<<

RAMP3 determines rapid recycling of atypical receptor-3 for guided angiogenesis

Duncan I. Mackiea, Natalie R. Nielsena, Matthew Harrisb, Smriti Singha, Reema B. Davisa, Danica Dya, Graham Laddsb, and Kathleen M. Carona,1

aDepartment of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and bDepartment of Pharmacology, University of Cambridge, CB2 1PD Cambridge, United Kingdom

Edited by Robert J. Lefkowitz, Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC, and approved October 21, 2019 (received for review April 2, 2019) Receptor-activity–modifying (RAMPs) are single of RAMPs with most GPCR families suggests that they may have transmembrane-spanning proteins which serve as molecular chap- expanded interacting partners (12, 13). erones and allosteric modulators of G-–coupled receptors The subfamily is composed of 24 different (GPCRs) and their signaling pathways. Although RAMPs have been GPCRs that each bind a diverse array of chemokine and previously studied in the context of their effects on Family B GPCRs, ligands, leading to a wide variety of signaling events and physi- the coevolution of RAMPs with many GPCR families suggests an ological functions, ranging from innate immunity, developmental expanded repertoire of potential interactions. Using biolumines- , and HIV infection to cancer metastasis (14–16). The cence resonance energy transfer-based and cell-surface expression redundancy of ligands binding to multiple chemokine receptors approaches, we comprehensively screen for RAMP interactions underscores the importance, and also the complexity, of chemo- within the chemokine receptor family and identify robust interac- kine signaling. As one example, the establishment of extracellular tions between RAMPs and nearly all chemokine receptors. Most chemokine gradients for directed cell migration relies on the notably, we identify robust RAMP interaction with atypical chemo- concerted, spatiotemporal functions of both typical signaling che- kine receptors (ACKRs), which function to establish chemotactic gra- mokine receptors with atypical chemokine receptors (ACKRs), dients for directed cell migration. Specifically, RAMP3 association with sometimes referred to as “decoy” receptors (17). Unlike typical atypical chemokine receptor 3 (ACKR3) diminishes

chemokine receptors which elicit a downstream signaling pathway CELL BIOLOGY (AM) ligand availability without changing G-protein coupling. in response to ligand, ACKRs do not signal through G proteins Instead, RAMP3 is required for the rapid recycling of ACKR3 to but rather bind and degrade ligands rapidly from the extracellular the plasma membrane through Rab4-positive vesicles following either AM or SDF-1/CXCL12 binding, thereby enabling formation milieu. Considering this high level of complexity in ligand binding of dynamic spatiotemporal chemotactic gradients. Consequently, and signaling, we hypothesized that RAMPs may interact with genetic deletion of either ACKR3 or RAMP3 in mice abolishes chemokine receptors to modulate their complex signaling, traf- directed cell migration of retinal angiogenesis. Thus, RAMP asso- ficking, and physiological properties. Here, we developed a mul- ciation with chemokine receptor family members represents a timodal screening platform which identified robust interactions molecular interaction to control receptor signaling and trafficking between RAMPs and the family of chemokine receptors. We properties. further found that RAMPs play essential roles in determining the internalization, trafficking, and recycling of ACKR3, ultimately receptor-activity–modifying proteins | G-protein–coupled receptors | influencing guided angiogenesis in the postnatal retina. chemokine receptors | guided cell migration | endosomal sorting Significance eceptor-activity–modifying proteins (RAMPs) are a family Rof single-pass transmembrane proteins with 3 mammalian G-protein–coupled receptors (GPCRs) exist within multi-protein members: RAMP1, RAMP2, and RAMP3, each encoded by a complexes on the surface of cells in order to respond to a wide distinct and corresponding (1–3). The RAMPs were first variety of extracellular stimuli such as , mi- identified by virtue of their requirement for promoting the for- gratory cues, hormones, , and odors. In this study, we ward translocation of the -receptor–like receptor (CLR) discover and characterize an expanded repertoire of GPCRs that from the cytoplasm to the plasma membrane (3). Moreover, their interact with receptor-activity–modifying proteins (RAMPs)—a association with this receptor imparts ligand-binding specificity, class of proteins that can modulate the type and consequences such that a RAMP1-CLR oligomer preferentially binds the neu- of extracellular signals to GPCRs. Specifically, we find that RAMP ropeptide, calcitonin-gene–related peptide (CGRP), while the interaction with chemokine GPCRs is essential for enabling these RAMP2-CLR or RAMP3-CLR oligomers display higher affinity receptors to bind and degrade extracellular migratory cues and for a related vasodilatory peptide, adrenomedullin (AM). Recent thereby establish gradients for directed cellular migration. In the cryogenic electron microscopy resolution of the molecular inter- absence of these critical proteins, the process of blood vessel action between RAMP1 and CLR exemplifies how RAMPs im- sprouting within the postnatal retina is dysfunctional. part ligand-binding specificity (4), which has been exploited in the Author contributions: D.I.M., N.R.N., M.H., R.B.D., G.L., and K.M.C. designed research; design of the Food and Drug Administration-approved antibody- D.I.M., N.R.N., M.H., S.S., R.B.D., and D.D. performed research; D.I.M., M.H., G.L., and based therapy against a G-protein–coupled receptor (GPCR) for K.M.C. contributed new reagents/analytic tools; D.I.M., N.R.N., M.H., S.S., R.B.D., D.D., the treatment of migraine (5). Subsequent studies have further G.L., and K.M.C. analyzed data; and D.I.M., N.R.N., M.H., G.L., and K.M.C. wrote the paper. defined critical roles for RAMPs to impart biased downstream sig- The authors declare no competing interest. naling and intracellular trafficking dynamics to numerous GPCRs. This article is a PNAS Direct Submission. However, these pleiotropic effects of RAMPs have mostly been Published under the PNAS license. studied in the context of a few receptors in the Family B/ 1To whom correspondence may be addressed. Email: [email protected]. family, including the (CTR), , This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. (6, 7), CRF1 (8, 9), and VPAC1/2 (8) and the Family A 1073/pnas.1905561116/-/DCSupplemental. receptors CaSR (10) and GPR30/GPER1 (11). The strong coevolution First published November 11, 2019.

