中文 / ENGLISH

Contact us

PHONE:0512-57037155
FAX:0512-55186984
MOBILE:15190183483
E-MAIL:cyclochem@163.com
QQ:173218523 675855172
ADDRESS:江苏昆山经济开发区太湖南路89号

The current position:Home -> NewsNews details

Methyl-_-cyclodextrin induces vasopressin-independent apical accumulation

2011-05-22 08:40:35

Methyl-_-cyclodextrin induces vasopressin-independent apical accumulation

of aquaporin-2 in the isolated, perfused rat kidney

Leileata M. Russo, Mary McKee, and Dennis Brown

Program in Membrane Biology and Division of Nephrology, Department of Medicine,

Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts

Submitted 2 November 2005; accepted in final form 26 January 2006

Russo, Leileata M., Mary McKee, and Dennis Brown. Methyl-

_-cyclodextrin induces vasopressin-independent apical accumulation

of aquaporin-2 in the isolated, perfused rat kidney. Am J

Physiol Renal Physiol 291: F246 –F253, 2006. First published

January 31, 2006; doi:10.1152/ajprenal.00437.2005.—Vasopressin

increases urine concentration by stimulating plasma membrane accumulation

of aquaporin-2 (AQP2) in collecting duct principal cells,

allowing bulk water flow across the collecting duct from lumen to

interstitium down an osmotic gradient. Mutations in the vasopressin

type 2 receptor (V2R) cause hereditary X-linked nephrogenic diabetes

insipidus (NDI), a disease characterized by excessive urination and

dehydration. Recently, we showed that inhibition of endocytosis by

the cholesterol-depleting drug methyl-_-cyclodextrin (m_CD) induces

plasma membrane accumulation of AQP2 in transfected renal

epithelial cells overexpressing epitope-tagged AQP2. Here, we asked

whether m_CD could induce membrane accumulation of AQP2 in

situ using the isolated, perfused kidney (IPK). By immunofluorescence

and electron microscopy, we show that AQP2 was shifted from

a predominantly intracellular localization to the apical membrane of

principal cells following 1-h perfusion of Sprague-Dawley rat kidneys

with 5 mM m_CD. Quantification of staining revealed that the

intensity of AQP2 was increased from 647 _ 114 (control) to 1,968 _

299 units (m_CD; P _ 0.001), an effect similar to that seen after

perfusion with 4 nM dDAVP (1,860 _ 298, P _ 0.001). Similar

changes were observed following m_CD perfusion of kidneys from

vasopressin-deficient Brattleboro rats. No effect of m_CD treatment

on the basolateral distribution of AQP3 and AQP4 was detected.

These data indicate that AQP2 constitutively recycles between the

apical membrane and intracellular vesicles in principal cells in situ

and that inducing apical AQP2 accumulation by inhibiting AQP2

endocytosis is a feasible goal for bypassing the defective V2R signaling

pathway in X-linked NDI.

diabetes insipidus; collecting duct; endocytosis; immunocytochemistry;

aquaporin recycling

AQUAPORIN-2 (AQP2) is a hydrophobic _29-kDa protein that is

composed of six transmembrane segments and cytoplasmically

located NH2 and COOH termini (2). Expressed in principal

cells of the kidney collecting duct (CD), AQP2 plays a major

role in whole body water homeostasis through water reabsorption

that results in urine concentration (6, 8). This function is

achieved by the insertion of AQP2 into the apical membrane of

principal cells, where it acts as a hydrophilic pore allowing the

osmotic flow of water into the cell. Water then exits basolaterally

into the interstitium via aquaporins-3 and/or -4 (12, 35)

and perhaps also AQP2 in some kidney regions (9, 17a, 25).

Dysfunction of the AQP2 membrane accumulation process

results in whole body deregulation of water homeostasis,

contributing to excessive urination and dehydration, a disease

known as diabetes insipidus (DI) (10, 24, 26, 31). AQP2

distribution and function are chiefly regulated by the antidiuretic

hormone arginine vasopressin (AVP), which is released

by the posterior pituitary gland in response to high-serum

osmolarity and/or blood volume reduction (17). Circulating

AVP binds to the vasopressin type 2 receptor (V2R), located

on the basolateral side of principal cells of the CD, causing

intracellular cAMP levels to increase. This activates protein

kinase A (PKA) (27), resulting in the phosphorylation of AQP2

at residue S256. This phosphorylation is necessary for the

AVP-induced redistribution of AQP2 from a vesicular to a

membrane localization (13, 18), allowing water reabsorption to

occur in the CD. AQP2 membrane accumulation can also occur

after an increase in cytoplasmic cGMP levels; protein kinase G

(PKG) is also capable of phosphorylating AQP2 on residue

S256 (4).

