aw, a...“ s‘ 5.5.! 1?. ($2 .r 1.»- L‘ .5 . . J .. unit? xx ”dist“. an. .4 e331 .; . me}. 15... .4. smawrw...» .01. ‘. o\ «M. , ”a: $xhuuw. . 52.1.1 (v!) v. “MGR". ,énuhgi‘ ..4 ~ ' . ' W 5‘ LIBRARIES MICHIGAN STATE UNIVERSITY EAST LANSING, MICH 48824-1048 This is to certify that the dissertation entitled ROLE FOR RECEPTOR ACTIVITY MODIFYING PROTEINS IN THE TRAFFICKING OF THE ADRENOMEDULLIN RECEPTOR presented by Jennifer Melinda Bomberger has been accepted towards fulfillment of the requirements for the Doctoral degree in Physiology W Maidr Professor’s Signature /0 %j 3’05“ 0 Date MSU is an Affirmative Action/Equal Opportunity Institution PLACE IN RETURN Box to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE > ‘OI AY 1 4 2008 ROLE FOR RECEPTOR ACTIVITY-MODIFYING PROTEINS (RAMPs) IN THE TRAFFICKING OF THE ADRENOMEDULLIN RECEPTOR By Jennifer Melinda Bomberger A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Physiology 2005 ABSTRACT ROLE FOR RECEPTOR ACTIVITY MODIFYING PROTEINS IN THE TRAFFICKING OF THE ADRENOMEDULLIN RECEPTOR By Jennifer Melinda Bomberger Adrenomedullin (AM) is a vasodilatory peptide with effects in numerous physiological systems. AM exerts its effects through binding to a heterotrimeric G-protein-coupled receptor complex (calcitonin receptor-like receptor (CL-R) and single-transmembrane accessory protein, receptor activity modifying protein (RAMP)). CL-R is unique in that it requires association with the accessory protein, RAMP, for efficient receptor expression at the plasma membrane and for determination of receptor phenotype. A heterodimer of CL-R and RAMP] yields a calcitonin gene-related peptide (CGRPl) receptor, whereas complexing of CL-R with the RAMP2 or RAMP3 isoforms characterizes a fully functional AM receptor (AMI and AM2 receptors, respectively). Curiously, although RAMP expression differs from tissue to tissue, the receptor complex is coupled to Go.s in all systems studied to date, regardless of the RAMP isoform associated or ligand stimulation (AM or CGRP). Thus, it is the overall hypothesis of this thesis that although RAMP isoforms do not regulate the type of G-protein coupled to the receptor complex, the isoform of RAMP associated with CL-R does regulate the life cycle of the receptor complex. Thus, this thesis focuses on the role of the RAMPs in regulation of the desensitization, internalization, recycling and degradation of the CL-R/RAMP receptor complex. In particular, this study focuses on the differential trafficking of the AMI and AM2 receptor subtypes by RAMP2 and RAMPB, respectively. Renal cell culture systems, both heterologous expression and native, are the model systems utilized to study the differential receptor trafficking in this project. Using mutagenesis approaches, data from this investigation demonstrates that association of CL-R with RAMP2 or RAMP3 isoforms could result in differential receptor phosphorylation patterns with prolonged agonist stimulation. The Serine 421 residue on CL-R is critical to the desensitization and internalization of the CL-R/RAMPZ heterodimer, while Threonine 423 on CL-R is required for efficient CL-R/RAMP3 receptor complex desensitization and internalization. While Serine 421 is a putative phosphorylation site for PKA, Threonine 423 is a putative phosphorylation site for PKC. Selective inhibitors against these kinases also inhibited CL-R/RAMPZ (by H-89) and CL-R/RAMP3 (by Ro 32-0432) desensitization and internalization. Differences were also observed in the interaction of AM receptor subtypes with NaVI-F Exchanger Regulatory Factor-l (NHERF-l). Upon prolonged AM exposure, NHERF-l was found to inhibit the internalization, but not desensitization, of the CL—R/RAMP3 receptor complex. The effect of NHERF-l was specific for the AM2 receptor. This inhibition of internalization was dependent on PDZ interaction 'of RAMP3 with NHERF-l and NHERF-1 interaction with MERM proteins in the actin cytoskeleton. Finally, a model for differential post- endocytic targeting of the AM receptor subtypes was proposed from studies in HEK 293, Rat2 fibroblast, and rat mesangial cells. Through adenylate cyclase and CAMP accumulation assays, whole-cell competition binding, and immunofluorescence microscopy experiments it was demonstrated that RAMP3 interacts with NSF via a PDZ recognition sequence at the C-terminus of RAMP3. This was an effect specific for the CL-R/RAMP3 receptor complex. This RAMP3- NSF interaction is proposed to target the AM2 receptor complex for a recycling pathway after agonist-induced internalization, whereas expression of the AMI receptor complex, unable to interact with NSF, will be targeted for degradation after agonist-induced endocytosis. These findings report a novel function for the RAMP3 in post-endocytic receptor trafficking and provide the first difference between the RAMP2 and RAMP3 isoforms in the trafficking of the AMlR and AMZR. In addition, this data demonstrates a functional difference between the AMlR (CL-R+RAMP2) and AMZR (CL-R+RAMP3) receptor complexes, in spite of very similar second messenger systems and the physiological responses thus far identified. T 0 Mike and my parents, for your unwavering support and encouragement, I am eternally grateful. iv ACKNOWLEDGEMENTS Successful completion of this dissertation is the result of the combined efforts of many individuals. I would like to express my gratitude to my graduate committee members: Drs. Nambi Aiyar, Laura McCabe, Kathleen Gallo, Walter Esselman, and William Spielman. Their continued guidance and assistance during my graduate education and thesis research proved indispensable. I would like to thank Drs. McCabe, Gallo, and Esselman for their time dedicated to the review and direction of my thesis research project. I am particularly indebted to Dr. Aiyar for his training and assistance in establishing the radioligand binding methodology in our laboratory. I would especially like to thank Dr. William Spielman, my major advisor, for his continual encouragement and mentoring during my training. I would also like to extend a special thanks to my friends and colleagues in the Spielman lab and the Department of Physiology, particularly Dr. Narayanan Parameswaran, Dr. Wojciech Nowak, and Carol Hall. I am especially thankful to Nara for the endless support, critical analysis, and scientific guidance imparted to me. Finally, I am very appreciative of the office and computer support staff of the Department of Physiology for the friendly interactions and continual assistance they provided to me. In particular, I would like to sincerely thank Michelle Duchene for always taking the time to lend a helping hand. TABLE OF CONTENTS List of Tables ........................................................................................ viii List of Figures ......................................................................................... ix Key to Abbreviations ............................................................................... xii 1. Introduction ......................................................................................... l 2. Literature Review ................................................................................... 3 2.1. Adrenomedullin....................................................... ................... 3 2.1.1. Adrenomedullin gene and protein structure .............................. 3 2.1.2. Location of adrenomedullin gene and protein expression. . . . . . . . ......4 2.1.3. Biological actions of adrenomedullin .................................... 5 2.1.4. Adrenomedullin expression in pathophysiological states ............. 9 2.1.5. Effects of adrenomedullin gene alteration and adrenomedullin gene delivery .............................................................. 12 2.2. Adrenomedullin Receptor Complex ................................................ 13 2.2.1. Discovery of adrenomedullin receptor complex ....................... 13 2.2.2. RAMP gene and protein structure ....................................... 17 2.2.3. Tissue and cell specific RAMP expression ............................. 19 2.2.4. Mechanism of RAMP-receptor interaction ............................. 21 2.2.5. Additional RAMP-interacting receptors ................................ 24 2.2.6. Regulation of RAMP gene expression .................................. 26 2.2.7. Receptor Component Protein (RCP) .................................... 29 2.3. GPCR Life-cycle Regulation ........................................................ 30 2.3.1. GPCR life-cycle ............................................................ 30 2.3.2. Regulation of receptor desensitization .................................. 33 2.3.3. Regulation of receptor intemalization/endocytosis ................... 35 2.3.4. Regulation of receptor recycling vs. degradation ..................... 38 2.3.5. Protein-protein interaction domains/motifs ............................ 41 2.3.6. Na+/H+ Exchanger Regulatory Factor-1 (NHERF-l) ................ 47 2.3.7. N-ethylmaleimide Sensitive Factor (NSF) .............................. 49 3. Characterization of AM-stimulated desensitization response in transfected HEK 293 cells .......................................................................................... 52 3. l . Introduction ............................................................................ 52 3.2. Materials and Methods ............................................................... 53 3.3. Results/ Discussion ................................................................... 57 vi 4. Differential regulation of adrenomedullin receptor desensitization and internalization by receptor activity-modifying proteins .............................. 68 4.1 . Introduction ............................................................................ 68 4.2. Materials and Methods ............................................................... 70 4.3. Results .................................................................................. 76 4.4. Discussion ............................................................................ 109 5. RAMP isoform-specific regulation of adrenomedullin receptor trafficking by NHERF-l .................................................................................. l 13 5. 1 . Introduction ........................................................................... 1 13 5.2. Materials and Methods .............................................................. 115 5.3. Results ................................................................................. 122 5.3.1. Role of NHERF-l in Intemalization of the CL-R/RAMP complex .................................................................. 122 5.3.2. RAMP isoform-specific regulation of CL-R/RAMP receptor complex trafficking ..................................................... 123 5.3.3. Role of PDZ interactions in trafficking of the CL-R/RAMP3 complex .................................................................. 124 5.3.4. Mechanism of inhibition of CL-R/RAMP3 internalization by NHERF-l ....................................................... 127 5.3.5. NHERF’s role in receptor trafficking in primary human proximal tubule epithelial cells ................................................... 128 5.4. Discussion ............................................................................ 156 6. Novel function for receptor activity-modifying proteins in post-endocytic receptor trafficking ........................................................................ 160 6. 1. Introduction ........................................................................... 160 6.2. Materials and Methods .............................................................. 162 6.3. Results ................................................................................. 169 6.3.1. Role of NSF in Resensitization of the CL-R/RAMP complex. . ...169 6.3.2. RAMP isoform-specific regulation of CL—R/RAMP receptor complex trafficking ..................................................... 172 6.3.3. Role of PDZ interactions in trafficking of the CL—R/RAMP3 complex .................................................................. 172 6.3.4. RAMP3 and NSF regulation of receptor trafficking in unaltered cell lines .................................................................. 175 6.4. Discussion ............................................................................ 201 7. Summary and Conclusions ................................................................... 205 7.1. Specific aims, major hypotheses, and results of the study ..................... 205 7.2. Limitations of the study ............................................................ 208 7.3. Positive outcomes of the study .................................................... 209 References .......................................................................................... 21 1 vii LIST OF TABLES Table 1. Biological actions of adrenomedullin .................................................. 8 Table II. RAMP isoform and CL-R receptor expression in renal cell lines ............... 21 Table III. Regulation of expression of RAMP isoforms and CL-R in various pathophysiologies and animal models .......................................... 28 Table IV. Terminology for the GPCR life-cycle ............................................... 33 Table V. Binding properties for homologous competition binding of rAM on intact HEK-293T cells expressing CL-R-GF P and RAMP3 +/- NSF or NHERFI ........................................................................... 67 viii LIST OF FIGURES Figure l. Depiction of GPCR life-cycle ......................................................... 32 Figure 2. Representative desensitization experiments of AM receptors in HEK 293 Cells ................................................................................. 59 Figure 3. Desensitization dose response of AM receptors in HEK 293 cells ............... 61 Figure 4. Desensitization time course for AM receptors in HEK 293 cells ................ 63 Figure 5. Competition curves for rAM on intact HEK- 293T cells, expressing CL- R and RAMP3, with [”51] rAM .................................................... 65 Figure 6. Scatchard plot for homologous competition binding of rAM on intact HEK 293T cells, expressing CL—R and RAMP3, with [”51] rAM ................. 66 Figure 7. Desensitization and internalization of the AMI and AM2 receptor in HEK 293 cells ...................................................................... 84 Figure 8. Illustration of cluster mutations of CL-R to investigate role of phosphorylation on AM receptor desensitization and internalization. . ....86 Figure 9. Role of phosphorylation on AM] and AM2 receptor desensitization and internalization (cluster mutations) .............................................. 87 Figure 10. Role of phosphorylation on AM] receptor desensitization and internalization (point mutations) ................................................ 91 Figure l 1. Role of phosphorylation on AM2 receptor desensitization and internalization (point mutations) ................................................ 94 Figure 12. Role of phosphorylation of residues in third intracellular loop of CL-R on AMI and AM2 receptor desensitization ....................................... 97 Figure 13. Role of phosphorylation of RAMPs on AM] and AM2 receptor desensitization and internalization .............................................. 99 Figure 14. Role of kinases in AMI receptor desensitization and internalization. . . . . 103 Figure 15. Role of kinases in AM2 receptor desensitization and internalization. . . . . 105 Figure 16. Role of kinases in AMI and AM2 receptor desensitization and internalization ..................................................................... 107 ix Figure 17. Figure 18. Figure 19. Figure 20. Figure 21. Figure 22. Figure 23. Figure 24. Figure 25 Figure 26. Figure 27. Figure 28. Figure 29. Figure 30. Figure 31. Figure 32. Figure 33 The role of NHERF-l in the trafficking of the AM2R (CL-R+RAMP3) in HEK 293 cells .................................................................... 131 Localization of CL-R, RAMP3, and NHERF-1 in HEK 293 cells .....l34 The role of NHERF-l in post-endocytic sorting of CGRPI (CL-R+R1) and AM] receptor ............................................................... 135 The effect of RAMP3 PDZ motif and NHERF-1 PDZ domain mutations on the trafficking of the AM2R (CL-R+RAMP3) ........................... 139 Localization of CL-R, RAMP3A145-8, and NHERF -1 in HEK 293 cells during an internalization experiment .......................................... 143 Interaction studies of RAMP3 with NHERF -1 .................................. 144 Mechanism of NHERF-l inhibition of internalization of CL-R/RAMP3 receptor complex ................................................................. 145 Trafficking of AM—R in human proximal tubule cells (hPTE cells) .......... 149 . Effect of RAMP3 and NHERF-1 RNA interference on internalization of AM2-receptor in human proximal tubule cells ........................... 151 Mechanism of NHERF -1 inhibition of internalization of CL-R/RAMP3 receptor complex in hPTE cells ................................................ 154 Trafficking of AM-R in human proximal tubule cells (hPTE cells) .......... 155 Effect of N-ethylmaleimide, an NSF inhibitor, on resensitization of CAMP accumulation in rat mesangial cells (RMCs) ........................ 178 The sorting of the AM2R (CL-R+RAMP3) in HEK 293 cells ................ 179 The role of NSF in the sorting of the AM2R (CL-R+RAMP3) in HEK 293 cells .................................................................... 181 Localization of CL-R, RAMP3, and NSF in HEK 293 cells during a recycling experiment .......................................................... 183 The role of NSF in post-endocytic sorting of CGRPl (CL-R+Rl) and AMI receptor ............................................................... 185 . The effect of RAMP3 PDZ motif deletion on the trafficking of the AM2R (CL-R+RAMP3) ........................................................ 189 Figure 34. Figure 35. Figure 36. Figure 37. Figure 38. Figure 39. Localization of CL-R, RAMP3A145-8, and NSF in HEK 293 cells .......... 191 Effect of PDZ motif substitution on the C-terminus of RAMP2 on recyclingresensitization of CL-R/RAMP complex ......................... 192 Effect of RAMP3 PDZ motif point mutations on the recycling/resensitization of the CL-R/RAMP3 complex in the presence of NSF .................................................................. 194 Interaction studies of RAMP3 with NSF .......................................... 196 Effect of RAMP3 RNA interference on resensitization of AM2-R in rat mesangial cells ................................................................... 197 AM-R trafficking in Rat2 fibroblast cells ........................................ 200 xi HEK 293 HIF HRE KEY TO ABBREVIATIONS Adrenomedullin Adrenomedullin receptor (composed of CL-R+RAMP2)* Adrenomedullin receptor (composed of CL-R+RAMP3)* a-amino-3-hydroxy—5-methyl-isoxazolepropionate Adrenomedullin receptor (generic) Amylin Amylin receptor (composed of CTR+RAMP1)* Amylin receptor (composed of CTR+RAMP3)* Angiotensin II Arginine-vasopressin Bz-adrenergic receptor Adenosine 3’,5’ cyclic monophosphate Calcitonin gene-related peptide CGRP receptor (composed of CL-R+RAMP1)* Cystic fibrosis transmembrane conductance regulator Cycloheximide Calcitonin receptor-like receptor * Calcitonin Calcitonin receptor Cytochalasin D Deoxycorticosterone acetate 8-opioid receptor Ezrin/moesin/radixin domain Endothelin- l Drosophila enabled/vasodilator-stimulated phosphoprotein homology 1 Glyceraldehyde-3-phosphate dehydrogenase G protein accessory sorting protein Glomerular filtration rate G protein-coupled receptor GPCR-regulated kinase Human aortic endothelial cells Human embryonic kidney 293 cells Hypoxia inducible factor Hypoxia response element * Nomenclature recommended by International Union of Pharmacology (IUPHAR) XXXII (as described by Poyner et al. Pharmacol Rev 54: 233-246, 2002.) This naming system extends to all of the established receptors for the calcitonin family of peptides. xii HUVEC KOR L-NAME LPS LV MAPK NEM NHE NHERF NSF NO PAMP PDGF PDGF-R sz PG PI P13K PKA PKC PLCB PP2A PSD-95 PTH PTH-R Q-PCR RAEC RAMP RBF RCP RMC RTK RT-PCR SH2/3 SHR SNAP SNARE UTRS VSMC Human umbilical vein cells tc-opioid receptor Nco-nitro-L-arginine methyl ester Lipopolysaccharide Left ventricle Mean arterial pressure Mitogen-activated protein kinase N-ethylmaleimide Na+/H+ exchanger Na+/H+ exchanger regulatory factor N-ethylmaleimide sensitive factor Nitric oxide Proadrenomedullin N-tenninal 20 peptide Platelet-derived grth factor PDGF receptor PSD-95/Discs Large/Zona occludens-l Prostaglandin Phosphotidyl inositol Phosphotidyl inositol 3-kinase Protein kinase A (CAMP protein kinase) Protein kinase C Phospholipase CB Protein phosphatase 2A Post-synaptic density-95 protein Parathyroid hormone Parathyroid hormone receptor Quantitative-polymerase chain reaction Rabbit aortic endothelial cells Receptor activity-modifying protein Renal blood flow Receptor component protein Rat mesangial cells Receptor tyrosine kinase Reverse transcriptase- polymerase Chain reaction Src Homology domain 2/3 Spontaneously hypertensive rat Soluble N-ethylmaleimide-sensitive factor attachment protein Soluble N-ethylmaleimide-sensitive factor attachment protein receptor Untranslated regions Vascular smooth muscle cells xiii 1. Introduction Recent discovery of receptor activity modifying proteins (RAMPS) has enhanced our understanding of the mechanisms known to regulate the signaling of the G protein- Coupled receptor (GPCR) superfamily. RAMPS (1-3) have been demonstrated to associate with the calcitonin (CTR) and calcitonin receptor-like (CL-R) receptors and differentially regulate the ligand selectivity of the receptors. Although RAMPS enable differential receptor phenotypes, the signaling elicited through receptor activation is quite similar, namely Guts activation to activation adenylate cyclase enzymes, leading to CAMP accumulation and protein kinase A activation. Similar biological responses are even seen, regardless of ligand stimulation. For this reason, it has been hypothesized that the RAMP isoforms have additional regulatory roles, thus far unidentified. This thesis research investigates the role of RAMPS in the regulation of the CL-R/RAMP receptor complex after agonist exposure and receptor activation, in particular, during the processes of desensitization, internalization, and receptor trafficking from the endosomes. This brief introductory Chapter is followed by a comprehensive literature review in the second chapter. The literature review provides an in-depth discussion of the past and current scientific literature surrounding the topic of this thesis project, including the biology of adrenomedullin, the Characterization of the CL-R/RAMP receptor complex, the protein-protein interactions involved in GPCR Signaling, and the regulation of the GPCR life-cycle. Chapters three through five are organized according to the proposed Specific aims of this study, and each consist of an introduction, experimental data, and Specific discussion relevant to the individual aim. Chapter three investigates if phosphorylation of the receptor complex regulates the process of agonist-stimulated desensitization and internalization of the receptor complex. An emphasis is placed on whether the desensitization and internalization are differentially regulated by RAMP2 vs. RAMP3 in complex with the receptor. The fourth Chapter examines protein-protein interactions that regulate the internalization of the CL-R/RAMP receptor complex. The role of NHERF-l in regulation of the CL-R/RAMP complex internalization and the mechanism for this regulation are investigated. The fifth chapter describes protein- protein interactions that regulate the trafficking of the CL-R/RAMP complex after agonist-stimulated receptor endocytosis. This Chapter illustrates differential receptor complex trafficking by the RAMP2 and RAMP3 isoforms, suggesting a mechanism for the differential trafficking of the CL-R/RAMP complex observed in different cell lines. A model for RAMP expression determining receptor fate after agonist-induced internalization is proposed. Concluding this thesis, chapter six, offers a list of the major hypotheses tested and the experimental results obtained in each study. Also discussed are the limitations of this study and the positive outcomes of the thesis work, focusing on the contributions of these findings to the field of receptor biology and future directions. 2. LITERATURE REVIEW: 2.1. Adrenomedullin. 2.1.1. Gene and protein structure. Adrenomedullin (AM) was initially isolated and Characterized by Kitamura and colleagues from a human pheochromocytoma [1]. It was found to have the ability to increase cyclic AMP levels in rat platelets and exerted strong hypotensive effects through vasodilatory activity in the resistance vessels. AM levels were also measured in the circulation with radioimmunoassays. Later, the same laboratory published the sequence of the AM gene [2]. The AM gene is located in a single locus of Chromosome 11, is comprised of 4 exons and 3 introns, and is flanked at the 5’ end by RNA polymerase II responsive TATA, CAAT, and GC boxes. The AM gene also encodes binding sites for activator protein-2 (AP-2) [3], a cyclic AMP-regulated enhancer [4], nuclear factor— KB [5], hypoxia-inducible factor-l (HIF -1) [6], hypoxia response elements (HRES) [7], and a binding site for steroidogenic factor-1 (SF-1) [8]. This multitude of binding sites for regulatory factors indicates a complex participation of the factors in the regulation of the AM gene. The AM gene encodes a 185 amino acid precursor protein termed preproadrenomedullin (preproAM), which is processed to yield the AM polypeptide and another bioactive molecule, proadrenomedullin N-terminal 20 peptide (PAMP) [9]. AM is a member of the CGRP family of peptides because of its structural homology to calcitonin gene-related peptide (CGRP), a potent vasodilator in the central nervous system and peripherally acting neurotransmitter. AM has low sequence homology with CGRP, but strong structural homology. The 52 amino acid AM peptide shares an N-terminal ring structure, formed by one intramolecular disulfide bond (between residues 16 and 21), and an amidated carboxy-terminal end with the additional members of the CGRP family of peptides: calcitonin (CT), CGRP, amylin (AMY), and newly discovered interrnedin [10, 11]. The structural integrity of the members of this family of peptides is crucial for their biological activity [12, 13]. For example, the carboxy-tenninal portion of the peptides, lacking the N-terminal ring structure, serves as selective peptidyl competitive antagonists to the respective fitll-length counterparts. Peptide fragment AM22-52 serves as a competitive blocker for AM, while a-CGRP3-37 and [3- CGRP3-37 are receptor blockers for CGRP receptors [13-15]. Given that these antagonists are peptide fragments and the limitations peptide inhibitors can impose with experimental manipulations, much effort is being placed in the development of non- peptide, selective antagonists. Thus far, BIBN4096BS, WO98/11128 (Compound 1), and SB-273779 have been Characterized as highly selective, non-peptide antagonists for subtypes of the CGRP receptor [16-18]. Similar antagonists have yet to be developed for the AM receptor. 2.1.2. Location of adrenomedullin gene and protein expression In the current literature, AM expression has been established to be ubiquitous in a wide diversity of tissues. Demonstrated with high-sensitivity radioimmunoassays and/or by immunohistochemical studies, AM expression has been reported in the adrenal medulla, heart, aorta, kidney, lung, brain, pancreas, skin, and additional tissues [1, 19- 24]. AM protein and/or mRNA expression has been detected in a multitude of cell types, as well. Some examples are cardiac myocytes [6], vascular smooth muscle cells [25, 26], endothelial cells [27], renal mesangial cells [28-30], renal proximal, distal, and collecting tubular cells [31-33], pulmonary cells [34], and various human tumor cell lines [7, 35- 37]. Expression of AM in such diverse tissues suggests the multifunctional role for AM. 2.1.3. Biological actions of adrenomedullin To date, the best-described biological actions of adrenomedullin are in the cardiovascular system. Most well-Characterized is surely the hypotensive effect of AM, which is both very potent and long-lasting. However, AM’S additional biological actions are very diverse, acting in almost any system tested, and therefore extend beyond the scope of discussion in this literature review. I will focus on the actions of AM in the cardiovascular and renal systems, the systems of particular interest to the laboratory of my thesis research. Following this discussion can be found a chart with a brief description of AM’S wide assortment of actions in additional systems (Tablel). In the vascular beds of humans, rats, cats, dogs, and sheep, AM studies have Shown a relaxation of the resistance vessels to attain a long-term drop in the mean arterial blood pressure (MAP) [38-43]. In most instances, this vasodilation is capable of attenuation by L-NAME, suggesting an involvement of a nitric oxide (N O)-dependent pathway for the effect [44-46]. Some variations have been reported in cases of different species or localization of AM stimulation [47-49]. In addition to decreasing blood pressure, AM administration will increase heart rate. NO-dependent mechanisms have also been found to be utilized by AM to dilate the renal vasculature and to mediate diuretic and natriuretic responses in the kidney [50]. In terms of the microvasculature, AM’S effects are very similar to those in the larger resistance vessels, with AM acting on CGRP or AM receptors to elicit relaxation via either activation of adenylate cyclase, release of NO, or activation of potassium channels. In addition to its potent vasomotor activity, AM plays critical roles in cell growth regulation. Its effects on cell growth and apoptosis depend predominantly on the cell type and the experimental conditions under examination. For example, AM has been well-Characterized to promote proliferation in a number of human tumor cell lines, as well as Swiss 3T3 fibroblasts, human oral keratinocytes, rat gastric epithelial cells, and human retinal pigment epithelial cells [51-55]. Conversely, in several cardiorenal cell types AM has been reported to be anti-proliferative. These cell types include rat vascular smooth muscle cells, hypertrophy in cultured myocytes and fibroblasts, rat mesangial cells, and human proximal tubule epithelial cells [56-59]. In parallel to reports for the vascular cell types, AM is expressed by a wide variety of renal cells where it is capable of yielding numerous biological effects. Based on reverse transcriptase- polymerase chain reaction (RT-PCR) studies in the rat nephron segments, Owada et al. have detected AM expression in the glomerulus, cortical collecting duct, outer medullary collecting duct, and intermedullary collecting duct, but not in the proximal convoluted tubule or medullary thick ascending limb [30]. In opposition, a different study, reported by Jensen et al., describes mRNA expression for AM and AM-R colocalized in renal vessels, glomeruli, and inner medullary collecting ducts. AM mRNA was also detected in proximal tubules, whereas AM-R mRNA was found in distal convoluted tubules [60]. The previous study also detected high levels of expression of AM in the rat mesangial cells, as well. Based on the wide expression patterns of AM in the kidney, it is not surprising that AM exerts profound effects on renal function. Studies by Ebara and colleagues demonstrated that intrarenal infusion of AM at concentrations suboptimal for heart rate and blood pressure alterations resulted in an increase in renal blood flow (RBF), total urine output, and urinary sodium excretion. At higher concentrations of AM, MAP shows marked decreases, while GFR is now increased, vasodilation of efferent and afferent arterioles occurs, and distal tubular sodium reabsorption is further decreased, further increasing sodium excretion [30, 32, 45, 61]. Later studies established these AM- stimulated renal vasodilatory and natriuretic effects were NO-dependent [45, 46, 62]. In contrast to these studies and accepted AM actions in the kidney, Leclerc et al. described a CAMP-dependent sodium-sparing capacity of AM, by a mechanism of regulating the Na+/H+ exchangers of the distal tubules [63]. Generalized Biological Actions of AM: Vascular Effects: > AM administration results in sustained hypotension via NO, CAMP, and/or PG generation (depends on vascular bed) [13, 38, 44, 64] > Positive ionotrophic and Chronotrophic effects on coronary artery [65-67] dilation Renal Effects: . > Intrarenal infusion of AM produces increases in RBF, GFR, Na+ [45, 68, 69] excretion, and urine flow > Inhibits PDGF-induced (MAPK-dependent) mesangial cell [70, 71] proliferation > Stimulates intrarenal renin release [72] Effects in Bone: > Promotes osteoblast grth and protein synthesis (yielding increased area of mineralized bone) [73, 74] Effects in Lung: > Inhaled AM reduces histamine-, acetylcholine-, and antigen-induced bronchoconstriction [12, 64] Effects in Endocrine System: > Inhibits ACTH release [75, 76] > Inhibits insulin secretion (increases blood glucose) [23] > Inhibits aldosterone production/secretion [77] Effects in Central Nervous System: > Central stimulation of the sympathetic nervous system (increases blood pressure and heart rate) [78, 79] > AM administration inhibits thirst drive and salt appetite, attenuates AVP release, and is pro-anorexic [80-82] Table 1. Biological actions of adrenomedullin. 2.1.4. Adrenomedullin expression in pathophysiological states Because of AM’S diverse biological actions, acting in almost any system tested, it is not surprising that AM’s role in pathophysiologies is quite extensive and extends beyond the scope of discussion in this literature review. I will focus on the involvement of AM in pathologies of the cardiovascular and renal systems, the systems of particular interest to the laboratory of my thesis research. Hypertension Plasma levels of AM have been demonstrated to be elevated in patients with primary arterial hypertension, with even higher levels in patients with complications of hypertension, such as left ventricular hypertrophy and nephrosclerosis [83]. AM gene expression is increased in the Dahl salt-sensitive rats fed a high Na+ diet [84], but the most dramatic elevation of AM gene expression is in the spontaneously hypertensive rats (SHR) treated with deoxycorticosterone acetate (DOCA) and fed a high Na+ diet (DOCA-salt SHR) and the stroke-prone SHR [85-87]. These two animal models also Show increased CL-R and RAMP2/3 gene expression. It has been suggested that the up- regulation of the cardiac AM system in hypertension is a protective mechanism, acting to decrease myocardial overload with the vasodilatory and natriuretic properties of AM, as well as limiting further myocardial hypertrophy and remodeling due to the proliferative regulation by AM. Heart Failure AM plasma concentration is increased in patients with congestive heart failure, and this elevation is correlative with disease severity [88, 89]. AM concentration in myocardial tissue obtained from heart transplant recipients with severe heart failure is higher than in donors, suggesting that increased plasma AM levels in heart failure patients is a result of increased myocardial production of AM [90]. In addition, AM, CL- R, RAMP2, and RAMP3 gene expression is markedly up-regulated in different animal models of heart failure (induced by volume or pressure overload) [91-95]. AM up- regulation may be acting in a protective manner, in this situation, to increase myocardial contraction due to its positive ionotrophic properties, or as a compensatory mechanism to decrease cardiac preload and afterload. Additional protective aspects of AM expression are a decrease in myocardial remodeling, due to AM’S role in attenuating myocyte hypertrophy, proliferation of myocardial fibroblasts, and production of extracellular matrix. Finally, AM acts in accordance with natriuretic peptides to counteract the vasoconstriction and sodium retention by renin-angiotensin-aldosterone, endothelin, and the sympathetic nervous system. AM infusion in heart failure patients has had beneficial effects, including an increase in cardiac output and natriuresis, reduction of peripheral resistance, pulmonary capillary wedge pressure, left ventricular end diastolic pressure and plasma aldosterone, and an increase in ejection fraction [96]. Some studies have suggested, though, that the vasodilatory and natriuretic properties of AM are impaired in heart failure, with no mechanism for the abated function apparent at the present time [41, 97]. Atheroscierosfi AM levels in the plasma of patients with chronic ischemic stroke are elevated and correlate with the extent of carotid artery atherosclerosis [98]. In addition, AM has been detected in macrophages found within atherosclerotic plaques [99]. Due to AM’s inhibitory effect on migration and proliferation of vascular smooth muscle cells, 10 my: . inhibition of endothelial cell apoptosis, and anti-inflammatory activity, it could be hypothesized that AM could have beneficial effects in atherosclerosis[100]. AM’S role in atherosclerosis, in Classic models of the disease, has yet to be tested. Data does exist that indirectly suggests AM may have atheroprotective roles, including enhanced intimal thickening observed following arterial injury in AM” mice and attenuation of restenosis with AM gene overexpression following balloon-induced or cuff-induced arterial injury in rodents [101-103]. Myocardial Infarction During the acute phase of myocardial infarction, plasma AM levels have been demonstrated to be increased, reaching their maximum after 2-3 days and returning to baseline after a duration of about 3 weeks [104]. Increased AM levels are associated with hemodynamic impairment. Evidence for this is a positive correlation between plasma AM and central venous pressure, left ventricular and diastolic pressure, and pulmonary capillary wedge pressure, and negative correlations of plasma AM with ventricular ejection fraction [105]. AM, CL-R, and RAMP2 expression increases in ischemic and non-ischemic myocardium following coronary artery ligation in the rat [106, 107]. AM gene expression is also shown to be increased with hypoxia, due in part, to an oxidative Stress mechanism [108]. AM production by myocytes may also be stimulated in the infracted region by mechanical stretch, angiotensin II, and proinflammatory cytokines. Due to AM’S effects of local coronary vasodilation and reduction of oxidative stress- induced myocardial cell injury, it is believed that AM is playing a protective role in myocardial ischemia [109, 110]. Supporting this concept, AM overexpression before ischemia/reperfusion injury will decrease the superoxide anion generation in the 11 hypoperfused myocardium, decrease the infart area, inhibit apoptosis of cardiomyocytes, and reduce the number of ventricular fibrillation incidents [111]. Additionally, when AM is infused at suboptimal levels for effect on blood pressure, it retains the ability to inhibit cardiac remodeling following experimental myocardial infarction in the rat [112]. Renal Diseases In patients with Chronic renal failure, plasma AM levels gradually increase. This is thought to be due to decreased peptide Clearance, but an elevation due to chronic volume overload cannot be disqualified. In various types of glomerulonephritis, while plasma AM levels are increased, AM excretion is decreased [113, 114]. Hypoxia is known to up-regulate AM gene expression, suggesting that AM may protect the kidney from ischemia-reperfusion injury [115]. In fact, it has been shown that in AM +/' mice, the AM deficiency aggravates histological lesions and functional impairment in experimental ischemic acute renal failure, while AM overexpression has been shown to be protective against these effects [116]. 