12
Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris H. Robben a,b , Marleen L. A. Kortenoeven a , Mozes Sze a , Chris Yae c , Graeme Milligan b , Viola M. Oorschot e , Judith Klumperman e , Nine V. A. M. Knoers d , and Peter M. T. Deen a,1 Departments of a Physiology and d Human Genetics, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, 6500HB Nijmegen, The Netherlands; c Vantia Ltd., SO16 7NP Southampton, United Kingdom; b Molecular Pharmacology Group, Division of Neuroscience and Molecular Pharmacology, Faculty of Biomedical and Life Sciences, University of Glasgow, G12 8QQ Glasgow, United Kingdom; and e Department of Cell Biology, Institute of Biomembranes, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands Edited by Peter C. Agre, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, and approved June 1, 2009 (received for review January 15, 2009) Binding of the peptide hormone vasopressin to its type-2 receptor (V2R) in kidney triggers a cAMP-mediated translocation of Aqua- porin-2 water channels to the apical membrane, resulting in water reabsorption and thereby preventing dehydration. Mutations in the V2R gene lead to Nephrogenic Diabetes Insipidus (NDI), a disorder in which this process is disturbed, because the encoded, often intrinsically functional mutant V2 receptors are misfolded and retained in the endoplasmic reticulum (ER). Since plasma membrane expression is thought to be essential for V2R activation, cell permeable V2R antagonists have been used to induce matu- ration and rescue cell surface expression of V2R mutants, after which they need to be displaced by vasopressin for activation. Here, however, we show that 3 novel nonpeptide V2R agonists, but not vasopressin, activate NDI-causing V2R mutants at their intracellular location, without changing their maturation and at a sufficient level to induce the translocation of aquaporin-2 to the apical membrane. Moreover, in contrast to plasma membrane V2R, degradation of intracellular V2R mutants is not increased by their activation. Our data reveal that G protein-coupled receptors (GPCRs) normally active at the plasma membrane can be activated intracellularly and that intracellular activation does not induce their degradation; the data also indicate that nonpeptide agonists constitute highly promising therapeutics for diseases caused by misfolded GPCRs in general, and NDI in particular. GPCR pharmacological chaperones signaling ER retention osmoregulation I n mammals, G protein-coupled receptors (GPCRs) form the largest family of plasma membrane receptors that transduce information from hundreds of extracellular signals to intracel- lular messengers. Not surprisingly, mutations in GPCR genes are the cause of many genetic diseases, including retinitis pigmen- tosa (1), hypogonadotropic hypogonadism (2), obesity (3), hy- pothyroidism (4), and X-linked nephrogenic diabetes insipidus (NDI) (5). Disease-causing mutations in GPCR genes can be divided in 5 classes, based on their cell-biological fates (6). Class II comprises more than 40% of gene mutations and causes so-called ‘‘conformational diseases,’’ which lead to misfolded receptors that are recognized and retained by the quality control system of the endoplasmic reticulum (ER). Often, these proteins are targeted for degradation by the proteasome (7). The vasopressin type-2 receptor (V2R) is a prototype GPCR in such studies. In states of hypernatremia or hypovolemia, arginine-vasopressin (AVP) binding to the V2R of renal col- lecting duct principal cells induces a cAMP-signaling cascade, resulting in translocation of aquaporin-2 (AQP2) water channels to the apical membrane and water reabsorption from pro-urine (antidiuresis). Patients with V2R gene mutations suffer from X-linked congenital NDI, a disease in which the antidiuretic response to AVP is lacking, leading to polyuria and polydipsia, with the risk of severe dehydration, especially in infancy. In cell-culture studies, nearly all missense V2R mutants in NDI appear misfolded, ER-retained, and unstable. However, many of these V2R mutants appear intrinsically-functional, because they exert a vasopressin-induced cAMP response when, due to over expression, some V2R mutants are expressed in the plasma membrane (8). Therefore, the inability of intracellularly- retained V2R mutants to escape the ER quality control and traffic to the basolateral membrane is fundamental to the disease. Like many other GPCRs, V2R is thought to function only when expressed in the plasma membrane. Therefore, to work toward a putative therapeutic treatment of NDI, nonpeptide V1R and V2R antagonists have been used, which can interact with and stabilize ER-retained V2R mutants and to rescue their cell surface expression. There, they could be activated following incubation with and displacement by high concentrations of dDAVP (9–12). Interestingly, however, the pharmaceutical industry has re- cently developed nonpeptide V2R-specific agonists because these drugs can be orally administered to treat incontinence and bed wetting. Since with oral administration, these drugs have to pass the intestinal cell layer to be taken up in our vascular system, we reasoned that they must be able to pass cell membranes. If so, they then may also facilitate the transport of V2R mutants in NDI to the cell surface, where they can directly activate the mutant receptors instead of having the need for displacement by vasopressin as with antagonists. Here, we tested how one recently developed and 2 novel V2R-specific nonpeptide agonists, but not the peptidic analogue of AVP (dDAVP), induce V2R mutants in NDI to generate cAMP at their intracellular site and thereby restore the normal physiological response, i.e., the translocation of AQP2 to the apical membrane. As such, our data reveal that nonpeptide agonists are highly promising novel therapeutics to treat diseases due to misfolded GPCRs in general, and NDI due to V2R mutations in particular. Results Activation of the V2R Pathway by NonPeptide Agonists. The chem- ical structures of the recently developed nonpeptide agonist Author contributions: J.H.R., M.L.A.K., G.M., J.K., N.V.A.M.K., and P.M.T.D. designed re- search; J.H.R., M.L.A.K., M.S., and V.M.O. performed research; C.Y. contributed new re- agents/analytic tools; J.H.R., M.L.A.K., M.S., and V.M.O. analyzed data; and J.H.R., M.L.A.K., G.M., J.K., N.V.A.M.K., and P.M.T.D. wrote the paper. The authors declare no conflict of interest. This article is a PNAS Direct Submission. 1 To whom correspondence should be addressed. E-mail: [email protected]. This article contains supporting information online at www.pnas.org/cgi/content/full/ 0900130106/DCSupplemental. www.pnas.orgcgidoi10.1073pnas.0900130106 PNAS July 21, 2009 vol. 106 no. 29 12195–12200 PHYSIOLOGY

Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

  • Upload
    vantram

  • View
    236

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

Intracellular activation of vasopressin V2 receptormutants in nephrogenic diabetes insipidus bynonpeptide agonistsJoris H. Robbena,b, Marleen L. A. Kortenoevena, Mozes Szea, Chris Yaec, Graeme Milliganb, Viola M. Oorschote,Judith Klumpermane, Nine V. A. M. Knoersd, and Peter M. T. Deena,1

Departments of aPhysiology and dHuman Genetics, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, 6500HBNijmegen, The Netherlands; cVantia Ltd., SO16 7NP Southampton, United Kingdom; bMolecular Pharmacology Group, Division of Neuroscience andMolecular Pharmacology, Faculty of Biomedical and Life Sciences, University of Glasgow, G12 8QQ Glasgow, United Kingdom; and eDepartment of CellBiology, Institute of Biomembranes, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands

Edited by Peter C. Agre, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, and approved June 1, 2009 (received for reviewJanuary 15, 2009)

Binding of the peptide hormone vasopressin to its type-2 receptor(V2R) in kidney triggers a cAMP-mediated translocation of Aqua-porin-2 water channels to the apical membrane, resulting in waterreabsorption and thereby preventing dehydration. Mutations inthe V2R gene lead to Nephrogenic Diabetes Insipidus (NDI), adisorder in which this process is disturbed, because the encoded,often intrinsically functional mutant V2 receptors are misfoldedand retained in the endoplasmic reticulum (ER). Since plasmamembrane expression is thought to be essential for V2R activation,cell permeable V2R antagonists have been used to induce matu-ration and rescue cell surface expression of V2R mutants, afterwhich they need to be displaced by vasopressin for activation.Here, however, we show that 3 novel nonpeptide V2R agonists,but not vasopressin, activate NDI-causing V2R mutants at theirintracellular location, without changing their maturation and at asufficient level to induce the translocation of aquaporin-2 to theapical membrane. Moreover, in contrast to plasma membrane V2R,degradation of intracellular V2R mutants is not increased by theiractivation. Our data reveal that G protein-coupled receptors(GPCRs) normally active at the plasma membrane can be activatedintracellularly and that intracellular activation does not inducetheir degradation; the data also indicate that nonpeptide agonistsconstitute highly promising therapeutics for diseases caused bymisfolded GPCRs in general, and NDI in particular.