DI has several distinct causes. Hereditary forms may be

“nephrogenic (NDI)” (the result of mutations in the AQP2 or

V2R genes) or “central (CDI)” (the result of mutations in the

AVP gene). Several mutations in the AQP2 gene have been

identified and they commonly lead to the mistargeting of AQP2

to subcellular organelles rather than the plasma membrane,

resulting in AQP2 degradation (23). X-linked NDI is caused by

mutations in the V2R and is much more common than autosomal

NDI in which AQP2 is defective. The AQP2 protein

expressed in X-linked NDI patients as well as in patients with

CDI is probably functional, but the inability of the kidney to

respond to AVP or the lack of active AVP hormone precludes

AQP2 accumulation on the plasma membrane of principal

cells.

In cases where AVP itself is dysfunctional, the symptoms of

CDI may be treated by administration of 1-desamino(8-Darginine)

vasopressin or dDAVP, a synthetic structural analog

of AVP that has a high specificity for the V2R (1). Treatment

of CDI with dDAVP is generally successful; however, dDAVP

in combination with some drugs may cause dilutional hyponatremia

(12a). Of course, dDAVP is not effective in most cases

where the V2R receptor is mutated or unresponsive to AVP,

although high doses of AVP may be effective in a few

instances where the V2R has a lower, but not absent, affinity

for the hormone. To date, therefore, the treatment of NDI

resulting from V2R mutations is limited and relies largely on

reducing urine flow by (seemingly paradoxical) treatment with

thiazide diuretics with Address for reprint requests and other correspondence: L. M. Russo, concomitant sodium restriction.

Program in Membrane Biology, Division of Nephrology, Massachusetts General

Hospital/Harvard Medical School, Simches Research Center, 185

Cambridge St, Rm 8100, Boston, MA 02114 (e-mail: leileata.russo@

receptor.mgh.harvard.edu).

The costs of publication of this article were defrayed in part by the payment

of page charges. The article must therefore be hereby marked “advertisement

in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Am J Physiol Renal Physiol 291: F246–F253, 2006.

First published January 31, 2006; doi:10.1152/ajprenal.00437.2005.

F246 0363-6127/06 $8.00 Copyright © 2006 the American Physiological Society http://www.ajprenal.org

Downloaded from ajprenal.physiology.org on December 4, 2007

However, some recent studies are emerging that suggest

alternative strategies for therapeutic intervention in NDI. These

are based on our increasing understanding of the signal transduction

and trafficking pathways that result in the membrane

accumulation of AQP2. Previous work in vitro showed that

AQP2 is constitutively recycled between the plasma membrane

and intracellular vesicles, even in the absence of AVP (22, 32).

The relative rates of endo- and exocytosis of AQP2 were

proposed to be critical in determining the amount of AQP2 in

the plasma membrane (14, 25). After reaching the cell surface,

AQP2 is subsequently internalized by clathrin-mediated endocytosis

(32). The depletion of plasma membrane cholesterol by

methyl-_-cyclodextrin (m_CD) prevents the formation and

budding of clathrin-coated pits, resulting in the acute blockade

of endocytosis (29). With the use of this strategy, a very rapid

plasma membrane accumulation of AQP2 was demonstrated in

AQP2-transfected LLC-PK1 and inner medullary CD (IMCD)

cells in culture. Furthermore, inhibiting endocytosis with dominant-

negative (K44A) dynamin also resulted in membrane

accumulation of AQP2 in cultured cells (32).

The aim of our present study was to determine whether the

endocytosis inhibitor m_CD would cause vasopressin-independent

AQP2 accumulation in principal cells of the functioning,

intact rat kidney. This would indicate that AQP2 is

constitutively recycling not only in transfected cell models in

vitro but also in CD principal cells in situ. However, m_CD

Fig. 1. Methyl-_-cyclodextrin (m_CD) treatment

increases apical aquaporin-2 (AQP2) expression in

the isolated, perfused kidney (IPK): immunofluorescence

microscopy. Sprague-Dawley kidneys were

perfused under control conditions, in the presence of

5 mM m_CD, or with 4 nM dDAVP for 60 min.

Kidneys were then fixed, sectioned, and immunostained

using anti-AQP2 antibodies. For all treatments,

examples of tubules sectioned transversely

from the inner stripe of the outer medulla (A, C, E)

and longitudinally in the inner medulla/papilla (B,

D, F) are illustrated. Under control conditions (A,

B), AQP2 has a cytosolic distribution in principal

cells of the collecting duct. In the inner stripe

sections (A, C, E), AQP2-negative intercalated cells

appear as dark, unstained “holes” in the epithelial

layer. After perfusion with 5 mM m_CD (C, D),

AQP2 shows an increased apical localization in

principal cells of the inner stripe and inner medulla.

After perfusion with 4 nM dDAVP, a similar and

expected increased apical localization of AQP2 in

medullary collecting ducts is seen in the IPK preparation.

However, dDAVP also induced a clear

basolateral localization of AQP2 in the inner medulla

(F and arrows, inset) but not in the inner stripe

(E) principal cells. Bar _ 40 _m.