2.1.5. Effects of adrenomedullin gene alteration and adrenomedullin gene delivery Two labs have produced AM knockout mice, and while some differences exist, AM’S role in development is very evident. Both labs demonstrated that homozygosity of the AM knockout is embryonically lethal. The first report disrupted the AM gene so that neither AM or PAMP were produced [117, 118]. The homozygous AM knockout embryos showed poorly developed vitelline vessel vasculogenesis, hemorrhages, myocardial hypertrophy, and hydrops fetalis. These mice typically died between embryonic day 13.5 and 14.5. The heterozygous AM +/' mice are able to survive to 12 adulthood and are fertile, but they are characterized arterial hypertension due to NO deficiency. The second group to generate AM knockout mice did so by placing a stop codon at the starting point for the AM coding sequence in the preproAM gene [119]. This resulted in mice that did not express AM, but Show normal levels of PAMP. While the homozygous AM mutation in these mice was also lethal (although these AM-/- mice did not show the same placental and vascular defects or hydrops fetalis), some differences were seen in the heterozygous knockout mice. These animals also reached adulthood and were fertile, but in this case, the AM +/- mice showed normal blood pressure regulation. In experimental hypertension induced with angiotensin H administration and high-salt diet, the AM+/- mice showed similar increases in blood pressure as wild-type mice, but did Show greater organ damage, due to greater hypertrophy of the left ventricle and coronary arteries, and higher vascular oxidative stress. This data indicates a role for AM in protecting against end-organ damage in Na+-induced hypertension, independent of BP regulation. When comparing the two animal models of AM knockout, it could be hypothesized that PAMP plays a primary role in blood pressure regulation (through NO- dependent mechanisms), but it must be considered that both animal models were generated using different genetic backgrounds. 2.2. Adrenomedullin Receptor Complex 2.2.1. Discovery of adrenomedullin receptor complex, The numerous biological activities of AM (discussed previously, Table 1) had been suggested to be mediated by a cell-surface receptor. Until recently, identification of 13 this receptor had been quite elusive, due to conflicting reports in both pharmacological inhibition and Cloning studies. While AM’S vasodilatory response was inhibited in some tissues with the CGRP antagonist, CGRP3-37, in other tissues no effect was seen, even at concentrations that potently inhibited the CGRP-mediated vasodilatory response [42, 120-122]. This observation led to the hypothesis that an AM-specific receptor existed. The AM-specific vasodilation was observed, for example, in guinea pig pulmonary artery, hypotensive effects in Long-Evans rats, control of aldosterone production in rat adrenals, and other AM-mediated actions [123-126]. Meanwhile, other groups demonstrated AM-stimulated responses that were sensitive to both CGRP and AM peptidal inhibitors (CGRP3-37 and AM22-52, respectively) [127]. Unfortunately, the molecular cloning attempts of the AM receptor initially yielded data equally confusing as the pharmacological inhibition studies. The AM receptor was first reported to have been Cloned from the rat lung by Kapas et al. This receptor was capable of binding 125I-AM and eliciting an elevation in CAMP in response to AM stimulation when transfected into COS-7 cells [128]. This receptor and a canine receptor, identified a few months later by Kapas and Clark, RDC-l, showed considerable homology to a previously described orphan receptor, termed L1 or GIOd [129-131]. Part of the confusion arose when subsequent attempts by other laboratories to replicate and further these studies, resulted in an inability of these labs to reproduce the previously reported observations [ 132-134]. Later an alternative for the AM receptor was proposed in studies by Aiyar and colleagues. They identified a previous orphan receptor, now termed calcitonin receptor- like receptor (CL-R), which exhibited well-characterized CGRP1 receptor pharmacology 14 with a weak AM cross-reactivity. When this human CL—R was expressed in human embryonic kidney (HEK 293) cells, a robust increase in CGRP-stimulated CAMP accumulation was observed, as was Specific ”SI-CGRP binding and dose-dependent inhibition of CAMP production by CGRP3-37 [135]. Similar results were seen for rat and porcine CL-R [136, 137]. Propagating the classification of CL-R as an orphan receptor, additional labs were unable to reproduce the results of Aiyar and colleagues in different cell lines [43, 138, 139]. CL-R was Characterized as a seven transmembrane-Spanning G- protein-coupled receptor, but its native ligand remained unidentified. Based on structural and amino acid homology, CL-R was Classified as a family B GPCR (Secretin family of GPCRS). Other members of this GPCR family include receptors for secretin, gastric inhibitory peptide, glucagon, pituitary adenylate cyclase activating hormone, vasoactive intestinal peptide, grth hormone releasing hormone, parathyroid hormone, and calcitonin. CL-R is comprised of 461 and 464 amino acids and shares 50% and 54% amino acid sequence identity with the rat and human calcitonin receptors, respectively [43, 139]. Reforming the study of this receptor system, a critical study was published in 1998 by McLatchie et al. that Clarified the elusiveness of the AM and CGRP receptors, while also providing an entirely novel form of GPCR regulation. Utilizing a Xenopus oocyte system that expressed endogenous CGRP receptor and an exogenous CAMP- sensitive cystic fibrosis transmembrane regulator (CFTR), they systematically screened complementary RNAs derived from the SK-N-MC (human neuroblastoma cell line endogenously expressing CGRP receptor characteristics) cell’s DNA library [134, 140]. This screen yielded Cloning of a 148 amino acid protein, termed receptor activity 15 modifying protein-l (RAMP-l), that was capable of significant CGRP-mediated CAMP accumulation in the oocytes. Yet when expressed in HEK 293T, COS-7, or Swiss 3T3 cells, this protein was unable to elicit a CGRP-stimulated CAMP response. Dual expression of CL-R and RAMP-1 was found to restore the CGRP-mediated CAMP production in oocytes and HEK 293T cells. This presumably was the result of these cells not expressing endogenous CL-R, so both the receptor and RAMP expression were required for the CAMP response to be observed. Database search identified two additional isoforms of RAMPS (RAMP-2 and RAMP-3), which together the RAMP proteins only Show approximately 31% sequence identity. Repeating similar experiments with RAMP-2/3 in oocytes, as performed for RAMP-1, yielded a quite surprising result. For the first time, a GPCR showed a differing receptor phenotype when expressed with different accessory proteins. Namely, when CL-R was expressed with RAMP-1, a CGRP receptor was produced, whereas when CL-R was expressed with RAMP-2 or RAMP-3 the receptor was responsive to AM (AMI receptor and AM2 receptor, respectively) [134]. These results were confirmed by a number of additional labs in human endothelial and vascular smooth muscle cells, rat osteoblast-like UMR-106 and COS-7 cells, and Drosophila Schneider 2 cells [141-143]. This was a revolutionizing concept for the GPCR field. Notably, while RAMP2 and RAMP3 share only approximately 30% sequence identity, they generate virtually pharmacologically and biologically identical AM receptors when CO-expressed with CL-R in HEK 293T cells [144]. Until recently published by our laboratory, no other laboratories detected differences in receptor phenotype or signaling between the AMlR and AM2R. Additionally, it was presumed 16 that differences in receptor regulation must exist between the two receptor isoforms. A report from our lab recently described the differential regulation of RAMP2 and RAMP3 expression by platelet-derived grth factor (PDGF) and lipopolysaccharide (LPS) in rat glomerular mesangial cells [145, 146]. Moreover, RAMP expression studies in animal disease models Show variable RAMP2 and RAMP3 mRNA expression (see section 2.2.6). 2.2.2. RAMP gene and protein structure RAMP-1 and RAMP-2 were initially cloned from human neuroblastoma cell (SK- N-MC) DNA library, whereas RAMP-3 was isolated from the human spleen. Comparison of sequences of the RAMPS with the genomic map predicted their Chromosomal location, as well as gene organization. RAMP-1 was shown to reside on Chromosome 2, the RAMP-2 gene on chromosome 17, and RAMP-3 gene is on chromosome 7 [134]. A scan of the human genome revealed no more sequences similar to the RAMPS [147]. RAMP-1 and -3 share some similarities in their gene composition, being comprised of three exons divided by large introns. The RAMP-l and -3 genes are large in comparison to that of RAMP-2 (approximately 24 kilobases vs. 5 Kb, respectively). RAMP-2 has four exons, but shorter introns regions. All three genes have similar localization of the 5’UTR and the signal peptide sequence on the first exon and the C-terminal and transmembrane domains on the last exon of the corresponding RAMP genes [148]. Despite the relatively low amino acid sequence identity between the RAMP isoforms (approximately 30%), the hydrophobicity plot analysis suggested a substantial l7 similarity of protein structure. The sequence similarity of the RAMP isoforms between species is quite well conserved, at approximately 90% between mouse and rat. Sequence identity between the rodent and human sequences is 70, 65, and 85% for the RAMP], RAMP2, and RAMP3, respectively [149, 150]. RAMP-l and RAMP-3 are 148 amino acid proteins, while RAMP2 is composed of 175 amino acids, but all isoforms are made up of large extracellular domains, an approximately 20 amino acid transmembrane domain, and a roughly 10 amino acid intracellular domain [134]. The RAMPS have a molecular weight of only approximately 14-17 molecular weight. Several sequences and/or residues are conserved over the RAMP isoforms and different species, indicating an importance in regulatory functions or preservation of secondary structure. Included are four cysteine residues located in the extracellular domain and two sequences localized to the N-terminal and C-terrninal regions, respectively: DPPXX and LVVWXSK [134]. Several consensus Sites for N-glycosylation have also been identified on RAMP2 and RAMP3. Four N-glycosylation Sites identified on human, mouse and rat RAMP3 are conserved in the mouse RAMP2. Suggesting differential post-translational modifications of the RAMP isoforms, RAMP] has no consensus sites for N-glycosylation [151]. Protein kinase A and C phosphorylation consensus sites also are present on the C- terrninal intracellular domains of RAMP] and RAMP3, but not RAMP2 [150, 152]. Our laboratory has also recently reported a function for the previously-identified PDZ recognition motif on the extreme C-terminus of RAMP3, but not RAMP] or RAMP2, that is responsible for protein-protein interactions important in the regulation of trafficking of the receptor complex [153, 154]. Further investigation is Clearly required 18 to determine the functional consequences of the discussed conserved regulatory sequences. 2.2.3. Tissue and cell Specific RAMP expression RAMP mRNA distribution has been analyzed in several species to date, including mouse, rat, and human, with differences existing, but general expression patterns have been established. RAMP-l has shown predominant expression in the heart, brain, skeletal muscle, thymus, spleen, fat, and kidney; RAMP-2 expression is abundant in the heart, aorta, kidney, spleen, fat, and lung. RAMP-3 shows the widest distribution, but tends to exhibit prevalent expression in the kidney, heart, brain, and lung [134, 148-150, 155,156] In addition to tissue distribution, the RAMPS’ expression has been studied in a variety of cell types. Differential expression of RAMP isoforms in cell types has been reported, especially in the initial characterization of the CGRP and AM receptors, leading to the identification of CL-R, a previous orphan receptor capable of Signaling through both ligands, depending on RAMP expression. Endogenous expression of RAMPS in a particular cellular background has been well-documented to affect the functional character of the observed receptor subtype. Endogenous expression of the RAMPS also affects the functional receptor observed when overexpressing RAMP isoforms in a cell type. For example, rabbit aortic endothelial cells (RAECS) endogenously express RAMP-2, but not RAMP-l, and are selectively responsive to AM and blockade by AMzz- 52, but not CGRP3-37. While transfection of hRAMP-l into RAECs will yield a CGRP response that is selectively inhibited by CGRP3-37, overexpression of hRAMP-l and 19 hRAMP-3 into the RAECS results in decrease of CGRP-mediated CAMP response. This work suggests a greater affinity of the rabbit CL-R for RAMP-3 as compared to RAMP- ]. However, the overexpression of hRAMP-3 does not alter the responsiveness of the CL-R in RAECS to AM, suggesting an intraspecies preference for RAMP isoforms [157]. Variable RAMP isoform-CL receptor affinity has been demonstrated in additional cell types, namely UMR 106-06 cells and COS-7 cells [139, 142]. RAMP distribution has also been elucidated in several organ systems to date, namely the central nervous system and the kidney. In the central nervous system, using in situ hybridization to detect expression, RAMP] mRNA was predominantly expressed in the cortex, caudate putamen and olfactory tubercles; RAMP2 mRNA was most abundant in hypothalamus; and RAMP3 expression was restricted to the thalamic nuclei [158]. Notably, in specific brain areas only a single RAMP isoform was often detected, suggesting mutual exclusivity in expression. Of particular interest to our laboratory, renal RAMP mRNA expression has been evaluated. RAMP-2 and RAMP-3 mRNA has been detected in abundance in the rat kidney, showing similar expression patterns in both the cortical and medullary parts of the kidney [159]. Using quantitative RT-PCR, Totsune and colleagues measured the mRNA expression levels of RAMP-2, RAMP-3, and CL-R in the kidney of normal Munich-Wistar rats. RAMP-2 expression was 26.5i1.9 mmol per mole of GAPDH and RAMP-3 mRNA levels were 7.73:0.4 mmol per mole of GAPDH. CL-R was detected in the kidney in this study, but at significantly lower concentrations [160]. Our lab has characterized RAMP and CL-R mRNA expression in several renal cell lines. Please see the chart below for RAMP isoform and CL-R distribution (mRNA expression) in the various renal cell lines. The differential 20 expression of the RAMPS (especially RAMP-2 and RAMP-3) in the different regions of the kidney suggests additional, yet to be identified, regulatory roles for the RAMPS. CELL LINE RAMP-l RAMP-2 RAMP-3 CL—R Human primary proximal tubule cells + + + + Rat mesangial cells + + + + Rat kidney fibroblasts + + + Rabbit cortical collecting duct cells + + Table II. RAMP isoform and CL-R receptor expression in renal cell lines. 2.2.4. Mechanism of RAMP-receptor interaction Several studies have now established that RAMPS initially interact with their receptor partners in the ER/Golgi and maintain this association throughout the life-cycle of the receptor complex. The RAMP and receptor are trafficked together to through the endocytic pathway to the recycling endosome or lysosome after agonist activation of the receptor [153, 161-164]. The interaction of the RAMP isoform with CL-R results in modification of the terrnina] glycosylation of the receptor [134, 163]. RAMP-l and RAMP-2 are both known to modulate CL-R through glycosylation, although only the RAMP-2/CL-R complex requires glycosylation for expression of the receptor complex at the cell surface [163]. While glycosylation may determine cell surface expression for the receptor complex, glycosylation is not required for determination of receptor phenotype by the RAMPS. 2] Experiments in insect cells, where glycosylation state of the receptor is not altered, Show no alteration in ligand recognition or receptor activation, as compared to similar studies in mammalian cell lines [14]]. Evidence exists that indicates that the N-terminal, extracellular domain of the RAMP is critical to determination of receptor phenotype, while the transmembrane domain is important in the stabilization of the RAMP-receptor interaction [144]. Chimera studies with the RAMP-1 and RAMP-2 extracellular domains suggest that the N-terminal regions of the RAMPS determined ligand selectivity of CL-R [165]. This work surprisingly suggests that the extracellular domains of RAMPS are sufficient to maintain a fully functional receptor for CGRP. Chimera of the extracellular domain of RAMP-1 with the transmembrane and intracellular domains of PDGF-R only showed a ten-fold decrease in potency for CGRP signaling and binding. However, the extracellular domain alone showed a greatly diminished (approximately 4000-fold) responsiveness to CGRP. These studies aren’t entirely consistent with data reported by Steiner et al. with respect to the transmembrane domain of RAMP-1 [166]. Steiner and colleagues reported that deletion of only a portion of the RAMP-1 transmembrane domain caused a dramatic decrease in the potency of CGRP, regardless of functional trafficking of the receptor to the cell surface. Further work will be needed, possibly studying the extracellular and transmembrane interactions of RAMP-2 and -3 as well, to determine the interactions necessary for receptor trafficking, activation, and heterodimer stability. AS mentioned above, a role has been indicated for the transmembrane domain of RAMPS in the stabilization of the RAMP interaction with CL-R and the calcitonin receptor (CTR). Chimera experiments with RAMP-1 and RAMP-2 defined the 22 extracellular domain as having the responsibility for receptor phenotype determination. Because of a selective recognition of amylin by RAMP-1 or RAMP-3 complexed with the CTR, RAMP-1 and -2 chimeras is an excellent tool to delineate the domains responsible for agonist binding, receptor activation, and heterodimer stability. Levels of amylin binding, induced with the wild-type RAMPS, were paralleled by the chimeras according to the transmembrane domain/C-terminus present in the heterodimer [140]. This led to the theory that the transmembrane domain of the RAMPS was the primary interaction site for the RAMP and receptor (at least for the CTR). This model may be rather simplistic or receptor-specific, though, in light of additional recent studies on the N-terrninus of RAMP-1 that have identified a stretch of aromatic residues within RAMP- ] that are probably important in the interaction between RAMP-1 and the CL-R. These N-terrninal residues (F93, YIOO, or F101) act independently of determination of receptor phenotype [167]. While not altering the ECSO value for the CGRP-mediated CAMP response, mutation of any of the above aromatic residues to alanine rendered the receptor complex unable to express at the cell surface and bind agonist (CGRP). In addition, recent work by Sexton and colleagues, with a more diverse set of RAMP-U2 chimeras, indicates a strong induction of amylin binding, even in the presence of the transmembrane domain of RAMP-2 [168]. Further work is required to establish the regions responsible for interaction between the RAMP and receptor, with differences possibly existing for different receptor partners of the RAMPS. Segments of all three RAMPS have been identified that are required for generation of a functional receptor phenotype. On RAMP-2 the extracellular region from amino acid 77 to 101, in particular residues 86-92, are critical for adrenomedullin receptor 23 phenotype when the RAMP is expressed with CL-R [169]. The equivalent region on RAMP-3, residues 59-65, is also required for AM receptor phenotype, although no homology between the crucial sequences of RAMP-2 and RAMP-3 exists. This suggests that this sequence plays a structural role in the RAMPS or this region possesses an allosteric effect on CL-R conformation. Data from RAMP-l deletion studies suggests a role for these short sequences in the extracellular region on the proper conformation of CL-R to bind ligand [167]. Deletion of the short N-tennina] sequence from RAMP-1, residues 101-103, abolishes the induction of CGRP receptor phenotype by the receptor complex, but if the residues are replaced with alanine residues, the potency of CGRP remains unaltered. Disulfide bond formation also appears critical to ligand binding and receptor activation for the RAMPS and their heterodimeric partners (CL-R and CTR). While four cysteine residues are conserved between all three RAMP isoforms, RAMP-1 and RAMP- 3 contain an additional 2 conserved cysteine residues [170]. Data from Flahaut et al. suggests that all cysteine residues on RAMP-3 are crucial for ligand binding and receptor » activation. It could be speculated that the additional disulfide bond formation on RAMP- ] and RAMP-3 allow for the increased affinity of the RAMP-l/CL-R and RAMP-3/CL-R (to a lesser extent) receptor complexes for CGRP. 2.2.5. Additional RAMP-interacting receptors One of the first questions raised after the discovery of RAMPS and their role in trafficking and ligand selectivity of CL-R was if additional receptors can interact with the RAMPS. Given the Significant sequence identity between the CTR and CL-R, the CTR 24 was an obvious first Choice. Strong evidence exists to establish that RAMP association is not required for CTR binding and activity induced by calcitonin [161, 17]]. However, RAMP-l and RAMP-3 isoforms were found, by several independent laboratories, to interact with the CTR to form a high affinity amylin receptor. These results were confirmed in exogenous systems in COS-7 and rabbit aortic endothelial cells and with an endogenous CTR in Chinese hamster ovary (CHO)—Kl cells [161, 172]. RAMPS have a relatively ubiquitous distribution, with at least one RAMP expressed in most tissues examined to date. Noteworthy, this distribution extends beyond that of the Characterized receptor partners (CL-R and CTR), suggesting a more widespread role for RAMPS in receptor regulation [134, 148, 149]. The discovery of the RAMPS led to an interest in the potential of the RAMPS to interact with additional GPCRS to determine cell surface expression or receptor phenotype, especially for receptors that had been thus far difficult to Characterize. RAMPS were thought to maybe he the missing link in pairing numerous orphan receptors with their ligands. A recent study by Christopoulos et al. identified additional interacting receptors for the RAMPS by screening the capacity of the receptors to traffic the RAMPS to the cell surface [134, 161, 172, 173]. Screened in this study were 10 of the GPCRS of the family B of GPCRS, the family to which CL-R and CTR belong. Of the 10 GPCRS tested, 6 of the receptors were capable of trafficking at least one of the RAMP isoforms to the cell surface [174]. In addition to CL-R and CTR, the newly identified RAMP-interacting receptors include the vasoactive intestinal peptide/pituitary adenylate cyclase activating peptide (VPAC)-l receptor, parathyroid hormone receptor (-1 and -2), and the glucagon receptor. While the CL-R, CTR, and VPAC-l receptors interact strongly with all three RAMP isoforms, the 25 PTHIR and glucagon receptor only interact with RAMP-2, and PTH2R only interacts with RAMP-3. The VPAC2, glucagon-like peptide (-1 and -2), and growth honnone- releasing hormone receptors were unable to interact with any of the RAMP isoforms [174]. This study, while suggesting a more widespread role for the RAMPS in receptor regulation, also reveals additional specificity of RAMP-receptor interactions. Of note, with the exception of CL-R, RAMP association with interacting receptors is not required for Chaperoning of the receptors. The most compelling consequence of the newly-discovered RAMP-receptor interactions is the augmentation of VPAC] receptor-mediated signaling [174]. RAMP-2 interaction with VPACl-R does not alter the agonist binding properties or CAMP signaling of the receptor, however RAMP-2 association with the VPACl-R does enhance the receptor-mediated phosphoinositol hydrolysis pathway. While no Change is seen in the EC50 for the agonist- mediated PI hydrolysis, the maximal VPACI-mediated PI signaling (Emax) is increased. The physiological relevance of this observation has yet to be defined. The data reported in this project suggests additional roles RAMPS may be fulfilling with their interacting receptors. Beyond roles in cell surface trafficking and receptor phenotype determination, results from this study propose roles for RAMPS in sorting of receptors after ligand activation to determine the receptor’s fate. 2.2.6. Regulation of RAMP gene expression The Changes in RAMP gene expression have been studied under many disease models, physiological Changes, and drug treatment. Below is a table summarizing the findings of these studies, focusing on the cardiovascular and renal system Changes (Table 26 III adapted from review by Udawela et al.) [168]. Dramatic expression pattern alterations are seen, indicating an important role for dynamic RAMP regulation in these systems. These results suggest the potential for RAMP regulation as a means of modulating some of the pathophysiological conditions associated with the RAMP- interacting GPCRS. 27 32:25.35; - m - Model Tissue R1 R2 R3 CL-R AM Ref. Exp. Exp. Exp. Exp. Exp. Heart Failure (HF) in Atrium N/D T T N/D TT TT [94] rats (coronary artery Ventricle N/D T N/D T TTT ligation) Kidney N/D -- N/D -- -- Rat ischemic heart Non-ischemic N/D T N/D N/D T [ 107] failure (left coronary left ventricle ligation) Ischemic LV N/D T T T N/D N/D T T T LV hyptertrophy to LV at N/D T T T T T T T T T [93] heart failure in rat (KCl hypertrophy Lnjection) LV at HF N/D TT T TT T T T TT T Chronic HP in rats Atria T -- TT T -- -- [91] (aortic banding) Ventricles T T -- T T -- -- Myocardial ischemia in Myocardium N/D T N/D N/D TTT [175] rats (isoproterenol Aorta N/D T N/D N/D T T induced) Blood pressure changes PVN N/D ll N/D N/D -- [176] in rat Nucleus of N/D ll N/D N/D -- Increase by solitary tract phenylephrine PVN N/D -- N/D N/D I I Decrease by Nucleus of MD TT T N/D N/D -- nitroprusside solitary tract Hypertensive rat [85] Salt loaded Renal cortex N/D 111 TT 1 -- Renal medulla N/D I I -- -- -- DOCA treated- salt Renal cortex N/D 1 -- -- -- loaded Renal medulla N/D 11 T T -- T T DOCA- salt loaded LV N/D T T T T [86] Chronic salt loading in Adrenal gland T T T -- T T T T [177] rat Kidney -- -- n t n SHR LV N/D T -- T T [87] Ang 11 treatment in rat Cardiomyocytes TT -- TT -- N/D [178] Endothelin treatment Cardiomyocytes -- l L T T [179] Hypoxic rat Lung T T T -- T -- N/D [180] Hypoxia [MR-32 N/D 111 N/D N/D T T [18 1] NB-96 N/D -- N/D N/D T T Renal injury in rat Kidney (remnant N/D -- 11 11 -- [160] (mass ablation) tissue) Renal injury in rat Kidney T T T T T -- T T -- [150] (obstructive nephropathy) Renal fibrosis in rat Kidney T T T LT -- T -- [1 82] Table III. Regulation of expression of RAMP isoforms and CL-R in various pathophysiologies and animal models. (N/D, not done; --, no Change). 28 2.2.7. Receptor Component Protein (RCP) During the process of trying to Clone the CGRP receptor, Dickerson and colleagues identified a novel 148 amino acid protein that appeared to play a role in the regulation of CGRP signaling [183]. This protein was identified with a system Similar to that used to identify the receptor activity modifying proteins, a system utilizing the PKA activation requirement of CFTR to register CAMP responses to particular ligands [134]. In this case, CGRP stimulation allowed the Cloning of a protein later named CGRP receptor component protein (RCP). RCP, now cloned in several species, Shows highly conserved (82%) protein sequence homology between species thus far sequenced [184, 185]. While RCP is detected primarily in membrane fractions prepared from cells, it shows no amino acid structure for hydrophobic regions or consensus motifs for lipid attachment [186]. This data concluded that RCP is a cytosolic, yet peripheral membrane protein. RCP has also been shown to be immunoprecipitated with RAMP] and CL-R in complex from NIH3T3 cells or lysates of the cerebellum [186, 187]. There are no conserved glycosylation sites in the RCP sequence, but several conserved protein phosphorylation sites do exist [184, 185]. This raises the possibility of regulation of RCP function in an in vivo setting. RCP expression has been studied in several in vivo settings, with expression correlating to tissues known to contain CGRP receptors. CGRP is present in the spinal cord in primary afferent fibers and mediates nociception [188]. In these tissues, RCP expression is found juxtaposed to that of the CGRP receptors [189]. CGRP receptors are also known to be expressed on the walls of blood vessels, in this situation mediating 29 vasodilation, and again RCP expression is found in this tissue, correlating with a role for RCP in CGRP signaling and biological effects [184]. The role of RCP was tested in NIH3T3 cells, a cell line that endogenously expresses the CGRP receptor system, as well as RCP. RCP expression was inhibited in these cells, to test the affect, with antisense constructs for RCP. The antisense inhibited the expression of RCP to that below detection with Western blot. Loss of RCP in the NIH3T3 cells did not affect either the affinity or the receptor density of CGRP receptor in the antisense cells, as determined by radioligand binding with 125I-CGRP [186, 187]. Interestingly, the RCP antisense cells showed a 70% reduction in CAMP response, as compared to wild-type NIH3T3 cells. Recent results from the Dickerson laboratory have suggested a role for RCP in adrenomedullin signaling, as well [190]. Lack of RCP expression did not appear to have any effect on the signal transduction for other GPCRS, including the Bz-adrenergic receptor or the A21, adenosine receptor [186, 187]. These results suggested that RCP was not working in a Chaperoning capacity, but had more influence on coupling of the receptor to its signaling pathway. The model proposed by Dickerson et al. for a functional CGRP receptor includes the CL-R receptor, RAMP], and now RCP. More work is needed to establish the role of RCP in the coupling of the CGRP receptor to its respective G protein. 2.3. GPCR Life-cycle Regulation 2.3.1. GPCR life-cycle G protein-coupled receptors (GPCRS) are known to convey environmental signals to the intracellular environment via heterotrimeric G proteins to affect the cellular 30 behavior in response to a stimulus. Their form of regulation had in the past been thought of as Short-term control, whereas more recently they have been shown to play roles in longer-terrn regulation of such physiological processes as proliferation, apoptosis, cellular migration, and hypertrophy [191, 192]. Their long-term regulation is partially a result of the desensitization and endocytosis processes and the role of these processes in the intracellular signaling. This attenuation of signaling and removal of the receptor from the plasma membrane can decrease the receptor numbers capable of signaling for minutes to hours, depending on the cell type and receptor. Thus, desensitization is the process of an attenuation of Signaling as a result of prolonged exposure of the receptor to its agonist (see Table IV and Figure 1 below). Desensitization of a receptor often signals for the removal of the receptor from the plasma membrane by the pinching off of the plasma membrane and internalization of the receptor, termed endocytosis. Many forms of regulation control these processes, some of which being phosphorylation of the receptor intracellularly, interaction of the receptor with additional proteins (caveolin, arrestin molecules), and interaction of the receptor with the endocytic machinery (Clathrin, dynamin, adaptor protein-2 (AP-2), etc.). The receptors, regardless of the type, are often endocytosed into the early endosomes where their fate is determined. From this point receptors are targeted for recycling to the plasma membrane or sorted for degradation in the lysosomes. Protein-protein interactions and post-translational modifications are being elucidated that serve as sorting Signals from the early endosome, to target proteins for either recycling to the plasma membrane to promote receptor Signaling or shuttling of the receptor to the lysosomes for proteolytic degradation and a down-regulation of the 31 receptor. The factors that determine the regulation of the GPCR life-cycle will be discussed in the following sections. Figure 1. Depiction of GPCR life-cycle (modeled for prototypical GPCR life-cycle of the B2- adrenergic receptor) [193]. 32 Term: Definition: Desensitization Attenuation in receptor Signaling in response to prolonged agonist exposure Endocytosis Uptake by a cell of material from the environment by invagination (infolding) of its plasma membrane Intemalization Transportation of cells or soluble material into the cell via a vacuole/vesicle Resensitization Return of unbound receptor to plasma membrane, ready for ligand stimulation and receptor Signaling Recycling Targeting of receptor from endosome for return pathway to plasma membrane for continued signaling Degradation Targeting of receptor from endosome for pathway to lysosomes/proteosomes to be degraded Down-regulation Trafficking of receptor for internalization and degradative pathway to promote decrease of available receptors at plasma membrane Table IV: Terminology for steps in GPCR life-cycle. 