GPCR � pharmacological chaperones � signaling � ER retention �osmoregulation

In mammals, G protein-coupled receptors (GPCRs) form thelargest family of plasma membrane receptors that transduce

information from hundreds of extracellular signals to intracel-lular messengers. Not surprisingly, mutations in GPCR genes arethe cause of many genetic diseases, including retinitis pigmen-tosa (1), hypogonadotropic hypogonadism (2), obesity (3), hy-pothyroidism (4), and X-linked nephrogenic diabetes insipidus(NDI) (5). Disease-causing mutations in GPCR genes can bedivided in 5 classes, based on their cell-biological fates (6). ClassII comprises more than 40% of gene mutations and causesso-called ‘‘conformational diseases,’’ which lead to misfoldedreceptors that are recognized and retained by the quality controlsystem of the endoplasmic reticulum (ER). Often, these proteinsare targeted for degradation by the proteasome (7).

The vasopressin type-2 receptor (V2R) is a prototype GPCRin such studies. In states of hypernatremia or hypovolemia,arginine-vasopressin (AVP) binding to the V2R of renal col-lecting duct principal cells induces a cAMP-signaling cascade,resulting in translocation of aquaporin-2 (AQP2) water channelsto the apical membrane and water reabsorption from pro-urine(antidiuresis). Patients with V2R gene mutations suffer fromX-linked congenital NDI, a disease in which the antidiuretic

response to AVP is lacking, leading to polyuria and polydipsia,with the risk of severe dehydration, especially in infancy. Incell-culture studies, nearly all missense V2R mutants in NDIappear misfolded, ER-retained, and unstable. However, many ofthese V2R mutants appear intrinsically-functional, because theyexert a vasopressin-induced cAMP response when, due to overexpression, some V2R mutants are expressed in the plasmamembrane (8). Therefore, the inability of intracellularly-retained V2R mutants to escape the ER quality control andtraffic to the basolateral membrane is fundamental to thedisease.

Like many other GPCRs, V2R is thought to function onlywhen expressed in the plasma membrane. Therefore, to worktoward a putative therapeutic treatment of NDI, nonpeptideV1R and V2R antagonists have been used, which can interactwith and stabilize ER-retained V2R mutants and to rescue theircell surface expression. There, they could be activated followingincubation with and displacement by high concentrations ofdDAVP (9–12).

Interestingly, however, the pharmaceutical industry has re-cently developed nonpeptide V2R-specific agonists becausethese drugs can be orally administered to treat incontinence andbed wetting. Since with oral administration, these drugs have topass the intestinal cell layer to be taken up in our vascular system,we reasoned that they must be able to pass cell membranes. If so,they then may also facilitate the transport of V2R mutants inNDI to the cell surface, where they can directly activate themutant receptors instead of having the need for displacement byvasopressin as with antagonists.

Here, we tested how one recently developed and 2 novelV2R-specific nonpeptide agonists, but not the peptidic analogueof AVP (dDAVP), induce V2R mutants in NDI to generatecAMP at their intracellular site and thereby restore the normalphysiological response, i.e., the translocation of AQP2 to theapical membrane. As such, our data reveal that nonpeptideagonists are highly promising novel therapeutics to treat diseasesdue to misfolded GPCRs in general, and NDI due to V2Rmutations in particular.

ResultsActivation of the V2R Pathway by NonPeptide Agonists. The chem-ical structures of the recently developed nonpeptide agonist

Author contributions: J.H.R., M.L.A.K., G.M., J.K., N.V.A.M.K., and P.M.T.D. designed re-search; J.H.R., M.L.A.K., M.S., and V.M.O. performed research; C.Y. contributed new re-agents/analytic tools; J.H.R., M.L.A.K., M.S., and V.M.O. analyzed data; and J.H.R., M.L.A.K.,G.M., J.K., N.V.A.M.K., and P.M.T.D. wrote the paper.

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

1To whom correspondence should be addressed. E-mail: [email protected].

This article contains supporting information online at www.pnas.org/cgi/content/full/0900130106/DCSupplemental.

www.pnas.org�cgi�doi�10.1073�pnas.0900130106 PNAS � July 21, 2009 � vol. 106 � no. 29 � 12195–12200

PHYS

IOLO

GY

Page 2: Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

OPC51 (13) and 2 novel nonpeptide V2R agonists VA88 andVA89 are shown in supporting information (SI) Fig. S1 A andtheir characteristics summarized in Table S1. In vivo, AVPbinding to the V2R triggers a cAMP signaling cascade, whichincreases AQP2 expression and induces its redistribution fromvesicles to the apical membrane. To characterize the activity ofthe nonpeptide V2R agonists, we used Madin-Darby Caninekidney (MDCK)-V2R cells, which show a subcellular localiza-tion and regulation of human V2R as anticipated to occur in vivo(14). Intracellular cAMP measurements revealed dose-responsecurves with similar maximal activities for dDAVP, VA88, andOPC51 (Fig. S1B, P � 0.05 at 1 �M agonist). However, the EC50values for VA88 (11.9 � 1.0 nM) and OPC51 (13.6 � 0.7 nM)were approximately 10-fold higher than for dDAVP (1.1 � 0.4nM). Nontransfected MDCK cells did not respond to any of theagonists (not shown).

To test whether the compounds increase AQP2 expression,polarized collecting duct (mpkCCD) cells, which show AVP-induced expression of endogenous AQP2 (15), were incubatedfor 4 days with 0.1 or 10 nM of the agonists. Subsequentimmunoblotting of the cell lysates revealed AQP2 expressionwith all 3 nonpeptide agonists and dDAVP at 10 nM, and withdDAVP and VA89 at 0.1 nM (Fig. S1C). Moreover, cell surfacebiotinylation experiments (Fig. S1D) and confocal laser scanningmicroscopy (CLSM) (Fig. S1E Left) revealed that AQP2, ex-pressed upon treatment with dDAVP or the nonpeptide ago-nists, localized in the apical membrane. With removal of theligands for 2 hours, AQP2 was internalized into vesicles (Fig. S1EMiddle), whereas subsequent reapplication of dDAVP or thenonpeptide agonists re-established the localization of AQP2 inthe apical membrane (Fig. S1E Right), indicating that all non-peptide agonists can also redistribute AQP2 from vesicles to theapical membrane. These data reveal that all 3 nonpeptideagonists activate human V2R with similar efficacy, although withlower potency, compared to dDAVP and that they regulateAQP2 expression and trafficking similar to dDAVP.

Many V2R Mutants in NDI Are Intrinsically Functional. With transientover-expression of V2R mutants in cells, a fraction of theER-retained V2R mutants localizes to the plasma membrane,which can then be used to determine the intrinsic functionality(i.e., ability to bind agonist and induce a cAMP response) ofER-retained V2R mutants. To test this for V2R mutants in NDI,GFP-tagged forms of WT-V2R, 6 V2R mutants associated withNDI (L44P, Y128S, I130F, S167T, Y280C, P322S), and thenonfunctional mutant V2R-S167L (8) were over-expressed inCOS cells and stimulated with 1 �M dDAVP or 1 �M VA88,VA89, or OPC51. With all compounds, cAMP levels weresignificantly increased for WT-V2R (5–6-fold; P � 0.01) and allV2R mutants (3–4-fold; P � 0.01), except V2R-S167L (shownfor dDAVP [Fig. 1A] and VA88 [Fig. 1B]). The variations inresponse between V2R mutants are likely intrinsic to the mutantproteins, because immunoblotting revealed similar expressionlevels for WT-V2R and its mutants (Fig. 1 A and B). These datashow that 6 out of 7 V2R mutants in NDI are intrinsicallyfunctional.