Report

AQUAPORIN-2 RECYCLING IN THE ISOLATED, PERFUSED KIDNEY F247

AJP-Renal Physiol VOL 291 JULY 2006 www.ajprenal.org

Downloaded from ajprenal.physiology.org on December 4, 2007

targets and binds cholesterol, and its use in vivo is limited due

to its potential toxicity and its ability to lyse erythrocytes.

Therefore, we examined the effect of m_CD on AQP2 localization

using the ex vivo isolated, perfused kidney technique

(IPK). This technique allows the kidney to function on an

artificial closed circuit, hence removing extra renal influences,

making the IPK an invaluable tool for the investigation of

drugs or compounds that may be toxic or display complex

interactions in vivo (33). The results obtained confirm previous

in vitro studies and demonstrate that AQP2 constitutively

recycles to the apical membrane, a process that is

halted by the depletion of cholesterol using m_CD and

which results in the accumulation of AQP2 on the plasma

membrane (22). This provides proof-of-principle evidence

that specific targeting of m_CD (or other agents that inhibit

endocytosis) to CDs may provide a novel form of treatment

for DI resulting from dysfunctional V2R or AVP or as an

alternative to dDAVP treatment in cases where its use is

contraindicated.

MATERIALS AND METHODS

IPK preparation. All animal experiments carried out were approved

by the Institutional Committee on Research Animal Care, in

accordance with National Institutes of Health (NIH) Guide for the

Care and Use of Laboratory Animals. Male Sprague-Dawley rats or

male Brattleboro rats weighing 250–270 g were used. To prevent the

interference of oxytocin release on AQP2 distribution during surgery

(19), Brattleboro rats (which lack vasopressin) were deprived of water

overnight before the experiment. Rats were anesthetized by an intraperitoneal

injection of 45 mg/kg body wt of pentobarbital sodium

(Nembutal; Abbott Laboratories, Chicago, IL). PBS containing 10%

mannitol and 200 IU heparin (final volume 1 ml; Sigma, St. Louis,

MO) was injected into the femoral vein. To prevent dehydration, 1.5

ml of 0.9% NaCl were also injected into the femoral vein immediately

before surgery. A laparotomy was performed and the right ureter was

catheterized with polyethylene tubing PE/1 (Scientific Commodities).

The right renal artery was cannulated via the superior mesenteric

artery and the kidney was removed by en bloc dissection. The whole

procedure was completed within 10 min. The kidney perfusion pressure

was maintained at 95–100 mmHg with a peristaltic pump monitored

using a calibrated aneroid manometer. The kidney was perfused

with a recirculating Krebs-Henseleit buffer containing 5% BSA,

Fig. 2. Apical accumulation of AQP2 in cortical collecting ducts (CCDs) is

induced by m_CD and dDAVP. Sprague-Dawley kidneys were perfused under

control conditions (CON), in the presence of 5 mM m_CD or with 4 nM

dDAVP for 60 min. Kidneys were then fixed, sectioned, and immunostained

using anti-AQP2 antibodies. Under control conditions (A), AQP2 has a

predominantly cytosolic distribution but some cells have an apical distribution

in the cortical collecting duct. After perfusion with 5 mM m_CD (B), AQP2

has an increased apical localization in the collecting duct. After perfusion with

4 nM dDAVP (C), a similar and expected increased apical localization of

AQP2 in the collecting ducts is seen in the IPK preparation. G, glomerulus.

Bar _ 50 _m.

Fig. 3. Apical accumulation of AQP2 in outer medullary collecting ducts is

induced by m_CD and dDAVP. Sprague-Dawley kidneys were perfused under

control conditions, in the presence of 5 mM m_CD, or with 4 nM dDAVP for

60 min. Kidneys were then fixed, sectioned, and immunostained using anti-

AQP2 antibodies. Under control conditions (A), AQP2 has a predominantly

cytosolic distribution. After perfusion with 5 mM m_CD (B), AQP2 shows an

increased apical localization in the collecting duct. After perfusion with 4 nM

dDAVP (C), the expected increased apical localization of AQP2 in the

collecting ducts is seen in the IPK preparation. Bar _ 40 _m.

Report

F248 AQUAPORIN-2 RECYCLING IN THE ISOLATED, PERFUSED KIDNEY

AJP-Renal Physiol VOL 291 JULY 2006 www.ajprenal.org

Downloaded from ajprenal.physiology.org on December 4, 2007

glucose, essential amino acids, and oxygen radical scavengers as

previously described (34). The perfusate was filtered using a 0.45-_m

filter and was oxygenated with 95% O2-5% CO2, pH 7.4, and

maintained at 37°C throughout the entire procedure. The kidney was

first equilibrated on the apparatus for 10 min and was then perfused

for a 60-min time period. Urine and plasma samples were collected at

40 and 60 min. Urine osmolality was measured using a Wescor Vapor

Pressure Osmometer 5520 (Wescor, Logan, UT).

m_CD and dDAVP experiments were carried out using exactly the

same procedure except that the perfusate contained 5 mM m_CD

(Sigma) or 4 nM dDAVP (Sigma).