www.pnas.org/cgi/doi/10.1073/pnas.1905561116 PNAS | November 26, 2019 | vol. 116 | no. 48 | 24093–24099 Downloaded by guest on October 1, 2021 Results Identification of Chemokine Receptor: RAMP Interactions Using Bioluminescence Resonance Energy Transfer and Fluorescence- Activated Cell Sorting. We established a bioluminescence reso- nance energy transfer (BRET)-based screening platform to iden- tify RAMP-GPCR protein interactions by transiently coexpressing a constant amount of GPCR-rLuc protein with increasing amounts of RAMP-YFP protein in HEK293T cells. The canonical RAMP- interacting receptor CLR was used as a positive control and the noninteracting beta-2 (β2ADR) served as a negative control in order to empirically establish a systematic and multivariate process for identifying and scoring RAMP-GPCR interactions. To evaluate each interaction, the first major dis- criminator applied was a threshold of Bmax > 0.100 (Fig. 1 and SI Appendix, Table S1). All potential interactions failing to reach this minimum signal level were deemed negative, as indicated by salmon shading. Next, we applied a best-fit comparison for lin- earity versus hyperbolic curve fitting for all curves with a Bmax > 0.100. Interactions with a linearity R2 greater than the hyperbolic R2 were considered poor, indicated by yellow shading. Finally, the remaining curves which satisfied the hyperbolic curve fit test were further divided based on their BRET50 values. Hyperbolic curves with a BRET50 > 10 were considered good interactions and colored blue, while curves with BRET50 < 10 were considered the strongest possible candidates and shaded green. All chemokine receptors exhibited interactions with at least one RAMP. RAMP3 interactions were the most frequent and strongest (24/24 receptors), followed by RAMP2 (20/24) and RAMP1 (15/24) (Fig. 1). Flow cytometry was performed as an orthogonal assay to verify if BRET-based protein-protein interactions translated to effects on RAMP surface expression upon cotransfection with each receptor (SI Appendix, Fig. S1A). In the absence of a GPCR, FLAG- and HA-RAMP1 and RAMP2 displayed minimal plasma membrane expression, indicative of endogenous intracellular localization. Both FLAG- and HA-RAMP3 exhibited endoge- nous plasma membrane expression, an effect also observed in previous studies (18). Importantly, RAMP3 plasma membrane expression could be further enhanced by coexpression with CLR or CTR. CCR5, CXCR2, CX3CR1, and CMKLR1 exhibited highly significant RAMP interactions in both BRET-based and cell-surface expression assays. Interestingly, we noted that 3 of the members of the subfamily of confirmed ACKRs (ACKR1–3, but not ACKR4) interacted robustly with RAMP3 and, unlike RAMP1/2, reduced RAMP3 cell-surface expression compared to RAMP3 expressed alone. To confirm that the observed effects on RAMP cell-surface expression were not due to overexpression, FLAG-RAMPs were titrated with a fixed concentration of re- ceptor. We observed a saturable level of cell-surface expression above vector alone for FLAG-RAMP1 with CCR5 and FLAG- RAMP2 with CXCR2, with the plateau corresponding to a 1:1 RAMP:receptor ratio (SI Appendix,Fig.S1B). For FLAG-RAMP3, whichisabletotraffictotheplasmamembranealone,ACKR3 restricted its cell-surface expression in a saturable manner. ACKR3 has been reported to more broadly localize to early endosomes prior to ligand stimulation (19). Therefore, a possible explanation Fig. 1. BRET screening of the chemokine receptor family reveals heterodimers for the reduced FLAG-RAMP3 plasma membrane localization with RAMPs. ΔBRET was determined using HEK293T cells for each receptor: upon coexpression with ACKR3 is that the ACKR3-RAMP3 RAMP pair and plotted as a function of the total fluorescence/total lumi- complex resides largely intracellularly or, alternatively, is targeted nescence ratio. Curves were calculated using one site-binding (hyperbola) for degradation. This mechanism may also apply to other GPCRs and representative saturation isotherms are displayed for each receptor. (A) that reduce FLAG-RAMP3 plasma membrane expression. Inter- A systematic, multicomponent approach was used to score interactions. First, > estingly, but not unexpectedly, the results from the BRET and all interactions failing to reach a requirement of Bmax 0.1 were deemed fluorescence-activated cell sorting (FACS) experiments reveal negative (salmon). Next, a comparison of fits between hyperbolic and linear 2 > 2 some differences in specific RAMP-GPCR interactions. The models was used where Linear R Hyperbolic R was deemed a poor in- teraction (yellow). Finally, the remaining interactions were deemed good or BRET assay provides a measure of total protein-protein inter- strong based on the BRET50 values—BRET50 > 10 (blue) or BRET50 < 10 actions regardless of cellular location, whereas FACS is one of a (green), respectively. (B) Curves are representative of n = 3 to 4 independent number of techniques that measure the extent to which GPCRs experiments for each RAMP-receptor interaction, with average data reported alter RAMP expression at the cell surface. Thus, a comparative in SI Appendix, Table S1.