Subcellular Localization of V2R Mutants in Polarized MDCK Cells. Next,we determined the localization of V2R mutants in NDI. In vivo,the V2R is present in the basolateral membrane of polarizedcollecting duct cells and expressed at low levels. As theseconditions are nicely mimicked in stably transfected MDCK-V2R cells ((14) and Fig. S2 A), the V2R mutants were alsoexpressed in MDCK cells. In contrast to WT-V2R, the V2Rmutants -L44P, -I130F, -S167T, -Y128S, -Y280C, and -P322S aremisfolded and mainly retained in the ER, as they colocalize toa great extent with the ER-marker Protein Disulfide Isomerase(PDI; Fig. S2 A).

Glycosylated membrane proteins en route to the plasmamembrane change their high-mannose glycosylation, added tothem in the ER lumen, for complex glycosylation as they pass themedial-trans Golgi compartment (16). While high-mannoseglycosylated V2R are 60 to 62 kDa, mature receptors are 75 kDaand resistant to endoglycosidase H (endo H), while they remainsensitive to protein N-glycosidase F (PNGase F). WT-V2R inMDCK cells is expressed as a mature 75 kDa protein, becauseit is insensitive to endoH and reduced to its O-glycosylated formof 65 kDa with PNGase F digestion (Fig. 2A). In contrast, theV2R mutants are expressed as 62 kDa immature proteins thatare quantitatively equally sensitive to digestion with EndoH andPNGase F (Fig. 2 A), indicative of localization in the earlysecretory pathway.

To investigate further whether V2R mutants were essentiallyabsent from the plasma membrane, cells expressing WT-V2R orits L44P and P322S mutants were subjected to cell surfacebiotinylation. As shown in Fig. 2B, only mature WT-V2R wasdetected at the cell surface, whereas no immature receptors weredetected for either WT or the mutants.

To analyze the subcellular localization of the V2R mutants inmore detail, EM analysis was performed on empty MDCK cellsor cells expressing WT-V2R, V2R-P322S, -L44P, or -Y128S (Fig.S2B) and signals were quantified for WT-V2R and V2R-P322S(Table 1). Because the ER in MDCK cells is fragmented andtherefore not easy to discern, MDCK cells expressing WT-V2Ror V2R-P322S were double labeled for the ER-marker PDI andV2R (Fig. S2C). Consistent with LM studies (14), WT-V2R is

Fig. 1. Activation of wild-type and mutant V2R by peptidic and nonpeptidicagonists. (A) Intrinsic activity of V2R mutants in NDI. COS-M6 cells transientlyexpressing WT-V2R-GFP or its mutants were treated for 10 min. with 1 �MdDAVP or 1 �M VA88, (B) washed, lysed, and cAMP levels measured asdescribed above. Untreated samples were set to 1 and significantly-increasedcAMP levels (P � 0.05; student’s t test) compared to mock-transfected cells areindicated by an asterisk. In parallel, cell equivalents were analyzed by immu-noblotting. Protein masses are indicated in kDa.

Fig. 2. Subcellular localization of WT-V2R and V2R mutants in NDI. (A) MDCKWT-V2R, V2R-L44P, or -P322S protein samples were digested with Endogly-cosydase H or Protein N-glycosydase F. Subsequently, protein samples andtheir respective undigested controls were analyzed by immunoblotting. (B)MDCK cells expressing WT-V2R, V2R-L44P, or -P322S were subjected to cellsurface biotinylation and equalized total lysate and cell surface samples wereanalyzed by immunoblotting.

12196 � www.pnas.org�cgi�doi�10.1073�pnas.0900130106 Robben et al.

Page 3: Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

mainly present in the plasma membrane and endosomes/lysosomes. In contrast, V2R-P322S localizes predominantly tothe endoplasmic reticulum and Golgi region. However, low levelsof mutant receptors were also found in endosomes and in theplasma membrane (Table 1). These data reveal that in MDCKcells, V2R mutants associated with NDI are expressed in animmature form and are mainly retained in the ER and pre-Golgicompartments, although a small fraction of V2R mutant is foundin post-Golgi compartments, including the plasma membrane.

Nonpeptide Agonists, but Not dDAVP, Rescue V2R Mutants’ Function-ing. Because WT-V2R and its mutants had clearly differentsubcellular localizations and the mutants were essentially absentfrom the plasma membrane, we determined the ability of thenonpeptide agonists to generate a cAMP response in our V2Rmutant-expressing MDCK cell lines. Incubation with the Emaxconcentrations of dDAVP (100 nM) or the nonpeptide agonists(1 �M; Fig. 1B) for only 10 min induced an approx. 7–9-fold overbasal cAMP response in MDCK-V2R cells, whereas no effectwas observed in mock-transfected MDCK cells (Fig. 3). Admin-istration of 100 nM dDAVP did not induce a significant cAMPresponse in cells expressing the V2R mutants. However, incu-bation with 1 �M VA88, VA89, or OPC51, significantly in-creased cAMP in MDCK cells expressing V2R-L44P, -Y128S,-Y280C, -P322S, (Fig. 3; P � 0.01), -I130F, and -S167T (data notshown). Similar cAMP data were obtained when the cells wereincubated with the agonists while cell surface V2Rs were con-tinuously blocked with a peptidic V2R antagonist (not shown).These data indicated that nonpeptide agonists, but not dDAVP,can activate intracellularly-retained V2R mutants in NDI.

Nonpeptide Agonists Do Not Affect the Localization or Maturation ofV2R Mutants. Nonpeptide antagonists induce the maturation andtrafficking of V2R mutants from the ER to the basolateralmembrane (10–12, 17). Moreover, activation of WT-V2R byAVP induces its internalization to lysosomes (14). Because thenonpeptide agonists activate the ER-retained V2R mutants, weinvestigated their effects on the localization and maturation ofthese mutants. For this, WT and mutant V2R-expressing MDCKcells were treated for 16 h with 100 nM dDAVP, or 1 �M of thenonpeptide agonists or the CPAn SR1. Subsequent CLSManalysis revealed that none of the 3 nonpeptide agonists nordDAVP changed the ER localization of V2R-L44P or -Y128S(Fig. 4A and Fig. S3). In contrast and as reported (11), SR1clearly increased the basolateral membrane localization of thesemutants. However, WT-V2R was internalized from the basolat-eral membrane by dDAVP and the nonpeptide agonists, but notby SR1 (Fig. 4A Bottom and Fig. S3), which shows that theunchanged localization of the V2R mutants with the nonpeptideagonists was not due to the absence of agonist activity of thecompounds.

Because trafficking of V2R-GFP to the Golgi complex andbeyond, but also of V2R mutants when incubated with SR1,increases its mass from 62 to 75 kDa (11), cells treated as abovewere also subjected to immunoblot analyses. Consistent with ourCLSM data, none of the nonpeptide agonists induced matura-tion of the V2R mutants, as only the immature 62 kDa form wasdetected. In contrast, incubation with SR1 shifted the mass of theV2R mutants from 62 to 75 kDa (Fig. 4B; shown for V2R-L44P,-Y128S, and -P322S). For WT-V2R, treatment with VA88,VA89, or OPC51 did not significantly affect the level of matu-ration. In line with the agonistic activity, however, they causedan increase of a 47 kDa degradation product for V2R-GFP (Fig.4B), which was also observed in LLC-PK1-V2R cells treated withdDAVP (18). Together, these data indicate that activation bynonpeptide agonists does not change the localization or matu-ration of the V2R mutants.