Tissue fixation and immunostaining. Following perfusion on the

IPK apparatus, the kidney was immediately flushed with paraformaldehyde

lysine periodate (PLP) fixative and then immersion-fixed

overnight in PLP, as described previously (3). Tissue was then

extensively washed in PBS and cryoprotected in 30% sucrose. Tissue

was embedded in OCT and 5-_m-thick cryosections were cut. Sections

were treated with 1% SDS in PBS, an antigen retrieval technique

(7), and then blocked using a solution of 1% BSA in PBS. Previously

characterized primary antibodies used were against the COOH terminus

of AQP2 (30), AQP3, or AQP4 (15). Secondary antibodies used

were goat anti-rabbit IgG coupled to CY3 or FITC (Jackson Immuno-

Research Laboratories, West Grove, PA).

Staining was analyzed using a Nikon E800 fluorescence microscope

equipped with a Hamamatsu Orca CCD camera and IP Lab

Spectrum acquisition and image analysis software, or with a Zeiss

Axioplan microscope equipped with a Radiance 2000 confocal laserscanning

system (Bio-Rad). For quantification, digital images were

acquired using the Nikon microscope with a _40 objective. All

images from different slides were collected with the same microscope

setting. Perpendicularly sectioned cells from the inner stripe/inner

medulla boundary region were chosen at random and a line was drawn

through the cell from the basolateral to apical pole, passing through

the region of the nucleus. The line was analyzed for intensity and the

intensity at the level of the apical pole was recorded. Twenty-five

individual cells were quantified in each of three to four kidneys from

each group (75–100 points in total for each group). Analysis was

performed on a Macintosh computer using the public domain NIH

Image program (developed at the U.S. NIH and available on the

Internet at http://rsb.info.nih.gov/nih-image/).

Electron microscopy and immunogold staining. For electron microscopy

and immunogold staining, small pieces of PLP-perfused rat kidney

(_1 mm3) were dehydrated through a graded series of ethanol to 100%

ethanol. They were then infiltrated with LR White resin (Electron Microscopy

Sciences, Fort Washington, PA) and were placed in fresh LR

White for a few hours and embedded in gelatin capsules at 50°C

overnight. For the immunostaining, thin sections were cut on a Reichert

Ultracut E ultramicrotome and were collected on formvar-coated nickel

grids. The grids were incubated on drops of 1% BSA/PBS _ 5% normal

goat serum (Sigma) for 10 min at room temperature as a blocking step.

They were then placed on drops of either DAKO antibody diluent

(negative control, DAKO, Carpinteria, CA), or on drops of primary

antibody diluted in DAKO diluent for 1 h, at room temperature. Following

several rinses on drops of PBS, the grids were incubated on drops of

gold-conjugated secondary anti-rabbit IgG (Ted Pella, Redding, CA) for

1 h at room temperature. Finally, the grids were rinsed several times on

drops of distilled water and stained for 5 min on drops of aqueous uranyl

acetate (EMS). They were rinsed on drops of distilled water, dried, and

examined in a JEOL 1011 transmission electron microscope at 80 kV,

and imaged digitally.

Statistical analysis. All data are expressed as means _ SE where n

is the number of measurements. Statistical analysis was performed

using Student’s t-test where P _ 0.05 was taken as statistically

significant. Statistical analysis of apical intensity was carried out by

performing a one-way ANOVA followed by Student’s t-test.

RESULTS AND DISCUSSION

Following perfusion of intact, isolated kidneys with 5 mM

m_CD for 60 min, immunocytochemical analysis of AQP2 revealed

a striking redistribution of AQP2. It relocated from a

predominantly cytoplasmic location in outer medullary (inner

stripe) principal cells of the nontreated IPK (Fig. 1A) to an apical

distribution in the m_CD-treated kidney (Fig. 1C), an effect

similar to that of dDAVP (Fig. 1E). This marked apical redistribution

of AQP2 induced by both m_CD and dDAVP was also

evident in CDs of the cortex and the outer stripe (Figs. 2 and 3).

In the middle portion of the papilla, AQP2 was also relocated to

the apical pole of principal cells following m_CD treatment,

whereas dDAVP induced not only apical accumulation, but also

basolateral localization of AQP2 in this portion of the CD (Fig. 1,

B, D, F), as previously described in vivo (36). In addition,

basolateral AQP2 was also seen in a population of cortical

connecting segments (data not shown), a phenomenon that has

also been described earlier (9). The AQP2 redistribution was also

evident by electron microscopy (Fig. 4, A-C), where AQP2 gold

labeling showed an increased apical plasma membrane localization

following m_CD (Fig. 4B) and dDAVP (Fig. 4C) treatment.