24094 | www.pnas.org/cgi/doi/10.1073/pnas.1905561116 Mackie et al. Downloaded by guest on October 1, 2021 integration of results for these 2 methodologies provides the interactions do not influence the downstream G-protein or β-arrestin most informed interpretation for the biological functions of responses of ACKR3 to either AM or SDF-1/CXCL12 ligands. these putative RAMP-GPCR interactions. ACKR3-RAMP3 Coexpression Scavenges and Attenuates AM Signaling. ACKR3 Interacts with RAMP2/3 without Affecting or We next established cell-based cAMP-EPAC reporter assays to β-Arrestin Coupling. We were particularly intrigued by the puta- distinguish the cell-intrinsic and cell-autonomous functions of tive ACKR3-RAMP interaction because, using genetic mouse ACKR3 in AM ligand scavenging via activation of the CLR-RAMP3 models, we previously showed that the developmental pheno- receptor heterodimer. For example, HEK293T cells transfected types of precocious cardiomyocyte and lymphatic hyperplasia in with CLR, RAMP3, N-ethylmaleimide-sensitive factor (NSF), − − Ackr3 / mice are attributable to gain-of-function in the AM and the EPAC reporter responded in a dose-dependent manner ligand (20), which elicits its signaling through the canonical to AM stimulation [EC50 = 1.6 nM ± logEC50 = 0.11 (Fig. 3A, RAMP-interacting receptor heterodimers CLR-RAMP2 and purple circles)], and this signaling could be significantly attenuated CLR-RAMP3. Therefore, we hypothesized that ACKR3 may with a large effect at high AM concentrations, evident by the also utilize RAMPs to modulate its ligand-scavenging activities. development of a biphasic dose–response curve fit, by the To address this, we further validated the ACKR3-RAMP3 pro- cotransfection of ACKR3 [EC50_1 = 1nM± logEC50_1 = 0.20 tein interaction by observing the colocalization of Myc-ACKR3 and EC50_2 = 3.1 μM ± logEC50_2 = 0.46, (Fig. 3A,orange and HA-RAMP3 at the plasma membrane of nonpermeabilized squares)] This was consistent with our previous results showing HEK293T cells by confocal microscopy (Fig. 2A) and within the reduced pERK activation by AM in the presence of ACKR3 cytoplasm by proximity ligation assay (Fig. 2B), to an extent (20). Because we found that RAMPs did not induce ACKR3- similar to that observed for the CLR-RAMP–positive controls. mediated cAMP production in response to AM (SI Appendix, Consistent with the established scavenging properties of ACKRs, Fig. S2 A and B), we reasoned that the reduced potency of AM at we used the BRET-based biosensor EPAC (21) to confirm that the CLR-RAMP3 receptor is caused by reduced bioavailability RAMP1, -2, or -3 could not induce activation of Gαs(SI Appendix, of AM ligand in the presence of the ACKR3-scavenging re- Fig. S2A) nor of Gαi/o (SI Appendix,Fig.S2B) in response to ceptor. To confirm this, and to provide a model for the cell- logarithmic dosage of AM or SDF-1/CXCL12 ligands. Further- autonomous scavenging effects of ACKR3 on the AM ligand, we more, we did not detect any effects of RAMPs on the AM- or employed a coculture system in which reporter cells expressing SDF-1/CXCL12–induced recruitment of β-arrestin-1-YFP or CLR-RAMP3-EPAC were cocultured with cells expressing either β-arrestin-2-YFP to ACKR3-rLuc, as evidenced by nonlinear ACKR3-RAMP3-NSF or glucagon-like peptide 2 receptor (GLP2R) regression best-fit curves of BRET activation (SI Appendix, Fig. as a nonscavenging control cell (Fig. 3B). Upon stimulation with CELL BIOLOGY S2 C and D). Both the AM and SDF-1/CXCL12 ligand caused AM, the CLR-RAMP3-EPAC reporter cells cocultured with the the recruitment of β-arrestin-1 and β-arrestin-2 to ACKR3, al- nonscavenging GLP2R-expressing cells stimulated maximal cAMP though SDF-1/CXCL12 showed more efficacy at lower doses than production with a potency of 0.52 nM ± logEC50 = 0.05 (black cir- AM (SI Appendix,Fig.S2C and D). Collectively, these data es- cles). In contrast, when the CLR-RAMP3-EPAC reporter cells were tablish RAMPs as interacting partners of ACKR3, but these cocultured with ACKR3-RAMP3-NSF–expressing cells, there was a

