785, 1995 endocytotically active kidney cells. Proc Natl Acad Sci USA 95:
100. Chou CL, Ma T, Yang B: Fourfold reduction of water perme- 8075– 8080, 1998
ability in inner medullary collecting duct of aquaporin-4 knock- 116. Friedrich T, Breiderhoff T, Jentsch TJ: Mutational analysis dem-
out mice. Am J Physiol 274: C549 –C554, 1998 onstrates that ClC-4 and ClC-5 directly mediate plasma mem-
101. Hasegawa H, Ma T, Skach W: Molecular cloning of a mercurial- brane currents. J Biol Chem 274: 896 –902, 1999
insensitive water channel expressed in selected water-transport- 117. Loo DD, Hirayama BA, Meinild AK: Passive water and ion
ing tissues. J Biol Chem 269: 5497–5500, 1994 transport by cotransporters. J Physiol 518: 195–202, 1999
102. Jung JS, Bhat RV, Preston GM: Molecular characterization of an 118. Ramón y Cajal S: Advice for a Young Investigator, translated by
aquaporin cDNA from brain: Candidate osmoreceptor and reg- Swanson N, Swanson LW, Cambridge, MA, MIT Press, 1999
ulator of water balance. Proc Natl Acad Sci USA 91: 13052– 119. Borgnia M, Nielsen S, Engel A, Agre P: Cellular and molecular
13056, 1994 biology of the aquaporin water channels. Annu Rev Biochem 68:
103. Nielsen S, Nagelhus EA, Amiry-Moghaddam M: Specialized 425– 458, 1999

You might also like