However, a marked basolateral accumulation of AQP2 was noted

after dDAVP, but not m_CD treatment in the middle portion of

Fig. 4. Immunogold electron microscopy showing that m_CD increases apical

membrane expression of AQP2 in the IPK. Sprague-Dawley kidneys were

perfused in control conditions, in the presence of 5 mM m_CD, or with 4 nM

dDAVP for 60 min as in Fig. 1. Kidneys were then fixed and processed in LR

White embedding resin for electron microscopy analysis using immunogold

labeling for AQP2. A: AQP2 is found in a mainly subapical location (some

indicted by arrowheads) in the control IPK, although some apical AQP2 (some

indicated by arrows) is also detectable. B: apical plasma membrane AQP2

expression is greatly increased in the m_CD-treated IPK (some indicated by

arrows). C: apical AQP2 distribution is also increased in the dDAVP-treated

IPK (some indicated by arrows). Bar _ 0.5 _m.

Report

AQUAPORIN-2 RECYCLING IN THE ISOLATED, PERFUSED KIDNEY F249

AJP-Renal Physiol VOL 291 JULY 2006 www.ajprenal.org

Downloaded from ajprenal.physiology.org on December 4, 2007

the inner medulla (not shown). Morphologically, the CDs from all

IPK groups looked similarly intact, and epithelial cells were well

preserved when analyzed by electron microscopy, demonstrating

that the change in AQP2 distribution using m_CD treatment was

not accompanied by gross cellular alterations. In support of this,

both AQP3 and AQP4 maintained their usual tight basolateral

localization in both the nontreated control and m_CD-treated IPK

(Fig. 5, A-D).

Analysis of AQP2 distribution in the apical pole of principal

cells from CDs along the inner stripe/inner medullary border

(where the effect was the most pronounced) using NIH Image

revealed the apical AQP2 redistribution to be statistically

significant in both m_CD-treated group (P _ 0.001) and

dDAVP-treated group (P _ 0.001) compared with the nontreated

control IPK (Fig. 6). Both treatments resulted in an

almost threefold increase in apical membrane staining intensity

in this system. However, despite this increase in apical AQP2,

no significant change in urine concentrating ability was observed

[330 _ 11 (n _ 4) control IPK vs. 362 _ 6.8 (n _ 3)

dDAVP-treated IPK]. This is consistent with the results of

Lieberthal et al. (21), who demonstrated that the addition of

dDAVP resulted in the partial production of concentrated urine

by the IPK only when erythrocytes (40–45% hematocrit) were

included in the circulating medium (20). This maneuver was

not possible in the present set of experiments, because m_CD

efficiently lyses erythrocytes, which can lead to tubular necrosis

due to the release of heme proteins (5).

To determine whether the same effects of m_CD could be

obtained in a vasopressin-deficient model in which AQP2

levels are considerably lower than in normal rats (28), Brattleboro

rats which lack functional AVP were used in a parallel

study to determine the effect of m_CD on AQP2 distribution.

Our data revealed a similar redistribution of AQP2 from a

cytoplasmic localization to an apical localization following

5 mM m_CD perfusion in these animals (Fig. 7, A and B).

Similar to the results from Sprague-Dawley rats, AQP3 distri-

Fig. 5. m_CD has no effect on AQP3 and AQP4

localization in the IPK: confocal immunofluorescence

microscopy. Sprague-Dawley kidneys were

perfused under control conditions or in the presence

of 5 mM m_CD for 60 min. Kidneys were then

fixed and immunostained using anti-AQP3 and anti-

AQP4 antibodies. A and C: AQP3 distribution under

control IPK conditions demonstrates the expected

basolateral distribution of AQP3 (A) and AQP4 (C)

in principal cells of the inner medullary collecting

duct. B and D: AQP3 and AQP4 distribution in 5

mM m_CD IPK also demonstrates basolateral staining

for AQP3 (B) and AQP4 (D) comparable to that

observed in the control IPK. This result demonstrates

that m_CD did not interfere with the normal

basolateral expression of AQP3 or AQP4. Bar _

10 _m.

Fig. 6. Effect of m_CD on the apical intensity of AQP2 immunostaining in the

IPK. Quantitation of AQP2 distribution in the IPK was analyzed using images

similar to those shown in Fig. 1, in control conditions, after 5 mM m_CD

treatment, and after perfusion with 4 nM dDAVP (see MATERIALS AND METHODS).

Collecting ducts from the inner stripe/inner medulla boundary were

selected for quantification. The data revealed significant and comparable

increases in AQP2 apical intensity following 60-min perfusion with either

5 mM m_CD perfusion (P _ 0.001) or 4 nM dDAVP (P _ 0.001) compared

with the control IPK. Data are expressed as arbitrary units of fluorescence

intensity. *P _ 0.001, n _ 3 for control and dDAVP perfusions and n _ 4 for

m_CD perfusion.

Report

F250 AQUAPORIN-2 RECYCLING IN THE ISOLATED, PERFUSED KIDNEY

AJP-Renal Physiol VOL 291 JULY 2006 www.ajprenal.org

Downloaded from ajprenal.physiology.org on December 4, 2007

bution (Fig. 7, C and D) and AQP4 distribution (not shown)

were also unaffected by the perfusion with m_CD in the

Brattleboro rat kidney. This finding demonstrates that even in

the absence of preexposure to AVP in vivo, AQP2 can be

sequestered to the apical membrane of principal cells by

exposure to m_CD.