2.3.2. Regulation of receptor desensitization A prolonged exposure of cells to a particular ligand results in an attenuation of responsiveness to subsequent stimulation with that ligand. This phenomenon is called desensitization, which can be homologous or heterologous. Homologous desensitization is Classified as a decrease in the response that is specific for the stimulated receptor. Because G-protein regulated kinases (GRKS) are activated and only phosphorylate GPCRS in the agonist-bound state, GRKS are capable of attenuating receptor signaling by homologous desensitization [194, 195]. An attenuation of receptor signaling that is the result of second messenger Signaling, and in principal not specific for the activated 33 receptor, is termed heterologous desensitization. Protein kinase A and C are kinases activated through second messenger signaling of GPCRS, and therefore can activate heterologous receptor desensitization [196]. The most comprehensive study of desensitization, thus far, has been in elucidating the molecular mechanisms of this process for the B-adrenergic receptors [195, 197]. Using concepts from the examination of desensitization of the B-adrenergic receptors, it has been Shown in numerous membrane-bound receptor systems, particularly GPCRS, that phosphorylation is an integral step in the attenuation of receptor signaling, termed desensitization. Virtually all GPCRS thus far studied have specific serine, threonine, and/or tyrosine amino acid residues in the third intracellular loop or C-terminus of the receptor that require phosphorylation for the efficient desensitization of the receptor. In most cases, phosphorylation of these key residues allows for interaction with non-visual arrestins. This interaction disrupts the coupling of the receptor to the G-protein, thus halting signaling of the receptor through the G-protein uncoupling. In addition, B-arrestin interaction with the GPCR promotes Clathrin-mediated receptor internalization. Without phosphorylation of required residues on the third intracellular loop or C-terminus of the GPCR, signaling will be maintained and desensitization of receptors is attenuated, or prevented. The mechanism of desensitization of CL-R has been studied in several cell lines to date. Desensitization of CL-R was found to involve protein kinase A activation in rat mesangial cells (RMCS) stimulated with AM, SK-N-MC (neuroblastoma cell line) cells stimulated with CGRP, and vascular smooth muscle cells (VSMCS) stimulated with CGRP [198-200]. Whereas, in HEK 293 cells, desensitization of the CGRP receptor 34 (CL-R/RAMPI) has been shown to be dependent on the activity of G-protein receptor kinase(GRK)-6 [201]. In HEK 293 cells, CL-R has been shown to be phosphorylated when stimulated with agonist (AM or CGRP) via in vivo phosphorylation assays [202]. Given the published data for the B-adrenergic receptor and additional GPCRS supporting a phosphorylation-dependent mechanism for receptor desensitization, and the above- described studies with CL-R, it is hypothesized that phosphorylation of CL-R is required for receptor desensitization after prolonged agonist exposure. The studies in this project will focus on the requirement of phosphorylation in the desensitization process for the AM receptor subtypes. With respect to physiological effects of the desensitization process, the inhibition of the process of desensitization for particular GPCRS has been Shown to elicit protective therapeutic effects. Leflcowitz et. al. have Shown that the inhibition of B-adrenergic receptor kinase (kinase that phosphorylates the B-adrenergic receptor to cause desensitization) in the heart can delay the development of heart failure in multiple animal models, in some cases even restoring cardiac function [203, 204]. Others have also shown inhibition of desensitization of the p-opioid receptor to be beneficial in preventing morphine tolerance [205, 206]. Because of the potential therapeutic effect of prolonged AM signaling in a system where it exerts protective effects, understanding the regulation of the desensitization process for the AM receptor is crucial. 2.3.3. Regulation of receptor internalization/endocytosis Predominantly, endocytosis of G-protein coupled receptors is enhanced by agonist binding, whereas many nutrient receptors, such as the transferring and LDL receptors are 35 constitutively endocytosed regardless of ligand occupancy status [207-209]. The best- characterized pathway for receptor internalization is mediated by Clathrin-coated vesicles. Often, clathrin-mediated internalization of mammalian GPCRS requires agonist stimulation and interaction with [3-arrestin molecules. The B-arrestin proteins not only disrupt G-protein coupling with the receptor, but also serve as adaptors to link the receptor to the endocytic machinery [210, 211]. Clathrin-mediated endocytosis generally also requires the activity of dynamin GTPaseS for proper vesicle formation and cleavage from the plasma membrane [212]. A second pathway for receptor internalization is now emerging. Data now confirms that receptors are internalized via non-coated vesicles, such as the flask-shaped caveolae and other pinocytic mechanisms [207, 212]. Caveolin-mediated internalization also requires the activity of dynamin, but internalization mechanisms independent of dynamin have even been suggested [212, 213]. In fact, internalization of a GPCR by different endocytic pathways in single cell has even been demonstrated for the Cholecystokinin (CCK) receptor [214]. Caveolin-mediated endocytosis involves receptor complex localization in a lipid raft domain enriched in caveolin proteins. These domains are termed caveolae. Intemalization of receptors by the different pathways may be determined by the phosphorylation state of the receptor. The [3 l-adrenergic receptor has been demonstrated undergo agonist-stimulated internalization via caveolae when phosphorylated by protein kinase A and internalization by Clathrin-Coated pit endocytosis when phosphorylated by GRKS during the desensitization process [215]. The mechanism of internalization may also have an important effect on the endocytic sorting of the receptors. The receptor tyrosine kinase, TGF-B, is degraded when internalized by a 36 caveolae-mediated mechanism, but is recycled to promote receptor signaling when internalized by Clathrin-Coated pit endocytosis [216]. NHERF-l, PDZ domain-containing protein discussed previously, has been shown to associate with the epidermal growth factor receptor (EGFR), a receptor tyrosine kinases (RTK) to regulate receptor internalization. In this case, NHERF-l association acts to stabilize the receptor at the plasma membrane and decrease the endocytosis rate of the receptor [217]. NHERF-l has also been shown to attenuate the constitutive endocytosis of the PTHl-R by a mechanism of tethering the receptor to the actin cytoskeleton through protein-protein interactions [218]. Using an endogenous system, I have also shown in this project that NHERF-l is essential to ‘hold’ the receptor-complex at the membrane, the absence of which leads to internalization of the receptor complex. These studies suggest a critical role for protein-protein interactions and NHERF-1 in regulation of receptor internalization and maintenance of receptor numbers at the plasma membrane. While many GPCRS utilize similar mechanisms for endocytosis, the functional consequence of endocytosis varies from receptor to receptor. Intemalized receptors that are trafficked through a rapid recycling pathway are restored to the plasma membrane in a functional state to achieve resensitization. On the other hand, receptors that are internalized and targeted to late endosomes and lysosomes experience proteolytic degradation, thus promoting attenuation of receptor signaling and down-regulation of the receptor. 37 2.3.4. Regulation of receptor recycling vs. degradation After internalization, factors influencing the sorting of receptors in the early endosome are largely unknown, but some of the critical players are beginning to be identified for the GPCRS. Receptor ubiquitination, interaction of the receptor with PDZ domain-containing proteins, and/or interaction of the receptor with additional, newly- identified proteins has been shown to be required in some GPCR systems for efficient targeting of the GPCR for either degradation or recycling pathways [219-221]. Agonist-induced endocytosis is a means of regulating signaling for a multitude of membrane-bound receptors, particularly for G-protein coupled receptors. For some receptors in distinct cell backgrounds, internalization is a means of rapid recycling, characterized by dephosphorlyation and dissociation from agonist in intracellular vesicles for restoration at the plasma membrane in a functional state to achieve resensitization. On the other hand, some receptors that are internalized and targeted to late endosomes and lysosomes experience proteolytic degradation, thus promoting down-regulation of the receptor at the plasma membrane and attenuation of receptor signaling. Much effort is currently being placed on the signal that sorts the internalized receptors for recycling or degradation. Ubiquitination of GPCRS has recently been shown to contribute to the sorting fate of the receptors for degradation. Ubiquitination is a process that involves the addition of multiple ubiquitin molecules on targeted lysine residues of a protein marked for degradation by an ubiquitinating enzyme complex. This addition of ubiquitin subunits to a particular protein acts as a degradation sorting signal for the degradation machinery [219, 222]. Much of the mechanism in the recognition of the degradation signal is unknown, but the mechanism for the addition of ubiquitin 38 molecules to targeted lysine residues on proteins has been determined. Ubiquitin addition to a protein is a result of the coordinated activity of three enzymes: the ubiquitin- activating enzyme (E1), ubiquitin—carrying enzyme (E2), and the ubiquitin ligase (E3). Marchese and Benovic have recently shown that ubiquitination of the Chemokine receptor, CXCR4, by the ubiquitin E3 ligase, atrophin-interacting protein 4 (AIP4), is mediated by lysine residues in the C-terminus of the receptor. In this study, mutation of lysine residues to arginine on the C-terminus of the receptor inhibits ubiquitination and subsequent receptor degradation, but did not alter the internalization of the receptor upon agonist binding [219, 223]. Irnmunoprecipitation with an ubiquitin antibody also showed that the receptor was ubiquitinated, as well. In addition to the CXCR4 receptor, the [32- AR has been shown to be targeted for degradation upon ubiquitination of the receptor. Leflcowitz’s group also showed that rapid ubiquitination of associated B-arrestin plays a role in the internalization of the ubiquitinated BZ-AR. Mutations of lysine residues on the C-terminus of the receptor not only inhibited the degradation of the receptor, but also promoted the recycling of the receptor back to the plasma membrane after agonist- induced internalization [220]. It has been shown in several GPCR systems that interactions with PSD-95/Discs- large/ZO-l homology (PDZ) domain containing proteins are responsible for the efficient targeting of the receptor after internalization [224-226]. This is a relatively new area of research in the GPCR field, but it is currently thought that the presence of a PDZ recognition sequence on the C-terminus of the receptor is the sorting Signal for the interaction of the receptor with the PDZ domain-containing protein and targeting of the receptor for recycling to the plasma membrane from the early endosome. Recycling of 39 the [32-Adrenergic receptor (BZ-AR) is dependent on the interaction of B2-AR, via its PDZ motif, with a protein called N-ethylmaleimide sensitive factor (NSF) [224, 225]. In addition, via the PDZ domain, PDZ domain-containing protein called Na+/H+ Exchange Regulatory Factor (NHERF) interaction with the tc-opioid receptor has recently been Shown to increase the recycling efficiency of this particular GPCR [226]. It has also been shown that the 8-opioid receptor (DOR) is targeted for degradation following agonist- induced internalization, yet when a Chimera of the DOR and the C-terminal PDZ motif from the BZ-AR are treated with DOR agonist, the receptor is rapidly recycled to the plasma membrane afier endocytosis [221, 227]. This suggests that the lack of the PDZ recognition motif in the C-terminus of the DOR inhibits binding of the DOR with proteins to target the receptor for recycling, and subsequently the receptor is degraded. Recent data suggests that the C-terminus of the DOR interacts with a novel protein, termed G-protein accessory sorting protein (GASP), to target the receptor for degradation [22]], suggesting that sorting for degradation is not the default pathway for GPCRS, but is also a tightly regulated decision. While for most GPCRS the PDZ motif seems to predominantly be required for receptor recycling after agonist-induced endocytosis, the CXCR4 receptor requires an intact PDZ motif on the receptor to be targeted to the lysosomes for degradation. This PDZ recognition motif falls in the region found to be required for proper receptor ubiquitination for degradation. Disruption of this domain decreases the targeting of the receptor to the lysosomes from the early endosomes after internalization [219]. Clearly, more work is required to begin to understand the complex regulation of the sorting of GPCRS for the endosome after agonist-induced internalization. 4O As described, the fate of the GPCR is quite variable after agonist-induced internalization, with multiple GPCRS shown to both recycle and degrade, depending on the cell type and agonists interacting with the receptor. In the case of CL-R, the receptor fate differs, depending on the cell type where the receptor is expressed. It has been shown that CL-R/RAMP complex will degrade upon prolonged agonist exposure and receptor endocytosis in HEK 293 cells and Rat2 fibroblast cells, while in rat mesangial cells (RMCS) the receptor complex is effectively recycled after internalization [162, 198, 228]. The mechanism that regulates the pathway to which the receptor complex is targeted after agonist-induced internalization was previously unknown. Work from this project has proposed a model that specific RAMP isoform expression characterizes the cell to either recycle or degrade the AM receptor complex after prolonged agonist stimulation. 2.3.5. Protein-protein interaction domains/motifs Protein-protein interactions are being Shown routinely in to regulate numerous biological processes, tying together crucial partners in signaling complexes. This method of regulation has become commonplace the GPCR field. The number of protein-protein interaction domains is ever-growing and for the purpose of this literature review, I will focus on the most common interactions that have been shown to regulate GPCRS (i.e. 8H2, SH3, EVHI, WW domains). I will concentrate, in particular, on the post-synaptic density 95, discs large, zonula occludens-l (PDZ) domain interactions. This dissertation project focuses, in large part, on the role of PDZ interactions in the regulation of the life- CyCle and trafficking of the adrenomedullin receptor. 41 SrC-homology 2 (SH2) domains are modules of ~100 amino acids that bind to Specific phosphor-tyrosine (pY)-containing peptide motifs. Conventional SH2 domains have a conserved pocket that recognizes pY, and a more variable pocket that binds 3-6 residues C-terrninal to the pY, granting specificity. Phosphopeptides of optimal sequence bind to SH2 domains with dissociation constants of ~50-500 nM [229, 230]. The SH2 domain is embedded in a wide variety of metazoan proteins that regulate functionally diverse processes, and for this reason, SH2 domains must display sufficiently high off- rates for rapid and reversible signal transduction. SH2 domain interactions are responsible for the regulation of such varied processes as scaffolding, kinases, cytoskeletal regulation, phosphatases, phosphoinositide signaling, transcription, and ubiquitination, to name just a few [229]. Ste plays an active role in the agonist-induced desensitization of betaZ-adrenergic receptors via SH2 domain interactions. Src binds, via its SH2 domain, to the beta2-adrenergic receptor on an agonist-mediated phosphor- tyrosine residue, thereby allowing Src to phosphorylate and activate G-protein-coupled receptor kinase 2 (GRK2), a response obligate for agonist-induced desensitization [231]. Src-homology 3 (SH3) domains generally bind to Pro-rich peptides that form a left-handed helix, with the minimal consensus Pro-X-X-Pro. Each Pro is usually preceded by an aliphatic residue. Each of these aliphatic-Pro pairs binds to a hydrophobic pocket on the SH3 domain. From this, two Classes of SH3 domains have been defined (Class I and Class 2) which recognize RKXXPXXP and PXXPXR motifs, respectively. The ligand can, in principle, bind in either orientation. Such peptides usually bind to the SH3 domain with a Kd in the mM range [232, 233]. Dopamine (D4) receptor has recently been reported to bind SH3 domains of proteins, such as Grb2, at its proline-rich putative .42 third cytoplasmic loop. GPCRS have also been demonstrated to bind via internal SH3 domains, namely the P2Y(2) receptor, binds Src via an agonist-dependent SH3 domain interaction that facilitates Src activation, which recruits the EGF R into a protein complex with the P2Y(2) receptor and allows Src to efficiently phosphorylate the EGFR. The functional significance of these interactions is presently unknown [234]. WW domains are small 38 to 40 amino acid residue modules that have been implicated in binding to Pro-rich sequences. WW domains and SH3 domains can potentially bind overlapping sites, with both binding a left-handed poly-proline type II helix. In addition, the Pin] WW domain functions as a phosphoserine- or phosphothreonine-binding module. In its function, the WW domain shares elements of SH3 and SH2 domains by recognizing proline-rich ligands and, in some cases, by being regulated by phosphorylation. The domain name is derived from two conserved Trp residues spaced 20 to 22 residues apart within the consensus sequence. The dissociation constants (Kd) for WW—ligand complexes lie in the high nM to low mM range for proline-rich ligands, and in the low mM range for phospho-SP- or phospho-TP- containing ligands [233, 235]. W domains have attracted attention because the signaling complexes they mediate have been implicated directly or indirectly in several human diseases including muscular dystrophy, Alzheimer’s and Huntington’s diseases, and, more recently, cancer. Drosophila enabled/vasodilator-stimulated phosphoprotein homology 1 (EVHl) domains are 115 residue protein-protein interaction modules which provide essential links for their host proteins to various signal transduction pathways. Like Src homology 3, WW and GYF domains and profilin, EVH] domains recognize and bind specific 43 proline-rich sequences [233]. The binding is of low affinity, but tightly regulated by the high specificity encoded into residues in the proteinzpeptide interface [236]. Many EVHl-containing proteins are associated closely with actin-based structures and are involved in re-organization of the actin cytoskeleton. EVH] domains are also present in proteins enriched in neuronal tissue, thus implicating them as potential mediators of synaptic plasticity, linking them to memory formation and learning. For example, homer, an EVH] domain protein forms an adaptor system that regulates coupling of group 1 metabotropic glutamate receptors with intracellular inositol trisphosphate receptors and is modified by neuronal activity [237]. The discovery of the PDZ domain was based on the recognition of sequence repeats in several proteins. The first initial of these three proteins (Postsynaptic density 95, Discs large, Zonula occludens-l) fashioned the name PDZ domain. PDZ domains are quite widespread throughout the metazoans, yet the several yeast species thus far sequenced Show surprisingly few PDZ domains. Classification of PDZ domains, based on their structure and their recognition peptide sequences, has been more difficult than originally thought. Bezprozvanny and Maximov proposed Classifying PDZ domains into 25 groups, based on the nature of the residues in two positions, the (18] position and the position immediately after the [3B strand [238]. Instead, classification of PDZ domains has been focused primarily on their specificity for C-tenninal peptides. Class I PDZ domains recognize a C-tenninal sequence of X-S or T-X-V or L. Examples of proteins containing these interaction sites include the NMDAZAB receptor, the BZ-adrenergic receptor, 5-Catenin. The PDZ domain- containing proteins in this Class, sharing this interaction include PSD-95, Erbin, and 44 NHERF [239-241]. The second Class of PDZ domains recognizes a C-terrninal peptide with the sequence of X-w-X-w. Interacting proteins with Class II PDZ domains include neurexin, syndecan, and EphB2. Examples of members of the PDZ Class II domain- containing proteins are CASK, PICK], and syntenin [242-245]. The final Class of PDZ domains is Class III and classified by a X-D or E-X-w recognition sequence. The melatonin receptor interacts with the Class III PDZ domain-containing protein, nNOS [246]. In many cases, as we have observed, PDZ domain interactions are constitutive, with binding affinities in the 1 to 10 pM range. There are demonstrations for regulation of this interaction, as well. For example, the interaction of the Bz-adrenergic receptor with NHERF has been shown to be dependent on the receptor activation by agonist [240]. In addition, phosphorylation has been shown to regulate some PDZ domain interactions. For example, the interaction between the inwardly rectifying 1K+ Channel Kit 2.3 and PSD-95 is disrupted by PKA-dependent, serine phosphorylation of the channel at the -2 position [247]. Also, the Bz-adrenergic receptor, when serine-phosphorylated at its -2 position by GRKS, is unable to interact with NHERF [224]. Solving the structural basis of the PDZ domain interaction was critical in determining the specificity with which the PDZ domain-containing proteins bound their ligands. PDZ domains are comprised of 80 to 90 amino acids that make up Six [3 strands (BA-BF) and two a-helices (CIA and (1B), arranged in a compact globular structure. Several PDZ domains, both complexed with their ligands and ligand-free, have been solved for their three-dimensional structure to clarify the mechanism of the PDZ domain interactions. In short, the binding of the peptide ligand to the PDZ domain takes place in 45 an elongated surface groove as an antiparallel B strand interacts with the BB strand and the (1B helix [248]. PDZ domains have been shown in interact with their peptides in four different types of interactions: recognition of C-terminal motifs in peptides, recognition of internal motifs in peptide binding partners, PDZ-PDZ interactions, and recognition of lipids [249]. PDZ domains were originally thought to simply scaffold signaling complexes and/or receptors at the cell surface. It has become evident that PDZ domain interactions regulate a multitude of functions in cells of many types. A few examples to be discussed briefly in this review are: adaptors for tyrosine kinase receptors, epithelial polarity, and mediators of protein networks. PDZ interactions of Na+/H+ Exchanger Regulatory Factor (NHERF) have been shown to regulate the Clustering and autophosphorylation of the platelet-derived growth factor (PDGF) receptor. More potent receptor autophosphorylation is seen when the receptor is complexed with NHERF, leading to a more robust activation of MAPK signaling [250]. In terms of epithelial polarity, NHERF interactions also determine the expression of the Na+/H+ Exchanger-3 (NHE3) in the polarized proximal tubule cells and tissue. NHE3 is an antiporter responsible for the majority of the Na+ reabsorption in the proximal tubule, with the Na+/H+ exchange dependent on proper NHERF interactions to localize the transporter on the correct side of the epithelial cell for Na+ reabsorption [251]. Protein networks maintained by PDZ domain interactions are demonstrated many times over at the neural synapse. For example, through PDZ interactions, the NMDA receptor is tied to the Ca2+-ATPase Channel, the kainate receptor, the Shaker K+ Channel, and the metabotropic glutamate receptor. To portray the complexity of these interactions, in the literature the 46 postsynaptic NMDA receptor has been reported to have 17 primary interactions leading to 385 interactions with secondary proteins [249] . A major Challenge for the future will be to build models of multiprotein networks and explain the complexity and regulation of these “multiprotein machines”. 2.3.6. Na+/H+ Exchanger Regulatory Factor-l (N HERF-l) NHERF-1 was initially identified as a regulator of the Na+/H+ exchanger (N HE), mediating the activity of the transporter through CAMP-induced inhibition of NHE3. Through the identification of numerous targets of NHERF-1, some membrane-bound receptors and transporters, the role for this protein has been expanded to that of a scaffolding protein in membrane physiology, as well. NHERF-1 has been characterized as a 55 KDa protein containing two tandem PDZ domains, type I and type H domains, respectively [251]. NHERF-1 has also been shown to associate with members of the ezrin/radixin/moesin family of actin-binding proteins, and thus has the ability to link PDZ domain-interacting proteins to the actin cytoskeleton [252]. The two PDZ domains and MERM domain also allow NHERF to facilitate the formation of multiprotein signaling complexes. For example, NHERF-l is known to bind NHE3 and the PTHl-receptor, and through interactions with cytoskeletal proteins ties NHE3 to an activated PKA that phosphorylates and inactivates NHE3. Disruption of any of the previous interactions will abate NHE3 inactivation and allow improper Na+/H+ exchange [253]. Yeast two-hybrid experiments identified an additional protein that bound the C- terrninal region of NHE3. This protein shared structural homology with NHERF-1, and was thus termed NHE3 kinase A-regulated protein, or NHERF-2. Differential expression 47 of NHERF-1 and -2 has been observed throughout the kidney. NHERF-1 was detected in the proximal tubules, while NHERF-2 was detected in the glomeruli, peritubular capillaries, collecting duct principal cells, and low levels in the proximal tubules [254]. The differential expression suggests distinct physiological roles for each protein in the kidney. NHERF expression has been detected in tracheal, pancreatic, intestinal, and kidney epithelia. Determined by immunofluorescence microscopy, MERM cytoskeletal proteins are typically localized at the apical membranes of polarized cells. Because of their interactions with MERM proteins, both NHERF-1 and -2 are localized primarily in the apical membranes of kidney cells [255]. This apical localization is critical for the scaffolding of signaling complexes to function properly in a physiological setting. For example, Changes in the cystic fibrosis transmembrane regulator (CFTR) trafficking are seen in the majority (~90%) of humans with cystic fibrosis. Mutants of CFTR lacking the C-terminal PDZ interaction motif are unable to interact with NHERF and therefore Show altered trafficking of the mutant CF TR to basolateral membranes, in contrast to the wild-type CFTR expression at apical membranes [256]. Studies have implicated the interaction of CFTR with NHERF-l with the correct apical localization and proper intracellular trafficking of CFTR, and lack of NHERF-l interaction with contributing to the disease process. Several studies have shown NHERF-1 to play differing roles in various cellular processes, including receptor trafficking [257, 25 8]. NHERF-1 has been demonstrated to bind the extreme carboxy-tenninus of several G-protein coupled receptors (GPCRS), namely the Bz-adrenergic receptor, the K-opioid receptor, PTH-R, and the P2Y purinergic 48 receptor [154, 259]. Agonist exposure promotes NHERF-l association with the B2- adrenergic and the Ic-opioid receptors. NHERF-1 association with these receptors enhances the recycling of the receptors after agonist stimulation [240, 259]. In addition to GPCRS, NHERF-1 has also been shown to associate with the epidermal growth factor receptor (EGFR), a receptor tyrosine kinases (RTK). In this case, NHERF-1 association acts to stabilize the receptor at the plasma membrane and decrease the endocytosis rate of the receptor [217]. While utilizing different mechanisms, collectively these data suggest that NHERF-l interaction with both RTKS and G-protein-coupled receptors is mandatory to enhance the portion of receptors present at the cell surface. 2.3.7. N-etbylmaleimide Sensitive Factor (NSF) NSF is a 76 KDa protein with functions in vesicle exocytosis and membrane fusion events. NSF is composed of three domains: an N-terminal NSF-N domain followed by two ATP-binding domains, NSF-D] and NSF-D2. The N-terminal NSF-N domain is critical for binding to the SNAP-SNARE complex during vesicle fusion and exocytosis. Contained in the two ATP-binding domains is a 230-250 amino acid motif that is Characteristic of the ATPaseS Associated with various cellular Activities (AAA) family of ATPases, and the orientation of this domain classifies NSF as a Class II AAA protein [260]. NSF forms a homo-hexamer and requires binding of ATP for its active conformation. NSF is commonly known to interact with aSNAP (soluble NSF attachment protein) and SNARE (soluble N-ethy]maleimide-sensitive factor attachment protein receptor) proteins to form the 208 particle, a complex that plays a critical role in 49 intracellular membrane fusion and exocytosis [261-263]. Monomeric a-SNAP binds to NSF only upon binding SNARES. The C-terminal 45 amino acids of a-SNAP are required for binding and for stimulation of NSF’S ATPase activity [264-266]. The ATPase activity of NSF is responsible for the dissociation of a-SNAP and the SNARES to complete the fusion of a vesicle with the cell membrane. Factors influencing the sorting of receptors in the early endosomes are largely unknown, but interactions with PSD-95/Discs-large/ZO-l homology (PDZ) domain proteins are being suggested to regulate the receptor-targeting after internalization [259, 267, 268]. The life cycle of the B2-adrenergic receptor (B2-AR) was reported to be altered in the presence of NSF [268]. It has been shown that the B2AR interacts with NSF via a PDZ type I recognition motif (-DSLL) at its extreme C-terminus. Binding of the B2AR with NSF enhances the recycling of the B2AR alter agonist-stimulated receptor internalization. In addition, binding of NSF to the Glu2 subunits of the AMPA receptor was demonstrated to be crucial for the recycling of the a-amino-3-hydroxy-5-methyl- isoxazolepropionate (AMPA) receptor [269, 270]. NSF binds the Gluch subunit, but binding is enhanced by binding of another PDZ domain protein, PICK]. PICK], Gluch, and NSF form a tripartite complex that also binds a-SNAP to increase the disassembly of the complex upon vesicle fusion with the membrane [271]. NSF plays a Chaperoning role for SNARES in the majority of membrane fusion events in a cell, but when targeting membrane receptors for recycling, NSF acts independently of the SNARE complex to promote rapid resensitization of the receptors at the plasma membrane [261-263]. A report, published from our laboratory recently, demonstrated a role for NSF in the sorting of the AM2 receptor at the endosome for a recycling pathway. In this case, NSF ’s 50 interaction with an accessory protein (RAMP3) directs the trafficking of a receptor from the sorting endosome [153]. NSF has been shown to interact with another protein critical to receptor trafficking, the B-arrestin molecule. B-arrestin is responsible for two steps in the desensitization process for many GPCRS. First, the B-arrestin binds the receptor, upon receptor phosphorylation, and disrupts further signaling by the G-protein. Second, the B- arrestin molecule interacts with proteins to target many receptors for Clathrin-mediated endocytosis [272, 273] . B-arrestin binding to NSF appears to play a role in the turnover of the receptor at the plasma membrane. Overexpression of NSF causes an increase in the receptor Clearance from the plasma membrane after agonist-stimulation of the B2- adrenergic receptor [274]. At the present time, there is no obvious sequence that could be described as the NSF binding motif. It will be important to determine, through mutational analysis, the domains on NSF important for these protein-protein interactions. 51 3. Characterization of AM-stimulated desensitization response in transfected HEK 293 cells 3.1. Introduction The recent discovery of receptor activity modifying proteins (RAMPS) new alternatives in the regulation of G-protein coupled receptors (GPCRS). RAMPS are characterized as single transmembrane-spanning accessory proteins required for the function of an orphan GPCR, now termed the calcitonin receptor-like receptor (CL- R)[275]. Through structural homology, three RAMP isoforms (1-3) have been identified and are synthesized as distinct gene products. RAMPS are required for the cell surface expression, as well as for ligand selectivity of CL-R [275, 276]. A heterodimer of RAMP] and CL-R yields a calcitonin gene-related peptide-l (CGRP-1) receptor, whereas RAMP2 or -3 coexpressed with CL-R produces adrenomedullin receptors, AM] and AM2 receptors, respectively [277, 278]. Both AM and CGRP are multi-functional peptides with many overlapping functions, ranging from potent vasodilation to proliferation regulation to regulation of salt and water balance [279]. It has been suggested in the literature that differential expression of RAMP isoforms regulates both physiological and pathological states. Upon agonist binding, the CL-R/RAMP receptor complex causes cyclic AMP activation in most systems, regardless of whether the ligand is AM or CGRP. In addition, the receptor complex is phosphorylated by protein kinases and subsequently undergoes desensitization and internalization in response to a prolonged agonist stimulation [280]. Intemalization of all three RAMP isoforms with CL-R has been reported to be clathrin- mediated and dynamin-dependent. In addition, the CL-R/RAMP] receptor complex is 52 known to internalize with agonist pretreatment in a complex with the B-arrestin molecule and all three components colocalize to the endosomes. From this stage, the receptor complex is targeted for a recycling or degradation pathway, depending on the cell type and RAMP expression profile. The findings in this study characterized the AM-stimulated CAMP accumulation and desensitization of the two AM receptor subtypes in response to AM treatment. These experiments represent pilot studies performed to determine agonist doses and time points for remaining studies in this thesis. 3.2. Materials and Methods 3.2.1. Materials: Adrenomedullin was purchased from Bachem Bioscience, Inc. (King of Prussia, PA). 125I-labeled adrenomedullin was purchased from Amersham Biosciences Corp. (Piscataway, NY). Cell culture media, fetal bovine serum, penicillin/streptomycin, trypsin-EDTA were purchased from GibcoBRL® (Grand Island, NY). All other reagents were of highest quality available. 3.2.2. Cell Culture and Transfection protocols: HEK-293T cells (obtained fi'om ATCC) are maintained in DMEM low glucose media containing 10% FBS, 1% penicillin-streptomycin. Transfection of HEK293T was performed using Lipofectamine Plus protocol (Invitrogen Life Technologies, Carlsbad, CA). Cells were transfected with the DNA and Lipofectamine Plus as per manufacturer’s protocol. Cells were collected for assays after 48 hours of transfection. 53 3.2.3. RAMP cloning and expression: Full length CDNA of human RAMPS], 2 and 3 and bovine CL-R were described before [281, 282]. CL-R, Cloned into Nl-EGFP and also in pCDNA3.l expression vectors, was used for transfection in HEK 293T cells. 3.2.4. Desensitization assays: 48 hours post-tranfection cells were pretreated with or without 10 nM AM in DMEM containing 0.2% BSA for indicated time periods (up to 4 hr). After agonist exposure, cells were washed three times with Dulbecco’s phosphate buffered saline (dPBS, Gibco BRL) containing 0.2% BSA and either frozen for membrane preparation for adenylate cyclase assays or used immediately for intact-cell radioligand binding. Desensitization Protocol: Removal of AM, Cells AM pretreatment followed by washing transfected1 starts 1H\ R pt cti ity ass or a v assay Mrs 1 h" (CAMP accumulation) and receptor binding No Pretreat (Full Stimulation) 1 hr AM Pretreat (Desensltlzatlon) 3.2.5. Receptor binding: Homologous competition radioligand binding assays were performed as described by Aiyar et a1 and as established in our laboratory [283]. HEK 293T cells were transfected and approximately 200,000 cells/well seeded in poly-D- 54 lysine precoated 24-well plates (BD Biosciences, Palo Alto, CA). 48 hours post- tranfection cells were treated for desensitization or resensitization assays as described above. After agonist exposure, cells were washed three times with dPBS buffer containing 0.2% BSA then incubated with increasing concentrations (1 pM to 100 nM) of competing ligand (rAM) and 175-250 pM 12“’I-rAM for 30 min at 37°C. After incubation, plates were washed three times with ice-cold assay buffer and the reactions were terminated by the addition of 2M NaOH. Cells were then harvested and associated radioligand activity is counted on a y-counter. All binding assays were performed in duplicate, with each experiment repeated at least three times. Nonspecific binding was determined in the presence of 100 nM of unlabeled rAM. Data was analyzed by LIGAND (assuming radioligand and competitor both bind reversibly to a single binding site; MacLigand, Version 4.97, NIH, Bethesda, MD) using the following equation: Bmax 0 [hot ligand] KD + [hot ligand] + [cold ligand] Where B represents specific binding, [hot ligand] the single concentration of [1251]rAM studied, [cold ligand] the concentration of unlabeled rAM competing with the radiolabe] for AM receptor binding, Bmax the maximum number of binding Sites and KD the equilibrium dissociation constant (the equation was solved where the “cold ligand” ICso = [hot ligand] + KD). Analysis of all binding data was performed by computer-assisted nonlinear least square fitting using GraphPad PRIZM version 4 (GraphPad Software, San Diego, CA). Binding sites/cell was calculated and data was expressed as percent of the control. 