Nonpeptide Agonist Effects on V2R Expression and Stability. Non-peptide antagonists stabilize the expression of intracellularly-retained V2R mutants in NDI and promote their maturation (10,11), whereas activation of WT-V2R by agonists induces itslysosomal degradation (14, 18). As the nonpeptide agonistsactivate the V2R mutants, but do not induce their maturation,we investigated their effects on V2R mutant expression andstability. Semiquantification of the signals in Fig. 4B revealedthat 16 h treatment with any of the 3 nonpeptide agonists did notsignificantly affect the expression levels of the 62 and 75 kDaforms of WT-V2R, but did increase the signal of its 47 kDadegradation product (P � 0.01). Moreover, treatment with theagonists did not significantly change the V2R-L44P or V2R-P322S expression levels (Fig. 4B), nor those of V2R-I130F,-S167T, and Y280C. Only for V2R-Y128S, all 3 compoundssignificantly (P � 0.03) increased the expression of this mutant(Fig. 4B).

The unchanged expression levels observed in Fig. 4B are abalance between V2R synthesis and degradation, which may

Fig. 3. Functionality of retained V2R mutants upon treatment with non-peptide agonists. Confluent WT-V2R, V2R-L44P, -Y128S, -Y280C, or -P322S cellswere left untreated or treated for 10 min with IBMX alone or in combinationwith 100 nM dDAVP, 1 �M VA88, VA89, or OPC51. Subsequently, cells werelysed and cAMP levels were measured. Untreated samples were set to 1, andrelative changes are shown. Triplicate samples were measured, and experi-ments were performed at least in 3-fold. Asterisks indicate significantly in-creased cAMP levels (P � 0.05) compared to IBMX alone.

Table 1. Electron microscopic analysis of cells expressingGFP-tagged WT-V2R or V2R-P322S

WT V2R V2R-P322S

ER 5 � 1.2 36 � 3.5Golgi region 2 � 0.3 21 � 1.8Golgi stack 2 � 0.4 9 � 1.4PM 53 � 1.8 10 � 1.1Endosomes 31 � 2.7 17 � 2.3Lysosomes 6 � 1.8 2 � 0.7Undefined 1 � 0.2 6 � 0.7

Numbers indicate the percentages of total labeling for the indicated com-partments. Two different grids were scanned resulting in 3,123 gold particlesfor WT V2R and 2,251 for V2R-P322S. ER, endoplasmic reticulum; PM, plasmamembrane.

Robben et al. PNAS � July 21, 2009 � vol. 106 � no. 29 � 12197

PHYS

IOLO

GY

Page 4: Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

both be increased with the agonists. As a lack of V2R mutantdegradation with nonpeptide agonists activation may be clini-cally beneficial, we tested whether the agonists increase degra-dation of the ER-retained mutants. Cells were treated with thenonpeptide agonists for 6 h in the absence or presence of theprotein synthesis inhibitor cycloheximide. Subsequent immuno-blotting revealed that with cycloheximide, immature WT-V2Rexpression markedly decreased, mature V2R expression slightlydecreased, and expression of the 47 kDa degradation productremained unchanged (Fig. 4C), indicating general degradation.Cotreatment with the agonists VA88, VA89, or OPC51, how-ever, further decreased the 62 and 75 kDa signals and increasedthe 47 kDa signal. Together with their localization in lateendosomes/lysosomes (Fig. 4A), this indicates that the nonpep-tide agonists induce the lysosomal degradation of WT V2R.

Analysis of the V2R mutants showed that also their generalexpression was reduced by cycloheximide. Coincubation withany of the agonists, however, did not lead to a further decreaseof their expression, nor did the 47 kDa signal appear (Fig. 4C).This indicated that, although the nonpeptide agonists activatethe retained V2R mutants, they do not induce their degradation.

Activation of Retained V2R Mutants by Nonpeptide Agonists Inducesthe Translocation of AQP2 to the Apical Membrane. To relieve NDIin patients, the cAMP response generated from V2R mutantsupon activation by nonpeptide agonists should be sufficient toactivate PKA and allow re-distribution of AQP2 to the plasmamembrane. To test this in vitro, MDCK-V2R-L44P cells wereinfected with recombinant AQP2 lentiviruses, treated with ago-nists for 2 h, and analyzed by immunocytochemistry. In line withthe NDI phenotype, AQP2 localizes to intracellular vesicles inMDCK-V2R-L44P cells treated with 100 nM dDAVP (Fig. 5A

Upper). In contrast, coincubation with the adenylate cyclaseactivator forskolin, or 1 �M of the nonpeptide agonists VA89(Fig. 5A), VA88, or OPC51 (Fig. S4) induced redistribution ofAQP2 from vesicles to the apical and, to some extent, basolateralregion of the cells. Note the absence of AQP2 redistribution inVA89-treated cells lacking V2R-L44P (indicated by arrows).

To determine whether V2R-L44P activation actually resultedin AQP2 redistribution into the apical membrane, MDCK-V2R-L44P and MDCK-V2R control cells were treated as above andsubjected to apical cell surface biotinylation. Consistent with theimmunocytochemical data, AQP2 immunoblotting of the apicalcell surface proteins from MDCK-V2R-L44P cells revealed asignificant increase in apical membrane AQP2 following treat-ment with forskolin or any of the nonpeptide agonists, but notwith dDAVP (Fig. 5 B and C). In contrast, in MDCK-V2R cells,a significantly increased apical membrane expression of AQP2was observed with all agonists, including dDAVP. These differ-ences were not due to variations in AQP2 expression levels,because these were similar (Fig. 5B, lysates). These data indicatethat cAMP generated by the intracellular activation of V2R-L44P is sufficient to induce redistribution of AQP2 from vesiclesto the apical membrane.

Fig. 4. Agonist action on the localization, maturation and stability of V2Rmutants. (A) Localization. Confluent MDCK WT-V2R or V2R-L44P cells wereleft untreated or treated with 100 nM of the peptide agonist dDAVP, 1 �M thenonpeptide agonists VA88, or the nonpeptide antagonist SR1 for 16 h fol-lowed by CLSM analysis. (B) V2R Maturation and stability. MDCK WT-V2R,V2R-L44P, -Y128S, or -P322S cells were left untreated, treated with the non-peptide agonists VA88, VA89 or OPC51, or treated with the nonpeptideantagonist SR1 for 16 h. Subsequently, receptor proteins were analyzed byimmunoblotting. (C) Stability. MDCK WT-V2R, V2R-L44P, -Y128S or -P322S cellswere left untreated, treated with 50 �M cycloheximide alone, or supple-mented with 1 �M VA88, VA89 or OCP51 for 6 h. Subsequently, receptorproteins were analyzed by immunoblotting.

Fig. 5. Agonist induced translocation of AQP2. MDCKII-V2R-L44P cells in-fected with an AQP2-lentivirus were incubated overnight with 50 �M indo-methacin. (A) Then, cells were supplemented with 100 nM dDAVP, 10 �Mforskolin (Forsk.), or 1 �M VA89 for 2 h. Next, the cells were subjected toimmunocytochemistry using rabbit anti-AQP2 antibodies. (B) In parallel, MD-CKII-V2R-L44P and WT-V2R cells were pretreated as under (A) and subjectedto cell surface biotinylation. The 15 �g samples of total cell lysates andequalized cell surface fractions (biotin) were analyzed by immunoblottingusing anti-AQP2 antibodies. (C) The cell-surface data from (B) were semiquan-tified using densitometry. Samples indicated with an asterisk are significantlydifferent from the control (P � 0.05, student’s t test).

12198 � www.pnas.org�cgi�doi�10.1073�pnas.0900130106 Robben et al.