Fig. 2. Cellular distribution of ACKR3-RAMP3 and translocation to the plasma membrane. (A) HEK293T cells transfected with HA-RAMP alone or with Myc- CLR or Myc-ACKR3 as indicated. Addition of CLR and ACKR3 resulted in an increase in the detection of GPCR-RAMP at the plasma membrane. HA-tagged RAMP2 and RAMP3 exhibited very low levels of GPCR-independent localization. n = 3 for each con- dition. (B) PLA showed significant increases in signal for CLR with RAMP3 and ACKR3 with RAMP3 when compared with GPCR alone transfected controls. Blue is the nuclear staining and green is beta catenin to stain the membrane. (Right) The number of PLA signals per cell that correspond to the number of associations or heterodimer complexes for GPCR with RAMP3. Each color represents an individual ex- periment, n = 3, counting 7 to 21 individual cells per n. Statistical significance was determined by an unpaired t test. Error bars represent SEM of the means. (Scale bar, 10 μm.)

Mackie et al. PNAS | November 26, 2019 | vol. 116 | no. 48 | 24095 Downloaded by guest on October 1, 2021 Fig. 3. The ACKR3-RAMP3-NSF complex attenuates adrenomedullin signaling and regulates the ACKR3 receptor’s rapid recycling. (A) Expression of ACKR3inthesameHEK293TcellsastheCLR-RAMP3heterodimer resulted in a marked decrease in cAMP EPAC biosensor signal, indicating reduced

potency and a resulting biphasic dose–response (EC50 for −ACKR3 = 1.6 nM, ± logEC50 = 0.11 to +ACKR3 EC50_1 = 1nM,± logEC50_1 = 0.20 and EC50_2 = 3.1 μM, ± logEC50_2 = 0.46 with Hill slope for −ACKR3 = 0.58 ± 0.08 and nHill slope for +ACKR3 nH1 = −0.92 ± 0.40 and nH2 = −1.17 ± 0.99). (B) Coculturing the CLR-RAMP3-EPAC reporter cells with HEK293T cells expressing ACKR3-RAMP3-NSF resulted in a loss of AM potency as represented by the

rightward shift in the EC50 from 0.52 nM ± logEC50 = 0.05 to 1.3 nM ± logEC50 = 0.08 (Hill slope for +GLP2R = 0.86 ± 0.08, Hill slope for +ACKR3 = 0.50 ± 0.05). For both A and B, curves and statistical significance were determined by nonlinear regression with a comparison of fits (F-test); n = 6 in duplicate. (C) Immunofluorescence confocal microscopy shows ACKR3-RAMP3 colocalized at the plasma membrane. HEK293T cells were not treated with ligand and fixed, treated with AM for 1 h and fixed, or treated with AM for 1 h and washed/allowed to recover for 4 h and fixed. After ligand stimulation, ACKR3 and RAMP3 internalized and showed colocalization with NSF intracellularly. ACKR3 in the presence of RAMP3 and NSF (Left column) resulted in the ACKR3-RAMP3 complex localizing to the plasma membrane after the 4-h recovery phase. ACKR3 in the absence of NSF (Middle column) or RAMP3 (Right column) did not recycle to the plasma membrane after removal of ligand and the 4-h recovery. Images are representative of 3 independent experiments. (Scale bar, 10 μm.)

significant loss in potency to 1.3 nM ± logEC50 = 0.08, as demon- to scavenge AM ligand in both cell-intrinsic and cell-autonomous strated by the rightward shift in the dose–response curve (red settings, thereby attenuating AM signaling and further validating squares). These data demonstrate the ability of ACKR3-RAMP3 our previous studies using genetic mouse models.

24096 | www.pnas.org/cgi/doi/10.1073/pnas.1905561116 Mackie et al. Downloaded by guest on October 1, 2021 ACKR3 Rapid Recycling and Lysosomal Trafficking Are Dependent on RAMP3 and NSF. An inherent characteristic for establishing and maintaining chemotactic gradients for guided cell migration within discrete spatiotemporal boundaries is the rapid and dy- namic depletion of extracellular ligands from the nonmigrating region (22). Elegant zebrafish studies focused on primordial cell migration in response to SDF-1/CXCL12 gradients have impli- cated an important function for ACKR3 in this regard (23, 24). However, the molecular partners that enable ACKR3 to rapidly and cell autonomously scavenge ligands from the extracellular compartment remain unknown (25). RAMP3, by virtue of its C- terminal PSD-95/Discs-large/ZO-1 homology (PDZ)-recognition motif associates with NSF. This has previously been shown to facilitate the rapid recycling and resensitization of CLR to the plasma membrane following ligand-dependent internalization (26). Using confocal imaging, we confirmed these original findings for CLR following a 4-h recovery after removal of AM ligand (SI Appendix, Figs. S3 and S4). Similarly, we found that HEK293T cells transfected with ACKR3, RAMP3, and NSF displayed rapid internalization of ACKR3 following 1 h of AM ligand stimulation, with subsequent recycling of ACKR3 to the plasma membrane after 4 h of ligand removal and cycloheximide treatment (Fig. 3 C, Left column, and SI Appendix, Fig. S5). In contrast, in the absence of either RAMP3 or NSF, although ACKR3 ligand-stimulated internalization occurred at 1 h, the receptor failed to recycle to the plasma membrane and remained within intracellular vesicles after 4 h of recovery (Fig. 3 C, Middle and Right columns, and SI Appendix,Fig.S5). These data demonstrate the requirement of