The Sprague-Dawley-derived IPK perfused with 5 mM

m_CD displayed a decrease in kidney flow rate and urine

production (Table 1), although perfusion pressure was not

significantly different from nontreated control IPKs (Table 1).

Urine osmolality showed a tendency to be higher in the

m_CD-treated rat kidneys (as also seen for the dDAVP-treated

rats), although this was not statistically significant (Table 1).

As mentioned above, this finding is in agreement with previous

data suggesting that the IPK has a compromised concentrating

ability (20, 21). Although attempts were also made to analyze

the effect of m_CD on endocytosis through the injection of a

concentrated bolus of FITC dextran or horseradish peroxidase

(HRP) into the renal artery, we were unable to visualize the

endocytosis of these markers in any of the three groups studied.

Previous reports indicate that the IPK has disrupted sodium

handling as well as progressive ischemic injury of the medullary

thick ascending limb contributing to a decreased concentrating

ability (20, 21). In the present study, this decreased

concentrating ability also hampered efforts to analyze endocytosis

of the fluid phase markers FITC-dextran and HRP as they

did not achieve a high enough concentration in the renal tubule

to permit ready visualization.

Our data demonstrate for the first time the ability of m_CD

to sequester AQP2 in the apical membrane of CD principal

cells in the intact ex vivo functioning kidney. The addition of

m_CD to the IPK perfusate was able to significantly increase

the level of apical AQP2 in the CD in all kidney regions,

without affecting the expression and localization of AQP3 or

AQP4. However, the effect was most pronounced in the inner

stripe and the initial two-thirds of the inner medulla. A similar

effect was also found in kidneys from the vasopressin-deficient

Brattleboro rat. These data are consistent with the idea that

Fig. 7. m_CD increases apical AQP2 expression in

IPK from vasopressin-deficient Brattleboro rats.

Brattleboro rat kidneys were perfused under control

conditions and in the presence of 5 mM m_CD for

a total of 60 min. Kidneys were then fixed and

stained using anti-AQP2 antibodies. A: AQP2 in the

control IPK has a subapical distribution in principal

cells of the inner stripe collecting duct. B: after

perfusion with 5 mM m_CD, AQP2 has an increased

apical localization, similar to that seen in

Sprague-Dawley rat kidneys (see Fig. 1). C: AQP3

displayed a basolateral distribution in both control

IPK collecting ducts (C) and in the presence of

5 mM m_CD (D). Bar _ 10 _m.

Table 1. Physiological parameters for IPK perfusion of Sprague-Dawley rat kidneys

Time

Pressure, mmHg Flow Rate, ml/min Urine Volume, ml Plasma Osmolarity, mosM Urine Osmolarity, mosM

C M C M C M C M C M

40 min 94_2.5 99_0.5 21.8_2.3 7.0_1.5* 0.92_0.3 0.19_0.1† 321_5 326_2.5 320_19 354_6.5

20 min 96_2.5 100_1 20.7_2.4 6.7_1.6* 0.65_0.2 0.23_0.1† 322_5 327_1.5 330_11 365_13.5

Values are means _ SE, n _ 4 for each group. Where time is perfusion time, C is control isolated, perfused kidney (IPK) and M is methyl-_-cyclodextrin

IPK. *P _ 0.001, †P _ 0.05 compared with control IPK.

Report

AQUAPORIN-2 RECYCLING IN THE ISOLATED, PERFUSED KIDNEY F251

AJP-Renal Physiol VOL 291 JULY 2006 www.ajprenal.org

Downloaded from ajprenal.physiology.org on December 4, 2007

AQP2 constitutively recycles between cytoplasmic vesicles

and the apical plasma membrane of principal cells of the

kidney in situ, in the absence of vasopressin, a process that is

regulated at least in part by clathrin-mediated endocytosis (22,

32). However, the observation that dDAVP but not m_CD can

also induce basolateral accumulation of AQP2 in some regions

of the CD implies that while they are similar, the mechanisms

of action of the hormone and the drug may not completely

overlap.

Although we previously reported that AQP2 constitutively

recycles in LLC-PK1 and IMCD cells in culture (22, 32), the

fact that this constitutive recycling process occurs in situ is an

important finding for the development of future therapies for

NDI. The ability of AQP2 to be sequestered in the apical

membrane is paramount for the CD to exert its water reabsorption

function in the kidney. Although m_CD is toxic in vivo,

our present findings provide solid support for the idea that

specifically blocking apical endocytosis in the CD of nephrogenic

DI patients may eventually provide a plausible form of

therapy for NDI.

ACKNOWLEDGMENTS

We thank G. Hawthorn for excellent technical support. We also thank Dr.

W. D. Comper (Monash University, Australia) for kindly providing the IPK

apparatus that was used in these studies.