Page 5: Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

DiscussionNonpeptide Agonists Activate Intracellularly-Retained Mutant V2RProteins in NDI. One known and 2 novel nonpeptide V2R agonistsshowed all of the characteristics of V2R agonists, because theyactivated V2R to generate cAMP, caused internalization ofV2R-GFP following binding, and induced AQP2 synthesis andredistribution from vesicles to the apical membrane (Fig. S1, Fig.4 and 5).

The general concept that activation of GPCRs activated bycell-permeable ligands needs to occur at the plasma membraneis challenged by our data with these nonpeptide V2R agonists:While 6 out of 7 V2R mutants tested were intrinsically functionalreceptors (Fig. 1 A and B), MDCK cells stably expressing these6 functional V2R mutants were unresponsive to the cell-impermeable dDAVP (Fig. 3). Moreover, all V2R mutantstested mainly localize to the ER-Golgi complex (Fig. S2; Table1). Apparently, the low levels of V2R mutant detected in theplasma membrane (Table 1) are too low to induce a detectableresponse. In contrast to dDAVP, however, all 3 nonpeptideagonists induced a significant cAMP response with MDCK cellsexpressing the 6 functional V2R mutants, (Fig. 3), even whenblocking the plasma membrane V2R. Because dDAVP andnonpeptide agonists concentrations were used at equimolarconcentrations, the difference in effect is not due to higheractivity of the nonpeptide agonists, but most likely due to ahigher propensity of the nonpeptide agonists to penetrate thecell membrane. As such, our data reveal that a GPCR thatnormally functions at the cell membrane can generate its cor-responding signaling cascade from an intracellular location.Moreover, nonpeptide agonists can activate intrinsically-functional but disease-causing V2R mutants from these intra-cellular sites.

While the functional data above are clear, the lack of signalingfrom plasma membrane V2R-P322S is surprising. The mostlikely explanation is the low number of mutant receptors in theplasma membrane. Indeed, while 10% localizes in the plasmamembrane, the total abundance of V2R-P322S in our EManalysis was considerably less than of WT-V2R (Table 1), whichcan be attributed to their reduced stability (8). Alternatively,plasma membrane V2R-P322S may signal less efficient thanWT-V2R.

Recent observations that the �2-adrenergic receptor assem-bles with G�s and adenylate cyclase in the ER (19, 20) and thatthe naturally cell permeable agonist estrogen induces signalingfrom the ER-localized GPR30 (21, 22) indicate that nonpeptideagonists may activate the V2R mutants to generate cAMP whilelocated in the ER. However, considering the expression of someV2R mutants beyond the ER-Golgi complex, this remains to beestablished.

Intracellular Activation of V2R Mutants in NDI Does Not Change TheirLocalization or Stability. Even after prolonged incubation, none ofthe nonpeptide agonists induce any appreciable shift in local-ization of the V2R mutants, nor change their state of maturation(Fig. 4). In contrast, the nonpeptide V2R antagonist SR1 clearlyincreased V2R mutant expression at the cell surface and inducedmaturation (Fig. 4)(10–12, 17). The absence of translocation ofV2R mutants to the cell surface is further supported by the factthat cAMP responses are obtained within 10 min followingagonists addition, whereas it takes V2R antagonists 4–8 h toinsert V2R mutants in the plasma membrane (11, 23). Our dataare different from those of Petaja-Repo et al. (24), who foundthat nonpeptide agonists were able to stabilize the expressionand maturation of ER-retained wild-type �-opioid receptor(DOR). This indicates that nonpeptide agonist-induced matu-ration and rescue of cell surface expression is receptor-, muta-tion- and/or agonist-specific. Since in our study all V2R mutants

tested remained unaffected in their localization and maturation,a difference in receptor is most the most likely explanation.

At present, it is unknown whether intracellular receptors aredesensitized similar to their cell surface counterparts uponactivation. While agonist-induced lysosomal targeting and deg-radation reveals that WT-V2R is down-regulated in MDCKcells, nonpeptide agonists did not increase degradation of theintracellularly-retained V2R mutants in NDI (Fig. 4). It remainsto be established which step of the normal desensitization stepsis abrogated with the mutant V2R proteins and whether this isgenerally observed for intracellular GPCRs.

High Mannose-Glycosylated V2R Mutants Are Retained in, but NotConfined to, the ER-Early-Golgi Compartment. A paradigm is thatintegral membrane proteins on their way to the plasma mem-brane exchange their high-mannose (ER) glycosylation for com-plex-glycosylated moieties when passing through the Golgi com-plex. However, although all intracellularly-retained V2Rmutants in NDI localize to a great extent in the early secretorypathway (Table 1), are only high-mannose glycosylated, and areequally sensitive to endoH and PNGaseF digestion (Fig. 2 A), ourEM analysis revealed that a considerable pool of V2R-P322Slocalized to organelles beyond this stage. In transient expressionstudies (e.g., Fig. 1), non- or high-mannose glycosylated mem-brane proteins beyond the Golgi complex are commonly found,which is attributed to the inability of the ER quality controlsystem to retain the high number of membrane proteins synthe-sized. This, however, does not explain the observation in ourstably-transfected MDCK cells, because WTV2R is fully ma-tured (Fig. 2) and expressed at a higher level than the V2Rmutants.

Interestingly, of several other membrane proteins, includingthe cystic fibrosis transmembrane conductance regulator(CFTR), protein phosphatase CD45, and the epithelial sodiumchannel (ENaC), post-Golgi expression of immature forms havebeen found for endogenously expressed proteins or in stably-transfected cells (25–27). Possibly, low levels of proteins bypassthe Golgi and may directly be delivered to post-Golgi compart-ments, or all proteins traffic to the plasma membrane via theconventional pathway, but a population of these proteins areresistant to posttranslational processing (26). In line with this,misfolding of the V2R mutants may affect proper exchange ofsugar moieties in the Golgi complex, or a small fraction maybypass this organelle in our transfected MDCK cells. Our datado show, however, that expression of only the immature glyco-sylated form of a mutant membrane protein is an indicator ofretention in the ER or early Golgi compartment, but does notnecessarily exclude that a small fraction localizes beyond theGolgi complex. While it remains unknown whether GPCRmutants traffic beyond the Golgi complex in humans, traffickingof immature membrane proteins beyond the Golgi complex hasbeen observed for endogenously expressed membrane proteinsand may thus also apply to V2R mutants in vivo.

Potential Implications of the Use of V2R Nonpeptide Agonists for NDIPatients. The lack of V2R activation in NDI patients with V2Rmutations causes reduces AQP2 expression levels and abrogatesAQP2 translocation to the apical membrane in renal collectingduct principal cells (28). Our data indicate that the nonpeptideV2R agonists constitute highly promising therapeutics for thetreatment of NDI resulting from intracellularly-retained V2Rmutants. First, the nonpeptide agonists are unlikely to causemajor side effects, because they are highly selective for V2R (29).Nevertheless, monitoring of the V2R-specific extra-renal releaseof coagulation factors will be essential in patients. Secondly,these agonists induce a cAMP response mediated by ER-retained V2R mutants (Fig. 3) and are able to induce thecAMP-dependent expression of endogenous AQP2 (Fig. 1C).

Robben et al. PNAS � July 21, 2009 � vol. 106 � no. 29 � 12199

PHYS

IOLO

GY

Page 6: Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

Thirdly, the cAMP response generated by the intracellularlyactivated V2R mutants appears sufficiently strong to mediate thecritical step in renal water reabsorption, the trafficking of AQP2proteins from vesicles to the plasma membrane (Fig. 5).