RAMP3-NSF for the rapid recycling of the ACKR3 to the plasma CELL BIOLOGY membrane where it can evoke its ligand-scavenging activities. Following this same experimental paradigm, we were curious to address whether RAMP3-NSF expression could influence the fate of ACKR3 to different endosomal-sorting pathways. To address this, we tracked the ligand-activated internalization of tagged ACKR3 to fluorescently labeled, Rab-positive endosomes in the presence or absence of RAMP3-NSF. As shown in Fig. 4 A, Left columns, and SI Appendix, Fig. S6, ACKR3 rapidly in- Fig. 4. RAMP3 rescues internalized ACKR3 from Rab7a-positive late endo- ternalized to Rab4-positive vesicles following 1 h of treatment of somes, diverting the receptor to Rab4-positive rapid recycling vesicles resulting AM ligand and was subsequently recycled to the plasma mem- in resensitization after AM treatment. (A) HEK293T cells transfected with RFP- brane after 4 h of recovery in the presence of RAMP3-NSF. Rab4 and ACKR3 ±RAMP3-NSF were not treated with AM, treated with Conversely, in the absence of RAMP3-NSF, AM-stimulated 100 nM AM for 1 h and fixed, or treated and allowed to recover for 4 h. After 1 h ACKR3 internalized to non–Rab4-positive vesicles and never of AM treatment, ACKR3 +RAMP3-NSF are internalized and show returned to the cell surface (Fig. 4 A, Right columns, and SI colocalization with Rab4 intracellularly (Middle row, first column and Inset). In the ACKR3 –RAMP3-NSF condition, ACKR3 is internalized but colocaliza- Appendix, Fig. S6). Moreover, when cells were transfected with a tion with Rab4 does not occur (Middle row, third column and Inset). After a GFP-Rab7a lysosomal marker, AM-activated ACKR3 was 4-h recovery time post AM treatment, in the ACKR3 +RAMP3-NSF cells, spared from the lysosomal degradation pathway and resensitized ACKR3 and RAMP3 show distribution at the plasma membrane of the cell, to the plasma membrane in the presence of RAMP3-NSF (Fig. 4 demonstrating recycling of the receptor complex (Bottom row, first column B, Left columns, and SI Appendix, Fig. S7). In the absence of and Inset). In the ACKR3 –RAMP3-NSF cells, after the 4-h recovery, ACKR3 is RAMP3-NSF, however, ACKR3 was predominately sorted to- not present at the plasma membrane, and the receptor remained intracel- ward the lysosomal pathway, where its localization within the lu- lular (Bottom row, third column and Inset). (B) Conversely, HEK293T cells men of Rab7a-positive lysosomes and its absence from the plasma transfected with GFP-Rab7a and ACKR3 ±RAMP3-NSF were treated with AM + membrane was particularly evident 4 h after AM stimulation (Fig. as stated above. After 1 h of AM treatment, ACKR3 RAMP3-NSF are in- ternalized and show limited/no colocalization with Rab7a intracellularly 4 B, Right columns, and SI Appendix,Fig.S7). Importantly, we (Middle row, first column and Inset). However, in the ACKR3 –RAMP3-NSF found the same effects of RAMP3-NSF on ACKR3 sorting when condition, ACKR3 is internalized and shows robust localization in the lu- the receptor was activated by SDF-1/CXCL12 (SI Appendix,Figs. men of Rab7a-postive vesicles (Middle row,thirdcolumnandInset). After a S8–S10), indicating a ligand-unbiased requirement of RAMP3-NSF 4-h recovery time post AM treatment, in the ACKR3 +RAMP3-NSF cells, for the sorting of ACKR3 to rapidly recycling Rab4-positive ACKR3 and RAMP3 show distribution at the plasma membrane of the cell, endosomes, while being spared from lysosomal degradation. demonstrating recycling of the receptor complex (Bottom row, first column It has been established that the PDZ recognition sequence in and Inset). In the ACKR3 –RAMP3-NSF cells, after the 4-h recovery, ACKR3 is the C′-terminal tail of RAMP3 plays a critical role in the not present at the plasma membrane but is detected in the lumen of recycling of the canonical CLR-RAMP3-NSF complex (26). To Rab7a-late endosomes (Bottom row, third column and Inset). Images are representative of 3 independent experiments. (Scale bar, 10 μm.) test the hypothesis that the RAMP3 type I PDZ recognition motif is a molecular determinate of NSF-ACKR3 receptor trafficking, we generated a RAMP3ΔPDZ expression vector. The deletion of the PDZ motif (-DTLL) disrupted the ligand- ligand specificity. Interestingly, we also noted that the recep- stimulated, selective endosomal sorting of the ACKR3-RAMP3 tor complex instead localized to Rab11 slow-recycling endo- receptor complex (SI Appendix,Fig.S11, for AM ligand, and somes, which was previously not detected with wild-type (WT) SI Appendix,Fig.S12, for CXCL12 ligand) irrespective of RAMP3.