GRANTS

This work was supported by National Institutes of Health Grant DK-38452.

Dr. L. M. Russo is the recipient of a Juvenile Diabetes Research Foundation

Postdoctoral Fellowship. The Microscopy Core facility of the MGH Program

in Membrane Biology receives additional support from the Boston Area

Diabetes and Endocrinology Research Center (DK-57521) and the Center for

the Study of Inflammatory Bowel Disease (DK-43341).

REFERENCES

1. Andersson KE and Arner B. Effects of DDAVP, a synthetic analogue of

vasopressin, in patients with cranial diabetes insipidus. Acta Med Scand

192: 21–27, 1972.

2. Bai L, Fushimi K, Sasaki S, and Marumo F. Structure of aquaporin-2

vasopressin water channel. J Biol Chem 271: 5171–5176, 1996.

3. Beaulieu V, Da Silva N, Pastor-Soler Brown CR, Smith PJ, Brown D,

and Breton S. Modulation of the actin cytoskeleton via gelsolin regulates

vacuolar H-VATPase recycling. J Biol Chem 280: 8452–8463, 2005.

4. Bouley R, Breton S, Sun T, McLaughlin M, Nsumu NN, Lin HY,

Ausiello DA, and Brown D. Nitric oxide and arterial natriuretic factor

stimulate cGMP-dependent membrane insertion of aquarorin 2 in renal

epithelial cells. J Clin Invest 106: 1115–1126, 2000.

5. Braun SR, Weiss FR, Keller AI, Ciccone JR, and Preuss HG. Evaluation

of renal toxicity of heme proteins and their derivatives: a role in the

genesis of acute tubule necrosis. J Exp Med 131: 443–460, 1970.

6. Brown D. Ins and outs of aquaporin-2 trafficking. Am J Physiol Renal

Physiol 284: F893–F901, 2003.

7. Brown D, Lydon J, McLaughlin M, Stuart-Tilley A, Tyszkowski R,

and Alper S. Antigen retrieval in cryostat tissue sections and cultured

cells by treatment with sodium dodecyl sulfate (SDS). Histochem Cell Biol

105: 261–267, 1996.

8. Brown D and Nielsen S. Cell biology of vasopressin action. In: The

Kidney (7th ed.), edited by Brenner BM. Philadelphia, PA: Saunders,

2003, p. 573–597.

9. Coleman RA, Wu DC, Liu J, and Wade JB. Expression of aquaporins

in the renal connecting tubule. Am J Physiol Renal Physiol 279: F874–

F883, 2000.

10. Deen PMT and Brown D. Trafficking of native and mutant mammalian

MIP proteins. In: Aquaporins: Current topics in membranes, edited by

Hohmann S, Nielsen S, and Agre P. New York: Academic, 2001, p.

235–276.

12. Ecelbarger CA, Terris J, Frindt G, Echevarria M, Marples D, Nielsen

S, and Knepper MA. Aquaporin-3 water channel localization and regulation

in rat kidney. Am J Physiol Renal Fluid Electrolyte Physiol 269:

F663–F672, 1995.

12a.Electronic Medicines Compendium. http://emc.medicines.org.uk/emc/assets/

c/html/displaydoc.asp?documentid_4217#CONTRAINDICATIONS.

13. Fushimi K, Sasaki S, and Marumo F. Phosphorylation of serine 256 is

required for cAMP-dependent regulatory exocytosis of the aquaporin-2

water channel. J Biol Chem 272: 14800–14804, 1997.

14. Gustafson CE, Katsura T, McKee M, Bouley R, Casanova JE, and

Brown D. Recycling of aquaporin 2 occurs through a temperature- and

bafilomycin-sensitive trans-Golgi-associated compartment. Am J Physiol

Renal Physiol 278: F317–F326, 2000.

15. Huang Y, Tracy R, Walsberg GE, Makkinje A, Fang P, Brown D, and

Van Hoek AN. Absence of aquaporin-4 water channels from kidneys of

the desert rodent Dipodonys merriami merriami. Am J Physiol Renal

Physiol 280: F794–F802, 2001.

17. Inoue T, Nonoguchi H, and Tomita K. Physiological effects of vasopressin

and arterial natriuretic peptide in the collecting duct. Cardiovasc

Res 51: 470–480, 2001.

17a.Kamsteeg EJ, Bichet DG, Konings IB, Nivet H, Lonergan M, Arthus

MF, van Os CH, and Deen PM. Reversed polarized delivery of an

aquaporin-2 mutant causes dominant nephrogenic diabetes insipidus.

J Cell Biol 163: 1099–1109, 2003.

18. Katsura T, Gustafson CE, Ausiello DA, and Brown D. Protein kinase

A phosphorylation is involved in regulated exocytosis of aquaporin-2 in

transfected LLC-PK1 cells. Am J Physiol Renal Physiol 272: F817–F822,

1997.