While administration of the V1R antagonist SR49059 topatients was the first in vivo proof of principle that nonpeptideantagonists can relieve NDI in patients (17), the direct activationof functional ER-retained V2R mutants observed here indicatesthat treatment with nonpeptide V2R agonists is likely superiorover nonpeptide antagonists because rescue of cell surfaceexpression of the V2R mutants and subsequent displacement ofthe antagonists by endogenous AVP is circumvented. Consid-ering the low stability of ER-retained folding mutants (8), thelack of increased V2R mutant degradation upon activation bynonpeptide agonists provides an additional advantage for theirtherapeutic use. Determination of their in vivo therapeutic valuefor NDI, however, awaits approval for their use and testing inpatients, because mice lacking functional V2Rs die soon afterbirth (30) and OPC51 (803), VA (9990)88, and VA(9990)89-likecompounds are still in early phase clinical trials. However, theobserved decrease in urine output with OPC51803 in Brattle-boro rats, which have functional V2Rs but lack AVP, alreadyreveals the in vivo activity of nonpeptide V2R agonists on anormal functioning renal concentrating mechanism (29) andunderscores the potential of nonpeptide V2R agonists as ther-apeutic agents for NDI patients due to retained, but intrinsically-functional, V2R mutants.

In conclusion, our data show that a GPCR, which normallyfunctions in the plasma membrane, can generate a signalingcascade from an intracellular location to induce the receptor’sphysiological response. Moreover, the activation of intracellularV2R mutants by our nonpeptide agonists does not lead toincreased degradation. As approximately 40% of GPCR mutantsin diseases are misfolded and retained intracellularly, our dataindicate that nonpeptide agonists are ideal therapeutics to treatpatients suffering from one of the many ‘‘conformational dis-

eases’’ due to misfolded and retained GPCRs in general, andNDI due to misfolded V2R mutants in particular.

Materials and MethodsLigands. The nonpeptide V2R agonists VA999088 and VA999089 were kindlyprovided by Dr. Haigh (Vantia Ltd.), OPC51803 was kindly provided by Dr.Komuro (Otsuka Pharmaceutical Co., Tokushima, Japan). The V2R nonpeptideantagonist SR121463B was provided by Dr. Serradeil-Le Gal (Sanofi Syn-thelabo Recherche, Toulouse, France). dDAVP and the peptidic antagonist[Adamantaneacetyl1, O-Et-D-Tyr2, Val4, Aminobutyryl6, Arg8,9]-Vasopressinwere from Sigma. Their abbreviations and relevant characteristics are sum-marized in Table S1 and fully described in SI Materials and Methods.

Expression Constructs. With site-directed mutagenesis (Stratagene), the NDI-related mutations Y128S, Y280C, and P322S were introduced into the hV2RcDNA and the mutation-containing fragments were subcloned into the cor-responding sites of pEGFP-N1-V2R as described in SI Materials and Methods.

To generate a recombinant lentivirus encoding AQP2, the AQP2 cDNA wascloned into pLV-PGK (Tronolab) to yield pLV-PGK-AQP2. Viruses were gener-ated in HEK293 cells and titrated as described in SI Materials and Methods.

Cell Culture and Biochemical Assays. MDCK cell lines stably expressing GFP-tagged WT-V2R and mutants in NDI were generated as described (8, 14) andculture and transfection of COS cells was performed as described (8). cAMPmeasurements, cell surface biotinylation assays, Western blotting, and immu-nocytochemistry were performed as described (8, 14). These assays and theantibodies used are described in more detail in SI Materials and Methodssection.

ACKNOWLEDGMENTS. We thank Koji Komuro (Otsuka Pharmaceutical Co.,Tokushima, Japan) for kindly supplying OPC51. We thank Dr. Robert Haigh(Vantia Ltd., Chilworth, U.K.) for kindly supplying VA88 and VA89 and Dr.Claudine Serradeil-Le Gal (Sanofi Synthelabo Recherche, Toulouse, France) forkindly supplying SR1. We thank Rene Scriwanek and Marc van Peski forpreparation of the electron micrographs. J.K. and P.M.T.D. are recipients ofVICI grants 918.56.611 and 865.07.002 of the Netherlands Organization forScientific Research. We acknowledge the Dutch Kidney Foundation (PC 104,C06.2164), RUNMC and Netherlands Organization for Scientific Research(865.07.002) and Coordination Theme 1 (Health) of the European Communi-ty’s 7th Framework Program (HEALTH-F2–2007-201590, entitled EUNEFRON).

1. Illing ME, Rajan RS, Bence NF, Kopito RR (2002) A rhodopsin mutant linked to auto-somal dominant retinitis pigmentosa is prone to aggregate and interacts with theubiquitin proteasome system. J Biol Chem 277:34150–34160.

2. De Roux, N., et al. (1997) A family with hypogonadotropic hypogonadism and muta-tions in the gonadotropin-releasing hormone receptor. N Engl J Med 337:1597–1602.

3. Vaisse C, Clement K, Guy-Grand B, Froguel P (1998) A frameshift mutation in humanMC4R is associated with a dominant form of obesity. Nat Genet 20:114.

4. Biebermann H, Gruters A, Schoneberg T, Gudermann T (1997) Congenital hypothyroidismcaused by mutations in the thyrotropin receptor gene. N Engl J Med 336:1390–1391.

5. Van den Ouweland AMW, et al. (1992) Mutations in the Vasopressin Type-2 ReceptorGene (Avpr2) Associated with Nephrogenic Diabetes-Insipidus. Nat Genet 2:99–102.

6. Robben JH, Knoers N VAM, Deen PMT (2006) Cell biological aspects of the vasopressintype-2 receptor and aquaporin 2 water channel in nephrogenic diabetes insipidus.Am J Physiol Renal Physiol 291:F257–F270.

7. Vembar SS, Brodsky JL (2008) One step at a time: Endoplasmic reticulum-associateddegradation. Nat Rev Mol Cell Biol 9:944–957.

8. Robben JH, Knoers . . . . , Deen PMT (2005) Characterization of vasopressin V2 receptormutants in nephrogenic diabetes insipidus in a polarized cell model. Am J Physiol RenalPhysiol 289:F265–F272.

9. Bernier V, et al. (2004) Functional Rescue of the Constitutively Internalized V2 Vaso-pressin Receptor Mutant R137H by the Pharmacological Chaperone Action of SR49059.Mol Endocrinol 18:2074–2084.

10. Morello JP, et al. (2000) Pharmacological chaperones rescue cell-surface expression andfunction of misfolded V2 vasopressin receptor mutants. J Clin Invest 105:887–895.

11. Robben JH, Sze M, Knoers N, Deen PMT (2007) Functional rescue of vasopressin V2receptor mutants in MDCK cells by pharmacochaperones: Relevance to therapy ofNephrogenic Diabetes Insipidus. Am J Physiol 292:F253–F260.

12. Tan CM, Nickols HH, Limbird LE (2003) Appropriate polarization following pharmaco-logical rescue of V2 vasopressin receptors encoded by X-linked nephrogenic diabetesinsipidus alleles involves a conformation of the receptor that also attains matureglycosylation. J Biol Chem 278:35678–35686.

13. Nakamura S, et al. (2000) Characterization of a novel nonpeptide vasopressin V2-agonist, OPC-51803, in cells transfected human vasopressin receptor subtypes. Br JPharmacol 129:1700–1706.

14. Robben JH, Knoers NVAM, Deen PMT (2004) Regulation of the vasopressin V2 receptorby vasopressin in polarized renal collecting duct cells. Mol Biol Cell 15:5693–5699.

15. Hasler U, et al. (2002) Long-term regulation of aquaporin-2 expression in vasopressin-responsive renal collecting duct principal cells. J Biol Chem 277:10379–10386.

16. Sadeghi HM, Innamorati G, Birnbaumer M (1997) Maturation of receptor proteins ineukaryotic expression systems. J Recept Signal Transduct Res 17:433–445.

17. Bernier V, et al. (2006) Pharmacologic chaperones as a potential treatment for X-linkednephrogenic diabetes insipidus. J Am Soc Nephrol 17:232–243.