Mackie et al. PNAS | November 26, 2019 | vol. 116 | no. 48 | 24097 Downloaded by guest on October 1, 2021 −/− −/− Loss of Guided Cell Migration in Ackr3 and Ramp3 Mice during endothelial tip cells within the developing retina, thereby establishing Retinal Angiogenesis. To determine whether RAMP3-mediated its spatial positioning within the retina to maintain angiogenic gradients fating of ACKR3 to the rapid recycling endosomal pathway could (28). Predictably, a 50% reduction in the expression of the scavenging + − impact the scavenging properties of the receptor in a physiological receptor in Ackr3 / animals resulted in a significant reduction in the context, we turned to postnatal retinal angiogenesis as a model system number of endothelial tip cells within the retinas of postnatal day 3 of guided cell migration (27). In this context, angiogenic cues, like mice compared to control littermates (Fig. 5 B and E). Although the − − SDF-1/CXCL12 and AM, are enriched within peripheral astrocytes vast majority of Ackr3 / mice die at postnatal day 1, we were fortunate and serve as chemotactic gradients for guided angiogenesis of retinal to obtain and characterize a single surviving animal which displayed a vasculature by stimulating tip cells and filopodia (28) Fig. 5 A, D,and profound reduction in tip-cell number (Fig. 5E) and effacement of the G). Previous studies have defined the presence of ACKR3 within retinal angiogenic front. Importantly, we observed the same attenua- − − trailing arterioles and, concomitantly, its notable absence from leading tion of guided angiogenesis in Ramp3 / animals (29) compared to

Fig. 5. Vascular development is disrupted upon genetic deletion of ACKR3 or RAMP3 in vivo through a RAMP3-mediated rapid recycling mechanism. (A–C) Representative images of whole-mount immunofluorescence staining of postnatal day 3 retinal vasculature using isolectin B4 in control, AM overexpressing + − − − − − (Admhi/hi), ACKR3 heterozygous (Ackr3 / ), ACKR3 knockout (Ackr3 / ), and RAMP3 knockout (Ramp3 / ) mice. Statistics were evaluated using n = 3 to 7 mice. (Scale bars, 200 μM.) (D–F) Analysis and quantification of the retinal tip cells detected at the vascular periphery in control and gene targeted mice. Colored dots represent quantitation from different retinal quadrants of individual animals. Statistical significance was determined by an unpaired, 2-tailed t test with an n between 3 and 7 mice, as indicated in the figure. Error bars represent SEM of the means. (G) Cartoon depicting gradient-guided migration of vascular endothelial tip cells in response to normal chemotactic gradients (Top) established by ACKR3/RAMP3 (yellow/green) and abnormal gradients (Bottom)inthe absence of ACKR3/RAMP3 ligand scavenging. (H) Model of RAMP3-mediated endosomal sorting of ACKR3. (I) Ligand binding of ACKR3 leads to endocytosis of receptor. (II) ACKR3 is internalized to early endosomes, where it is colocalized with RAMP3 and NSF. (III) The PDZ-binding motifs of RAMP3 and NSF are required for the Rab4-positive rapid recycling endosome-dependent and RAMP3-facilitated recycling of internalized ACKR3 to the cell surface via a putative interaction with a PDZ-domain–containing protein. In the absence of RAMP3, ACKR3 is localized to Rab7a-positive late endosomes which controls the trafficking between late endosomes and lysosomes. (IV) ACKR3 scavenging is enabled by the rapid resensitization of ACKR3 to the cell surface by RAMP3.

24098 | www.pnas.org/cgi/doi/10.1073/pnas.1905561116 Mackie et al. Downloaded by guest on October 1, 2021 control littermates (Fig. 5 C and F). Additionally, we observed a sig- Additional studies that employ a wide range of biochemical, phar- − − nificant decrease in tip-cell filopodia in Ramp3 / animals (WT vs. macological, and cellular assays to elucidate the effects that each − − Ramp3 / ;20.7± 1.34 vs. 16.9 ± 0.90, P = 0.023) and a trend toward RAMP has on the ligand binding, biased functional selectivity, or + − + − decreased tip-cell filopodia in Ackr3 / animals (WT vs. Ackr3 / ; trafficking of other chemokine GPCRs should provide valuable 24.4 ± 1.45 vs. 22.5 ± 1.14, P = 0.333). These findings support an future exploration and opportunity. essential physiological function for AM-gradient guided cell migration through the scavenging activities of ACKR3 and RAMP3 (Fig. 5G). Materials and Methods Cell culture, BRET assays, flow cytometry, immunofluorescence confocal micros- Discussion copy, whole-mount immunohistochemistry, in situ proximity ligation assay (PLA), Collectively, these data significantly expand the repertoire of GPCRs receptor scavenging, internalization, resensitization, and trafficking assays per- that interact with RAMPs and provide at least one example of how a formed in this study are provided in SI Appendix, Materials and Methods.