55 3.2.6. Adenylate cyclase assays: Cyclase activity was done as described before with slight modifications [146, 284]. Cells were harvested from P100 or P60 plates and homogenized in Tris HCl (10mM)/EDTA (10mM) buffer. Membranes were prepared by homogenization and centrifugation in Tris HC] (50mM)/MgCl (10mM) buffer. Fina] concentration of 20 pg of protein/assay tube was obtained. Membranes were incubated for 15 min at 30°C with appropriate concentrations of drugs (lOOnM AM, 10nM Forskolin) and assay mix containing ATP regeneration system and a32P-ATP. After the reaction was stopped (with stop solution containing 3H-cAMP) contents of the assay tubes were passed through Dowex and subsequently through alumina columns to separate the degradation products of ATP, by washing the dowex with water and alumina with imidazole. Elution profile was done to determine the amount of water and imidazole needed to wash and elute the products. Product eluted from alumina column was counted for the presence of 3H-CAMP and GBZP-CAMP in a B-counter. Each experiment was done in triplicates and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. Basal cyclase activity and forskolin stimulation did not Show statistically significant differences between treatments. 3.2.7. CAMP accumulation assays: HEK 293 cells were seeded on a 24-well plate until reaching 80-90% confluency, then incubated in serum-free media overnight before experiment. Desensitization experiments were carried out as described in Materials and Methods section, with cells pretreated with 10nM rAM and subsequently Challenged for 10 min at 37°C with appropriate concentrations of drugs (IOOnM AM, 10nM Forskolin) in the presence of 200uM 3-isobutyl-1-methylxanthine. Determination of CAMP level 56 d. all (163 was measured using the biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in HEK 293 cells were calculated using a standard curve ranging from 10 to 10 4 frnol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. Basal cyclase activity and forskolin stimulation did not Show statistically significant differences between treatments. 3.2.8. Statistics: Data are presented as mean i S.E.M. Single group comparisons exercised a paired Student’s t-test method. Statistical significance was set at P<0.05. 3.3. Results/Discussion We and other groups have previously Shown that the AM] receptor (CL-R + RAMP2) and AM2 receptor (CL-R + RAMP3 complex) undergo agonist-stimulated desensitization, internalization, and degradation in HEK 293 cells [153, 276, 285]. An initial 100 nM AM Challenge to HEK 293 cells expressing CL-R and RAMP2 or RAMP3 results in an approximate 9 and 8-fold increase, respectively, in CAMP accumulation over basal. Pretreatment of HEK 293 cells expressing CL-R and RAMP2 or RAMP3 with 10 nM AM for one hour and subsequent challenge with 100 nM AM resulted in desensitization of the CAMP accumulation response. AM pretreatment did not significantly alter the basal or forskolin-stimulated (10 nM) CAMP accumulation (Figure 2A, B). Because AM pretreatment was not shown to alter the basal or FK-Stimulated CAMP accumulation, the remaining CAMP accumulation and adenylate cyclase activity data throughout the thesis will be expressed as percent maximal response, or percent 57 ..a‘ forskolin stimulation. Dose response experiments experiments for receptor desensitization indicate that maximal desensitization is observed with a 10 nM AM pretreat and 100 nM AM Challenge (Figure 3A, B). These will be the AM doses used for pretreatment and challenge in the desensitization and resensitization assays employed throughout the remaining Chapters of the thesis. The time course for desensitization shows a maximal attenuation of CAMP accumulation between one and two hours of agonist pretreatment (Figure 4A, B), and for this reason a one hour agonist pretreatment is used throughout the remaining thesis Chapters to measure receptor desensitization. Homologous competitive binding experiments were optimized by altering ligand and competing ligand concentrations, temperature, and duration of binding experiments. Figure 5 depicts a representative competition curve for the whole-cell, homologous competitive binding experiments employed throughout this thesis. Receptor binding sites per cell were determined from scatchard plots, as described in Materials and Methods section. A representative scatchard plot is shown in Figure 6. Table V shows receptor affinity and receptor density calculations for HEK 293 cells expressing CL-R and RAMP3 with NSF or NHERF-l expression. This table depicts a representative experiment and indicates no change in receptor affinity with agonist pretreatment or with expression of additional proteins (NSF or NHERF-l). In addition, co-expression of NSF or NHERF-1 with CL-R/RAMP3 does not significantly alter receptor density in the HEK 293 cells. For the remaining homologous competitive binding experiments, these calculations will be followed and data expressed as percent of control. 58 - No Pretreat DAM Pretreat c 250- .0 ‘E g 5 200- l a , g :2; 1504 u U) 2 :5, 100- n- 0 ° 0. _ I __ 53‘ “8“ “0 Figure 2A: Representative desensitization experiment of AM] receptor in HEK 293 cells, measured by CAMP accumulation. HEK 293 cells were transiently transfected with CL-R and RAMP2 and pretreated with 10 nM AM for one hour. After agonist pretreatments, cells were washed repeatedly, and CAMP accumulation in response to 100 nM AM challenge was measured. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 10 " frnol of CAMP. CAMP accumulation is shown on the left y-axis, raw data shown. Representative experiment is shown in figure. 59 - No Pretreat 300- D AM Pretreat 200' 100- cAMP Accumulation (pmollmg protein) 6353‘ ”A (‘00 “\N (\D 9‘“ CK Figure 2B: Representative desensitization experiment of AM2 receptor in HEK 293 cells, measured by CAMP accumulation. HEK 293 cells were transiently transfected with CL-R and RAMP3 and pretreated with 10 nM AM for one hour. After agonist pretreatments, cells were washed repeatedly, and CAMP accumulation in response to 100 nM AM Challenge was measured. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 104 frnol of CAMP. CAMP accumulation is shown on the left y-axis, raw data Shown. Representative experiment is shown in figure. 60 + No Pretreat +AM Pretreat O O n I CAMP Accumulation (nmollmg protein) We Forskolin] '63 1 0- G I I U I I \ «A ‘m (a 9% a9 e o e e 9 03° “*0 “o 030 Figure 3A: Desensitization dose response of AM] receptor in HEK 293 cells. CL-R/RAMP2 complex (AMI receptor) desensitizes in HEK 293 cells after prolonged agonist stimulation. HEK 293 cells transiently transfected with CL-R and RAMP2 were treated for one hour with AM (10 nM), then washed repeatedly, and after repeated wash steps, cells were re-Challenged with 100 nM rAM for 15 minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 10 4 frnol of CAMP. CAMP accumulation is shown on the left y-axis, expressed as percent maximal response (% forskolin stimulation). Representative experiment shown in figure. 6] -I- No Pretreat +AM Pretreat Dd) .. <3?” CI“.— 33,3 _ x . age“ 0:0 or?“ 532110- >‘E C 33 c < Figure 3B: Desensitization dose response of AM2 receptor in HEK 293 cells. CL-R/RAMP3 complex (AM2 receptor) desensitizes in HEK 293 cells after prolonged agonist stimulation. HEK 293 cells transiently transfected with CL-R and RAMP3 were treated for one hour with AM (10 nM) and then washed and receptor desensitization (adenylate cyclase activity) was measured. Adenylate cyclase activity is shown on the left y-axis, expressed as percent maximal response (% forskolin stimulation). n 2 3 experiments. 62 [% Forskolin] N w -h R R R .3 C I l l cAMPAccumulation (nmollmg protein) 0. \ 6 D 6 0 D 0““0 l‘ '5 A 6 «7. I AM Pretreat (min.) Figure 4A: Desensitization time course for AM] receptor in HEK 293 cells. CL-R/RAMP2 complex (AMl receptor) desensitizes in a time-dependent manner in HEK 293 cells after agonist stimulation. HEK 293 cells transiently transfected with CL-R and RAMP2 were treated for one hour with AM (10 nM), then washed repeatedly, and after repeated wash steps, cells were re- challenged with 100 nM rAM for 15 minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 10 4 fmol of CAMP. CAMP accumulation is shown on the left y-axis, expressed as percent control (no pretreatment) sample. Representative experiment shown in figure. 63 CAMP Accumulation (nmollmg protein) [% Control] AM Pretreat (min) Figure 4B: Desensitization time course for AM2 receptor in HEK 293 cells. CL-R/RAMP3 complex (AM2 receptor) desensitizes in a time-dependent manner in HEK 293 cells after agonist stimulation. HEK 293 cells transiently transfected with CL-R and RAMP3 were treated for one hour with AM (10 nM), then washed repeatedly, and after repeated wash steps, cells were re- challenged with 100 nM rAM for 15 minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 10 ’ fmol of CAMP. CAMP accumulation is shown on the left y-axis, expressed as percent control (no pretreatment) sample. Representative experiment shown in figure. 64 125- 2’ '5 100. .E In 75. 5.3 . g 50- o. ”3 251 o\ c I I r I I I I 13 12 11 1O 9 8 7 6 log rADM Figure 5: Competition curves for rAM on intact HEK-293T cells expressing CL-R and RAMP3 with [”51] rAM as radioligand were generated as described in Materials and Methods section. Representative experiment for homologous competition binding employed throughout remaining thesis Chapters. 65 P a C: Bound/Free O 'o '9 P O ‘ l P o 0 0:1 0.2 0.3 0.4 P c Figure 6: Scatchard plot for homologous competition binding of rAM on intact HEK-293T cells expressing CL-R and RAMP3, with [”51] rAM as radioligand, generated as described in Materials and Methods section. Representative experiment for homologous competition binding employed throughout remaining thesis Chapters. 66 Experlment: Kd (nM) Ki (nM) Bmax cellslwell Binding % control [final/assay) siteslcell CL-R+RAMP3 J Control 4.00 2.66 0.122 200000 368123 100.00 1 hr Pretreat 3.44 2.29 0.078 231500 203848 55.37 4 hr Recovery 3.73 2.49 0.071 246800 173477 47.12 CL-R+RAMP3+N8F Control 3.44 2.29 0.093 209300 267894 100.00 1 hr Pretreat 5.35 3.57 0.031 189400 97324 36.33 4 hr Recovery 4.45 2.97 0.076 177200 259451 96.85 CL-R-I-RAMP3-1-NHERF1 Control 3.80 2.62 0.129 230000 336335 100.00 1 hr Pretreat 2.44 1.68 0.138 220000 363389 108.04 4 hr Recovery 2.76 1.90 0.108 200000 323876 96.30 Table V: Binding properties for homologous competition binding of rAM on intact HEK-293T cells expressing CL-R and RAMP3 +/- NSF or NHERF], with [”51] rAM as radioligand, generated as described in Materials and Methods section. No statistically Significant changes in receptor affinity or receptor density observed in homologous competitive binding experiments. Representative experiment for homologous competition binding to determine binding sites per cell, employed throughout remaining thesis Chapters. 67 4. Differential regulation of adrenomedullin receptor desensitization and internalization by receptor activity-modifying proteins. 4.1. Introduction The recent discovery of receptor activity modifying proteins (RAMPS) has raised new possibilities for modes of regulation of G-protein coupled receptors (GPCRS). RAMPS are Characterized as single transmembrane-spanning accessory proteins requisite to the function of an orphan GPCR, now termed the calcitonin receptor-like receptor (CL- R)[275]. Through structural homology, three RAMP isoforms (1-3) have been identified as distinct gene products. RAMPS are required for the cell surface expression, as well as for ligand selectivity of CL-R [275, 276]. A heterodimer of RAMP] and CL-R yields a calcitonin gene-related peptide-1 (CGRP-1) receptor, whereas coexpression of RAMP2 or -3 with CL-R produces adrenomedullin receptors, AM] and AM2 receptors, respectively [277, 278]. Both AM and CGRP are multi-functional peptides with many overlapping functions, ranging from potent vasodilation .to proliferation regulation to regulation of natriuresis and diuresis [279]. It has been suggested in the literature that differential expression of RAMP isoforms plays a regulatory role in both physiological and pathophysiological disease states. Moreover, the recent identification of RAMP interactions with additional members of the Class II GPCR family and RAMP expression in cell lines lacking CL-R have raised the possibility of additional functions for RAMPS in GPCR regulation [286]. In addition, we have recently reported novel roles for RAMPS, through protein-protein interactions with NSF and NHERF, in the trafficking of the AM receptor subtypes. 68 Upon agonist binding, the CL-R/RAMP receptor complex causes cyclic AMP activation in most systems, regardless of whether the ligand is AM or CGRP. In addition, the receptor complex is phosphorylated by protein kinases and subsequently undergoes desensitization and internalization in response to a prolonged agonist stimulation [280]. Intemalization of all three RAMP isoforms with CL-R has been reported to be Clathrin- mediated and dynamin-dependent. In addition, the CL-R/RAMP] receptor complex is known to internalize with agonist pretreatment in a complex with the B-arrestin molecule and all three components colocalize to the endosomes. From this stage, the receptor complex is targeted for a recycling or degradation pathway, depending on the cell type and RAMP expression profile. Phosphorylation has been demonstrated to be a requisite step in the desensitization process of the majority of GPCRS. The phosphorylation of the receptor, upon agonist exposure, allows interaction of the receptor with the B-arrestin molecules to promote an attenuation of receptor signaling, or receptor desensitization. Receptor desensitization can occur by two mechanisms, termed homologous and heterologous desensitization. Homologous desensitization is Classified as desensitization specific for the agonist-occupied receptor. This occurs as a result of G-protein coupled receptor kinase (GRK) specificity for only agonist-occupied receptors. On the other hand, heterologous desensitization occurs by activation of second messenger kinases, PKA and PKC for example, and has no Specificity for agonist-occupied receptors. Heterologous desensitization raises the possibility of “cross-desensitization” for unoccupied receptors. Upon ligand binding, it has been reported that CL-R undergoes phosphorylation, while the RAMP partner remains unphosphorylated. Moreover, PKA has been shown to 69 regulate the phosphorylation and desensitization of the CL-R/RAMP] receptor complex in SK—N-MC and vascular smooth muscle cells, while GRK-6 was reported to phosphorylate and promote desensitization of the CL-R/RAMP] complex in HEK 293 cells [200-202]. Kinases responsible for the phosphorylation of the CL-R/RAMP2 or -3 receptor complexes have yet to be specifically Characterized. The findings in this study reveal that different RAMP isoform association with CL—R may lead to differential phosphorylation of the receptor complex. The Ser 421 residue (a putative PKA phosphorylation site) on CL-R was found to be critical for the desensitization and internalization of the CL-R/RAMP2 receptor complex, whereas the Thr 423 residue (a putative PKA phosphorylation site) on the C-terminus of CL-R was found to be crucial for the CL-R/RAMP3 receptor desensitization and internalization. CL-R-R2 complex desensitization and internalization was blocked by H89, a PKA inhibitor, while CL-R-R3 complex desensitization and internalization was blocked by RO 32-0432, a PKC inhibitor. Taken together, these results suggest that in addition to the various roles of RAMPS in the regulation of CL-R/RAMP complex as reported by us and others, the isoform of RAMPS could also be involved in the differential phosphorylation of CL-R. 4.2. Materials and Methods 4.2.1. Materials: Adrenomedullin was purchased from Bachem Bioscience, Inc. (King of Prussia, PA). 1251-labeled adrenomedullin was purchased from Amersham Biosciences Corp. (Piscataway, NY). Cell culture media, fetal bovine serum, 70 penicillin/streptomycin, trypsin-EDTA were purchased from GibcoBRL® (Grand Island, NY). RAMP3 antibody was purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Anti-rabbit Cy3 secondary antibody was from Jackson Immunoresearch Laboratories (West Grove, PA). PKA (H-89) and PKC (Ro 32-0432) inhibitors were purchased from Sigma (St. Louis, MO). All other reagents were of highest quality available. 4.2.2. Cell Culture and Transfection protocols: HEK-293T cells (obtained from ATCC) are maintained in DMEM low glucose media containing 10% F BS, 1% penicillin-streptomycin. Rat-2 fibroblast cells (obtained from ATCC) are maintained in DMEM high glucose media containing 10% FBS, 1% penicillin-streptomycin. Transfection of HEK293T and Rat-2 fibroblast cells was performed using Lipofectamine Plus protocol (Invitrogen Life Technologies, Carlsbad, CA). Cells were transfected with the DNA and Lipofectamine Plus as per manufacturer’s protocol. Cells were collected for assays afier 48 hours of transfection. 4.2.3. RAMP cloning and expression: Full length CDNA of human RAMPS], 2 and 3 and bovine CL-R were described before [281, 282]. CL-R, cloned into Nl-EGFP and also in pcDNA3.1 expression vectors, was used for transfection in HEK 293T cells. 4.2.4. Desensitization assays: 48 hours post-tranfection cells were pretreated with or without 10 nM AM in DMEM containing 0.2% BSA for indicated time periods (up to 4 71 hr). After agonist exposure, cells were washed three times with Dulbecco’s phosphate buffered saline (dPBS, Gibco BRL) containing 0.2% BSA and either frozen for membrane preparation for adenylate cyclase assays or used immediately for intact-cell radioligand binding. Desensitization Protocol: Removal of AM, Cells AM Pretreatment [- followed by washing .......1 .... 7 ¥ Y I \ Receptor activity assay 48 ours 1 hr / (CAMP accumulation) and receptor binding No Pretreat 1 (Full Stimulation) 1 hr AM Pretreat (Desensitization) 4.2.5. Receptor binding: Homologous competition radioligand binding assays were performed as described by Aiyar et al and as established in our laboratory [283]. HEK 293T cells were transfected and approximately 200,000 cells/well seeded in poly-D- lysine precoated 24-well plates (BD Biosciences, Palo Alto, CA). 48 hours post- tranfection cells were treated for desensitization or resensitization assays as described above. After agonist exposure, cells were washed three times with dPBS buffer containing 0.2% BSA then incubated with increasing concentrations (1 pM to 100 nM) of competing ligand (rAM) and 175-250 pM 125I-rAM for 30 min at 37°C. After incubation, plates were washed three times with ice-cold assay buffer and the reactions were 72 terminated by the addition of 2M NaOH. Cells were then harvested and associated radioligand activity is counted on a y-counter. All binding assays were performed in duplicate, with each experiment repeated at least three times. Nonspecific binding was determined in the presence of 100 nM of unlabeled rAM. Data was analyzed by LIGAND (assuming radioligand and competitor both bind reversibly to a Single binding site; MacLigand, Version 4.97, NIH, Bethesda, MD) using the following equation: Bmax 0 [hot ligand] KD + [hot ligand] + [cold ligand] Where B represents specific binding, [hot ligand] the single concentration of [1251]rAM studied, [cold ligand] the concentration of unlabeled rAM competing with the radiolabel for AM receptor binding, Bmax the maximum number of binding sites and KD the equilibrium dissociation constant (the equation was solved where the “cold ligand” ICso = [hot ligand] + KD). Analysis of all binding data was performed by computer-assisted nonlinear least square fitting using GraphPad PRIZM version 4 (GraphPad Software, San Diego, CA). Binding sites/cell was calculated and data was expressed as percent of the control. 4.2.6. Adenylate cyclase assays: Cyclase activity was done as described before with slight modifications [146, 284]. Cells were harvested from P100 or P60 plates and homogenized in Tris HC] (10mM)/EDTA (10mM) buffer. Membranes were prepared by homogenization and centrifugation in Tris HCl (50mM)/MgCl (10mM) buffer. Final concentration of 20 pg of protein/assay tube was obtained. Membranes were incubated for 15 min at 30°C with appropriate concentrations of drugs (100nM AM, lOpM 73 Forskolin) and assay mix containing ATP regeneration system and a32P-ATP. After the reaction was stopped (with stop solution containing 3H-CAMP) contents of the assay tubes were passed through Dowex and subsequently through alumina columns to separate the degradation products of ATP, by washing the dowex with water and alumina with imidazole. Elution profile was done to determine the amount of water and imidazole needed to wash and elute the products. Product eluted from alumina column was counted for the presence of 3H-CAMP and a32P-CAMP in a B-Counter. Each experiment was done in triplicates and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. Basal cyclase activity and forskolin stimulation did not Show statistically significant differences between treatments. 4.2.7. CAMP accumulation assays: HEK 293 cells were seeded on a 24-well plate until reaching 80-90% confluency, then incubated in serum-free media overnight before experiment. Desensitization experiments were carried out as described in Materials and Methods section, with cells pretreated with 10nM rAM and subsequently Challenged for 10 min at 37°C with appropriate concentrations of drugs (100nM AM, 10nM Forskolin) in the presence of ZOOpM 3-isobutyl-l-methylxanthine. Determination of CAMP level was measured using the biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in HEK 293 cells were calculated using a standard curve ranging from 10 to 10 4 frnol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. Basal cyclase activity and forskolin stimulation did not Show statistically significant differences between treatments. 74 4.2.8. Mutagenesis procedure: Site-directed mutagenesis was performed using a PCR- based strategy that employs the pfu Turbo polymerase (Stratagene, La Jolla, CA). A pair of complementary oligonucleotides containing the appropriate point mutations in the sequence of RAMP or a premature stop codon at position 145 or 147 codon of RAMP-3 for deletion mutants were synthesized (Michigan State University Macromolecular structure facility). The PCR for the mutation was as follows: 94°C for 2 minutes; 30 cycles of 94°C for 30 sec., 50°C for 30 seC., 68°C for 8 min.; final cycle of 68°C for 8 minutes. PCR product was digested for 4 hours with DpnI enzyme (lnvitrogen) and transformed in to DH50t cells. Mutations were confirmed by automated sequencing (Michigan State University Genomic Technology Support Facility). Putative phosphorylation sites and kinase consensus sites were identified with NetPhosK Server 1.0 database (http://www.cbs.dtu.dk/services/NetPhosK). 4.2.9. Immunofluorescence microscopy: HEK 293 cells were transfected as described above and seeded at 24hr post-transfection onto collagen type I-Coated coverslips. Resensitization assays were performed as described and reactions were stopped by fixing cells in 4% paraforrnaldehyde for 30min. at room temperature. Samples were perrneablized with 0.1% v/v Triton X-100 in PBS and blocked overnight in 0.1% v/v Triton X-100 in PBS + 10% goat serum. Samples were incubated in primary antibody in blocking buffer for 2h at room temperature (NSF at 1:250 and RAMP3 at 1:200). Appropriate secondary antibodies were applied for 1h at room temperature (Goat anti- mouse Cy3 at 1:500 and Goat anti-rabbit Cy5 at 1:400). Coverslips were mounted in 75 Shandon Permafluor mounting medium and slides stored at 2-8°C until analysis. Cells were visualized on a Zeiss 210 laser confocal microscope at a zoom of 2. Images presented are representative single optical sections of a z-series taken from at least twenty fields per experiment and at least three individual experiments. Images in this thesis/dissertation are presented in color. 4.2.10. Statistics: Data are presented as mean i S.E.M. Single group comparisons exercised a paired Student’s t-test method. Statistical significance was set at P<0.05. 4.3. Results We and other groups have previously shown that the AM2 receptor (CL-R + RAMP3 complex) undergoes agonist-stimulated desensitization, internalization, and degradation in HEK 293 cells [153, 276, 285]. Furthermore, we have recently shown that in the presence of NSF, the AM2 receptor complex undergoes recycling, instead of following a degradation pathway, in HEK 293 cells [153]. We have also demonstrated that the presence of the adaptor protein, NHERF, in HEK 293 cells can inhibit the internalization, but not the desensitization, of the AM2 receptor complex through protein- protein interactions. Given the recognized role of RAMPS in the trafficking of the AM receptor subtypes, in this study we have examined the role of RAMPS in regulating the phosphorylation of the AM receptor subtypes in the desensitization and internalization processes. Pretreatment of HEK 293 cells expressing CL-R and RAMP2 or RAMP3 with 10 nM AM for one hour resulted in desensitization of the CAMP accumulation response 76 from approximately 40% (of forskolin stimulation) in untreated cells to 15% in AM- treated cells (Figure 7A, B). In addition, as shown in FigureIOC and 11C, respectively, CL-R/RAMP2 or CL-R/RAMP3 complex underwent agonist-induced internalization as determined by receptor binding (Figure 7A, B) and immunofluorescence microscopy. These findings are in agreement with those of Kuwasako et al. and as reported by us recently [153, 276]. Using site-directed mutagenesis, we mutated Clusters of putative phosphorylation residues in the C-terminus of CL-R to alanine residues and tested the effect of these mutations on desensitization and internalization of the receptor complexes (Figure 8). Mutant CL-R/RAMP receptor complexes showed similar levels of receptor stimulation and receptor complex expression levels as compared to wild-type CL- R/RAMP receptor complex, as determined by CAMP accumulation and whole-cell ligand binding experiments, respectively (Figure 9A, C; data not shown). Additionally, the CL- R Cluster mutants showed similar receptor stimulation and receptor complex expression when co-expressed with RAMP2 or RAMP3 (determined by CAMP accumulation and whole-cell binding assays, respectively). Interestingly, when desensitization assays were performed on the Cluster mutants of CL-R in complex with RAMP2 or RAMP3, the same cluster mutant of CL-R showed reduced levels of receptor desensitization when complexed with RAMP2 or RAMP3 (Figure 9A, C, respectively). Cluster mutant #2 of CL-R, when in complex with RAMP2 or RAMP3, showed no significant receptor desensitization with AM pretreatment, as compared to wild-type CL-R and Cluster mutants #1 and #3. In addition, when receptor complex internalization was measured with whole-cell binding experiments, AM pretreatment also failed to promote significant internalization of the Cluster mutant #2 of CL-R when it was complexed with RAMP2 or 77 RAMP3 (Figure 93, D, respectively). Wild-type CL-R and Cluster mutants #1 and #3 showed similar levels of receptor internalization with one hour AM pretreatment. Individual residues within the second Cluster of CL-R were then examined for their role in regulation of receptor desensitization and internalization of the CL-R/RAMP receptor complex. Site-directed mutagenesis was again employed to create the Single mutations of putative phosphorylation residues within the second Cluster of CL-R, determined to be critical for agonist-stimulated receptor complex desensitization and internalization, regardless of RAMP in association with CL-R. Point mutants of putative phosphorylation residues to alanine residues were CO-expressed with RAMP2 or RAMP3 in HEK 293 cells and desensitization and internalization assays were performed. Point mutants of CL-R when complexed with RAMP2 or RAMP3 Showed similar levels of receptor stimulation and receptor complex expression as compared to wild-type CL- RfRAMP complexes (determined by CAMP accumulation and whole-cell binding assays, respectively). Additionally, no differences in receptor stimulation or receptor complex expression were seen when CL-R point mutants were expressed with RAMP2 or RAMP3. Using CAMP accumulation assays to determine receptor desensitization, only Ser 421 mutant of CL—R, when complexed with RAMP2, Showed a loss of receptor desensitization as compared to wild-type CL-R/RAMP2 receptor complex (Figure 10A). Whole-cell binding and immunofluorescence microscopy determined Ser 421 of CL-R to also be critical to the agonist-stimulated internalization of the CL-R/RAMP2 receptor complex (Figure 10B, C). The additional point mutants of CL—R, when co-expressed with RAMP2, showed no significant Changes in agonist-induced receptor desensitization 78 or internalization as compared to wild-type CL-R/RAMP2 receptor complex (determined by CAMP accumulation and whole-cell binding assays, respectively). Meanwhile, co—expression of the point mutants of CL-R with RAMP3 showed a different putative phosphorylation residue critical to receptor desensitization and internalization. While all point mutants of CL-R, when complexed with RAMP3, showed similar levels of receptor stimulation and receptor complex expression as compared to wild-type CL—R/RAMP3 complexes (determined by CAMP accumulation and whole-cell binding assays, respectively), the Thr 423 mutant of CL-R showed a lack of significant receptor desensitization when pretreated with AM, compared to an approximate 50% attenuation of receptor signaling seen for the additional point mutants and wild-type CL-R co-expressed with RAMP3 (as measured by CAMP accumulation assays) (Figure 11A). In addition, mutation of Thr 423 of CL-R to alanine inhibited the agonist-stimulated receptor internalization when co-expressed with RAMP3 in HEK 293 cells (as measured by whole-cell binding and immunofluorescence microscopy) (Figure 11B, C, respectively). The additional point mutants of CL-R, when co-expressed with RAMP3, showed no significant Changes in agonist-induced receptor desensitization or internalization as compared to wild-type CL—R/RAMP3 receptor complex (determined by CAMP accumulation and whole-cell binding assays, respectively). These findings indicate that the two RAMP isoforms differentially regulate the CL-R receptor in the process of receptor desensitization and internalization. To determine if mutation of putative phosphorylation residues on the third intracellular loop of CL-R altered receptor desensitization or internalization when complexed with RAMP2 or RAMP3, Site-directed mutagenesis of these residues to 79 alanine was performed. Desensitization assays, measured by CAMP accumulation, showed no effect of third intracellular loop mutations on desensitization of CL- R/RAMP2 or CL-R/RAMP3 receptor complex. The receptor activity was attenuated to levels similar to wild-type CL-R, when mutant receptors were CO-expressed with RAMP2 or RAMP3 (Figure 12A, B). Third intracellular loop mutants of CL—R, in complex with RAMP2 or RAMP3, showed no difference in receptor expression levels at the plasma membrane (as measured with whole-cell binding) in untreated cells, as compared to wild- type CL-R/RAMP2 or CL-R/RAMP3 complex (data not shown). These results indicate that amino acid residues on the third intracellular loop of CL-R are not required for desensitization or internalization of the CL—R/RAMP2 or CL-R/RAMP3 receptor complex. It was then important to determine if putative phosphorylation residues on the RAMP isoforms contributes to the regulation of receptor complex desensitization and/or internalization. Putative phosphorylation residues on the C-terminus of the RAMP2 and RAMP3 isoforms were mutated to alanine residues and desensitization/internalization assays were performed. The phosphorylation mutant RAMPS co-expressed with wild- type CL-R showed no difference in receptor stimulation or receptor complex expression levels at the plasma membrane (as measured with CAMP accumulation and whole-cell binding, respectively) in untreated cells, as compared to wild-type CL-R/RAMP2 or CL- R/RAMP3 complex (Figure 13A-D). Desensitization of the receptor complex was unaltered with the RAMP phosphorylation mutants, in comparison to wild-type RAMP2 or RAMP3 in complex with CL-R (as measured by CAMP accumulation) (Figure 13A, C). In addition, the RAMP phosphorylation mutations were unable to modify the 80 internalization of the CL-R/RAMP2 or CL-R/RAMP3 receptor complexes (Figure 13B, D). No significant differences were seen in the desensitization or internalization patterns of the phosphorylation mutant RAMPS, when compared to wild-type RAMP2 or RAMP3 complexed with CL-R. These findings indicate that serine/threonine amino acid residues of the RAMPS do not appear to play a role in regulation of the desensitization and internalization processes of the CL-R/RAMP receptor complex. Data presented so far supports the hypothesis that different CL-R/RAMP isoform associations could result in differential phosphorylation of the AM receptor subtypes as a means of regulating desensitization and internalization of the receptor complexes. It was then important to identify the kinases regulating the desensitization and internalization of the different CL-R/RAMP2 and CL-R/RAMP3 receptor complexes. Sequence analysis of CL-R in the NetPhosK database (http://www.Cbs.dtu.dk/serviceS/NetPhosK) predicted consensus sites for PKA and PKC phosphorylation on amino acid residues Ser 421 and Thr 423 of CL-R, respectively. These amino acid residues were identified in earlier experiments within this study to regulate desensitization and internalization of the CL- R/RAMP2 and CL—R/RAMP3 receptor complexes, respectively. Therefore, it was hypothesized that inhibition of these kinases would block receptor desensitization and internalization of the appropriate AM receptor complex. The Ser 421 amino acid residue of CL-R that was determined to be critical to the agonist-stimulated desensitization and internalization of the CL-R/RAMP2 receptor complex was predicted to be regulated by protein kinase A. In order to test if this kinase was regulating the CL-R/RAMP2 receptor complex desensitization and internalization, a specific protein kinase A (PKA) inhibitor, H—89, was used to perform desensitization and 8] internalization assays. As a control, a protein kinase C (PKC) inhibitor, RO 32-0432, was also tested. Cells pretreated with the PKC inhibitor and AM showed similar levels of receptor desensitization and internalization as cells pretreated with only AM (Figure 14A, B). On the other hand, cells pretreated with the PKA inhibitor in the presence of AM showed an inhibition of both receptor desensitization and internalization of the CL- R/RAMP2 receptor complex (Figure 14A, B). No Significant differences were observed in receptor CAMP stimulation and receptor complex expression levels in cells co- expressing CL-R and RAMP2 that were pretreated with or without the PKA and PKC inhibitors. These findings indicate that PKA, but not PKC, is involved in CL-R/RAMP2 desensitization and internalization, presumably by phosphorylating Ser 421 of CL-R. PKC and PKA inhibitors were then used to test their effects on the desensitization and internalization patterns of the CL-R/RAMP3 receptor complex. Because the Thr 423 on CL-R was a PKC consensus site, it was predicted that the PKC inhibitor would alter the desensitization and internalization of the receptor complex. CAMP accumulation and whole-cell binding assays showed that while the PKA inhibitor had no effect on the desensitization and internalization of the CL-R/RAMP3 receptor complex, the PKC inhibitor completely blocked the desensitization and internalization of the CL-R/RAMP3 complex (Figure 15A, B). These studies suggest that PKC, but not PKA, is involved in the desensitization and internalization of CL-R/RAMP3 receptor complex, seemingly by phosphorylation of Thr 423 of CL-R. To further Characterize the role of PKA and PKC in the desensitization and internalization of the AM] and AM2 receptors, respectively, the kinases were individually activated and tested for their ability to induce receptor desensitization and/or 82 internalization. No significant alterations were seen in the receptor expression levels or basal or forskolin-stimulated CAMP accumulation when transfected HEK 293 cells were pretreated with forskolin (10 TAM) or phorbol-l2-myristate-13-acetate (PMA, 1 nM) for the given time periods (data not shown). As would be predicted, activation of PKA with forskolin pretreatment resulted in efficient desensitization and internalization of the AMI, but not AM2 receptors (Figure 16A, B). PKA activation was capable of achieving levels of desensitization and internalization comparable to that induced by AM in cells transfected with CL-R and RAMP2. When transfected HEK 293 cells were pretreated with PMA, to activate PKC, desensitization and internalization was observed for the AM2, but not AM] receptors (Figure 16A, B). The PKC-induced desensitization and internalization observed for cells expressing CL-R and RAMP3 was comparable to that stimulated by AM. These findings are in agreement with the observations from the kinase inhibitor studies (Figures 14, 15). This data confirms a role for PKA in the desensitization and internalization processes for the AM] receptor (CL-R+RAMP2), and a role for PKC in the desensitization and internalization of the AM2 receptor. 