19. Lencer WI, Brown D, Ausiello DA, and Verkman AS. Endocytosis of

water channels in rat kidney: cell specificity and correlation with in vivo

antidiuresis. Am J Physiol Cell Physiol 259: C920–C932, 1990.

20. Lieberthal W, Stephens GW, Wolf EF, Rennke HG, Vasilevsky ML,

Valeri CR, and Levinsky NG. Effect of erythrocytes on the function and

morphology of the isolated perfused rat kidney. Renal Physiol 10: 14–24,

1987.

21. Lieberthal W, Vasilevsky ML, Valeri CR, and Levinsky NG. Interaction

between ADH prostaglandins in the isolated erythrocyte perfused rat

kidney. Am J Physiol Renal Fluid Electrolyte Physiol 252: F331–F337,

1987.

22. Lu H, Sun TX, Bouley R, Blackburn K, McLaughlin M, and Brown D.

Inhibition of endocytosis causes phosphorylation (S256)-independent

plasma membrane accumulation of AQP2. Am J Physiol Renal Physiol

286: F233–F243, 2004.

23. Marr N, Bichet DG, Hoefs S, Savelkoul PJ, Konings IB, De Mattia F,

Graat MP, Arthus MF, Lonergan M, Fujiwara TM, Knoers NV,

Landau D, Balfe WJ, Oksche A, Rosenthal W, Muller D, Van Os CH,

and Deen PM. Cell biologic and functional analyses of five new aquaporin-

2 missense mutations that cause recessive nephrogenic diabetes

insipidus. J Am Soc Nephrol 13: 2267–2277, 2002.

24. Morello JP and Bichet DG. Nephrogenic diabetes insipidus. Annu Rev

Physiol 63: 607–630, 2001.

25. Nielsen S, DiGiovanni SR, Christensen EI, Knepper MA, and Harris

HW. Cellular and subcellular immunolocalization of vasopressin-regulated

water channel in the rat kidney. Proc Natl Acad Sci USA 90:

11663–11667, 1993.

26. Nielsen S, Frokiaer J, Marples D, Kwon TH, Agre P, and Knepper

MA. Aquaporins in the kidney: from molecules to medicine. Physiol Rev

82: 205–244, 2002.

27. Nishimoto G, Zelenina M, Li D, Yasui M, Aperia A, Nielsen S, and

Nairn AC. Arginine vasopressin stimulates phosphorylation of aquaporin-

2 in rat renal tissue. Am J Physiol Renal Physiol 276: F254–F259,

1999.

28. Promeneur D, Kwon TH, Frokiaer J, Knepper MA, and Nielsen S.

Vasopressin V2-receptor-dependent regulation of AQP2 expression in

Brattleboro rats. Am J Physiol Renal Physiol 279: F370–F382, 2000.

29. Rodal SK, Skretting G, Garred O, Vilhardt F, van Deurs B, and

Sandvig K. Extraction of cholesterol with methyl-_-cyclodextrin perturbs

formation of clathrin-coated endocytic vesicles. Mol Biol Cell 10: 961–

974, 1999.

30. Sabolic I, Katsura T, Verbavatz JM, and Brown D. The AQP2 water

channel: effect of vasopressin treatment, microtubule disruption, and

disruption in neonatal rats. J Membr Biol 143: 165–175, 1995.

31. Sasaki S. Nephrogenic diabetes insipidus: update of genetic and clinical

aspects. Nephrol Dial Transplant 19: 1351–1353, 2004.

32. Sun TX, Van Hoek A, Huang Y, Bouley R, McLaughlin M, and Brown

D. Aquaporin-2 localization in clathrin-coated pits: inhibition of endocy-

Report

F252 AQUAPORIN-2 RECYCLING IN THE ISOLATED, PERFUSED KIDNEY

AJP-Renal Physiol VOL 291 JULY 2006 www.ajprenal.org

Downloaded from ajprenal.physiology.org on December 4, 2007

tosis by dominant-negative dynamin. Am J Physiol Renal Physiol 282:

F998–F1011, 2002.

33. Taft DR. The isolated perfused rat kidney model: a useful tool for drug

discovery and development. Current Drug Discovery Technologies 1:

97–111, 2004.

34. Tay M, Comper WD, Vassiliou P, Glasgow EF, Baker MS, and Pratt

L. The inhibitory action of oxygen radical scavengers on proteinuria and

glomerular heparan sulfate loss in the isolated perfused kidney. Biochem

Int 20: 767–778, 1990.

35. Terris J, Ecelbarger CA, Marples D, Knepper MA, and Nielsen S.

Distribution of aquaporin-4 water channel expression within rat kidney.

Am J Physiol Renal Fluid Electrolyte Physiol 269: F775–F785,

1995.

36. Van Balkom BW, van Raak M, Breton S, Pastor-soler N, Bouley R,

van der Sluijs P, Brown D, and Deen PM. Hypertonicity is involved in

redirecting the aquaporin-2 water channel into the basolateral, instead of

the apical, plasma membrane of renal epithelial cells. J Biol Chem 278:

1101–1107, 2003.