RAMP-GPCR interaction can dictate the physiological functions of + + − Animals. Adm /hi and Ackr3 / mice were maintained on a C57BL/6 genetic back- guided cell migration by governing receptor endosomal sorting and − − ground, and Ramp3 / mice were maintained on a 129/SvEv genetic background. recycling (Fig. 5H). We focused our efforts on the process of retinal − − Ackr3 / and Admhi/hi mice were generated by heterozygous intercrosses, re- − − angiogenesis because we, and others, have previously shown that both spectively. Ramp3 / mice were generated through homozygous crosses of ACKR3 ligands, SDF-1/CXCL12 and AM, and their respective cog- Ramp3−/− mice, and Ramp3+/+ isogenic mice were used as controls. A total of 6 mice nate signaling receptors, CXCR4 and CLR, are critical for driving across 2 Adm genotypes (3 Adm+/+ and 3 Admhi/hi), 11 mice across the 3 Ackr3 + + + − − − guided cell migration within this vascular bed (28, 30). However, genotypes (4 Ackr3 / ,6Ackr3 / ,and1Ackr3 / ),and14miceacross2Ramp3 + + − − ACKR3 also plays critical roles in the migration of GABAergic in- genotypes (7 Ramp3 / and 7 Ramp3 / ) were used in this study. This study was terneurons within the embryonic (1, 31) in the homing powered to attain statistical significance of P < 0.05 with a 90% probability between + + + + + − + + − − ofimmunecellstolymphoidtissuesandinthemigrationofcancer Adm / and Admhi/hi, Ackr3 / and Ackr3 / ,andRamp3 / and Ramp3 / mice. All cells (32, 33). Thus, it will be of interest to determine whether RAMP3 animal procedures and protocols were approved by the Institutional Animal also dictates the scavenging functions of ACKR3 within these other Care and Use Committee of the University of North Carolina, Chapel Hill. contexts. If so, the RAMP3-ACKR3 interface should provide a Data and Materials Availability. No datasets were generated or analyzed therapeutic target for modulating the actions of the CXCR4/CXCL12 during the current study. All correspondence and material requests should axis in anticancer and antiinflammatory strategies (34). be addressed to K.M.C. Indeed, the molecular interface formed between RAMP1 and CLR has recently been exploited in the design of the anti-CGRP ACKNOWLEDGMENTS. This work was supported by NIH Grants RO1- migraine drug, (5, 35). In our current study, we find that DK099156, RO1-HD060860, and RO1-HL129086 (to K.M.C.); American CELL BIOLOGY 23 of the 24 described chemokine receptors display moderate-to- Association Innovator Award 16IRG27260077 (to K.M.C.); NIH Grant F32- HL134279 (to D.I.M.); American Heart Association Grant 15POST25270006 (to strong molecular interactions with RAMPs. These findings identify R.B.D.); NIH Grant F31-HL143836 (to N.R.N.); Biotechnology and Biological unique and pharmacologically tractable avenues for the modulation Sciences Research Council (BBSRC) Grant BB/M00015X/2 (to G.L.); and BBSRC of chemokine function in a wide range of physiological processes. Doctoral Training Partnership Grant BB/JO14540/1 (to M.H.).