83 - CAMP Accumulation D Binding — -100 -75 i * ~50 I N 0| CAMP (nmollmg protein) [°/o Forskolin] licence %1 ("ea/saus) xeui Bugpuga No Pretreat AM Pretreat Figure 7A: Desensitization and internalization of the AM] and AM2 receptor in HEK 293 cells. CL-R/RAMP2 complex (AMI receptor) desensitizes and intemalizes in HEK 293 cells after prolonged agonist stimulation. HEK 293 cells transiently transfected with CL-R and RAMP2 were treated for one hour with AM (10 nM) and then washed and receptor desensitization (CAMP accumulation) and internalization (radioligand binding) were measured. CAMP accumulation is shown on the left y-axis, expressed as percent maximal response (% forskolin stimulation), and radioligand binding is shown on the right y-axis, expressed as percent control. * p S 0.05; n 2 4 experiments. 84 - CAMP Accumulation DBinding E 40- —~ -100 Q. s a O H —- 5g 30- -75 g”: as i as =0- 9 20- -50 g x E .2 :75 = <1 .3: '7" :9. 10- -25 “3 E 8 o 0' 0 5 No Pr'etreat AM Pfetreat Figure 7B: Desensitization and internalization of the AM] and AM2 receptor in HEK 293 cells. CL-R/RAMP3 complex (AM2 receptor) desensitizes and intemalizes in HEK 293 cells after prolonged agonist stimulation. HEK 293 cells transiently transfected with CL-R and RAMP3 were treated for one hour with AM (10 nM) and then washed and receptor desensitization (CAMP accumulation) and internalization (radioligand binding) were measured. CAMP accumulation is shown on the left y-axis, expressed as percent maximal response (% forskolin stimulation), and radioligand binding is shown on the right y-axis, expressed as percent control. * p S 0.05; n 2 4 experiments. 85 @renomedullin) Plasma membrane T324 53293 3/841'416 RAMP S419 S421 CLR Cluster #1 T423 34254426 Cluster #3 S428 Cluster #2 T431 8439 T450 Figure 8: Illustration of Cluster mutations of CL-R to investigate role of phosphorylation on AM receptor desensitization and internalization. Site-directed mutagenesis techniques, described in Materials and Methods section, were employed to mutate putative phosphorylation residues on C- tenninus of CL-R to alanine residues. Cluster mutants (1-3), as depicted in figure, are used in experiments reported in Figure 9A-D. 86 - CL-R+R2 D CL-R Cluster mutant 1 E21 CL-R Cluster mutant 2 D CL-R Cluster mutant 3 %‘ 60- ’6 . A?“ ”I 21% 40" 2.2” w * =\°20-I * * 'L 2 5 0- No Prbtreat AM Pfetreat Figure 9A: Role of phosphorylation on AM] and AM2 receptor desensitization and internalization (Cluster mutations). Amino acid residues 421-428 are critical to desensitization of AM] receptor. HEK 293 cells transiently transfected with CL-R/CL-R Cluster mutants and RAMP2 were seeded in 48-well plates and treated for one hour with AM (10 nM) and then washed and receptor desensitization (CAMP accumulation) was measured. After repeated wash steps, cells were re-Challenged with 100 nM rAM for 15 minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 104 fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p S 0.05; n 2 3 experiments. 87 - CL-R+RAMP2 I: CL-R Cluster mutant 1 1:11 CL-R cluster mutant 2 D CL-R cluster mutant 3 100- F — F l 75- 50- N 01 l Binding max (sites/cell) [% Control] 31' 3(- No Pr'etreat AM Pfetreat Figure 93: Role of phosphorylation on AM] and AM2 receptor desensitization and internalization (Cluster mutations). Amino acid residues 421-428 are critical to AM] receptor internalization. HEK 293 cells were CO-transfected with CL-R or CL-R cluster mutants and RAMP2. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour, wasth as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 3. 88 - CL-R+R3 E CL-R cluster mutant 1 D CL-R cluster mutant 2 . D CL-R Cluster mutant 3 010) ?O co 45 O O 1 n CAMP (nmollmg protein) [% Forskolin] N R jiiil No Pretreat AM Pretreat Figure 9C: Role of phosphorylation on AM] and AM2 receptor desensitization and internalization (Cluster mutations). Amino acid residues 421-428 are critical to desensitization of AM2 receptor. HEK 293 cells transiently transfected with CL-R/CL-R Cluster mutants and RAMP3 were seeded in 48-well plates and treated for one hour with AM (10 nM) and then washed and receptor desensitization (CAMP accumulation) was measured. After repeated wash steps, cells were re-Challenged with 100 nM rAM for 15 minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 104 fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p S 0.05; n 2 3 experiments. 89 - CL-R+RAMP3 D CL-R Cluster mutant 1 1:1 CL-R Cluster mutant 2 D CL-R Cluster mutant 3 g 125- l E H 100 177.9 g g 75' * * E U 50 * \° -1 EL 2 251 No Prbtreat AM Pfetreat Figure 9D: Role of phosphorylation on AM] and AM2 receptor desensitization and internalization (Cluster mutations). Amino acid residues 421-428 are critical to AM2 receptor internalization. HEK 293 cells were co-transfected with CL-R or CL-R Cluster mutants and RAMP3. 48h post-transfection. cells were pretreated with AM (10 nM) for one hour, washed as described in Materials and Methods. and receptor internalization was measured with whole-cell binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 3. 90 h o I I—1 - WT CL-R+R2 CL-R S421A 30' E i: CL-R Y422A 5;; i: CL-R T423A its: :3 CL-R S425A "4 mm CL-R T426A A O I CL-R S428A ’///////////////////////////A FWWFZZ‘?‘ fiwfi‘hm ,v , L2... CAMP (nmollmg protein) [% Forskolin] N R NoiPretreat AM Pretreat Figure 10A: Role of phosphorylation on AM] receptor desensitization and internalization (point mutations). Residue Ser 421 is crucial to desensitization of AM] receptor. HEK 293 cells transiently transfected with CL-R/CL-R point cluster mutants and RAMP2 were seeded in 48- well plates and treated for one hour with AM (10 nM) and then washed and receptor desensitization (CAMP accumulation) was measured. After repeated wash steps, cells were re- challenged with 100 nM rAM for 15 minutes and plates were frozen. Detemtination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 10 " fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p S 0.05; n 2 3 experiments. 91 100- N ‘1' [% Control] 0| R 25d Binding max (sites/cell) 1 :‘ .7 h. z . . ‘ .1, 44‘ ., 1;. f: "'-. . l is» ‘. i. , > * 4 1 ‘43; '5. 'H : l I 1. l l ‘ ' i 1 I} . '3‘ 4 '4. .I: .0 . . . 1' ,9; 2 . 3.: .. .4, ll.‘ 1 . 1'4, [pp , ii 4 .. 2: I1 :"‘ .14 u ; -. ,; il l »., 7: ill '-' _.,. . w . _/ l l w . .‘9 1 ,l -§ , . .;. i = , a. 7 ‘l‘. '4: 7 :» ‘v . J r. e 1. .2 1 . No Pretreat AM Pretreat - WT CL-R+ R2 1:31 CL-R S421A E CL-R Y422A E: CL-R T423A - CL—R S425A E21 CL-R T426A :21 CL-R S428A Figure 108: Role of phosphorylation on AM] receptor desensitization and internalization (point mutations). Residue Ser 421 is critical to AM] receptor internalization. HEK 293 cells were co- transfected with CL-R or CL-R point mutants and RAMP2. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour, washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n z 3. 92 WT CL-R RAMP2 ()Vcrluy No Prctrcat AM Pretreat CL-R S421A RAMP2 ()\Crla_\' No Pretreat AM Pretreat Figure 10C: Role of phosphorylation on AM] receptor desensitization and intemalization (point mutations). lmmunofluorescence microscopy shows failure of CL-R S421A/RAMP2 receptor complex to internalize after agonist stimulation. HEK 293 cells transfected with CL-R S421A- GFP and RAMP2 were pretreated with 10nM AM for lh. After pretreatment with AM, cells were washed, fixed, and components were visualized using anti-RAMP2 antibody (1:150) and detected with Cy3 secondary antibody (1:250), and CL-R is detected with an EGFP tag; overlays of staining patterns are shown in the far right panels. Images shown are representative of at least twenty fields imaged per experiment from at least three experiments. Bar scales on all images represent lOOum. 93 E 40- 3 - WT CL-R+R3 g? 30. p f i:iCL-R S421A m .3 ;,,_ i:i CL-R Y422A E if m CL-R T423A T: o 20‘ :iCL-R S425A g “t CL-R T426A IE- 10- - CL-R S428A 2 < o . No Pretreat AM Pretreat Figure 11A: Role of phosphorylation on AM2 receptor desensitization and internalization (point mutations). Residue Thr 423 is crucial to desensitization of AM2 receptor. HEK 293 cells transiently transfected with CL-R/CL-R point mutants and RAMP3 were seeded in 48-well plates and treated for one hour with AM (10 nM) and then washed and receptor desensitization (CAMP accumulation) was measured. After repeated wash 5th5, cells were re-Challenged with 100 nM rAM for 15 minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 10 ’ fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p 5 0.05; n 2 3 experiments. 94 % 100- , g u _ E _ E -WT CL-R+ R3 ,3 = 75. - g: * IZICL-R S421A 3.2 - * i: CL-R Y422A a: 8 50. EZICL-R T423A E g 1g ‘ - CL-R S425A EL- ‘ ; m CL-R T426A '2 25' :i 1:! CL-R S428A is 04 ‘ No Pretreat AM Pretreat Figure 11B: Role of phosphorylation on AM2 receptor desensitization and internalization (point mutations). Residue Thr 423 is critical to AM2 receptor internalization. HEK 293 cells were co- transfected with CL-R or CL-R point mutants and RAMP3. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour. washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using 125l—rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 3. 95 WT (‘L-R RAMP3 ()\ Ci‘lziy No Pretreat CL-R T423A AM Pretreat No Pretreat AM Pretreat Figure 11C: Role of phosphorylation on AM2 receptor desensitization and internalization (point mutations). lmmunofluorescence microscopy shows failure of CL-R T423A/RAMP3 receptor complex to internalize after agonist stimulation. HEK 293 cells transfected with CL-R T423A- GFP and RAMP3 were pretreated with 10nM AM for 1h. After pretreatment with AM, cells were washed, fixed, and components were visualized using anti-RAMP3 antibody (1:150) and detected with Cy3 secondary antibody (1:250), and CL-R is detected with an EGFP tag; overlays of staining patterns are shown in the far right panels. Images shown are representative of at least twenty fields imaged per experiment from at least three experiments. Bar scales on all images represent lOOum. 96 - No Pretreat 1:21AM Pretreat A 40- c E -' 30 > at '4: x * 2 E. g 20- :r. * .1. * 2 g IL H I [t B E- 10- E l 9. OJ _ l — d _— v? 019 v 939)“ ix" *9)“ is" *9}: is" *0“ Figure 12A: Role of phosphorylation of residues in third intracellular loop of CL-R on AM] and AM2 receptor desensitization. Phosphorylation of third intracellular loop residues of CL-R not vital in AM 1 receptor desensitization. HEK 293 cells transiently transfected with CL-R/CL-R point mutants and RAMP2 were seeded in 48-well plates and treated for one hour with AM (10 nM) and then washed and receptor desensitization (CAMP accumulation) was measured. After repeated wash steps, cells were re-Challenged with 100 nM rAM for 15 minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging fi'om 10 to 10 4 firm] of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p S 0.05; n 2 3 experiments. 97 - No Pretreat D AM Pretreat [°/o Forskolin] w e. ? 9' AC Activity (pmollminllmg protein) 25 0.: W" «P “V3 9.9. ‘5 9.? “a: k k 93‘ :c 0“” Figure 128: Role of phosphorylation of residues in third intracellular loop of CL-R on AM] and AM2 receptor desensitization. Third intracellular loop residues of CL-R not phosphorylated in desensitization of AM2 receptor. HEK 293 cells transiently transfected with CL-R/CL-R point mutants and RAMP3 were seeded in 48-well plates and treated for one hour with AM (10 nM) and then washed and receptor desensitization (CAMP accumulation) was measured. After repeated wash steps, cells were re-Challenged with 100 nM rAM for 15 minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 10 4 fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p _<_ 0.05; n 2 3 experiments. 98 - CL-R+RAMP2 E CL-R+RAMP2AS168,171A CAMP (nmollmg protein) [% Forskolin] N ‘P if] No Pr'etreat AM Pretreat 0d Figure 13A: Role of phosphorylation of RAMPS on AM] and AM2 receptor desensitization and internalization. Phosphorylation of residues on RAMP2 not essential to desensitization of AM] receptor. HEK 293 cells transiently transfected with CL-R and RAMP2/RAMP2 phosphorylation mutants were seeded in 48—well plates and treated for one hour with AM (10 nM) and then washed and receptor desensitization (CAMP accumulation) was measured. After repeated wash steps, cells were re-Challenged with 100 nM rAM for 15 minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 10 " fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p S 0.05; n 2 3 experiments. 99 - CL-R+RAMP2 D CL-R+RAMP2$168,171A 100- * 1'. Binding max (sites/cell) [°/o Control] 0| 8’ O I No Pr'etreat AM Pretreat Figure 138: Role of phosphorylation of RAMPS on AM] and AM2 receptor desensitization and internalization. Phosphorylation of RAMP2 is not critical to AMI receptor internalization. HEK 293 cells were co-transfected with CL-R and RAMP2/RAMP2 phosphorylation mutants. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour, washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using 12sl-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. "' p S 0.05; n 2 3. 100 - CL-R+RAMP3 D CL-R+RAMP3AS141T144,146A h C I N O n E'— . A O . n CAMP (nmollmg protein) [°/o Forskolin] N ‘P if] No Pr'etreat AM Pretreat O I Figure 13C: Role of phosphorylation of RAMPS on AM] and AM2 receptor desensitization and internalization. Phosphorylation of residues on RAMP3 not required for desensitization of AM2 receptor. HEK 293 cells transiently transfected with CL-R and RAMP3/RAMP3 phosphorylation mutants were seeded in 48-well plates and treated for one hour with AM (10 nM) and then washed and receptor desensitization (CAMP accumulation) was measured. Afler repeated wash steps, cells were re-Challenged with l00 nM rAM for 15 minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from ID to 104 frnol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p S 0.05; n 2 3 experiments. 10] - CL-R+RAMP3 E CL-R+RAMP3S141T144,146A 100- Binding max (sites/cell) [°/o Control] $ O I No Pfetreat AM Pretreat Figure 13D: Role of phosphorylation of RAMPS on AM] and AM2 receptor desensitization and internalization. Phosphorylation of RAMP3 is not vital for AM2 receptor internalization. HEK 293 cells were co-transfected with CL-R and RAMP3/RAMP3 phosphorylation mutants. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour, washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using I2SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 3. 102 - CL-R+RAMP2 El CL-R+RAMP2 (H-89) CL-R+RAMP2 (R0 32-0432) h C I .T. I. 09 O I A O I No Pretreat AM Pretreat CAMP (nmollmg protein) [% Forskoli ] N C Figure 14A: Role of kinases in AMI receptor desensitization and internalization. Phosphorylation of residues of CL-R for desensitization and internalization of AMI receptor is sensitive to H-89 in HEK 293 cells. Cells transiently transfected with CL-R and RAMP2 were seeded in 48-well plates and treated for one hour with AM (10 nM) with and without H-89 (1 pM) and Ro 32-0432 (3 uM) treatment, then washed, and receptor desensitization (CAMP accumulation) was measured. Afier repeated wash steps, cells were re-Challenged with 100 nM rAM for 15 minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 10 4 fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p S 0.05; n 2 3 experiments. 103 - CL-R+RAMP2 D CL-R + RAMP2 (H-89) IE: CL-R + RAMP2 (R0 32-0432) 125- I. § 7, 100' r- g: tag 75- x c u o E: 50. 3°“ :3 25- m l {31“} "'l' -r- No Pretreat AM Pretreat Figure 143: Role of kinases in AMI receptor desensitization and internalization. Phosphorylation of residues of CL-R for internalization of AMI receptor is sensitive to H-89 in HEK 293 cells. HEK 293 cells were co-transfected with CL-R and RAMP2. 48h post- transfection, cells were pretreated with AM (l0 nM) for one hour with and without H-89 (luM) and Ro 32-0432 (3uM) treatment, washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 3. 104 - CL-R+RAMP3 E CL-R+RAMP3 (H—89) CL-R+RAMP3 (R0 32-0432) A O I I. U C I .x O I CAMP (nmollmg protein) [% Forskolin] N c.’ O I_ No Pretreat AM Pretreat Figure 15A: Role of kinases in AM2 receptor desensitization and internalization. Phosphorylation of residues of CL-R for desensitization and internalization of AM2 receptor is sensitive to RO 32-0432 in HEK 293 cells. Cells transiently transfected with CL-R and RAMP3 were seeded in 48-well plates and treated for one hour with AM (10 nM) with and without H-89 (luM) and Ro 32-0432 (3},LM) treatment, then washed, and receptor desensitization (CAMP accumulation) was measured. After repeated wash steps, cells were re-Challenged with 100 nM rAM for IS minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from l0 to 10 4 fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p S 0.05; n 2 3 experiments. 105 - CL-R+RAMP3 [:1 CL-R+RAMP3 (RO 32-0432) n CL-R+RAMP3 (H-89) A 125- g l 7, 100- —~ g ..."'.." £3 75- X C a O * E 3 5o. 3"“ E 25- m , ' '1‘ -I- No Pretreat AM Pretreat Figure 158: Role of kinases in AM2 receptor desensitization and internalization. Phosphorylation of residues of CL-R for internalization of AM2 receptor is sensitive to R0 32- 0432 in HEK 293 cells. HEK 293 cells were co-transfected with CL-R and RAMP3. 48h post- transfection, cells were pretreated with AM (IO nM) for one hour with and without H-89 (luM) and Ro 32-0432 (3uM) treatment, washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using I25I-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. "‘ p S 0.05; n 2 3. 106 - CL-R+RAMP2 D CL-R+RAMP3 II N u o o l I 11 [°/o Forskolin] 2;; CAMP Accumulation (nmollmg protein) 04 _ 633‘ 6°" C9?“ C0" goq‘e le‘v‘e «3‘6 “P‘Q‘e V Figure 16A: Role of kinases in AM2 receptor desensitization and internalization. Desensitization and internalization of AMI receptor is dependent on PKA activation, while the AM2 receptor requires PKC activation. Cells transiently transfected with CL-R and RAMP2 or RAMP3 were seeded in 48-well plates and treated either for one hour with AM (10 nM) or for 30 min. with forskolin (10 nM) or for 10 min. with PMA (1 uM), then washed, and receptor desensitization (CAMP accumulation) was measured. After repeated wash steps, cells were re- Challenged with 100 nM rAM for IS minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in transfected HEK 293 cells were calculated using a standard curve ranging from 10 to 10 4 fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. "‘ p S 0.05; n 2 3 experiments. 107 - CL-R+RAMP2 E3 CL-R+RAMP3 125- 100- )— an N O 0| I I Binding Sites/Cell [°/o Control] «'3 O k Figure 168: Role of kinases in AM2 receptor desensitization and internalization. Desensitization and internalization of AMI receptor is dependent on PKA activation, while the AM2 receptor requires PKC activation. Cells transiently transfected with CL-R and RAMP2 or RAMP3 were seeded in 48-well plates and treated either for one hour with AM (10 nM) or for 30 min. with forskolin (IO uM) or for IO min. with PMA (1 uM), washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 3. 108 4.4. Discussion The process of agonist-promoted receptor desensitization and internalization is an indispensable self-regulation mechanism of the GPCRS. These processes allow receptor signaling to be Closely-monitored, providing a mechanism to promote receptor signaling or receptor down-regulation. Receptors preserve this maintenance in many different ways, with phosphorylation of the receptor ranking as one of the most common [196]. Receptor phosphorylation during the process of desensitization is thought to enable [3- arrestin molecule interaction with the receptor, thus uncoupling the receptor from its obligate G-protein and signaling for the assembly of the endocytic machinery for receptor internalization. In this study we examined the role of phosphorylation in the desensitization and internalization processes of the AM receptor subtypes. Putative phosphorylation residues were found to play a vital role in the process of desensitization and internalization of both AM receptor complexes. Notably, the two AM receptor subtypes were regulated by different amino acid residues on CL—R. Namely, the serine residue 421 of CL-R and threonine residue 423 of CL—R were responsible for the efficient receptor desensitization and internalization of the CL—R/RAMPZ and CL-R/RAMP3 receptor complexes, respectively. Protein kinase A inhibitor blocked only CL-R/RAMPZ desensitization and internalization, while a PKC inhibitor blocked only CL-R/RAMP3 desensitization and internalization, suggesting different kinases are involved in the phosphorylation of CL—R when different RAMP isoforms are associated. In addition, it was demonstrated the phosphorylation of the RAMP proteins or the third intracellular loop of CL-R was not required for AM receptor complex desensitization or internalization. 109 A prolonged exposure of cells to a particular ligand results in an attenuation of responsiveness to subsequent stimulation with that ligand. This phenomenon is called desensitization, which can be homologous or heterologous. Homologous desensitization is Classified as a decrease in the response to a particular ligand that is specific for the stimulated receptor. Because G-protein regulated kinases (GRKS) are activated and only phosphorylate GPCRS in the agonist-bound state, GRKS are capable of attenuating receptor signaling by homologous desensitization [194, 195]. An attenuation of receptor signaling that is the result of activation of second messenger signaling, and in principal not specific for the agonist-bound receptor, is termed heterologous desensitization. Protein kinase A and C are common kinases activated through second messenger signaling of GPCRS, and therefore can activate heterologous receptor desensitization [196]. While past studies in HEK 293 cells have identified homologous desensitization as a means of regulating CL-R, this was only Characterized for the CL-R/RAMPI receptor complex (CGRP-1 receptor) [20]]. Meanwhile, this study has identified heterologous desensitization to play a crucial role in the regulation of AM signaling in the HEK 293 cells, through both AM receptor subtypes. The current literature characterizes the CL-R/RAMP receptor complex to undergo heterologous desensitization in the majority of cell lines studied. Desensitization of CL- R was found to involve CAMP-dependent protein kinase (PKA) activation in rat mesangial cells (RMCS) stimulated with AM, SK-N-MC (neuroblastoma cell line) cells stimulated with CGRP, Rat2 fibroblast cells stimulated with AM, and vascular smooth muscle cells (V SMCs) stimulated with CGRP [198-200, 287]. No studies to date have reported regulation of desensitization of the CL-R/RAMP receptor complex by PKC. 110 Although, this kinase is found to promote heterologous desensitization of numerous other GPCRS, for example the D2 dopamine [288], H1 histamine [289], serotonin 5-HT(2A) [290], thromboxane receptor alpha [291], and alpha(2)-adrenergic receptors [292]. Data from this manuscript demonstrates a requirement in HEK 293 cells of PKA phosphorylation for AMI receptor desensitization/intemalization and PKC phosphorylation for AM2 receptor desensitization/internalization. Further studies are needed to define the implications of differential receptor phosphorylation to the AM signaling pathway. It is tempting to hypothesize that the different phosphorylation sites on CL-R when complexed with RAMP2 or RAMP3 may allow different protein-protein interactions that dictate down-stream receptor signaling or trafficking. It could be predicted that the differential phosphorylation of the AMI and AM2 receptors allow protein-protein interactions that allow the documented targeting of the two receptor complexes for different pathways after endocytosis, degradative vs. recycling pathways, respectively [153]. Understanding the mechanism of AM receptor desensitization is of great importance given the documented protective role of AM in various cardiovascular and renal disorders. In the cardiorenal disease states where AM is protective, circulating plasma levels of AM have been shown to be increased. For example, in Chronic glomerulonephritis, type I diabetes, and type II diabetes plasma AM levels are elevated [293-295]. In addition, AM delivery through adenoviral injection has been shown to decrease cardiac hypertrophy and renal damage in rat models of hypertension and improves cardiac function and prevents renal damage in streptozotocin-induced diabetic rats [296-300]. Because AM levels are chronically elevated in many of the above 111 cardiovascular and renal disorders, targeting receptor desensitization is a possible therapy for these pathophysiologies. The inhibition of the process of desensitization for additional GPCRS has been shown to elicit protective therapeutic effects for the disease states associated with the receptors. Leflcowitz et. al. have shown that the inhibition of [3- adrenergic receptor kinase (kinase that phosphorylates the B-adrenergic receptor to cause desensitization) in the heart can delay the development of heart failure in multiple animal models, in some cases even restoring cardiac function [203, 204]. Others have also shown inhibition of desensitization of the u—opioid receptor to be beneficial in preventing morphine tolerance [205, 206]. This is the first study to show that the RAMP isoforms are capable of dictating differential phosphorylation of CL-R to regulate the AM receptor complex agonist- stimulated desensitization and internalization. In addition, it has not previously been shown that different kinases are responsible for the phosphorylation of the two AM receptor subtypes (PKA for AMI receptor and PKC for AM2 receptor). This report indicates yet another novel form of regulation of the AM receptor life-cycle by the RAMP proteins. Given the recent report of RAMPS ability to interact with other receptors in the family 11 of GPCRS, RAMPS may be playing a more prevalent role in the regulation of GPCR life-cycle [286]. 112 5. RAMP isoform-specific regulation of adrenomedullin receptor trafficking by NHERF-1. 5.1 Introduction The recent discovery of receptor activity modifying proteins (RAMPS) has broadened the field of G-protein coupled receptor (GPCR) regulation. RAMPS were discovered as required accessory proteins to an orphan GPCR, now termed the calcitonin receptor-like receptor (CL-R) [275]. The three RAMP isoforms (1-3) are products of three distinct genes and yield unique single transmembrane accessory proteins. RAMPS are required for the plasma membrane expression and determination of receptor phenotype for CL-R [275, 276]. RAMPS have recently been found to associate with additional members of the Class 11 family of GPCRS [286]. Co-expression of RAMP] with CL-R yields a functional calcitonin gene-related peptide (CGRP) receptor, while co- expression of RAMP2 or -3 with CL-R produces a receptor responsive to adrenomedullin (AM) (AMI-R and AM2-R, respectively) [277, 278]. AM and CGRP are multi- functional peptides with many overlapping functions, ranging from potent vasodilation to proliferation to regulation of salt and water balance [279]. The RAMP isoforms have shown a differential expression patterns in different organ systems and in different pathophysiological states, suggesting a regulatory role for RAMPS in both physiological and pathophysiological situations. Furthermore, the identification of RAMP interactions with additional members of the Class II GPCR family and RAMP expression in cell lines lacking CL-R have raised the possibility of novel functions for RAMPS in GPCR regulation. 113 It has been shown in other GPCR systems that interactions with PSD-95/Discs- large/ZO-l homology (PDZ) domain proteins are responsible for altering the receptor- trafficking after agonist stimulation [259, 267, 268]. In particular, a protein called Na+/H” Exchange Regulatory Factor-1 (NHERF-1) has been shown to regulate the trafficking of the BZ-AR, K-OR, and PTH-R after agonist activation [240, 276, 301]. NHERF-1 is an adapter protein that is thought to tether membrane receptors to cytoskeletal proteins through PDZ interactions and interactions with MERM family of cytoskeletal proteins [257]. Comparable to the C-terminus of BZ-AR, PTH-R, CFTR, and PDGF-R, human RAMP3 C-terminus contains a type-I PDZ recognition motif, whereas CL-R, RAMPl or RAMP2 do not contain any PDZ recognition sequences [153, 154, 240, 250, 259, 302]. We hypothesized that RAMP3, via its interaction with NHERF-1, can regulate the trafficking of the CL-R/RAMP3 complex. We show here that while CL-R/RAMPI and CL-R/RAMP2 complexes do not interact with NHERF-1, CL—R/RAMP3 complex interacts with NHERF-l via the PDZ domain of NHERF-l [303]. Moreover, we show here that over-expression of NHERF-l in HEK-293 cells alters the trafficking pattern of the receptor complex to block the receptor’s internalization by tethering the receptor complex to the actin cytoskeleton via interactions between RAMP3 and NHERF-1 through a type I PDZ domain. Furthermore, we also demonstrate that in primary human proximal tubule cells (which express endogenous NHERF-l and CL-R/RAMPB), the CL- R/RAMP3 complex does not internalize upon agonist stimulation. Knocking down NHERF-1 or RAMP3 expression with RNAi causes the receptor to undergo 114 internalization upon agonist treatment, suggesting critical roles for both NHERF and RAMP3 in receptor internalization. 5.2. Materials and Methods 5.2.1. Materials: Adrenomedullin was purchased from Bachem Bioscience, Inc. (King of Prussia, PA). 1251-labeled adrenomedullin was purchased from Amersham Biosciences Corp. (Piscataway, NY). DMEM media, fetal bovine serum, penicillin/streptomycin, trypsin-EDTA were purchased from GibcoBRL® (Grand Island, NY). RAMP3 antibody was purchased from Santa Cruz Biotechnology (Santa Cruz, CA) and NHERF-1 (EBPSO) antibody was from Affinity Bioreagents (Golden, CO). Alexa 488-phalloidin was purchased from Molecular Probes (Eugene, OR). Anti-mouse Cy3 and anti-rabbit Cy5 secondary antibodies were from Jackson Immunoresearch Laboratories (West Grove, PA). All other reagents were of highest quality available. 5.2.2. Cell Culture and Transfection protocols: HEK-293T cells were obtained from ATCC and are maintained in DMEM low glucose media containing 10% FBS, 1%penicillin-streptomycin. Transfection of HEK293T cells was performed using Lipofectamine Plus protocol (Invitrogen). Cells were transfected with the DNA and Lipofectamine Plus as per manufacturer’s protocol. Cells were collected for assays after 48 hours of transfection. Human proximal tubule epithelial (hPTE) cells were acquired from Mediatech and maintained in appropriate media. Transfection of d-siRNA into hPTE cells was performed with Lipofectamine 2000 (Invitrogen) and cells incubated for 48 hours before assaying. 115 5.2.3. RAMP Cloning and expression: Full length CDNA of human RAMPS], 2 and 3 and bovine CL-R were described before [281, 282]. CL-R, Cloned into Nl-EGFP and also in pcDNA3.l expression vectors, was used for transfection in HEK 293T cells. 5.2.4. Desensitization and Resensitization assays: 48 hours post-tranfection cells were pretreated with or without 10 nM AM in DMEM containing 0.2% BSA for indicated time periods (up to 4 hr). After agonist exposure, cells were washed three times with Dulbecco’s phosphate buffered saline (dPBS, Gibco BRL) chtaining 0.2% BSA and either frozen for membrane preparation for adenylate cyclase assays or used immediately for intact-cell radioligand binding. For receptor resensitization assays, after agonist exposure, cells were washed and incubated for indicated time periods in DMEM containing 0.2% BSA and 5 11ng cycloheximide to allow receptor recovery. Desensltizatioanesensitlzation Protoc I: Removal 01AM, followed by Cello AM PNWOUMM {- weehlng. followed by Incubation wlthout AM ”ml M7 n.1, K ATL-L—Y'J—J Receptor eotlvlty essay 4m / 1 hr 24 hrs / (adenylate cyclase) and receptor blndlng et (MOV'W ”ml different tlme point- No Pretreat (Full Stlmulatlon) 1 hr AM Pretreat 2 I" (Desensitization) Recovery (Resensitization) 5.2.5. Receptor binding: Homologous competition radioligand binding assays were performed as described by Aiyar et a1 and as established in our laboratory [283]. HEK 293T cells were transfected and approximately 200,000 cells/well seeded in poly-D- 116 lysine pre-coated 24-well plates (BD Biosciences, Palo Alto, CA). 48 hours post- transfection cells were treated for desensitization or resensitization assays as described above. After agonist exposure, cells were washed three times with dPBS buffer containing 0.2% BSA then incubated with increasing concentrations (1 pM to 100 nM) of competing ligand (rAM) and 175-250 pM 125I-rAM for 30 min at 37°C. After incubation, plates were washed three times with ice-cold assay buffer and the reactions were terminated by the addition of 2M NaOH. Cells were then harvested and associated radioligand activity is counted on a y-counter. All binding assays were performed in duplicate, with each experiment repeated at least three times. Nonspecific binding was determined in the presence of 100 nM of unlabeled rAM. Data was analyzed by LIGAND (assuming radioligand and competitor both bind reversibly to a single binding site; MacLigand, Version 4.97, NIH, Bethesda, MD) using the following equation: Bmax 0 [hot ligand] KD + [hot ligand] + [cold ligand] Where B represents specific binding, [hot ligand] the single concentration of [1251]rAM studied, [cold ligand] the concentration of unlabeled rAM competing with the radiolabel for AM receptor binding, Bmax the maximum number of binding sites and KD the equilibrium dissociation constant (the equation was solved where the “cold ligand” ICso = [hot ligand] + KD). Analysis of all binding data was performed by computer-assisted nonlinear least square fitting using GraphPad PRIZM version 4 (GraphPad Software, San Diego, CA). Binding sites/cell was calculated and data was expressed as percent of the control. 117 5.2.6. Adenylate cyclase assays: Cyclase activity was done as described before with slight modifications [282, 284]. Cells were harvested from P100 or P60 plates and homogenized in Tris HCI (10mM)/EDTA (10mM) buffer. Membranes were prepared by homogenization and centrifugation in Tris HCl (50mM)/MgCl (10mM) buffer. Final concentration of 20 pg of protein/assay tube was obtained. Membranes were incubated for 15 min at 30°C with appropriate concentrations of drugs and assay mix containing ATP regeneration system and a32P-ATP. After the reaction was stopped (with stop solution containing 3H-CAMP) contents of the assay tubes were passed through Dowex and subsequently through alumina columns to separate the degradation products of ATP, by washing the dowex with water and alumina with imidazole. Elution profile was done to determine the amount of water and imidazole needed to wash and elute the products. Product eluted from alumina column was counted for the presence of 3H-CAMP and (13 2P-CAMP in a B-Counter. Each experiment was done in triplicates and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. Basal cyclase activity and forskolin stimulation did not show statistically significant differences between treatments. 5.2.7. CAMP accumulation assays: Human proximal tubule cells were seeded on a 24- well plate until reaching 80-90% confluency, then incubated in serum-free media overnight before experiment. Desensitization experiments were carried out as described in Materials and Methods section, with cells pretreated with 10nM rAM and subsequently Challenged for 10 min at 37°C with appropriate concentrations of drugs (100nM AM, 118 lOuM Forskolin) in the presence of ZOOuM 3-isobutyl-l-methylxanthine. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in human proximal tubule cells were calculated using a standard curve ranging from 10 to 10 4 fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. Basal cyclase activity and forskolin stimulation did not show statistically significant differences between treatments. 5.2.8. RNA Interference analysis: Gene-specific d-siRNA for lacZ (control), NHERF-I and RAMP3 were generated and purified using BLOCK-iT Dicer RNAi kit from Invitrogen (Carlsbad, CA). hPTE cells were transfected with d-siRNAs using Lipofectamine 2000 as per manufacturer’s instructions (Invitrogen). 48 hours after transfection cells were frozen for mRNA analysis, or used for CAMP accumulation assays or immunofluorescence microscopy. 5.2.9. Reverse transcriptase PCR (RT-PCR) analysis: RT-PCR analysis performed as described before [282].Total RNA was isolated from hPTEs using Trizol reagent (GIBCO BRL). After sodium acetate-ethanol precipitation and several ethanol washes, RNA was used as a template in a reverse transcriptase PCR amplification procedure. The RT-PCR reaction was carried out using Superscript One-Step RT-PCR with Platinum T aq (GIBCO BRL), in accordance with the manufacturer's specifications. Reactions were carried out, with a Perkin-Elmer model 9600 thermal cycler, in 50 ul of total reaction volumes subjected to the following conditions: 1) 94°C for 2 min (1 cycle); 3) 94°C for 119 30 s, 50°C for 30 s, 68°C for 30 s (30 cycles); and 4) 68°C for 7min (1 cycle). Products were separated by gel electrophoresis and subsequently visualized by ethidium bromide staining and ultraviolet illumination. Photographs of the gels were taken and digitalized with a UMAX Astra 2000P flat-bed scanner. 5.2.10. Mutagenesis procedure: Site-directed mutagenesis was performed using a PCR- based strategy that employs the pfu Turbo polymerase (Stratagene, La Jolla, CA). A pair of complementary oligonucleotides containing the appropriate point mutations in the sequence of RAMP/CL-R or a premature stop codon at position 145 or 147 codon of RAMP3 for deletion mutants were synthesized (Michigan State University Macromolecular structure facility). The PCR for the mutation was as follows: 94°C for 5 minutes; 30 cycles of 94°C for 30 sec., 50°C for 30 sec., 68°C for 8 min.; final cycle of 68°C for 8 minutes. PCR product was digested for 4 hours with DpnI enzyme (Invitrogen) and transformed in to DHSG. cells. Mutations were confirmed by automated sequencing (Michigan State University Genomic Technology Support Facility). 