18. Bouley R, et al. (2005) Downregulation of the vasopressin type 2 receptor aftervasopressin-induced internalization: Involvement of a lysosomal degradation path-way. Am J Physiol 288:C1390–C1401.

19. Dupre DJ et al. (2006) Seven transmembrane receptor core signaling complexes areassembled prior to plasma membrane trafficking. J Biol Chem 281:34561–34573.

20. Dupre DJ, Baragli A, Rebois RV, Ethier N, Hebert TE (2007) Signalling complexesassociated with adenylyl cyclase II are assembled during their biosynthesis. Cell Signal19:481–489.

21. Revankar CM, Cimino DF, Sklar LA, Arterburn JB, Prossnitz ER (2005) A transmembraneintracellular estrogen receptor mediates rapid cell signaling. Science 307:1625–1630.

22. Filardo EJ, Quinn JA, Frackelton AR, Jr, Bland KI (2002) Estrogen action via the Gprotein-coupled receptor, GPR30: Stimulation of adenylyl cyclase and cAMP-mediatedattenuation of the epidermal growth factor receptor-to-MAPK signaling axis. MolEndocrinol 16:70–84.

23. Wuller S, et al. (2004) Pharmacochaperones post-translationally enhance cell surfaceexpression by increasing conformational stability of wild-type and mutant vasopressinV2 receptors. J Biol Chem 279:47254–47263.

24. Petaja-Repo UE, et al. (2002) Ligands act as pharmacological chaperones and increasethe efficiency of delta opioid receptor maturation. EMBO J 21:1628–1637.

25. Planes C, et al. (2002) Hypoxia and beta 2-agonists regulate cell surface expression ofthe epithelial sodium channel in native alveolar epithelial cells. J Biol Chem 277:47318–47324.

26. Hughey RP, Bruns JB, Kinlough CL, Kleyman TR (2004) Distinct pools of epithelialsodium channels are expressed at the plasma membrane. J Biol Chem 279:48491–48494.

27. Baldwin TA, Ostergaard HL (2002) The protein-tyrosine phosphatase CD45 reaches the cellsurface via golgi-dependent and -independent pathways. J Biol Chem 277:50333–50340.

28. Kanno K, et al. (1995) Urinary excretion of aquaporin-2 in patients with diabetesinsipidus. N Engl J Med 332:1540–1545.

29. Nakamura S, et al. (2000) Antidiuretic effects of a nonpeptide vasopressin V2-receptoragonist, OPC-51803, administered orally to rats. J Pharmacol Exp Ther 295:1005–1011.

30. Yun J, et al. (2000) Generation and phenotype of mice harboring a nonsense mutationin the V2 vasopressin receptor gene. J Clin Invest 106:1361–1371.

12200 � www.pnas.org�cgi�doi�10.1073�pnas.0900130106 Robben et al.

Page 7: Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

Supporting InformationRobben et al. 10.1073/pnas.0900130106SI Materials and MethodsLigands. The nonpeptide V2R agonists VA999088 and VA999089(patent WO01/29005) were kindly provided by Dr. Haigh (Van-tia Ltd.), while OPC51803 (1) was kindly provided by Dr.Komuro (Otsuka Pharmaceutical Co. Ltd.). The V2R non-peptide antagonist SR121463B (2) was provided by Dr. Serra-deil-Le Gal (Sanofi Synthelabo Recherche). All compoundswere dissolved in dimethyl sulfoxide as 0.01 M stock solutionsand diluted in culture medium as indicated. dDAVP and thepeptidic antagonist [Adamantaneacetyl1, O-Et-D-Tyr2, Val4,Aminobutyryl6, Arg8,9]-Vasopressin were from Sigma. For read-ing convenience, VA999088, VA999089, OPC51803, andSR121463B are abbreviated as VA88, VA89, OPC51 and SR1,respectively, which is, together with relevant characteristics,summarized in the Table S1.

Expression Constructs. Expression constructs encoding the WTV2R or the NDI causing mutants –L44P, -I130F, -S167T, orS167L fused at their C-termini to enhanced green fluorescentprotein were as described (3). With site-directed mutagenesis(Stratagene), additional NDI-related mutations were introducedinto the human V2R cDNA sequence, using the primers GGT-GGGCATGTACGCGTCCTCCTCCATGATCCTGG (Y128S),GTGGTCGTCTGTGTGCTGTGCTGGGCCCCCTTCTTCC(Y280C), and CCTCAACAGCTGCACGAATTCCTGGATC-TATGC (P322S), and their complementary antisense primers on apEGFP-N1-V2R template. After digesting correct clones withPstI/HindIII, the mutation-containing fragments were subclonedinto the corresponding sites of pEGFP-N1-V2R. Sequence analysisconfirmed that only the desired mutations were introduced.

Recombinant Lentivirus. To generate a recombinant lentivirusencoding AQP2, the AQP2 cDNA was cut from pBSII-KS(�)AQP2 using SpeI and XhoI, and cloned into the correspond-ing sites of pRRL-CMV (4) (Tronolab) to yield pRRL-CMV-AQP2. Subsequently, pRRL-CMV-AQP2 was digested withMluI, blunted, and cut with NheI. The 994 bp insert containingthe entire AQP2 cDNA was cloned into the NheI, and theblunted AgeI site of pLV-PGK (4) (Tronolab) to yield pLV-PGK-AQP2. Recombinant lentiviruses were generated by co-transfecting pLV-PGK-AQP2, with the pMD2G, pMDLg, andpRSV-REV (4) (Tronolab) plasmids into HEK293T cells usingcalcium phosphate precipitation. Two and 3 days posttransfec-tion, recombinant viruses were collected from the supernatantby centrifugal concentration and subsequently titrated using anELISA based on measurement of viral DNA. 1 ng viral DNA on2,500 cells corresponds to a multiplicity of infection of 1.

Cell Culture, Transfection and Infection. MpkCCD cells were cul-tured as described (5). MDCK type II cells lacking endogenousV2R expression were kindly provided by Dr. Oksche (For-schungsinstitut fur Molekulare Pharmakologie, Berlin, Ger-many). MDCK-II cell maintenance, transfection and isolation ofclones were done as described for MDCK type I cells (6).Experiments were performed at least 2 times on 2 clones pertransfectant. Maintenance and transient transfection of COS-M6cells were done as described (3). For transient expression ofAQP2 in MDCK cells, 70–80% confluent cells were trypsinizedand mixed with the PGK-AQP2 lentivirus at a multiplicity ofinfection of 2. Cells were then seeded on semipermeable filtersat a density of 3 � 105 cells/cm2 in the presence of the lentivirusand incubated for 16 h at 37 °C. Subsequently, the medium wasreplaced with normal culture medium and the cells were grownfor an additional 2 days, treated as indicated, and subjected toapical cell surface biotinylation or immunocytochemistry asdescribed (6, 7).

Electron Microscopy. For immuno-electron microscopy, controland transfected MDCK cells were fixed in 4% formaldehyde andprepared for ultrathin cryosectioning as described (8). Immu-nogold labeling using anti-GFP antibodies (ABCAM, Cam-bridge, MA) was performed as described (9). NontransfectedMDCK cells did not show any significant label with the antiGFPantibody. For quantification, samples were selected for goodoverall morphology, after which counting was performed byrandomly scanning the sample. Each encountered gold particlethat was no further than 20 nm located from a membrane wasattributed to the membrane to which it was associated.

Immunoblotting and Immunocytochemistry. Polyacrylamide gelelectrophoresis, Western blotting, immunodetection, and semi-quantification of signals were performed as described (6, 10).Immunocytochemistry, CLSM, and data quantification wereperformed as described (3). Rabbit-anti GFP (11) and rabbit-anti AQP2 (12) antibodies were obtained as described.

cAMP Measurements. Measurement of intracellular cAMP intransiently-transfected COS-M6 cells or MDCK cells stably-expressing V2R (mutants) was performed as described (3).Triplicate samples were measured, and experiments were per-formed at least in 3-fold. Data were analyzed using GraphpadPrism 4 (Graphpad Software Inc.).