1. F. Saaber et al., ACKR3 regulation of neuronal migration requires ACKR3 phosphor- 19. B. Zhu et al., CXCL12 enhances human neural progenitor cell survival through a CXCR7- ylation, but not β-arrestin. Cell Rep. 26, 1473–1488.e9 (2019). and CXCR4-mediated endocytotic signaling pathway. Stem Cells 30,2571–2583 (2012). 2. K. R. Klein, B. C. Matson, K. M. Caron, The expanding repertoire of receptor activity 20. K. R. Klein et al., Decoy receptor CXCR7 modulates adrenomedullin-mediated cardiac modifying protein (RAMP) function. Crit. Rev. Biochem. Mol. Biol. 51,65–71 (2016). and lymphatic vascular development. Dev. Cell 30, 528–540 (2014). 3. L. M. McLatchie et al., RAMPs regulate the transport and ligand specificity of the 21. L. S. Barak et al., Pharmacological characterization of membrane-expressed human calcitonin-receptor-like receptor. Nature 393, 333–339 (1998). trace amine-associated receptor 1 (TAAR1) by a bioluminescence resonance energy 4. Y. L. Liang et al., Cryo-EM structure of the active, Gs-protein complexed, human CGRP transfer cAMP biosensor. Mol. Pharmacol. 74, 585–594 (2008). receptor. Nature 561, 492–497 (2018). 22. J. Bussmann, E. Raz, Chemokine-guided cell migration and motility in zebrafish de- 5. P. J. Goadsby et al., A controlled trial of erenumab for episodic migraine. N. Engl. J. velopment. EMBO J. 34, 1309–1318 (2015). – Med. 377, 2123 2132 (2017). 23. G. Venkiteswaran et al., Generation and dynamics of an endogenous, self-generated 6. C. Weston et al., Modulation of pharmacology by receptor activity- signaling gradient across a migrating tissue. Cell 155, 674–687 (2013). – modifying protein-2 (RAMP2). J. Biol. Chem. 290, 23009 23022 (2015). 24. B. Boldajipour et al., Control of chemokine-guided cell migration by ligand seques- 7. J. Cegla et al., RAMP2 influences glucagon receptor pharmacology via trafficking and tration. Cell 132, 463–473 (2008). – signaling. Endocrinology 158, 2680 2693 (2017). 25. N. Montpas et al., Ligand-specific conformational transitions and intracellular trans- 8. D. Wootten et al., Receptor activity modifying proteins (RAMPs) interact with the port are required for atypical chemokine receptor 3-mediated chemokine scavenging. VPAC2 receptor and CRF1 receptors and modulate their function. Br. J. Pharmacol. J. Biol. Chem. 293, 893–905 (2018). 168, 822–834 (2013). 26. J. M. Bomberger, N. Parameswaran, C. S. Hall, N. Aiyar, W. S. Spielman, Novel function 9. S. Bailey et al., Interactions between RAMP2 and CRF receptors: The effect of receptor for receptor activity-modifying proteins (RAMPs) in post-endocytic receptor traffick- subtypes, splice variants and cell context. Biochim. Biophys. Acta Biomembr. 1861, ing. J. Biol. Chem. 280, 9297–9307 (2005). 997–1003 (2019). 27. S. Rezzola et al., In vitro and ex vivo retina angiogenesis assays. Angiogenesis 17, 429– 10. A. J. Desai, D. J. Roberts, G. O. Richards, T. M. Skerry, Role of receptor activity mod- 442 (2014). ifying protein 1 in function of the sensing receptor in the human TT thyroid 28. G. A. Strasser, J. S. Kaminker, M. Tessier-Lavigne, Microarray analysis of retinal endo- carcinoma cell line. PLoS One 9, e85237 (2014). thelial tip cells identifies CXCR4 as a mediator of tip cell morphology and branching. 11. P. M. Lenhart, S. Broselid, C. J. Barrick, L. M. F. Leeb-Lundberg, K. M. Caron, G-protein- Blood 115,5102–5110 (2010). coupled receptor 30 interacts with receptor activity-modifying protein 3 and confers 29. R. Dackor, K. Fritz-Six, O. Smithies, K. Caron, Receptor activity-modifying proteins 2 sex-dependent cardioprotection. J. Mol. Endocrinol. 51, 191–202 (2013). 12. S. Barbash, E. Lorenzen, T. Persson, T. Huber, T. P. Sakmar, GPCRs globally coevolved and 3 have distinct physiological functions from embryogenesis to old age. J. Biol. – with receptor activity-modifying proteins, RAMPs. Proc. Natl. Acad. Sci. U.S.A. 114, Chem. 282 , 18094 18099 (2007). 12015–12020 (2017). 30. S. Sakimoto et al., An angiogenic role for adrenomedullin in choroidal neo- 13. E. Lorenzen et al., Multiplexed analysis of the secretin-like GPCR-RAMP interactome. vascularization. PLoS One 8, e58096 (2013). Sci. Adv. 5, eaaw2778 (2019). 31. P. Abe et al., CXCR7 prevents excessive CXCL12-mediated downregulation of CXCR4 – 14. S. J. Allen, S. E. Crown, T. M. Handel, Chemokine: Receptor structure, interactions, and in migrating cortical interneurons. Development 141, 1857 1863 (2014). antagonism. Annu. Rev. Immunol. 25, 787–820 (2007). 32. K. Hattermann, R. Mentlein, An infernal trio: The chemokine CXCL12 and its receptors 15. X. Blanchet, M. Langer, C. Weber, R. R. Koenen, P. von Hundelshausen, Touch of CXCR4 and CXCR7 in tumor biology. Ann. Anat. 195, 103–110 (2013). . Front. Immunol. 3, 175 (2012). 33. K. E. Quinn, D. I. Mackie, K. M. Caron, Emerging roles of atypical chemokine receptor 16. C. E. Hughes, R. J. B. Nibbs, A guide to chemokines and their receptors. FEBS J. 285, 3 (ACKR3) in normal development and physiology. 109,17–23 (2018). 2944–2971 (2018). 34. T. J. Schall, A. E. Proudfoot, Overcoming hurdles in developing successful drugs tar- 17. R. J. Nibbs, G. J. Graham, Immune regulation by atypical chemokine receptors. Nat. geting chemokine receptors. Nat. Rev. Immunol. 11, 355–363 (2011). Rev. Immunol. 13, 815–829 (2013). 35. L. Edvinsson, K. A. Haanes, K. Warfvinge, D. N. Krause, CGRP as the target of new 18. A. Christopoulos et al., Novel receptor partners and function of receptor activity- migraine therapies: Successful translation from bench to clinic. Nat. Rev. Neurol. 14, modifying proteins. J. Biol. Chem. 278, 3293–3297 (2003). 338–350 (2018).

Mackie et al. PNAS | November 26, 2019 | vol. 116 | no. 48 | 24099 Downloaded by guest on October 1, 2021