5.2.11. Immunofluorescence microscopy: HEK293 and hPTE cells were transfected as described above and seeded at 24hr post-transfection onto collagen type I-Coated coverslips. Desensitization assays were performed as described and reactions were stopped by fixing cells in 4% paraformaldehyde for 30min. at room temperature. Samples were perrneablized with 0.1% v/v Triton X-100 in PBS and blocked overnight in 0.1% v/v Triton X—100 in PBS + 10% goat serum. Samples were incubated in primary antibody in blocking buffer for 2h at room temperature (NHERF-1 at 1:250 and RAMP3 120 at 1:200). Appropriate secondary antibodies were applied for 1h at room temperature (Goat anti-mouse Cy3 at 1:500 and Goat anti-rabbit Cy5 at 1:400). Cytoskeletal staining was carried out using Alexa 488-phalloidin antibody at 1:75 (Molecular Probes). Coverslips were mounted in Shandon mounting medium and slides store at 2-8°C until analysis. Cells were visualized on a Zeiss 210 laser confocal microscope at a zoom of 2. Images presented are representative single optical sections of a z-series taken from at least twenty fields per experiment and at least three individual experiments. Images in this thesis/dissertation are presented in color. 5.2.12. Fusion protein overlays and western blotting: Overlay assays and western blotting performed as described before [153]. 10 pg of GST-fusion proteins were resolved on a 10% SDS-PAGE gel and transferred to nitrocellulose filters. Filters were blocked with 5% w/v fat-free milk powder in Tris-buffered saline with Tween 20 (TTBS: 20 mM Tris, pH 7.4, 500 mM NaCI, 0.1% v/v Tween 20) and incubated overnight at 4 °C in lysates of HEK293 cells with or without overexpression NHERF-l. Blots were then washed three times with TTBS buffer and incubated with anti-EBPSO (NHERF-1) monoclonal antibody for 2 h at room temperature. After three washes with TTBS, filters were incubated for 1h with horseradish peroxidase—conjugated goat anti-mouse secondary antibody (Gibco BRL®, Grand Island, NY), washed again with TTBS, soaked in Supersignal West Pico chemiluminescent substrate (Pierce) and exposed to x-ray film. Same protocol, with the exception of the overnight incubation with cell lysate, was followed for immunoblot analysis of RAMP3. 121 II-*— ——- r -I 5.2.13. Statistics: Data are presented as mean i S.E.M. Single group comparisons exercised a paired Student’s t-test method. Statistical significance was set at P<0.05. 5.3. Results 5.3.1. Role of NHERF-1 in Intemalization of the CL-R/RAMP complex We and others have previously shown that the AM2 receptor (CL-R + RAMP3 complex) undergoes agonist-stimulated desensitization, internalization, and degradation [153, 276, 285]. Furthermore, we have recently shown that in the presence of NSF, the AM2 receptor complex undergoes recycling, instead of following a degradation pathway, in HEK 293 cells [153]. In this study, we have examined the role of another protein, namely NHERF-1, on agonist-induced trafficking of the CL-R/RAMP3 complex. Pretreatment of HEK 293 cells expressing CL-R and RAMP3 with 10 nM AM for one hour resulted in desensitization of the adenylate cyclase response from 50% (of forskolin stimulation) in untreated cells to 28% in AM-treated cells (Figure 17A). In addition, as shown in Figure 178 and 18, CL-R/RAMP3 complex underwent agonist- induced internalization as determined by receptor binding and immunofluorescence microscopy. These findings are in agreement with those of Kuwasako et al. and as reported by us recently [153, 276]. However, over-expression of NHERF-1 with CL- R/RAMP3 resulted in a remarkable change in the agonist-induced receptor complex trafficking in HEK293 cells. HEK293 cells transfected with CL-R/RAMP3 and NHERF-1 showed similar levels of adenylate cyclase activity and desensitization patterns as compared to CL- R/RAMP3 alone (Figure 17A). But in the presence of NHERF-1, the receptor complex 122 failed to internalize with agonist pretreatment (Figure 17B, 18). To determine if NHERF-1 co-expression had changed the kinetics of internalization, agonist pretreatment was carried out for a time course extending to 4 hours. Four hours of agonist pretreatment still yielded a complete inhibition of receptor complex internalization in HEK293 cells co-expressing NHERF-l and the CL-R/RAMP3 complex, as compared to a continued internalization of the receptor complex in HEK293 cells lacking NHERF-l over-expression (Figure 17C). These results indicate that over-expression of NHERF-l alters the trafficking of the CL—R/RAMP3 receptor complex after AM-stimulated desensitization. 5.3.2. RAMP isoform-specific regulation of CL-R/RAMP receptor complex trafficking To determine if this effect of NHERF-1 was specific for RAMP3, the additional RAMPS (RAMPI or -2) were tested for their ability to act with NHERF-1 to alter the receptor complex trafficking. Interestingly, in contrast to RAMP3, presence of NHERF-I did not alter the internalization pattern of the CL-R/RAMPI or -2 receptor complexes. No significant differences were seen in the receptor numbers from whole-cell binding when CL-R was co-expressed with RAMPI, 2, or 3 (with and without NHERF-1). Desensitization patterns in cells transfected with CL-R+RAMP1 or CL—R+RAMP2 also remained unchanged in the absence or presence of NHERF-l (Figure l9A-D). These results indicate that RAMP3 must contain a molecular feature distinct from the other RAMPS that allowed its interaction and action with NHERF-1. 123 5.3.3. Role of PDZ interactions in trafficking of the CL-R/RAMP3 complex We have observed before that NSF regulated the CL-R/RAMP3 recycling by interacting specifically with the PDZ motif present at the extreme C-terminus of RAMP3. Neither CL-R, nor the other two RAMP isoforms (RAMPl or RAMP2), contain PDZ recognition motifs. To test the hypothesis that this domain is critical for interaction of the CL-R/RAMP3 complex with NHERF-l, the PDZ motif (-DTLL) on RAMP3 was deleted (RAMP3A145-8) and an internalization assay was performed. Deletion of this domain did not affect basal adenylate cyclase activity or the desensitization response of the CL- R/RAMP3 complex in response to AM, even in the presence of NHERF-l. In addition, the RAMP3 PDZ motif mutant (RAMP3AI45-8) showed no difference in receptor expression levels at the plasma membrane (as measured with whole-cell binding), as compared to wild-type CL-R/RAMP3 complex (in the presence/absence of NHERF-1). Unlike CL-R/RAMP3 complex, the CL—R/RAMP3A145-8 receptor complex was now capable of AM-induced internalization, as measured by whole-cell receptor binding and immunofluoresence microscopy (Figure 20A and 21). To further test the hypothesis that the absence of the PDZ motif on the RAMP2 accounts for the lack of ability of NHERF-1 to interact and therefore inhibit internalization, the PDZ motif of RAMP3, the amino acids -DTLL, were substituted on the C-terminus of RAMP2, in exchange for its original four C-terminal amino acids (- EAQA). The CL—R/RAMPZADTLL mutant complex expression levels were comparable to that of CL-R/RAMP2 complex, as determined by whole-cell binding and adenylate cyclase assays. Additionally, in the absence of NHERF-1, both CL-R/RAMPZ and CL- R/RAMPZADTLL showed similar levels of AM-stimulated internalization. However, C0- 124 expression of NHERF-1 with CL-R/RAMPZADTLL was now capable of inhibiting the internalization of the receptor complex with agonist pretreatment, as compared to wild- type CL-R/RAMPZ complex (Figure ZOB). These findings provide additional evidence that the PDZ motif on the RAMP3 interacts with NHERF-1, causing an inhibition of receptor internalization, despite normal desensitization. To identify the critical amino acids in the PDZ binding sequence that regulate the RAMP3/NHERF-1 interaction, site-directed mutagenesis was performed to mutate the individual amino acids of the PDZ motif to alanine. Mutations of the individual amino acids in the PDZ motif of RAMP3 did not affect the basal levels of receptor expression and function, as measured by whole-cell binding experiments and adenylate cyclase assays, respectively. In addition, the desensitization and internalization in the absence of NHERF-l were also similar between the wild-type and mutant CL-R/RAMP3 complexes. However, in the presence of NHERF-l, the CL-R/RAMP3T146A complex now underwent agonist-stimulated internalization, similar to when expressed without NHERF- ] (Figure 20C). The other point mutant RAMP3/CL-R complexes behaved like wild-type in the presence of NHERF-l, indicating Thr146 in the PDZ domain is critical for the PDZ interaction between RAMP3 and NHERF-1. As described before, NHERF-1 contains two PDZ domains through which it interacts with numerous proteins [258]. To determine which PDZ domain of NHERF-1 is responsible for the interaction with RAMP3, the two PDZ domains of NHERF-1 were deleted individually and agonist-induced internalization assays, employing whole-cell receptor binding, were performed. Cells expressing wild-type or mutant NHERF-1 with CL-R/RAMP3 receptor complex showed comparable levels of receptor expression and 125 function, as assessed by whole-cell receptor binding and adenylate cyclase assays, respectively. In addition, they showed similar desensitization patterns. However, internalization assays (with whole—cell binding) showed that the first PDZ domain of NHERF-l is responsible for the interaction with RAMP3. HEK 293 cells expressing the mutant NHERF-IAPDZI (lacking only the first PDZ domain) and CL-R/RAMP3 complex underwent AM-stimulated internalization compared to cells expressing the wild- type NHERF-1 (Figure 20D). Deletion of the second PDZ domain of NHERF-l (NHERF-IAPDZZ) had no effect on the internalization pattern of CL-R/RAMP3, as compared to wild-type NHERF-1 (Figure 20D). These findings further confirm that RAMP3 and NHERF-1 are interacting via their PDZ domains to inhibit internalization of the CL-R/RAMP3 receptor complex. To examine if the PDZ domain on RAMP3 is physically interacting with NHERF- ], overlay assays were performed. This was accomplished using GST-RAMP3 fusion proteins in an overlay assay with cell lysates of HEK293 cells overexpressing NHERF-l. Control experiments run with GST protein showed no detectable bands when incubated with NHERF-1 lysates and probed with an NHERF-1 antibody (Figure 22A). Irnportantly, GST- RAMP3 fusion proteins showed significant interaction with NHERF-l in the cell lysates of HEK293 cells overexpressing NHERF-1 in the overlay assay (Figure 22A). As a control, lysates of HEK 293 cells not over-expressing NHERF-1 showed no detectable bands when run with GST-RAMP3 in the overlay assay (Figure 22B). Additionally, when GST-RAMP3A145-8 fusion proteins were tested for interaction with NHERF-l using the above described overlay assay, no bands were detected, in contrast to wild-type GST-RAMP3 (Figure 22A). When blots used in the overlay assay were 126 stripped and probed for RAMP3, a band for RAMP3 was detected at the exact location as that of the NHERF-1 band detected in the overlay assay (Figure 22C). This data demonstrates a physical interaction between RAMP3 and NHERF-1, an interaction that is dependent on PDZ domain interactions of the two proteins and is capable of regulating CL-R/RAMP3 (AM2R) complex trafficking. 5.3.4. Mechanism of inhibition of CL-R/RAMP3 internalization by NHERF-1 The ERM domain of NHERF-1 is known to interact with MERM cytoskeletal proteins, allowing NHERF-1 to tether proteins to the actin cytoskeleton [304]. Hence, we hypothesized that the CL—R/RAMP3 receptor complex internalization is regulated by NHERF-1 through interactions of NHERF-l with cytoskeletal proteins. To test this hypothesis, we employed a mutant of NHERF-l with a deletion of its ERM domain [252]. Control experiments with the ERM domain mutants of NHERF-1 co-transfected with CL-R/RAMP3 showed similar levels of receptor expression and function, as measured by whole-cell binding and adenylate cyclase. As hypothesized, the ERM domain mutant of NHERF-l, when co-expressed in HEK 293 cells with CL-R/RAMP3 complex, was now capable of internalization after agonist pretreatment, differing from the wild-type NHERF-1 that inhibits receptor internalization (Figure 23A). These experiments were repeated and confirmed with confocal immunofluorescence microscopy, using phalloidin to label the actin cytoskeleton and test for NHERF-1 co- localization. While wild-type NHERF-l colocalized with the actin cytoskeleton (to presumably tether RAMP3 and therefore CL-R to the plasma membrane after agonist pretreatment), the ERM domain mutant of NHERF-l showed disrupted actin 127 cytoskeleton co-localization and therefore, did not block the internalization of RAMP3 (and CL-R) with agonist pretreatment (Figure 23B). These findings indicate that NHERF-1 inhibits the internalization of the CL-R/RAMP3 receptor complex by acting as an adaptor to tether the receptor complex to the actin cytoskeleton. To further test this mechanism for inhibiting internalization, an actin depolymerization agent (Cytochalasin D) was employed to disrupt the actin cytoskeleton and the ability of NHERF-1 to tether the CL-R/RAMP3 complex to the plasma membrane. Treatment of HEK293 cells expressing CL-R/RAMP3 in the presence and absence of NHERF-l had no effect on the adenylate cyclase activity or desensitization pattern of the receptor complex (Figure 23C). After treatment with Cytochalasin D, HEK293 cells expressing CL-R/RAMP3 and NHERF-1 were now capable of internalization. The Cytochalasin D experimental data further supports that the mechanism of inhibition of internalization of the CL-R/RAMP3 complex by NHERF-1 by tethering of the receptor complex to the plasma membrane by NHERF-1’s interaction with the actin cytoskeleton. 5.3.5. NHERF’s role in receptor trafficking in primary human proximal tubule epithelial cells To examine if our observations using the over-expressed system in HEK293 cells could be translated to a more physiological cell type, we Chose human proximal tubule cells to test our hypothesis. Because of the many roles of adrenomedullin and NHERF-1 in the kidney, we chose a human primary proximal tubule cell line to perform these studies. The human proximal tubule epithelial cells (hPTEs) were determined to express 128 CL-R, RAMP2, RAMP3, and NHERF-1 and show high levels of specific adrenomedullin binding and receptor stimulation by AM (data not shown). When pretreated with AM for a period of one hour, desensitization was observed with adenylate cyclase assays (Figure 24A). However, as we would predict, internalization was not observed (by whole-cell receptor binding assays) (Figure 24B). Intemalization was not observed when hPTE cells were pretreated with AM for up to four hours, indicating desensitization and internalization were not simply showing different kinetics. In order to test our hypothesis that RAMP3 and NHERF-1 were critical to the inhibition of internalization of the CL- R/RAMP3 receptor complex, RNA interference technology was employed to individually knockdown RAMP3 and NHERF-1. In both mRNA and protein expression studies, RAMP3 and NHERF-1 dramatically decreased in d-siRNA treated samples, while control experiments using lacZ knockdown showed no significant alteration in RAMP3 or NHERF-1 expression when compared to wild-type hPTE cells (Figure 25A, B; data not shown). Intemalization assays and whole-cell receptor binding assays were performed to determine the effect of RAMP3 and NHERF-1 RNA interference on receptor internalization in hPTE cells. Strikingly, when pretreated with agonist, hPTE cells with RAMP3 or NHERF-l RNA interference showed a regained ability to internalize the receptor complex, unlike the wild-type hPTE cells were internalization was inhibited (Figure 25C). This finding demonstrates that RAMP3 and NHERF-1 are both critical for the receptor trafficking of the CL-R/RAMP3 complex in hPTE cells, an unaltered cell line absent the issues of overexpression. To determine if the mechanism of inhibition of internalization in hPTE cells was similar to that in the HEK 293 cells, actin cytoskeletal tethering was examined in the 129 r“ A‘L l1 hPTE cells. hPTE cells were treated with Cytochalasin D, as was used in HEK 293 cells to disrupt the actin cytoskeleton, and internalization assays were performed. Cytochalasin D treatment did not alter the receptor expression levels in the hPTE cells, as measured by whole-cell receptor binding, from the levels in untreated cells. Interestingly, when cells were treated with Cytochalasin D and pretreated with AM for one hour, the receptor complex was now capable of internalization (Figure 26). This data supports the proposed model that NHERF-1 inhibits the internalization of the CL- R/RAMP3 receptor complex after agonist stimulation by tethering the receptor complex to the actin cytoskeleton via NHERF-1’s ERM domain interactions with the cytoskeleton and PDZ domain interactions with RAMP3. Finally, it was important to determine the effect of the inhibition of internalization on AM signaling in the hPTE cells. To this end, a resensitization assay was performed and CAMP accumulation was measured. AM pretreatment of the hPTE cells results in an attenuation of CAMP accumulation from 35% to 15% of maximal response, whereas allowed recovery time in the absence of agonist after the AM pretreatment was shown to recover AM signaling to similar levels as untreated cells. While it was shown in a previous Figure that AM pretreatment causes desensitization of the AM receptor signaling in the hPTE cells, the resensitization assay now showed that resensitization occurs in the hPTE cells, even in the absence of receptor internalization (Figure 24A, 27). This finding suggests the inhibition of internalization of the AM receptor in the hPTE cells is a mechanism for receptor resensitization, not requiring receptor internalization as a prerequisite. 130 'q F‘m ‘. “u I 50. - No Pretreatment CIAM Pretreatment 6F! E: 33 gr, * it —"'—~ * VL- a; Adenylate cyclase activi CL-R-I-RAMP3 CL-R+RAMP3+NHERF1 Figure 17A: The role of NHERF-l in the trafficking of the AM2R (CL-R+RAMP3) in HEK 293 cells. NHERF-1 does not alter desensitization of AM2R in transfected HEK 293 cells. HEK 293 cells transiently transfected with CL-R and RAMP3 with or without NHERF-l. At 48h post- transfection, cells were treated for one hour with AM (10 nM) and then washed and adenylate cyclase activity was measured. Alter agonist pretreatments, membranes were extracted and AC activity in response to 100 nM AM was measured. NHERF-l overexpression with AM2R had no significant effect on receptor desensitization. Experiments performed in triplicates and data expressed as percent maximal stimulation (% Forskolin). * p S 0.05; n 2 4. 131 - No Pretreatment D AM Pretreatment E5 ...1°°' ? 'o' .9 h 75- .6 8 go 50- * ‘6 $3 ” .5 " 25- m CL-R-I-RAMP3 CL-R-I-RAMP3-I-NHERF1 Figure 173: The role of NHERF-l in the trafficking of the AM2R (CL-R+RAMP3) in HEK 293 cells. NHERF-l inhibits internalization of AM2R in HEK 293 cells. HEK 293 cells transfected and pretreated with agonist as described in 17A. Receptor internalization measured by whole-cell competition binding assays using ”SI-rAM as ligand (cold rAM served as the competitor) and number of binding sites/cell was estimated using the GRAPHPAD PRISM software. “No pretrea ” represents samples at maximal radioligand binding that were not pre-incubated with agonist. “1h pretreat” represents samples pretreated with AM (10 nM) for one hour, washed as indicated in Methods section, and tested immediately after wash steps for radioligand binding. NHERF-l overexpression in cells expressing AM2R caused altered receptor trafficking to inhibit internalization of the receptor complex. * p S 0.05; n z 4 experiments. 132 .CL-R+RAMP3+NHERF1 150. IIICL-R+RAMP3 3 2A “3100- r— 05 :2 38 c * * --\° 22,50- * a A l 0 1 2 4 Pretreatment time (hrs) Figure 17C: The role of NHERF-l in the trafiicking of the AM2R (CL-R+RAMP3) in HEK 293 cells. Time course internalization of AM2-R in presence/absence of NHERF-l. Lengthening of agonist pretreatment does not alter inhibition of internalization of CL-R/RAMP3 complex with NHERF-l co-expression. HEK 293 cells transfected and pretreated with agonist for varying time points, as described in 17A. Receptor internalization measured by whole-cell competition binding assays using ”SI-rAM as ligand (cold rAM served as the competitor) and number of binding sites/cell was estimated using the GRAPHPAD PRISM software. * p S 0.05; n 2 3 experiments. 133 RAMP3 OVERLAY RAMP3 NHERF1 No Pretreat AM Pretreat No Pretreat AM Pretreat Figure 18: Localization of CL-R, RAMP3, and NHERF-1 in HEK 293 cells during an internalization experiment. After lh AM pretreatment. CL-R and RAMP3 are internalized in absence of NHERF-l co-expression. In cells co-expressing NHERF-l with CL-R/RAMP3 complex. internalization of receptor complex is blocked alter AM pretreatment. HEK 293 cells transfected with CL-R-GFP, RAMP3, and NHERF-1 were pretreated with 10nM ADM for 1h. Note: “1 hr pretreatment” indicates time just after AM pretreatment and wash steps. Cells were fixed and components were visualized using anti-RAMP3 antibody (1:200) and anti-NHERF-l antibody (1:250) with Cy5 anti-rabbit secondary antibody (1:400, in blue) and Cy3 anti-mouse secondary antibody (1:500, in red), respectively; CL-R-GFP is detected with an EGFP tag and shown in green; overlays of staining patterns are shown in the far right panels. Images shown are representative of at least twenty fields imaged per experiment from at least three experiments. Bar scales on all images represent lOOum. 134 - CL-R+RAMP1 D CL-R+RAMP1+NHERF1 3% 75‘ .I. o . < .9. _ 30 0 2 .5 8 3,3 — x . a: 2 2° 0 .5 E E o 3% 2.10: 2‘s 35 o- < No Pretreat CGRP Pretreat Figure 19A: The role of NHERF-l in post-endocytic sorting of CGRP] (CL-R+Rl) and AMI receptor. NHERF-l overexpression does not alter CGRP receptor desensitization after agonist stimulation. HEK 293 cells were transfected and pretreated as in figure 17A and then membranes were extracted. Adenylate cyclase activity was measured in membranes stimulated with 100 nM CGRP for 15 min. Experiments performed in triplicates and expressed as percent maximal stimulation (% Forskolin). * p S 0.05; n 2 3. 135 - CL-R+RAMP1 D CL-R+RAMP1+NHERF1 _ 100« fi 3 £ 2 g e "7’ E 50« * 5’0 .E o 'a o\ .E V m 0' No pretr'eatment CGRP Pretreatment Figure 198: The role of NHERF-l in post-endocytic sorting of CGRPl (CL-R+Rl) and AM] receptor. NHERF-l overexpression does not alter the trafficking of the CGRP receptor after agonist stimulation. HEK 293 cells were transfected with CL-R and RAMP], with or without NHERF-l. 48h post-transfection, cells were pretreated with CGRP (I 0 nM) for one hour, washed as described in Materials and Methods, and receptor internalization was measured with whole- Cell competition binding using l25l-rCGRP as the ligand and cold rCGRP as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. "‘ p _<_ 0.05; n 2 3. 136 - CL-R+RAMP2 D CL-R+RAMP2+NHERF1 I. «h C I [% Forskolin] N ‘3 Adenylate Cyclase Activity (pmollminlmg protein) No Pr'etreat AM Pr'etreat Figure 19C: The role of NHERF-l in post-endocytic sorting of CGRPI (CL-R+Rl) and AMI receptor. NHERF-I overexpression does not alter AMI receptor desensitization after agonist induction. HEK 293 cells were transfected and pretreated as in figure 17A and then membranes were extracted. Adenylate cyclase activity was measured in membranes stimulated with 100 nM AM for 15 min. Experiments performed in triplicates and expressed as percent maximal stimulation (% Forskolin). * p S 0.05; n 2 3. 137 - CL-R+RAMP2 E CL-R+RAMP2+NHERF 1 8 873 i :15 in . 5,8 50 CE ° '59\ 5" m No Pretfeatment AM Preti'eatment Figure 19D: The role of NHERF-l in post-endocytic sorting of CGRP] (CL-R+Rl) and AMI receptor. NHERF-l overexpression does not alter internalization of AMI receptor after receptor activation. HEK 293 cells were transfected with CL-R and RAMP2, with or without NHERF-1. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour, washed as described in Materials and Methods, and receptor internalization was measured with whole-cell competition binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. "' p S 0.05; n 2 3. 138 - CL-R+RAM P3 E CL-R+RAM P3+NHERF1 E3 CL-R+RAMP3A1 45-8+NHERF1 % 1oo- — l 2 2 F W O 3 1'.“- 75' * Ia c 0 2’0 50.. * ‘Q 2‘: a 25‘ No Pretfeatment AM Preti'eatment Figure 20A: The effect of RAMP3 PDZ motif deletion on the trafficking of the AM2R (CL- R+RAMP3). Deletion of RAMP3 PDZ motif allowed internalization of the CL-R/RAMP3 complex when co-expressed with NHERF-l. HEK 293 cells were co-transfected with CL-R, wild-type RAMP3 or RAMP3AI45-8, and NHERF-1. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour. washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. "‘ p S 0.05: n 2 3. 139 - CL-R+RAMP2 E CL-R+RAMP2+NHERF1 CL-R+RAMP2ADTLL+NHERF1 = 100- 8 a ‘0‘ 8°“ 8 b '6 g 60- 2’0 :5 °\° 40' .E V m 20‘ 01 No pretr'eatment AM pretreatment Figure 208: Effect of PDZ motif substitution on the C-terminus of RAMP2 on internalization of CL—R/RAMP complex. Substitution of PDZ motif (-DTLL) on the C—terminus of RAMP2 caused a Change in receptor trafficking to inhibit internalization of the receptor complex when co- expressed with NHERF-l. HEK 293 cells were co-transfected with CL-R, wild-type RAMP2 or RAMP2ADTLL, and NHERF-1. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour, washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 3. 140 - No pretreatment DAM pretreatment — 1oo- ¥ 5. l E? 8°‘ * '5 = 60- g8 E 20' l 0" I I I I '5 :6 v v '5 :b v~ v~ $9§°Q€§£s§ QvQI} °\°9«\§) e e «9’ e e a Figure 20C: Effect of RAMP3 PDZ motif point mutations on the internalization of the CL- R/RAMP3 complex in the presence of NHERF-l. Only point mutant RAMP3TI46A was critical in the inhibition of the internalization of the CL-R/RAMP3 complex seen when co-expressed with NHERF-l. HEK 293 cells were co-transfected with CL-R, wild-type RAMP3 or RAMP3 point mutants, and NHERF-1. 48h post-transfection. cells were pretreated with AM (10 nM) for one hour. washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using l25l-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 3.. 141 - NHERF ENHERFAPDZ‘I ENHERFAPDZZ 125' g =. 100- l m 0 w ‘,_l :3 b 75- m c ,‘ ..-. c» o * ‘ c U 50. "5 E: ’ :j .E 25- -' 2‘ m * ; No Pr'etreat 1 hr Pietreat Figure 20D: The effect of NHERF-l PDZ domain deletion on the trafficking of the AM2R (CL- R+RAMP3). Deletion of NHERF-l PDZl domain allowed internalization of the CL-R/RAMP3 complex. HEK 293 cells were co-transfeCted with CL-R, wild-type RAMP3, and wild-type or PDZ domain mutants of NHERF-l. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour, wasth as described in Materials and Methods, and receptor intemalization was measured with whole-cell binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 3. 142 RAMP3Af45-8 NHERF1 OVERLAY Figure 21: Localization of CL-R, RAMP3A14S-8. and NHERF-1 in HEK 293 cells during an No Pretreat AM Pretreat internalization experiment. Untreated and after one hour ADM (10nM) pretreatment, CL-R and RAMP3AI45-8 internalize similarly to the wild-type CL-R/RAMP3 complex in the absence of NHERF-l. Experiments performed as described in Figure 18. Fixed cells were stained with anti- RAMP3 antibody (1:200) and anti-NHERF-l antibody (1:250) with CyS anti-rabbit secondary antibody (1:400, in blue) and Cy3 anti-mouse secondary antibody (1:500, in red), respectively; CL—R-GFP is shown in green: overlays of staining patterns are shown in the far right panels. Images shown are representative of at least twenty fields imaged from at least three experiments. Bar scales on all images represent lOOum. 143 ' (M- 8' (.us'r- C' (.‘ST- (N. mun (.s'r- RAMP3 (.‘s'r- RAMP3 csr RAMP3 mm on RAMP3 ANS-8 (zs’r RAMP3 nus-s 70 KDa 50 KDa Figure 22: Interaction studies of RAMP3 with NHERF-l. Fusion proteins of GST-RAMP3 demonstrate a physical interaction with NHERF-l in an overlay assay and western blot analysis. lOug of GST, GST-RAMP3. and GST-RAMP3AI45-8 protein was run and separated on an SDS- PAGE gel and transferred to a nitrocellulose filter. The nitrocellulose filter was incubated overnight in lysate of HEK293 cells transfected with or without NHERF-I at 4°C. Filter was then washed and probed by immunoblot for NHERF-l (1:250). Identical filters were probed by immunoblot for RAMP3 (12400). A. GST. GST-RAMP3, and GST-RAMP3AI 45-8 overlay assay incubated in NHERF-l-transfected HEK 293 lysates and probed for NHERF-l. B, GST, GST- RAMP3, and GST-RAMP3AI45-8 overlay assay incubated in non-transfected HEK 293 lysates and probed for NHERF-l. C. lmmunoblot of GST, GST-RAMP3. and GST-RAMP3A145-8 probed for RAMP3. Shown are representative blots of at least four experiments. 144 - CL-R+RAMP3+NHERF1 D CL-R+RAMP3+NHERF1AERM _ 100- a T a ‘3 3°“ 3 .5 la 5 60" * 5’0 % °\° 40' .E V m 20- No pretr'eatment AM prefieatment Figure 23A: Mechanism of NHERF-l inhibition of internalization of CL-R/RAMP3 receptor complex. ERM domain mutant of NHERF-I blocks the inhibition of internalization of CL- R/RAMP3 complex seen with wild-type NHERF-l. HEK 293 cells were transfected with CL-R, RAMP3, and wild-type NHERF-l or ERM domain mutant NHERF-l. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour, washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. "‘ p S 0.05; n 2 3. 145 OVERLAY PHALLOIDIN No Pretreat AM Pretreat RAMP-3 NHERFAERM PHALLOIDIN OVERLAY No Pretreat AM Pretreat I46 Figure 238: Mechanism of NHERF-l inhibition of internalization of CL-R/RAMP3 receptor complex. Localization of RAMP3, NHERF-l (or NHERF-IAERM), and actin cytoskeleton in HEK 293 cells during an internalization experiment. Untreated cells show RAMP3 and wild-type NHERF-l distributed at the plasma membrane with NHERF-l colocalizing with the actin cytoskeleton, and after AM pretreatment, all component remain in the same distribution. ERM domain mutant of NHERF-l shows poor co-Iocalization with the actin cytoskeleton in both untreated and AM-pretreated conditions, and therefore, shows internalization of RAMP3 with agonist pretreatment, HEK 293 cells were transfected with CL-R, RAMP3, and wild-type or ERM domain mutant NHERF-l. Experiments performed as described in Figure 18. Fixed cells were stained with anti-RAMP3 antibody (1:200) and anti-NHERF-l antibody (1:250) with Cy5 anti-rabbit secondary antibody (1:400, in blue) and Cy3 anti-mouse secondary antibody (1:500, in red), respectively; actin cytoskeleton is stained with Alexa 488-phalloidin and is shown in green; overlays of staining patterns are shown in the far right panels. Images shown are representative of at least twenty fields imaged from at least three experiments. Bar scales on all images represent lOOum. 147 - Basal E Cyto D treated 100' 0| C, l Binding max (sites/cell) [% Control] No Pretreat AM Pretreat Figure 23C: Mechanism of NHERF-l inhibition of internalization of CL-R/RAMP3 receptor complex. Cytochalasin D treatment blocks the inhibition of internalization of CL-R/RAMP3 complex. HEK 293 cells were transfected with CL-R, RAMP3, and wild-type NHERF-1. 48h post-transfection, cells were pretreated Cytochalasin D (IOuM) for 15 min., then with AM (10 nM) for one hour, washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 3. 148 A O I [% Forskolin] N C.’ CAMP Accumulation (nmollmg protein) I No Pretreat 1 hr AM Pretreat Figure 24A: Trafficking of AM-R in human proximal tubule cells (hPTE cells). In hPTE cells, AM-R signaling desensitizes with AM pretreatment. hPTE cells seeded in 24-well plates were pretreated with IOnm rAM for lh were washed extensively to remove residual agonist and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in hPTE cells were calculated using a standard curve ranging from 10 to 104 fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. “ p S 0.05; n 2 4 experiments. 149 E 125- a: 2 3%,100- £3 75. x c a: O a: 50- “L. c :5 25] .E m 0 e “I “‘ gt“ AM Pretreat Figure 248: Trafficking of AM-R in human proximal tubule cells (hPTE cells). AM-R fails to internalize when incubated in AM for varying time points. hPTE cells were grown as described in Figure 24A. Receptor internalization measured by whole-cell competition binding assays using ”SI-rAM as ligand (cold rAM served as the competitor) and number of binding sites/cell was estimated using the GRAPHPAD PRISM software. “No pretreat” represents samples at maximal radioligand binding that were not pre-incubated with agonist. “1,2, and 4h pretreat” represents samples pretreated with AM (10 nM) for according time period, washed as indicated in Methods section, and tested immediately after wash steps for radioligand binding. * p S 0.05; n 2 3 experiments. In cells endogenously expressing AMR, internalization is inhibited, but desensitization in unaltered. 150 RAMP3 NHERF1 WT RNAi WT RNAi PCR: RAMP3 NHERF1 Figure 25A: Effect of RAMP3 and NHERF-1 RNA interference on internalization of AM2-R in human proximal tubule cells. RAMP3 and NHERF-1 RNA interference disrupts the inhibition of internalization of AM-R after agonist pretreatment. hPTE cells seeded in 48-well plates were transfected with d-siRNA of RAMP3 or NHERF-l and incubated for 48 hr to allow RNA knockdown. mRNA analysis by Q-PCR showed dramatically decreased levels of RAMP3 and NHERF-1 mRNA in RAMP3 and NHERF-1 RNA interference samples (respectively) as compared to wild type, while lacZ knockdown had no effect on RAMP3 or NHERF-l message levels (data not shown). RNA isolation and RT-PCR performed as described in Experimental Procedures section. n=3 experiments. 151 Wildelype hPTE cells NHERF Knockdown RAMP-3 Knockdown Figure 25B: Effect of RAMP3 and NHERF-1 RNA interference on internalization of AM2-R in human proximal tubule cells. RAMP3 and NHERF-1 RNA interference disrupts the inhibition of internalization of AM-R after agonist pretreatment. hPTE cells seeded in 48-well plates were transfected with d-siRNA of RAMP3 or NHERF-l and incubated for 48 hr to allow RNA knockdown. lmmunofiuorescence microscopy of hPTEs with RAMP3 or NHERF-l RNA interference demonstrated greatly decreased levels of RAMP3 and NHERF-1 protein expression in RAMP3 and NHERF-1 RNA knockdown cells, as compared to wild-type hPTE cells. lacZ knockdown had no effect on RAMP3 or NHERF-l protein expression in hPTEs (data not shown). Cells prepared as described in Experiment procedures section for immunofluorescence microscopy. Fixed cells were stained with anti-RAMP3 antibody ( 1:200) or anti-NHERF-I antibody (1:250) and detected with a Cy3 anti-rabbit secondary antibody (1:500). Images shown are representative of at least three experiments. 152 - Wild-type El RAMP-3 RNAi DNHERF RNAi ‘=‘ 125- a: 2 g H 100' a _ -- 0 19,35 75. x c * «I 0 * E 2 50- m\ .E L 25.. 1: .E j oi oi No Pr'etreat AM Pfetreat Figure 25C: Effect of RAMP3 and NHERF-1 RNA interference on internalization of AM2-R in human proximal tubule cells. RAMP3 and NHERF-1 RNA interference disrupts the inhibition of internalization of AM-R alter agonist pretreatment. hPTE cells seeded in 48-well plates were transfected with d-siRNA of RAMP3 or NHERF-l and incubated for 48 hr to allow RNA knockdown. AM-R is capable of internalization when RAMP3 or NHERF-l are knocked down with RNA interference. hPTE cells were grown as described in Figure 24A. Receptor internalization measured by whole-cell competition binding assays using ”SI-rAM as ligand (cold rAM served as the competitor) and number of binding sites/cell was estimated using the GRAPHPAD PRISM software. "No pretreat" represents samples at maximal radioligand binding that were not pre-incubated with agonist. "l pretreat” represents samples pretreated with AM (10 nM) for according time period, washed as indicated in Methods section, and tested immediately after wash steps for radioligand binding. * p S 0.05; n 2 3 experiments. 153 -Wild-type hPTE DCyto D treated hPTE x ”100' r— :23 C, 5 cl 5322' 75 .s '0 ° 2%‘3 50- "Vi. m 25- No Pretr'eatment AM Pretfeatment Figure 26: Mechanism of NHERF-I inhibition of internalization of CL-R/RAMP3 receptor complex in hPTE cells. Cytochalasin D treatment blocks the inhibition of internalization of CL- R/RAMP3 complex. hPTE cells were grown as described in Figure 24A. hPTE cells were pretreated Cytochalasin D (lOuM) for 15 min., then with AM (10 nM) for one hour, washed as described in Materials and Methods, and receptor internalization was measured with whole-cell binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. "‘ p S 0.05; n 2 3. 154 $ 30 15- CAMP Accumulation (nmollmg protein) [% Forskolin] OJ ‘6. 3““9 “c We“?! “Wm“ Figure 27: Trafficking of AM-R in human proximal tubule cells (hPTE cells). In hPTE cells, AM-R signaling desensitizes with AM pretreatment, but also resensitizes when then incubated in the absence of agonist. hPTE cells seeded in 24-well plates were pretreated with lOnm rAM for 1h were washed extensively to remove residual agonist, incubated for 4h in serum-free media in the absence of agonist and presence of Sug/ml cycloheximide, and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in hPTE cells were calculated using a standard curve ranging from 10 to 10 4 fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p S 0.05; n 2 4 experiments. 