Statistical Analysis. Statistical analysis was performed using stu-dent’s t-test. P-values � 0.05 were considered significant.

1. Nakamura S, et al. (2000) Characterization of a novel nonpeptide vasopressin V2-agonist, OPC-51803, in cells transfected human vasopressin receptor subtypes. Br JPharmacol 129:1700–1706.

2. Serradeil-Le Gal C, et al. (2002) Nonpeptide vasopressin receptor antagonists: Devel-opment of selective and orally active V1a, V2 and V1b receptor ligands. Prog Brain Res139:197–210.

3. Robben JH, Knoers NVAM, Deen PMT (2005) Characterization of vasopressin V2receptor mutants in nephrogenic diabetes insipidus in a polarized cell model. Am JPhysiol 289:F265–F272.

4. Mangeat B, Trono B (2005) Lentiviral vectors and antiretroviral intrinsic immunity. HumGene Ther 16:913–920.

5. Li Y, Shaw S, Kamsteeg EJ, Vanderwalle A, Deen PM (2006) Development of lithium-induced nephrogenic diabetes insipidus is dissociated from adenylyl cyclase activity.J Am Soc Nephrol 17:1063–1072.

6. Deen PMT, et al. (2002) Aquaporin-2: COOH terminus is necessary but not sufficient forrouting to the apical membrane. Am J Physiol 282:F330–F340.

7. Kamsteeg EJ, et al. (2006) Short-chain ubiquitination mediates the regulated endocy-tosis of the aquaporin-2 water channel. Proc Natl Acad Sci USA 103:18344–18349.

8. Slot JW, Geuze HJ (2007) Cryosectioning and immunolabeling. Nat Protocols 2:2480–2491.

9. Oueslati M, et al. (2007) Rescue of a nephrogenic diabetes insipidus-causing vaospres-sin V2 receptor mutant by cell-penetrating peptides. J Biol Chem 282:20676–20685.

10. Robben JH, Knoers NVAM, Deen PMT (2004) Regulation of the vasopressin V2 receptorby vasopressin in polarized renal collecting duct cells. Mol Biol Cell 15:5693–5699.

11. Cuppen E, et al. (2000) The zyxin-related protein TRIP6 interacts with PDZ motifs in theadaptor protein RIL and the protein tyrosine phosphatase PTP-BL. European J Cell Biol79:283–293.

12. Deen PMT, et al. (1994) Requirement of human renal water channel aquaporin-2 forvasopressin-dependent concentration of urine. Science 264:92–95.

Robben et al. www.pnas.org/cgi/content/short/0900130106 1 of 6

Page 8: Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

Fig. S1. Characterization of V2R agonists. (A) Chemical structures of (1) VA88, (2) VA89 and (3) OPC51. (B) Dose-response curves of dDAVP, VA88, and OPC51on V2R. MDCK WT-V2R cells were treated with increasing amounts of dDAVP (squares), VA88 (triangles), or OPC51 (diamonds) for 10 min in the presence of 1mM IBMX. Subsequently, cAMP levels were measured using a fluoremetric competition immunoassay kit. (C) V2R agonist-induced expression of AQP2. MpkCCDcells were left untreated (C) or treated with 10 or 0.1 nM of the nonpeptide agonists or dDAVP for 4 days. Next, cells were lysed and immunoblotted for AQP2.Protein masses are indicated in kDa. (D) Apical membrane insertion of AQP2 by V2R agonists. MpkCCD cells were left untreated (C), or treated for 4 days with1 nM dDAVP or the nonpeptide agonists. Next, cells were subjected to cell surface biotinylation and equalized total lysates and cell surface fractions wereanalyzed by immunblotting using AQP2 antibodies. (E) In parallel, cells were treated with agonists as under (D) (control), incubated without ligand for 2 h (noligand), followed by incubation with 100 nM dDAVP or 1 �M of the nonpeptide agonists for 1 h (1 h agonist). After each incubation point, cells were fixed, andimmunocytochemically stained for AQP2. Representative cross-sections of the cells are shown.

Robben et al. www.pnas.org/cgi/content/short/0900130106 2 of 6

Page 9: Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

Fig. S2. Intracellular retention of V2R mutants in NDI. (A) MDCK cells expressing the GFP-tagged WT-V2R, V2R-L44P, or V2R-Y128S were grown to confluenceon filters. Subsequently, the cells were fixed, subjected to immunocytochemistry using anti-PDI antibodies to stain the endoplasmic reticulum (Left), and analyzedby CLSM. (B) MDCK cells expressing GFP-tagged WT-V2R (A), V2R-P322S (B), V2R-L44P (C), or V2R-Y128S (D) were processed for cryo-immunogold labeling usinganti-GFP antibodies (10 nm gold particles). In WT-V2R expressing cells, a considerable amount of label is found in the microvilli (MV), whereas the nuclearenvelope (NE) and Golgi (G) region are relatively devoid of labeling. Cells expressing V2R mutants display less label in the microvilli, whereas relatively more isseen in the NE and Golgi region. (Scale bar, 200 nm.) n � nucleus. (C) Electron micrographs showing the occurrence of GFP-tagged WT-V2R (A) and V2R-P322S(B) in the endoplasmic reticulum (ER). Transfected MDCK cells were double-labeled for the ER marker PDI (10 nm gold particles) and GFP (15 nm gold). The labelfor PDI outlines the rather fragmented ER membranes in MDCK cells. Arrows point to ER cisternae containing GFP-tagged constructs of V2R-WT (A) or V2R-P322S(B). E � endosome; n � nucleus; NE � nuclear envelope; MV � microvilli. (Scale bars, 200 nm). For this labeling, mouse monoclonal antibodies against PDI(Stressgen) and rabbit anti-mouse antibody (Dakopatts) were used.

Robben et al. www.pnas.org/cgi/content/short/0900130106 3 of 6

Page 10: Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

Fig. S3. Peptide and nonpeptide agonist action on the localization, maturation, and stability of V2R mutants in NDI. MDCK cells expressing GFP-taggedV2R-Y128S were grown to confluence on filters and subsequently left untreated,or treated with 100 nM of the peptide agonist dDAVP, 1 �M the nonpeptideagonists VA88, VA89, OPC51 or the nonpeptide antagonist SR1 for 16 h. Subsequently, cells were fixed and analyzed by CLSM.

Robben et al. www.pnas.org/cgi/content/short/0900130106 4 of 6

Page 11: Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

Fig. S4. Agonist induced translocation of AQP2. MDCKII-V2R-L44P cells infected with a lentivirus containing an AQP2 expression cassette were incubatedovernight with 50 �M indomethacin. (A) Then, cells were kept on indomethacin supplemented with 100 nM dDAVP, 10 �M forskolin (Forsk.), 1 �M VA88, orOPC51 for 2 h. Next, the cells were fixed, permeablilized and subjected to immunocytochemistry using rabbit anti-AQP2 antibodies and subjected to CLSM.

Robben et al. www.pnas.org/cgi/content/short/0900130106 5 of 6

Page 12: Intracellular activation of vasopressin V2 receptor ... · Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists Joris

Table S1. Overview of the used compounds

Compound Abbreviation (Ant)agonist (Non-)peptide

1-desamino-8-D-AVP dDAVP Agonist PeptideVA999088 VA88 Agonist Non-peptideVA999089 VA89 Agonist Non-peptideOPC51803 OPC51 Agonist Non-peptideSR121463B SR1 Antagonist Non-peptide

The abbreviated name, ligand type, and (non-)peptidic nature of thecompounds tested are summarized

Robben et al. www.pnas.org/cgi/content/short/0900130106 6 of 6