155 5.4. Discussion Like other GPCRS, the adrenomedullin and CGRP receptors mediate various physiological actions in different cell types using a variety of mechanisms [305, 306]. The receptor undergoes classical lifecycle wherein it is phosphorylated upon agonist stimulation, leading to desensitization, internalization and either recycling or degradation depending on the cell type. However, unlike other GPCRS, the AM and CGRP receptors are regulated by single transmembrane accessory protein called RAMPS (RAMP1-3). In addition to the original observation by Foord’s group that RAMPS are necessary for the cell surface expression and specificity of CL-R to the ligands CGRP and AM, our observations reported recently demonstrated that RAMPS (particularly RAMP3) also has other roles that may regulate the lifecycle of the receptor complex. Specifically, RAMP3 interaction with NSF was found to be important for recycling the receptor after internalization. Absence of RAMP3 or inhibition of NSF resulted in receptor complex degradation [153]. The present study was undertaken to investigate other binding partners and additional roles for RAMP3 in CL-R/RAMP lifecycle. We show here using both heterologous expression, as well as endogenous systems, that RAMP3 interaction with NHERF-l is essential for regulating the internalization of the CL—R/RAMP3 complex. In addition, similar to our NSF study, the PDZ motif in RAMP3 is critical for the interaction with NHERF-l. In particular, we have found that the Thr146 in RAMP3 is critical for this interaction. Whether RAMP3 is phosphorylated at this site by any of the kinases, and if this is essential for NHERF-1 interaction, is not known. Several studies have shown NHERF-l to play differing roles in various cellular processes, including receptor trafficking [257, 258]. NHERF-1 has been demonstrated to 156 bind the extreme COOH terminus of several G-protein coupled receptors (GPCRS), namely the Bz-adrenergic receptor, the K-OpIOid receptor, and the P2Y purinergic receptor [154, 259]. Agonist exposure promotes NHERF-l association with the fig-adrenergic and the K-opioid receptors. Unlike our study, NHERF-1 association with these receptors enhances the recycling of the receptors after agonist stimulation [240, 259]. The DSLL motif in the C-terminus of adrenergic receptor was found to the critical for NHERF-l interaction. In the present study, we also found that the DTLL motif in RAMP3 is essential for the interaction with NHERF-l. Similar to the Thr in the DTLL motif, the Ser in the DSLL motif was found to be essential for the interaction with NHERF-1 [240]. In addition to GPCRS, NHERF-1 has also been shown to associate with the epidermal growth factor receptor (EGFR), a receptor tyrosine kinases (RTK). In this case, NHERF- 1 association acts to stabilize the receptor at the plasma membrane and decrease the endocytosis rate of the receptor [217]. Using an endogenous system we have also shown here that NHERF-l is essential to ‘hold’ the receptor-complex at the membrane, the absence of which leads to internalization of the receptor complex. While utilizing different mechanisms, collectively these data suggest that NHERF-l interaction with both RTKS and G protein-coupled receptors and RAMP3 is mandatory to enhance the portion of receptors present at the cell surface. Recent data suggests that RAMP3 can interact with receptors other than CL-R. It is important to determine if this novel role of RAMP3 in receptor trafficking is specific for CL-R or also for the other receptors RAMPS interact with, namely VPAC, PTHl- and 2-R, and glucagon receptors [286]. NHERF—l and -2 have been reported to interact with the PTHlR in a scaffolding capacity and tether the receptor to phospholipase C-B (PLCB) 157 and the actin cytoskeleton [301, 302]. In opossum kidney (OK) cells, NHERF-l mediates PTH-stimulated entry of extracellular calcium by a mechanism that is apically localized, PLC-dependent, pertussis toxin-sensitive, and requires an intact actin cytoskeleton. NHERF1 was also shown in inhibit the activation-independent internalization of the PTHIR in kidney distal tubule cells (when stably expressed) [218]. Given the ability of RAMP2 and RAMP3 to interact with the PTHlR and PTH2R, respectively, RAMP3 may be interacting with NHERF-l to play a trafficking role in these systems, as well. In addition to AM, Roh et al. have reported that interrnedin, a newly discovered peptide from the calcitonin gene peptide superfamily, can also bind the CL-R/RAMP3 complex [307]. It remains to be determined if this reported function for RAMP3 'is specific for AM, or if another peptide like intermedin could yield similar results. In our studies of interaction of RAMP3 with NSF and NHERF-1, the PDZ motif of RAMP3 was found to be critical for the interaction. Whether this motif binds additional proteins in a cell type specific manner remains to be examined. One could hypothesize that these predicted cell-type specific interactions would lead to regulation of the various events in the receptor life cycle. In addition to RAMP3, RAMPl and RAMP2 also regulate the expression of CL-R at the plasma membrane. Whether RAMP] and 2 bind other proteins similar to RAMP3 also remains to be examined. Conclusions: This study has shown that one of CL-R’s heterodimeric partners, RAMP3, is capable of altering the trafficking of the receptor complex after agonist stimulation by interacting with NHERF-l via its PDZ motif, and thus the actin cytoskeleton. We have 158 demonstrated here for the first time that NHERF-RAMP3 interaction dissociates receptor desensitization from internalization of the CL-R/RAMP complex. Additionally, this reveals a novel function for the RAMP accessory proteins in receptor trafficking and an additional difference between the AMlR and AM2R. With recent reports of RAMPS complexing and regulating GPCRS other than CL-R, future studies will focus on additional binding partners of RAMPS and how they regulate the various events in the GPCR life-cycle. 159 6. Novel function for receptor activity-modifying proteins in post- endocytic receptor trafficking. 6.1. Introduction The recent discovery of receptor activity modifying proteins (RAMPS) has raised new possibilities for modes of regulation of G-protein coupled receptors (GPCRS). RAMPS were discovered as accessory proteins indispensable to the function of an orphan GPCR, now termed the calcitonin receptor-like receptor (CL-R)[275]. Three RAMP isoforms (1-3) have been identified as distinct gene products that yield single transmembrane-spanning proteins. RAMPS are required for the plasma membrane expression, as well as for determination of receptor phenotype for CL-R (selective ligand recognition)[275, 276]. Coexpression of RAMP-1 with CL-R yields a calcitonin gene- related peptide-l (CGRP-l) receptor, while coexpression of RAMP—2 or -3 with CL-R produces adrenomedullin receptors, AM-l and AM-2 receptors, respectively [277, 278]. AM and CGRP are multi-functional peptides with many overlapping functions, ranging from potent vasodilation to proliferation regulation to regulation of salt and water balance [279]. Differential expression of RAMP isoforms has been hypothesized to play a regulatory role in both physiological and pathophysiological disease states. Moreover, the recent identification of RAMP interactions with additional members of the Class II GPCR family and RAMP expression in cell lines lacking CL-R have raised the possibility of novel functions for RAMPS in GPCR regulation [286]. Upon activation, the CL-R/RAMP receptor complex causes cyclic AMP activation in most systems, irrespective of whether the ligand is AM or CGRP. In addition, the receptor complex undergoes desensitization and internalization (via Clathrin- 160 mediated endocytosis) in response to a prolonged agonist stimulation [280]. Once internalized, the receptor complex either undergoes degradation or recycling, depending on the cell type. In HEK 293 cells the CL-R/RAMP complex has been shown to be targeted to the lysosomes for degradation, while in rat mesangial cells, the CL—R/RAMP receptor complex is sorted for dephosphorylation and resensitization (and presumably recycling) as a fully functional receptor [276, 285]. The mechanism that regulates the pathway to which the receptor complex is targeted after agonist-induced internalization remains unknown. Factors influencing the sorting of receptors in the early endosomes are largely unknown, but some of the critical players are beginning to be identified for the GPCRs. It has been shown in other GPCR systems that interactions with PSD-95/Discs-large/ZO- l homology (PDZ) domain proteins are responsible for altering the receptor-targeting after internalization [259, 267, 268]. The life cycle of the BZ—adrenergic receptor ([52- AR) was reported to be altered in the presence of a protein termed N-ethylmaleimide sensitive factor (NSF) [268]. It has been shown that the BZAR interacts with NSF via a PDZ type I domain (-DSLL) at its extreme C-terminus. In addition, binding of NSF to the Glu2 subunits of the AMPA receptor was also demonstrated to be crucial for the recycling of the a-amino-3-hydroxy-5-methyl-isoxazolepropionate (AMPA) receptor [269, 270]. NSF is a hexameric ATPase that plays a Chaperoning role for SNARES in the majority of membrane fusion events in a cell, but when targeting membrane receptors for recycling, NSF acts independently of the SNARE complex to promote rapid resensitization of the receptors at the plasma membrane [261-263]. 161 Similar to the C-terminus of B2-AR, human RAMP3 C-terminus has a type-I PDZ recognition sequence (-DTLL motif). CL-R, RAMP] or RAMP2 do not, however, contain any PDZ motifs. We hypothesized that RAMP3, via its interaction with NSF, regulates the trafficking of the CL-R/RAMP3 complex. We show here that while CL- R/RAMP] and CL-R/RAMP2 complexes do not interact with NSF, CL-R/RAMP3 complex interacts with NSF via the PDZ motif of RAMP3 [153]. Moreover, we demonstrate that over-expression of NSF in HEK-293 cells alters the life cycle of CL- R/RAMP3 complex from a degradative to recycling pathway via interactions of the PDZ motif of RAMP3 and NSF. These findings demonstrate that RAMP3, in addition to determining the receptor phenotype and allowing receptor membrane expression, is also significantly involved with the regulation of the turnover of the CL-R/RAMP complex. 6.2. Materials and Methods 6.2.]. Materials: Adrenomedullin was purchased from Bachem Bioscience, Inc. (King of Prussia, PA). 1251-labeled adrenomedullin was purchased from Amersham Biosciences Corp. (Piscataway, NY). N-ethylmaleimide was purchased from Sigma (St. Louis, MO). Cell culture media, fetal bovine serum, penicillin/streptomycin, trypsin- EDTA were purchased from GibcoBRL® (Grand Island, NY). RAMP3 antibody was purchased from Santa Cruz Biotechnology (Santa Cruz, CA) and NSF antibody was from Calbiochem® (La Jolla, CA). Anti-mouse Cy3 and anti-rabbit Cy5 secondary antibodies were from Jackson Immunoresearch Laboratories (West Grove, PA). All other reagents were of highest quality available. 162 6.2.2. Cell Culture and Transfection protocols: Rat mesangial cells were obtained from ATCC and are maintained in RMPI 1640 media containing 15% FBS and 1% penicillin-streptomycin. HEK-293T cells (obtained from ATCC) are maintained in DMEM low glucose media containing 10% FBS, 1% penicillin-streptomycin. Rat-2 fibroblast cells (obtained from ATCC) are maintained in DMEM high glucose media containing 10% FBS, 1% penicillin—streptomycin. Transfection of HEK293T and Rat-2 fibroblast cells was performed using Lipofectamine Plus protocol (Invitrogen Life Technologies, Carlsbad, CA). Cells were transfected with the DNA and Lipofectamine Plus as per manufacturer’s protocol. Cells were collected for assays after 48 hours of transfection. 6.2.3. RAMP cloning and expression: Full length CDNA of human RAMPS], 2 and 3 and bovine CL-R were described before [281, 282]. CL—R, Cloned into Nl-EGFP and also in pCDNA3.l expression vectors, was used for transfection in HEK 293T cells. 6.2.4. Desensitization and Resensitization assays: 48 hours post—tranfection cells were pretreated with or without 10 nM AM in DMEM containing 0.2% BSA for indicated time periods (up to 4 hr). After agonist exposure, cells were washed three times with Dulbecco’s phosphate buffered saline (dPBS, Gibco BRL) containing 0.2% BSA and either frozen for membrane preparation for adenylate cyclase assays or used immediately for intact-cell radioligand binding. For receptor resensitization assays, after agonist exposure, cells were washed and incubated for indicated time periods in DMEM containing 0.2% BSA and 5 jig/ml cycloheximide to allow receptor recovery. 163 Resensitization Protocol: Removal of AM. followed by Celia AM PNUOOUMM (’ washing, followed by incubation without AM transfected atarte j 1 - I I i 1 X Y W Receptor activity assay (adenylate cyclase) and ‘8 0"" 1 hr 2" h" ti receptor binding at (recovery me) different time polnta No Pretreat 1 h AM (Full Stimulation) ' Pretreat 2'4 h' (Desensitization) R°°°V°W (Resensitization) 6.2.5. Receptor binding: Homologous competition radioligand binding assays were performed as described by Aiyar et a1 and as established in our laboratory [283]. HEK 293T cells were transfected and approximately 200,000 cells/well seeded in poly-D- lysine pre-coated 24-well plates (BD Biosciences, Palo Alto, CA). 48 hours post— transfection cells were treated for desensitization or resensitization assays as described above. After agonist exposure, cells were washed three times with dPBS buffer containing 0.2% BSA then incubated with increasing concentrations (1 pM to 100 nM) of competing ligand (rAM) and 175-250 pM 125I-rAM for 30 min at 37°C. After incubation, plates were washed three times with ice-cold assay buffer and the reactions were terminated by the addition of 2M NaOH. Cells were then harvested and associated radioligand activity is counted on a y-counter. All binding assays were performed in duplicate, with each experiment repeated at least three times. Nonspecific binding was determined in the presence of 100 nM of unlabeled rAM. Data was analyzed by 164 LIGAND (assuming radioligand and competitor both bind reversibly to a single binding site; MacLigand, Version 4.97, NIH, Bethesda, MD) using the following equation: Bm,x - [hot ligand] KD + [hot ligand] + [cold ligand] Where B represents specific binding, [hot ligand] the single concentration of [1251]rAM studied, [cold ligand] the concentration of unlabeled rAM competing with the radiolabel for AM receptor binding, Bmax the maximum number of binding sites and KD the equilibrium dissociation constant (the equation was solved where the “cold ligand” ICso = [hot ligand] + KD). Analysis of all binding data was performed by computer-assisted nonlinear least square fitting using GraphPad PRIZM version 4 (GraphPad Software, San Diego, CA). Binding sites/cell was calculated and data was expressed as percent of the control. 6.2.6. Adenylate cyclase assays: Cyclase activity was done as described before with slight modifications [282, 284]. Cells were harvested fi'om P100 or P60 plates and homogenized in Tris HCl (10mM)/EDTA (10mM) buffer. Membranes were prepared by homogenization and centrifugation in Tris HCI (50mM)/MgC1 (10mM) buffer. Final concentration of 20 pg of protein/assay tube was obtained. Membranes were incubated for 15 min at 30°C with appropriate concentrations of drugs (100nM AM, lOpM Forskolin) and assay mix containing ATP regeneration system and (1.3 2P-ATP. After the reaction was stopped (with stop solution containing 3H-CAMP) contents of the assay tubes were passed through Dowex and subsequently through alumina columns to separate the degradation products of ATP, by washing the dowex with water and alumina with 165 imidazole. Elution profile was done to determine the amount of water and imidazole needed to wash and elute the products. Product eluted from alumina column was counted for the presence of 3H-CAMP and a32P-CAMP in a B-counter. Each experiment was done in triplicates and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. Basal cyclase activity and forskolin stimulation did not show statistically significant differences between treatments. 6.2.7. CAMP accumulation assays: Rat mesangial cells were seeded on a 48-well plate (and Rat2 fibroblast cells on 24-well plates) until reaching 80-90% confluency, then incubated in serum-free media overnight before experiment. Resensitization experiments were carried out as described in Materials and Methods section, with cells pretreated with 10nM rAM and subsequently challenged for 10 min at 37°C with appropriate concentrations of drugs (100nM AM, IOpM Forskolin) in the presence of 200pM 3- isobutyl-l-methylxanthine. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in cells were calculated using a standard curve ranging from 10 to 10 4 fmol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. Basal cyclase activity and forskolin stimulation did not show statistically significant differences between treatments. 6.2.8. RNA Interference analysis: Gene-specific d-siRNA for lacZ (control) and RAMP3 were generated and purified using BLOCK-iT Dicer RNAi kit from Invitrogen 166 (Carlsbad, CA). RMCs were transfected with d-siRNAs using Lipofectamine 2000 as per manufacturer’s instructions (Invitrogen). 48 hours after transfection cells were frozen for mRNA analysis, or used for CAMP accumulation assays or immunofluorescence microscopy. 6.2.9. Quantitative PCR analysis: Total RNA was isolated from RMCs using Trizol reagent (GIBCO BRL). After sodium acetate-ethanol precipitation and several ethanol washes, RNA was used as a template in a quantitative PCR amplification procedure. Quantitative PCR analysis was carried out with the LUX (Light Upon Extension) fluorogenic primer method, following the protocol in the manufacturer’s manual, as described by Nazarenko et al . 6.2.10. Mutagenesis procedure: Site-directed mutagenesis was performed using a PCR- based strategy that employs the pfu Turbo polymerase (Stratagene, La Jolla, CA). A pair of complementary oligonucleotides containing the appropriate point mutations in the sequence of RAMP or a premature stop codon at position 145 or 147 codon of RAMP-3 for deletion mutants were synthesized (Michigan State University Macromolecular structure facility). The PCR for the mutation was as follows: 94°C for 2 minutes; 30 cycles of 94°C for 30 sec., 50°C for 30 sec., 68°C for 8 min.; final cycle of 68°C for 8 minutes. PCR product was digested for 4 hours with DpnI enzyme (Invitrogen) and transformed in to DHSa cells. Mutations were confirmed by automated sequencing (Michigan State University Genomic Technology Support Facility). 167 6.2.1]. Immunofluorescence microscopy: HEK 293 cells were transfected as described above and seeded at 24hr post-transfection onto collagen type I-Coated coverslips. Resensitization assays were performed as described and reactions were stopped by fixing cells in 4% paraformaldehyde for 30min. at room temperature. Samples were perrneablized with 0.1% v/v Triton X-100 in PBS and blocked overnight in 0.1% v/v Triton X-100 in PBS + 10% goat serum. Samples were incubated in primary antibody in blocking buffer for 2h at room temperature (NSF at 1:250 and RAMP3 at 1:200). Appropriate secondary antibodies were applied for 1h at room temperature (Goat anti- mouse Cy3 at 1:500 and Goat anti-rabbit Cy5 at 1:400). Coverslips were mounted in Shandon Permafluor mounting medium and slides stored at 2-8°C until analysis. Cells were visualized on a Zeiss 210 laser confocal microscope at a zoom of 2. Images presented are'representative single optical sections of a z-series taken from at least twenty fields per experiment and at least three individual experiments. Images in this thesis/dissertation are presented in color. 6.2.12. Fusion protein overlays and western blotting: 10 pg of GST-fusion proteins were resolved on a 10% SDS-PAGE gel and transferred to nitrocellulose filters. Filters were blocked with 5% w/v fat-flee milk powder in Tris-buffered saline with Tween 20 (TTBS: 20mM Tris, pH 7.4, 500mM NaCl, 0.1% v/v Tween 20) and incubated overnight at 4 °C in lysates of HEK293 cells with or without overexpression NSF. Blots were then washed three times with TTBS buffer and incubated with anti-NSF monoclonal antibody for 2 h at room temperature. After three washes with TTBS, filters were incubated for 1h with horseradish peroxidase-conjugated goat anti-mouse secondary 168 antibody (Gibco BRL®, Grand Island, NY), washed again with TTBS, soaked in Supersignal West Pico chemiluminescent substrate (Pierce) and exposed to x-ray film. Same protocol, with the exception of the overnight incubation with cell lysate, was followed for immunoblot analysis of RAMP3. 6.2.13. Statistics: Data are presented as mean i S.E.M. Single group comparisons exercised a paired Student’s t-test method. Statistical significance was set at P<0.05. 6.3. Results 6.3.]. Role of NSF in Resensitization of the CL—R/RAMP complex Our lab has previously published that rat mesangial cells (RMCS) endogenously express the AMlR (CL-R + RAMP2) and the AM2R (CL-R + RAMP3) (8). This data was repeated and NSF expression was confirmed in the RMCs with RT-PCR and immunocytochemistry (data not shown). Our laboratory has also reported that pretreatment of rat mesangial cells (RMCS) with AM leads to an agonist-stimulated desensitization and internalization of the CL-R/RAMP complex. Phosphatase-dependent resensitization of AM responsiveness was also demonstrated after agonist-stimulated desensitization [285]. Measuring CAMP accumulation we repeated these results in this study (Figure 28). As a preliminary test to determine if NSF is involved in the resensitization of AM responsiveness, we used a pharmacological inhibitor of NSF, N- ethylmaleimide (NEM). In RMCs treated for 45 seconds with 50 pM NEM during the resensitization experiment, resensitization was blocked, as measured by CAMP 169 accumulation (Figure 28). NEM, however, did not affect basal CAMP accumulation or the desensitization response when compared to untreated cells, an important finding given the ability of NEM to interfere with Ga subunits (Figure 28). These results indicate that NSF plays a role in the sorting of the CL-R/RAMP complex following agonist-induced internalization in this endogenous CL-R/RAMP system where the receptor complex is recycled. To fully evaluate the molecular mechanisms of this observation, we used HEK 293 cells to examine the interaction of the CL—R/RAMP complex with NSF and the impact of this interaction on receptor trafficking. In contrast to RMCs, HEK 293 cells express very low endogenous levels of RAMPS. Kuwasako et al. have demonstrated that in HEK 293 cells overexpressing the CL-R/RAMP complex, agonist-induced internalization leads to receptor trafficking to a degradation pathway [276]. In this study the internalized CL-R/RAMP complexes were colocalized with LAMP-1, a lysosomal marker, to show the targeting of the receptor for the degradation pathway. Utilizing adenylate cyclase activity assays, whole-cell ligand binding, and immunofluorescence microscopy we confirmed these findings (Figure 29A,B). Pretreatment of HEK 293 cells transfected with CL-R and RAMP3 with 10 nM AM for one hour resulted in desensitization of the adenylate cyclase response from 50% (of forskolin stimulation) in untreated cells to 28% in AM-treated cells (Figure 29A, left axis). Even after the removal of agonist and incubation with buffer alone for indicated times through 4h, the adenylate cyclase response remained desensitized (Figure 29A, left axis), indicating a lack of resensitization. Consistent findings were obtained with whole- cell binding and immunofluorescence microscopy experiments (Figure 29A, right axis, 29B). 170 To determine if NSF overexpression could alter the receptor trafficking in this cell system, NSF was co-transfected with CL-R and RAMP-3, and resensitization and recycling assays were performed. Resensitization and recycling were monitored by adenylate cyclase activity assays and whole-cell competition binding, respectively. In addition, visualization of the trafficking of the receptor complex was performed by immunofluorescence microscopy. In the absence of NSF, pretreatment with AM for one hour resulted in desensitization of the adenylate cyclase response and internalization of the receptor complex. Upon removal of agonist and incubation with buffer alone for 4h, the adenylate cyclase response remained desensitized and the receptor complex remained internalized, indicating a lack of resensitization (Figure 30A, B). In contrast, when NSF was co-transfected in the cells, although the desensitization response (i.e. response after 1h agonist treatment) was not altered, the cells now underwent time-dependent resensitization (i.e. response after 1, 2, or 4h agonist removal) in response to AM (Figure 30A). Consistent findings were obtained with whole-cell binding and immunofluorescence microscopy experiments (Figure 30B, 31). Time course experiments indicated the time course for complete resensitization and recycling of the CL-R/RAMP3 receptor complex to be 4b in HEK 293 cells, as measured by adenylate cyclase, whole-cell binding, and immunofluorescence microscopy experiments (Figure 31). All subsequent experiments in HEK 293 cells use the 4h time point to determine receptor complex recycling and resensitization. These results indicate that the presence of NSF alters the intracellular sorting of the CL-R/RAMP3 receptor complex after AM- stimulated endocytosis. 171 6.3.2. RAMP isoform-specific regulation of CL-R/RAMP receptor complex trafficking To determine if this effect of NSF was specific for RAMP3, the additional RAMPS (RAMP—l or -2) were tested for their ability to act with NSF to alter the receptor complex life cycle. Interestingly, in contrast to RAMP3, presence of NSF did not alter the resensitization response or recycling pattern of the CL-R/RAMP-l or -2 receptor complexes. Both the activity and receptor number remained at desensitized levels in cells transfected with CL-R+RAMP1 or CL-R+RAMP2 (along with NSF) (Figure 32A-D). Both CL-R/RAMP] and CL-R/RAMP2 complexes showed no difference in receptor expression levels at the plasma membrane (as measured with whole-cell binding) in untreated cells, as compared to CL-R/RAMP3 complex. These results indicate that RAMP3 must contain a molecular determinant distinct from the other RAMPS that allowed its interaction and activity with NSF. 6.3.3. Role of PDZ interactions in trafficking of the CL-R/RAMP3 complex We hypothesized that the unique characteristic of RAMP3 that allowed its interaction with NSF to change receptor trafficking was a PDZ motif on its extreme C- terrninus. In order to establish if this domain is critical for interaction of the CL- R/RAMP3 complex with NSF, the PDZ motif (-DTLL) on RAMP3 was deleted. Deletion of this domain did not affect basal adenylate cyclase activity or the desensitization response of the CL-R/RAMP3 complex in response to AM, even in the presence of NSF (Figure 32). In contrast, the deletion of the PDZ motif (-DTLL) significantly affected the resensitization and recycling of the CL-R/RAMP3 receptor 172 BL“- complex in the presence of NSF. Both radioligand binding and adenylate cyclase assays showed a loss of recycling and resensitization, respectively, of the receptor complex when RAMP3A145-8 was expressed in the presence of NSF, as compared to the wild- type RAMP3 with NSF (Figure 33A, B). Mutant RAMP3 (RAMP3A145-8) showed no difference in receptor expression levels at the plasma membrane (as measured with whole-cell binding) in untreated cells, as compared to wild-type CL—R/RAMP3 complex. Additionally, results were confirmed when a recycling assay with RAMP3A145-8 was performed with immunofluorescence microscopy (Figure 34). To further test the hypothesis that the absence of the PDZ motif on the RAMP2 accounts for the lack of interaction of the CL-R/RAMP2 complex with NSF, and hence the inability of the CL-R/RAMP2 complex to follow a recycling pathway, the PDZ motif of RAMP3, the amino acids -DTLL, were substituted on the C-terminus of RAMP2, in exchange for its original four C-terminal amino acids (-EAQA). The RAMPZADTLL mutant showed similar levels of adenylate cyclase activity and whole-cell radioligand binding without pretreatment and after desensitization, as compared to wild type RAMP2 in control experiments (Figure 35). The RAMPZADTLL mutant also showed similar receptor expression levels at the plasma membrane (as measured with whole-cell binding) in untreated cells, as with the wild-type CL-R/RAMP2 complex. Resensitization assays (measured with adenylate cyclase activity) and recycling assays (measured by whole-cell binding) were performed in HEK 293 cells were transfected with CL-R, RAMPZADTLL, and NSF, as described previously. Similar to the CL- R/RAMP3 complex, the CL-R- RAMPZADTLL complex underwent resensitization and recycling in the presence of NSF, as assessed by adenylate cyclase and whole-cell 173 binding (Figure 35). These findings provide additional evidence that the PDZ motif on the RAMP3 is the site of interaction of the receptor complex with NSF, causing a Change in receptor trafficking, from a degradative to a recycling pathway. To further identify the critical amino acids in the PDZ recognition sequence which regulate the RAMP3/NSF interaction, point mutations of the amino acids of the RAMP3 PDZ motif to alanine were performed. The functional effects of the point mutations were analyzed with resensitization assays, measured by adenylate cyclase activity, and recycling assays, measured by whole-cell binding, as described before. Our results indicate that mutation of D145, T146, or L148 to alanine disrupted the RAMP3 interaction with NSF and inhibited the resensitization and recycling of the CL-R/mutant RAMP3 complex after AM-induced endocytosis in HEK 293 cells (Figure 36). Mutation of L147 to alanine had no effect on the resensitization or recycling of the receptor complex in the presence of NSF (Figure 36). Control experiments with the point mutations co-transfected with CL-R showed similar levels of adenylate cyclase activity and whole-cell binding as compared to wild-type RAMP3 without pretreatment and after desensitization. RAMP3 point mutants also showed no difference in receptor expression levels at the plasma membrane (as measured with whole-cell binding) in untreated cells, as compared to wild-type CL-R/RAMP3 complex. To examine if the PDZ motif on RAMP3 is interacting with NSF, overlay assays were performed. This was accomplished using GST-RAMP3 fusion proteins in an overlay assay with cell lysates of HEK 293 cells overexpressing NSF. Control experiments run with GST protein showed no detectable bands when incubated with NSF lysates and probed with an NSF antibody (Figure 37A). Importantly, wild-type RAMP3 174 fusion proteins showed interaction with NSF in the cell lysates of HEK 293 cells overexpressing NSF in the overlay assay (Figure 37A). In addition, RAMP3A145-8 fusion proteins, lacking the PDZ motif on RAMP3, showed no detectable bands when incubated with NSF lysates and probed with an NSF antibody (Figure 37A). Lysates of HEK 293 cells not over—expressing NSF showed no detectable bands when run with GST-RAMP3 in the overlay assay and probed for NSF (Figure 37B). When blots used in the overlay assay were stripped and probed for RAMP3, a band was detected in the GST— RAMP3 lane in the exact location as in the overlay assay when probed for NSF (Figure 37C). This data demonstrates an interaction between RAMP3 and NSF via the PDZ motif on RAMP3, an interaction that is capable of regulating CL-R/RAMP3 (AM2R) complex trafficking. 6.3.4. RAMP3 and NSF regulation of receptor trafficking in unaltered cell lines It was important to establish if our observations in the HEK 293 cells was transferable to unaltered cells lines. Reexamining the rat mesangial cells used in the first set of experiments, we employed RNA interference technology to knockdown RAMP3 expression. Having demonstrated the requirement of NSF in the cells for efficient receptor resensitization (Figure 28), the RAMP3 RNA interference experiment would determine if RAMP3 was also required for effective receptor resensitization. In both mRNA and protein expression studies, RAMP3 expression dramatically decreased, while control experiments using lacZ knockdown showed no significant alteration in RAMP3 expression when compared to wild-type cells (Figure 38A, B; data not shown). Resensitization assays were performed and CAMP accumulation was measured to 175 &\\"'r ' determine the effect of RAMP3 RNA interference on receptor resensitization in rat mesangial cells. RNA interference for RAMP3 showed similar levels of CAMP accumulation as compared to wild-type RMCs without pretreatment and after desensitization (Figure 38C). Strikingly, when allowed sufficient recovery time in the absence of agonist following desensitization, RMCs with RAMP3 RNA interference showed an inability to resensitize, unlike the wild-type RMCs (Figure 38C). This finding demonstrates that NSF and RAMP3 are both critical for receptor targeting for resensitization in rat mesangial cells, an unaltered cell line absent of the issues of overexpression. As a further test of our proposed model, we employed a cell line that does not express RAMP3, but does express the AMlR (CL-R and RAMP2). We hypothesized that, with the absence of RAMP3, this cell line would fail to resensitize following agonist pretreatment, and that expression of RAMP3 in these cells would allow a switch in receptor targeting for resensitization. Rat2 fibroblast cells have been shown by Choksi et al. to lack RAMP3 expression, but do express the AM1R(CL-R and RAMP2) [308]. We repeated this finding and confirmed the expression of NSF in Rat2 fibroblast cells with RT-PCR and immunocytochemistry (data not shown). We performed resensitization assays with the Rat2 fibroblast cells and measured resensitization by CAMP accumulation. The Rat2 fibroblast cells exhibited a decrease in CAMP accumulation after agonist pretreatment and failed to resensitize, as predicted, when allowed to recover in the absence of agonist for four hours. Interestingly, consistent with our model, RAMP3 expression in the Rat2 cells showed no alteration in CAMP accumulation without pretreatment and after desensitization when compared to wild-type Rat2 cells, but now 176 showed resensitization when allowed recovery time in the absence of agonist (Figure 39). Furthermore, expression of the RAMP3 PDZ motif mutant in the Rat2 cells was unable to cause receptor resensitization, while showing similar levels of CAMP accumulation as compared to wild-type Rat2 cells without pretreatment and following desensitization (Figure 39). RAMP3 and RAMP3A145-8 showed similar levels of transfection efficiency in the Rat2 cells, as measured by immunocytochemistry (data not shown). This data further confirms the crucial role of RAMP3 in the targeting of the AM2R for resensitization/recycling after agonist-stimulated desensitization. Additionally, it suggests that differential RAMP expression in cells may determine the sorting of the AM-R from the endosomes after internalization. 177 -Control 40- er; 50 pM NEM E 3 0 H 5E a: O § 13 g .9 5E a. E < o Figure 28: Effect of N-ethylmaleimide, an NSF inhibitor, on resensitization of CAMP accumulation in rat mesangial cells (RMCS). The lh pretreatment time point shows desensitization of CAMP accumulation response in cells with or without treatment with NEM. The 2 hr recovery time point shows resensitization is inhibited in cells treated with NEM, as compared to untreated RMCs. This indicates a role for NSF in the resensitization observed in RMC cells. RMC cells seeded in 24-well plates were pretreated with IOnm rAM for 1h were washed extensively to remove residual agonist and treated with 50 pM NEM for 45 seconds. Following NEM treatment. cells were washed repeatedly and incubated in serum-free media with Spg/ml cycloheximide for indicated times. After recovery time, RMC cells were re-Challenged with 100 nM rAM for 15 minutes and plates were frozen. Determination of CAMP level was measured using the Biotrak CAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. CAMP levels in rat mesangial cells were calculated using a standard curve ranging from 10 to 10 4 frnol of CAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p S 0.05; n 2 3 experiments. 178 Fig cor des ICCO eipn Show O, O I .AC Activity '120 DBinding -100 $ 1 '80 -4o 10- -2o 0‘ -0 Control 1h pretreat L 1 1 protein) {5 AC Activity (pmollminlmg 3 [% Forskolin] 3 a [Ionuoo %] (lies/sails) xewa Recovery time (hr) Figure 29A: The sorting of the AM2R (CL-R+RAMP3) in HEK 293 cells. CL-R/RAMP3 complex does not resensitize/recycle in HEK 293 cells after agonist-stimulated ‘ " " ‘ " " HEK 293 cells transiently transfected with CL-R and RAMP3 were treated for one hour with AM (10 nM) and then washed and receptor resensitization (adenylate cyclase activity) and recycling (radioligand binding) were measured after indicated recovery times in the absence of agonist. Adenylate cyclase activity is shown on the left y-axis, expressed as percent maximal response (% forskolin stimulation), and radioligand binding is shown on the right y-axis, expressed as percent control. * p S 0.05; n 2 3 experiments. 179 1hr Pretreat O 4 hr Recovery Figure 298: The sorting of the AM2R (CL-R+RAMP3) in HEK 293 cells. lmmunofluorescence microscopy shows failure of CL-R/RAMP3 receptor complex to recycle after agonist-stimulated internalization. HEK 293 cells transfected with CL—R-GFP and RAMP3 were pretreated with 10nM AM for lh. After pretreatment with AM, cells were washed and incubated in serum-free media with Spg/ml cycloheximide to allow receptor recycling for indicated times. Cells were fixed and components were visualized using anti-RAMP3 antibody (1:200) and detected with Cy5 secondary antibody (1:200), and CL-R is detected with an EGFP tag; overlays of staining patterns are shown in the far right panels. Images shown are representative of at least twenty fields imaged per experiment from at least three experiments. Bar scales on all images represent 50pm. 180 - CL-R+RAMP3 D CL-R+RAMP3+NSF A 125- % I 2 ,8 = 100- r '7: Fl: 53% 75' . :5 8 * * P .5 o 50. * * * at E z E 25- m ,1 _ _ _ _ __ “A 360* IN ‘1. b ' Recovery time (hr) Figure 30A: The role of NSF in the sorting of the AM2R (CL-R+RAMP3) in HEK 293 cells. NSF causes recycling of AM2R in HEK 293 cells. HEK 293 cells transiently transfected with CL-R and RAMP3 with or without NSF. At 48h post-transfection, cells were treated for one hour with AM (10 nM) and then washed and receptor recycling was measured after indicated recovery times in the absence of agonist. Receptor recycling measured by whole-cell competition binding assays using ”SI-rAM as ligand (cold rAM served as the competitor) and number of binding sites/cell was estimated using the GRAPHPAD PRISM software. “No pretreat” represents samples at maximal radioligand binding that were not pre-incubated with agonist. “1h pretreat” represents samples pretreated with AM (10 nM) for one hour, washed as indicated in Methods section, and tested immediately after wash steps for radioligand binding. “1 , 2, and 4 hr recovery” samples were pretreated with AM (10 nM) for one hour, washed, and allowed to recover for indicated times in media without agonist plus Spg/ml cycloheximide, then analyzed for radioligand binding. NSF overexpression in cells expressing AM2R caused altered receptor trafficking fiom degradation to recycling pathway. * p S 0.05; n 2 3 experiments. 181 - CL-R+RAMP3 :1 CL-R+RAMP3+NSF A 50' r- .E J; to * . "é’ E 40- [t ' ._ 0.3 * * > * . Egg 30- * . 1.. .‘ .5 LL ‘ 2 E .\° 20- i g T ;' i 3; 10' it' OJ I I I I ‘ o '8‘ " ‘1. 5‘ 00“ ‘6’66 I I 0 «’09 Recovery time (hr) Figure 30B: The role of NSF in the sorting of the AM2R (CL-R+RAMP3) in HEK 293 cells. NSF causes recycling of AM2R in HEK 293 cells. NSF causes resensitization of AM2R in transfected HEK 293 cells. HEK 293 cells transfected and pretreated with agonist as described in 29A. After agonist pretreatments, membranes were extracted and AC activity in response to 100 nM AM was measured. NSF overexpression with AM2R allowed time-dependent receptor resensitization. Experiments performed in triplicates and data expressed as percent maximal stimulation (% Forskolin). " p S 0.05; n 2 4. 182 No Pretreatment 1 hr Pretreatment 4 hr Recovery CL-R ..- RAMP3...- NSF I Overlay‘ I ,I 183 Figure 3]: Localization of CL-R, RAMP3, and NSF in HEK 293 cells during a recycling experiment. After lh AM pretreatment, CL-R and RAMP3 are internalized and show co- localization with NSF intracellularly. After 4h recovery time post-AM pretreatment, CL-R and RAMP3 show distribution at the plasma membrane of the cell, demonstrating recycling of the receptor complex. HEK 293 cells transfected with CL-R-GF P, RAMP3, and NSF were pretreated with 10nM AM for lh. After pretreatment with AM, Cells were washed and incubated in serum- free media with Spg/ml cycloheximide to allow receptor recycling for indicated times. Note: “1 hr pretreatment” indicates time just after AM pretreatment and wash steps, with no recovery time. Cells were fixed and components were visualized using anti-RAMP3 antibody (1:200) and anti- NSF antibody (1:250) with Cy5 anti-rabbit secondary antibody ( 1:400, in blue) and Cy3 anti- mouse secondary antibody (1:500, in red), respectively; CL-R-GFP is detected with an EGFP tag and shown in green; overlays of staining patterns are shown in the far right panels. Images shown are representative of at least twenty fields imaged per experiment fi'om at least three experiments. Bar scales on all images represent lOOpm. 184 - CL-R+RAMP1 E: CL-R+RAMP1+NSF = 1oo- .— a a 3 75- .53 a? .5 o 50- =9. 3} I 25- 1! E m . 0d a \1 so?” Figure 32A: The role of NSF in post-endocytic sorting of CGRP] (CL-R+Rl) and AM] receptor. NSF overexpression does not alter the trafficking of the CGRP receptor after internalization. HEK 293 cells were transfected with CL-R and R], with or without NSF. 48h post-transfection, cells were pretreated with CGRP (10 nM) for one hour, washed as described in Materials and Methods, allowed indicated recovery times in agonist-free media, and receptor recycling was measured with whole-cell competition binding using l2"'I-rCGRP as the ligand and cold rCGRP as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 3. 185 - CL-R+RAMP1 D CL-R+RAMP1+NSF l , N I» O O I I % Forskolin 3:3 AC Actiwty (pmollminlmg protein) O I Figure 328: The role of NSF in post-endocytic sorting of CGRPI (CL-R+R1) and AMI receptor. NSF overexpression does not cause CGRP receptor resensitization after agonist- stimulated endocytosis. HEK 293 cells were transfected and pretreated as in figure 29A and then membranes were extracted. Adenylate cyclase activity was measured in membranes stimulated with 100 nM CGRP for 15 min. Experiments performed in triplicates and expressed as percent maximal stimulation (% Forskolin). * p S 0.05; n 2 3. 186 - CL-R-I-RAMPZ 1:1 CL-R+RAMP2+NSF a 1001 —~ 3 3 W . g 3 75 * * 3e: l g 8 50- % .\° :2 25- in E m 0' u a e‘ 0'50 e‘“0 “O?‘ “8 ?‘ fi‘“ $06 Figure 32C: The role of NSF in post-endocytic sorting of CGRP] (CL-R+Rl) and AM] receptor. NSF overexpression does not cause recycling of AMI receptor after internalization. HEK 293 cells were transfected with CL-R and R2, with or without NSF. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour, washed as described in Materials and Methods, allowed indicated recovery times in agonist-free media, and receptor recycling was measured with whole-cell competition binding using I25I-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 4. 187 161 in me wit Mai - CL-R+RAMP2 E CL-R+RAMP2+NSF A 4oq .5: g .g 30- * e g .3 o . < z ‘3 2° 2 E “z E °\ 10.. E a. v o. — t x 99““ «0'536 39‘0““ $0 \“‘ “x“ Figure 32D: The role of NSF in post-endocytic sorting of CGRP] (CL-R+Rl) and AM] receptor. NSF overexpression does not cause AMI receptor resensitization after agonist-induced internalization. HEK 293 cells were transfected and pretreated as in figure 29A and then membranes were extracted. Adenylate cyclase activity was measured in membranes stimulated with 100 nM AM for 15 min. Experiments performed in triplicates and expressed as percent maximal stimulation (% Forskolin). * p S 0.05; n _>_ 3. 188 - CL-R+RAMP3+NSF [:1 CL-R+RAMP3A145-8+NSF c 150- 3 2 in "g " 100 as :5 o .5 ‘3 £9\ 50. x a E m 04 so?“ Figure 33A: The effect of RAMP3 PDZ motif deletion on the trafficking of the AM2R (CL- R+RAMP3). Deletion of RAMP3 PDZ motif blocked the recycling of the CL-R/RAMP3 complex when co-expressed with NSF. HEK 293 cells were co-transfected with CL-R, wild-type RAMP3 or RAMP3AI45-8, and NSF. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour, washed as described in Materials and Methods, allowed 4h recovery times in agonist-free media, and receptor recycling was measured with whole-cell binding using I25I-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM software. * p S 0.05; n 2 3. 189 - CL-R+RAMP3+NSF E CL-R+RAMP3A145-8+NSF * * 0' I I 6“ Figure 333: The effect of RAMP3 PDZ motif deletion on the trafficking of the AM2R (CL- 50- w 1k C O 1 a 3(- % Forskolin N ‘P AC Activity (pmollminlmg protein) R+RAMP3). Deletion of RAMP3 PDZ motif blocked the resensitization of the CL-R/RAMP3 complex afier agonist-stimulated desensitization when co-expressed with NSF. HEK 293 cells were transfected and pretreated as in figure 29A and then membranes were extracted as described in Materials and Methods section. Adenylate cyclase activity was measured in membranes stimulated with 100 nM AM for 15 min. Experiments performed in triplicates and expressed as percent maximal stimulation (% Forskolin). "‘ p S 0.05; n 2 3. 190 No Pretreatment 1 hr Pretreatment 4 hr Recovery Overlay Figure 34: Localization of CL-R, RAMP3AI45-8, and NSF in HEK 293 cells during a recycling experiment. Untreated and afler one hour AM (10nM) pretreatment, CL-R and RAMP3A145—8 internalize similarly to the wild-type CL-R/RAMP3 complex. Afier a 4h recovery time in the absence of agonist, CL-R and RAMP3A145-8 fail to recycle to the plasma membrane. Experiments performed as described in Figure 31. Fixed cells were stained with anti-RAMP3 antibody (1:200) and anti-NSF antibody (1:250) with Cy5 anti-rabbit secondary antibody (1 :400, in blue) and Cy3 anti-mouse secondary antibody (1:500, in red), respectively; CL-R-GFP is shown in green; overlays of staining patterns are shown in the far right panels. Images shown are representative of at least twenty fields imaged from at least three experiments. Bar scales on all images represent lOOpm. 191 - CL-R+RAMP2+NSF E CL-R+RAMP2ADTLL+NSF l Bmax (binding sites/cell) % Control 0| ‘? Figure 35A: Effect of PDZ motif substitution on the C-terminus of RAMP2 on recycling/resensitization of CL-R/RAMP complex. Substitution of PDZ motif (-DTLL) on the C- tenninus of RAMP2 cause a change in receptor trafficking phenotype from degradation to recycling of the receptor complex. HEK 293 cells were co-transfected with CL-R, wild-type RAMP2 or RAMPZADTLL, and NSF. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour, washed as described in Materials and Methods, allowed 4h recovery times in agonist-free media plus Sug/ml cycloheximide, and receptor recycling was measured with whole- cell binding using ”SI-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM sofiware. “‘ p S 0.05; n 2 4. 192 - CL-R+RAMP2+NSF E CL-R+RAMP2ADTLL+NSF Ts‘ .. l 2 .E 30 .— Q— - O a??? * * * a < 5 .\° 15' O E 3 0' I I I 01 «“0 «“3 0“0 Figure 358: Effect of PDZ motif substitution on the C-terminus of RAMP2 on recycling/resensitization of CL-R/RAMP complex. PDZ motif substituted on the C-terminal tail of RAMP2 cause resensitization of adenylate cyclase activity. HEK 293 cells were transfected and pretreated as in figure 29A and then membranes were extracted as described in Materials and Methods section. Adenylate cyclase activity was measured in membranes stimulated with 100 nM AM for 15 min. Experiments performed in triplicates and expressed as percent maximal stimulation (% Forskolin). "‘ p S 0.05; n 2 4. 193 - No Pretreatment D 1 hr Pretreatment [:34 hr Recovery Time Bmax (binding sites/cell) Figure 36A: Effect of RAMP3 PDZ motif point mutations on the recycling/resensitization of the CL-R/RAMP3 complex in the presence of NSF. All point mutants within the PDZ motif on RAMP3, except R3Ll47A, blocked the recycling of the CL-R/RAMP3 complex seen when co- expressed with NSF. HEK 293 cells were co-transfected with CL-R, wild-type RAMP3 or RAMP3 point mutants, and NSF. 48h post-transfection, cells were pretreated with AM (10 nM) for one hour, washed as described in Materials and Methods, allowed 4h recovery times in agonist-free media, and receptor recycling was measured with whole-cell binding using I25I-rAM as the ligand and cold rAM as the competitor. Number of binding sites/cell was estimated with GRAPHPAD PRISM sofiware. "‘ p S 0.05; n 2 3. I94 - No Pretreatment D 1 hr Pretreatment m 4 hr Recovery Time E 50' E 9E4o~ 5:3 ‘6Ee30‘ * 3:: <§.\° 20' OH 5 1o- 0" '5 v v v v s a? «,e ya e" e & «Lb Figure 368: Effect of RAMP3 PDZ motif point mutations on the recycling/resensitization of the CL—R/RAMP3 complex in the presence of NSF. Confirming the whole-cell binding data, all point mutations to the PDZ motif of RAMP3, except R3Ll47A, inhibited the resensitization of adenylate cyclase activity seen with wild-type RAMP3-CL-R complex and co-expression of NSF. HEK 293 cells were transfected and pretreated as in figure 29A and then membranes were extracted as described in Materials and Methods section. Adenylate cyclase activity was measured in membranes stimulated with 100 nM AM for 15 min. Experiments performed in triplicates and expressed as percent maximal stimulation (% Forskolin). * p S 0.05 two-tail; # p5 0.05 one-tail ; n 2 3-4. 195 a. b. c. GST GSTr GST- GST- RAMP3 GST RAMP} GST- RAMP3 GST RAMP3 .\145-8 GST RAMP3 A145-8 GST RAMP3 A145-8 70 KDa - -- so KDa Figure 37: Interaction studies of RAMP3 with NSF. Fusion proteins of GST-RAMP3 demonstrate interaction with NSF in an overlay assay and western blot analysis. lOug of GST, GST-RAMP3, and GST-RAMP3AI45-8 protein was run and separated on an SDS-PAGE gel and transferred to a nitrocellulose filter. The nitrocellulose filter was incubated overnight in lysate of HEK293 cells transfected with or without NSF at 4°C. Filter was then washed and probed by immunoblot for NSF (1:250). Identical filters were probed by immunoblot for RAMP3 (1:400). A) GST, GST-RAMP3, and GST-RAMP3AI45-8 overlay assay incubated in NSF-transfected HEK 293 lysates and probed for NSF. B) GST, GST-RAMP3, and GST-RAMP3A145-8 overlay assay incubated in non-transfected HEK 293 lysates and probed for NSF. C) lmmunoblot of GST, GST-RAMP3, and GST-RAMP3A145-8 probed for RAMP3. Shown are representative blots of at least three experiments. 196 - Wild-type 200. D RAMP3 Knockdown ..L Cl C e I. [% Forskolin] 3 C.’ cAMP Accumulation (nmollmg protein) 0| ‘? 0' I Preilea‘ “o Figure 38A: Effect of RAMP3 RNA interference on resensitization of AM2-R in rat mesangial cells. RAMP3 RNA interference disrupts the ability of AM—R to resensitize afier agonist pretreatment. RMC cells seeded in 24-well plates were transfected with d-siRNA of RAMP3 and incubated for 48 hr to allow RNA knockdown. Wild-type and d-siRNA transfected cells were pretreated with IOnm rAM for 1h and were washed extensively to remove residual agonist. Following washes, cells were incubated in serum-free media with 5 ug/ml cycloheximide for 2 hr. After recovery time, RMC cells were re-challenged with 100 nM rAM for 15 minutes and plates were frozen. cAMP levels were measured using the Biotrak cAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. cAMP levels in rat mesangial cells were calculated using a standard curve ranging from 10 to 10 4 fmol of cAMP. Each experiment was done in duplicate and repeated four times. Data is expressed as percent maximal response, % forskolin. * p S 0.05; n 2 4 experiments. I97 1.25- $ [Fold Wild-type] -° 3 5.5 tr RAMP3/GAPDH _o no ‘1' a Wild-type RAMP3 Knockdown P o o I Figure 38B: Effect of RAMP3 RNA interference on resensitization of AM2-R in rat mesangial cells. RAMP3 RNA interference disrupts the ability of AM-R to resensitize afier agonist pretreatment. RMC cells seeded in 24-well plates were transfected with d-siRNA of RAMP3 and incubated for 48 hr to allow RNA knockdown. mRNA analysis by RT-PCR showed dramatically decreased levels of RAMP3 mRNA in RAMP3 RNA interference sample as compared to wild type, while lacZ knockdown had no effect on RAMP3 message levels (data not shown). RNA isolation and Q-PCR performed as described in Experimental Procedures section. GAPDH expression measured for all samples for normalization. n=3 experiments. 198 Control RAMP3 Knockdown Anti-RAMP3 Figure 38C: Effect of RAMP3 RNA interference on resensitization of AM2-R in rat mesangial cells. RAMP3 RNA interference disrupts the ability of AM-R to resensitize after agonist pretreatment. RMC cells seeded in 24-well plates were transfected with d-siRNA of RAMP3 and incubated for 48 hr to allow RNA knockdown. lmmunofluorescence microscopy of RMCs with RAMP3 RNA interference demonstrated greatly decreased levels of RAMP3 protein expression in RAMP3 RNA knockdown cells, as compared to wild-type RMC cells. LacZ knockdown had no efiect on RAMP3 protein expression in RMCs (data not shown). Cells prepared as described in Experiment procedures section for immunofluorescence microscopy. Fixed cells were stained with anti-RAMP3 antibody (1:200) and detected with a Cy3 anti-rabbit secondary antibody (1 :500, in red). Images shown are representative of at least three experiments. Bar scales on all images represent 20pm. 199 - No transfection E RAMP3 -RAMP3A145-8 15- cAMP Accumulation (nmollmg protein) [%Forskolin] -r- '1- No pretreat 1hr pretreat 4hr recovery Figure 39: AM-R trafficking in Rat2 fibroblast cells. Rat2 fibroblast cells show a lack of receptor resensitization afier agonist pretreatment, whereas when RAMP3 is expressed in Rat2 cells, receptor resensitization is now observed. Expression of PDZ motif mutant of RAMP3 in Rat2 cells is unable to alter the receptor trafficking. Rat2 cells seeded in 24-well plates were pretreated with 100 nm rAM for 1h and were washed extensively to remove residual agonist. Following washes, cells were incubated in serum-free media with Sag/ml cycloheximide for indicated times. After recovery time, Rat2 cells were re-challenged with 1 mM rAM for 15 minutes and plates were frozen. Determination of cAMP level was measured using the Biotrak cAMP enzyme immunoassay system (Amersham Biosciences) according to the manufacturer's instructions. cAMP levels in Rat2 fibroblast cells were calculated using a standard curve ranging from ID to 10 4 frnol of cAMP. Each experiment was done in duplicate and repeated at least 3 times. Data is expressed as percent maximal response, % forskolin. * p S 0.05; n 2 3 experiments. 200 6.4. Discussion: While many GPCRS utilize similar mechanisms for endocytosis, the functional consequences of endocytosis vary from receptor to receptor. Intemalized receptors that are trafficked through a rapid recycling pathway are restored to the plasma membrane in a functional state to achieve resensitization. On the other hand, receptors that are internalized and targeted to late endosomes and lysosomes experience proteolytic degradation, thus promoting attenuation of receptor signaling and down-regulation of the receptor. The CL-R/RAMP system shows sorting of the receptor complex by both trafficking pathways in different cell lines. That is, in RMCs the CL-R/RAMP complex follows a post-endocytic recycling pathway, while in HEK 293 cells and Rat2 fibroblast cells the receptor complex undergoes degradation [276, 285]. Findings in this report demonstrate some of the cellular and molecular mechanisms responsible for the CL- R/RAMP receptor complex’s trafficking. We have shown that the PDZ type I motif at the C-terrninus of RAMP3 interacts with NSF to target the CL-R/RAMP3 complex for recycling after internalization. NSF is commonly known to interact with aSNAP (soluble NSF attachment protein) and SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins to form the 208 particle, a complex that plays a critical role in intracellular membrane fusion and exocytosis [261-263]. While other labs have demonstrated involvement of NSF with the trafficking of receptors, including the BZAR and AMPA receptors, this study is the first demonstration of NSF acting in concert with a heterodimeric partner of a receptor to alter the trafficking of the GPCR [268, 269]. Targeting of the receptor complex after agonist-stimulated internalization demonstrates a novel role for RAMP3. This new function for RAMP3 may explain the 201 altered RAMP expression patterns in certain animal models of disease. RAMP3 expression has been shown to be increased during the transition from left ventricular hypertrophy to heart failure in Dahl salt-sensitive and deoxycorticosterone acetate (DOCA)-salt spontaneously hypertensive rats and in the myocardium of rats with chronic heart failure, where recycling of the receptor complex would be advantageous for continued ligand responsiveness [309-311]. In fact, in various cardiorenal disease states where AM is protective, circulating plasma levels of AM have been shown to be increased. For example, in chronic glomerulonephritis, type I diabetes, and type II diabetes plasma AM levels are elevated [293-295]. In addition, AM delivery through adenoviral injection has been shown to decrease cardiac hypertrophy and renal damage in rat models of hypertension and improves cardiac function and prevents renal damage in streptozotocin-induced diabetic rats [296-300]. If indeed, AM is exerting protective effects against these diseases, then overcoming desensitization of the receptor complex would be important. It is of interest to determine if NSF expression or localization is altered in these disease states as well. Furthermore, studies identifying additional molecular mechanisms of CL—R/RAMP trafficking may be valuable for therapeutic targeting. While not only discovering a novel function for the RAMPS in post-endocytic trafficking, these findings also demonstrate a firnctional difference between the AMIR (CL-R+RAMP2) and AM2R (CL-R+RAMP3) receptor complexes, in spite of very similar second messenger systems and the physiological responses thus far identified. This study demonstrates the first difference between the RAMP2 and RAMP3 isoforms in the trafficking of the AMIR and AM2R. We suggest that this novel difference in 202 AMI and AM2 receptors may lead to either or both of the following two consequences: 1) there could be yet discovered roles of AM that differentially act through AMI and AM2R because of this novel role of RAMP3 or 2) in disease conditions, expression of RAMP3 may be preferentially stimulated to effect recycling of CLéR/RAMPB complex to affect physiological consequences of AM. This could result in a beneficial or harmful effect depending on the system in question (cardiorenal diseases vs. cancer, respectively). It is important to address the possibility of a species-specific effect, as the prototypic class I PDZ motif is present only in the human RAMP3 and not other species thus far sequenced. The human RAMP3 contains the classical PDZ class I motif (- DTLL), while the two rodent species sequenced (rat and mouse) have a'C-terminal amino acid sequence of -DRLL. The resensitization experiments in the rat mesangial cells demonstrate the importance of the rat RAMP3 for efficient receptor targeting for resensitization. Furthermore, the resensitization experiments in the Rat2 fibroblast cells illustrate the ability of human RAMP3 to alter receptor targeting in a rat cell line to promote receptor resensitization/recycling after agonist pretreatment. Taken together, these data suggest that the lack of complete conservation between species of the C- terrninal PDZ motif on RAMP3 does not alter our proposed model. Finally, recent data suggests that RAMP3 can interact with receptors other than CL-R. It is important to determine if this novel role of RAMP3 in receptor trafficking is specific for CL-R or also for the other receptors RAMPS interact with, namely VPAC, PTHI- and 2-R, and glucagon receptors [286]. In addition to AM, Roh et al. have reported that intermedin, a newly discovered peptide from the calcitonin gene peptide superfamily, can also bind the CL-R/RAMP3 complex [307]. If this reported function for 203 RAMP3 is specific for AM, or if another peptide like intermedin could cause this phenomenon, remains to be tested. This study has shown that CL-R’s heterodimeric partner, RAMP3, is capable of altering the trafficking of the receptor complex after endocytosis by interacting with NSF via its PDZ domain. This demonstrates a novel function for the RAMP accessory proteins and the first difference between the AMIR and AM2R. With recent reports of RAMPS complexing and regulating additional GPCRS, these findings may reveal a more widespread form of regulation of the GPCR life-cycle. 204 7. Summary and Conclusions 7.1. Specific aims, major hypotheses, and results of the study The major aim of this study was to investigate the role of the different RAMP isoforms in the regulation of the AM receptor complex life-cycle. It was hypothesized that through post-translational modifications and protein-protein interactions, the three RAMP isoforms differentially regulate the trafficking and signaling of the AM receptor. Hence, as summarized below, the following specific aims are addressed and the various hypotheses within these aims were tested, and the corresponding experimental results were obtained: Specific Aim #1: Phosphorylation of amino acid residues of AM receptor complex is involved in regulation of receptor desensitization and internalization. Hypothesis 1: Specific serine/threonine residues in the C-terminus or 3rd intracellular loop of CL-R are required for AM-R desensitization and internalization. Hypothesis 2: Specific serine/threonine residues in the intracellular domain of RAMPS are required for desensitization and internalization of AM-R. Hypothesis 3: Based on the isoform of RAMP associated with CL-R, different amino acid residues are important for receptor phosporylation, desensitization, and internalization. 205 Hypothesis 4: Different protein kinases regulate the phosphorylation of CL-R in presence of different RAMP isoforms. Results: a. Serine/threonine amino acid residues (Ser 421 for AMI receptor and Thr 423 for AM2 receptor) on the C-terminal tail of CL-R are required for CL-R/RAMP complex desensitization and internalization. b. Serine and threonine residues in the third intracellular loop of CL-R or intracellular domain of RAMPS are not required for receptor desensitization and internalization. c. Protein kinase A inhibitor blocks only CL-R/RAMPZ desensitization and internalization, while a PKC inhibitor blocks only CL-R/RAMP3 desensitization and internalization, suggesting different kinases are involved in the phosphorylation of CL—R when different RAMP isoforms are associated. Specific Aim #2: Protein-protein interactions of RAMPS with other proteins are involved in the regulation of agonist-stimulated internalization of the AM receptor. Hypothesis 1: RAMP3 (but not RAMP] or RAMP2) interacts with NHERF-1 to inhibit agonist-induced receptor internalization. Hypothesis 2: RAMP3 interacts with NHERF-l via a PDZ recognition motif on RAMP3 and a type I PDZ domain on NHERF-1. 206 Hypothesis 3: NHERF-1 inhibits internalization of the AM2 receptor (CL-R+RAMP3) via interactions with MERM proteins in the actin cytoskeleton. Results: a. NHERF-l specifically interacts with RAMP3 and causes an inhibition of agonist- induced receptor internalization, without affecting receptor desensitization. b. RAMP3 interacts with NHERF-l via a PDZ recognition motif on the extreme C- terminus of RAMP3 and a type I PDZ domain on the N-terminus of NHERF-l. c. NHERF inhibits internalization of AM2 receptor through interactions with the MERM actin cytoskeletal proteins via a C-terminal MERM domain on NHERF-1. Specific Aim #3: Protein-protein interaction of the RAMPS with various other proteins is involved in the regulation of AM receptor trafficking from the sorting endosome. Hypothesis 1: RAMP3, but not RAMPI or RAMP2, interacts with NSF to target the receptor complex for recycling pathway after agonist-stimulated internalization. Hypothesis 2: RAMP3 interacts with NSF via a PDZ recognition motif on RAMP3. Results: 3. RAMP3 (but not RAMP] or RAMP2) interacts with NSF to target AM receptor complex for a recycling pathway after agonist-induced internalization. 207 b. RAMP3 was determined to interact with NSF via a PDZ recognition motif on the extreme C-terminus of RAMP3. 7.2. Limitations of the study. I. The primary limitation of this study is that all experiments were performed in cell culture system, as opposed to using a whole-animal model system. The practicality of defining detailed molecular mechanisms required the use of cell culture systems. The cell culture systems utilized that were derived from the rat may not necessarily reflect the same regulatory mechanisms in human cells. The rat cell lines were chosen because of the specific RAMP isoform expression levels. Obtaining and maintaining human cell lines, both from the standpoint of cost and labor, was not practical. All changes in receptor signaling were elicited by exogenous administration of AM. Concentrations of AM used were within the established circulating levels of AM, yet it remains unknown what concentrations mesangial cells and proximal tubule cells are exposed to in vivo. Until physiological and pathological concentrations are determined in the vicinity of these two cell types, no arguments can be made regarding this issue. Changes in receptor trafficking were characterized using cAMP accumulation as the endpoint. Measuring the role of receptor trafficking 208 changes in regulation of a more physiological parameter (such as proliferation or migration) may provide additional information. 5. Chemical and pharmacological inhibitors were used to examine the roles of NSF and protein kinases. Given the limited specificity of chemical and pharmacological inhibitors, the results from these studies should be interpreted with some degree of caution. Nonetheless, it should be noted that the concentrations used in the studies have been published by others and characterized as specific within that range. In addition, where possible, supplementary controls were tested. 7.3. Positive outcomes of the study. This thesis research sought to examine differences in AM receptor signaling, dictated by the co-expression of the different RAMP isoforms. The three individual studies within the project characterized roles for the RAMP isoforms in differentially regulating the post-translational modifications and protein-protein interactions that regulate AM receptor complex signaling. While phosphorylation of CL-R had been demonstrated with prolonged agonist exposure, this study was the first to report specific amino acids of the receptorcritical for desensitization and internalization. In addition, it was not previously known that association of CL-R with different RAMP isoforms generated different phosphorylation patterns on the receptor, regardless of exposure to the same ligand (AM). It could be envisioned that this differential regulation of the receptor, directed by the RAMP isoforms, allows further protein-protein interactions that regulate receptor signaling and 209 trafficking. Future research investigating the interaction of differentially phosphorylated AM receptor complexes with different arrestin molecules could clarify a role for phosphorylation in the targeting of the receptor complex after agonist stimulation. The last two studies defined a role for the RAMPS in protein-protein interactions that regulate the trafficking and life-cycle of the receptor complex. Interactions of NHERF-1 and NSF with RAMP3 have been demonstrated, in different ways, to promote AM receptor signaling. The major finding of this thesis research that could potentially contribute to the greater field of receptor biology is the concept of RAMP expression dictating the receptor fate after agonist-stimulated desensitization. In light of the recent report of RAMPs’ interaction with other GPCRS, our observations may be applicable to additional receptor systems. Future studies investigating the role of RAMPS in the trafficking of other GPCRS could help establish this idea of more widespread receptor regulation. Given the documented expression of RAMPS in tissues devoid of CL-R expression, the possibility of additional regulatory roles for the RAMPS has been proposed. This thesis research has defined novel roles for the RAMP proteins in regulation of the AM receptor signaling and endocytic trafficking. Considering the established beneficial and harmful effects of AM signaling in different physiological systems (cardiorenal disease vs. cancer, respectively), it is important to have specific mechanisms to promote or repress AM signaling. In review of this study, we believe differential RAMP isoform expression provides a form of this indispensable receptor regulation. 210 REFERENCES 211 10. 11. Kitamura, K., et al., Adrenomedullin: a novel hypotensive peptide isolated from human pheochromocytoma. Biochemical and Biophysical Research Communications, 1993. 192(2): p. 553-60. Kitamura, K., et al., Cloning and characterization of CDNA encoding a precursor for human adrenomedullin. Biochemical and Biophysical Research Communications, 1993. 194(2): p. 720-5. Imagawa, M., R. Chiu, and M. Karin, Transcription factor AP—2 mediates induction by two diflerent signal-transduction pathways: protein kinase C and cAMP. Cell, 1987. 51(2): p. 251-60. Fink, J.S., et al., The CGT CA sequence motif is essential for biological activity of the vasoactive intestinal peptide gene cAMP-regulated enhancer. Proc Natl Acad Sci U S A, 1988. 85(18): p. 6662-6. Ishimitsu, T., et al., Transcriptional regulation of human adrenomedullin gene in vascular endothelial cells. Biochem Biophys Res Commun, 1998. 243(2): p. 463- 70. Cormier-Regard, S., S.V. Nguyen, and WC. Claycomb, Adrenomedullin gene expression is developmentally regulated and induced by hypoxia in rat ventricular cardiac myocytes. J Biol Chem, 1998. 273(28): p. 17787-92. Garayoa, M., et al., Hypoxia-induciblefactor-I (HIF -1 ) up-regulates adrenomedullin expression in human tumor cell lines during oxygen deprivation: a possible promotion mechanism of carcinogenesis. Mol Endocrinol, 2000. 14(6): p. 848-62. Montuenga, L.M., eta1., Expression of proadrenomedullin derived peptides in the mammalian pituitary: co-localization of follicle stimulating hormone and proadrenomedullin N—20 terminal peptide-like peptide in the same secretory granules of the gonadotropes. J Neurocndocrinol, 2000. 12(7): p. 607-17. Kitamura, K., et al., Complete amino acid sequence of porcine adrenomedullin and cloning of CDNA encoding its precursor. FEBS Lett, 1994. 338(3): p. 306-10. Hinson, J.P., S. Kapas, and D.M. Smith, Adrenomedullin, a multifunctional regulatory peptide. Endocr Rev, 2000. 21(2): p. 138-67. Roh, J ., et al., Intermedin is a calcitonin/calcitonin gene-related peptide family peptide acting through the calcitonin receptor-like receptor/receptor activity- modifizing protein receptor complexes. J Biol Chem, 2004. 279(8): p. 7264-74. 212 12. 13. 14. 15. l6. 17. 18. 19. 20. 21. 22. 23. Kitamura, K., et al., T he intermediate form of glycine-extended adrenomedullin is the major circulating molecular form in human plasma. Biochem Biophys Res Commun, 1998. 244(2): p. 551-5. Eguchi, S., et al., Structure-activity relationship of adrenomedullin, a novel vasodilatory peptide, in cultured rat vascular smooth muscle cells. Endocrinology, 1994. 135(6): p. 2454-8. Chiba, T., et al., Calcitonin gene-related peptide receptor antagonist human CGRP-(8-3 7). Am J Physiol, 1989. 256(2 Pt 1): p. E331-5. Longmore, J., et al., Effects of two truncated forms of human calcitonin-gene related peptide: implications for receptor classification. Eur J Pharmacol, 1994. 265(1-2): p. 53-9. Doods, H., et al., Pharmacological profile of BIBN4096BS, the first selective small molecule CGRP antagonist. Br J Pharmacol, 2000. 129(3): p. 420-3. Edvinsson, L., eta1., Characterisation of the effects of a non-peptide C GRP receptor antagonist in SK-N-MC cells and isolated human cerebral arteries. Eur J Pharmacol, 2001. 415(1): p. 39-44. Aiyar, N., et al., Pharmacology of SB-2 73 7 79, a nonpeptide calcitonin gene- related peptide 1 receptor antagonist. J Pharmacol Exp Ther, 2001. 296(3): p. 768-75. Ichiki, Y., et al., Distribution and characterization of immunoreactive adrenomedullin in human tissue and plasma. FEBS Lett, 1994. 338(1): p. 6-10. Sakata, J ., et al., Distribution and characterization of immunoreactive rat adrenomedullin in tissue and plasma. FEBS Lett, 1994. 352(2): p. 105-8. Satoh, F., et al., Immunocytochemical localization of adrenomedullin-like immunoreactivity in the human hypothalamus and the adrenal gland. Neurosci Lett, 1996. 203(3): p. 207-IO. Martinez, A., F. Cuttitta, and G. Teitelman, Expression pattern for adrenomedullin during pancreatic development in the rat reveals a common precursor with other endocrine cell types. Cell Tissue Res, 1998. 293(1): p. 95- 100. Martinez, A., et al., Regulation of insulin secretion and blood glucose metabolism by adrenomedullin. Endocrinology, 1996. 137(6): p. 2626-32. 213 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. Martinez, A., eta1., Expression of adrenomedullin and its receptor in normal and malignant human skin: a potential pluripotent role in the integument. Endocrinology, 1997. 138(12): p. 5597-604. Sugo, S., et al., Interleukin-l, tumor necrosis factor and lipopolysaccharide additively stimulate production of adrenomedullin in vascular smooth muscle cells. Biochem Biophys Res Commun, 1995. 207(1): p. 25-32. Sugo, S., eta1., Production and secretion of adrenomedullin from vascular smooth muscle cells: augmented production by tumor necrosis factor-alpha. Biochem Biophys Res Commun, 1994. 203(1): p. 719-26. Sugo, S., et al., Endothelial cells actively synthesize and secrete adrenomedullin. Biochem Biophys Res Commun, 1994. 201(3): p. 1160-6. Lai, K.N., et al., Gene transcription and synthesis of adrenomedullin by cultured human renal cells. Biochem Biophys Res Commun, 1998. 244(2): p. 567-72. Michibata, H., et al., Autocrine/paracrine role of adrenomedullin in cultured endothelial and mesangial cells. Kidney Int, 1998. 53(4): p. 979-85. Owada, A., et al., Microlocalization and effects of adrenomedullin in nephron segments and in mesangial cells of the rat. Am J Physiol, 1997. 272(6 Pt 2): p. F691-7. Sato, K., et al., Secretion of adrenomedullin by renal tubular cell lines. Nephron, 1998. 78(1): p. 9-14. Iougasaki, M., et al., Renal localization and actions of adrenomedullin: a natriuretic peptide. Am J Physiol, 1995. 268(4 Pt 2): p. F 657-63. Asada, Y., et al., Novel distribution of adrenomedullin-immunoreactive cells in human tissues. Histochem Cell Biol, 1999. 112(3): p. 185-9]. Martinez, A., et al., Expression of adrenomedullin in normal human lung and in pulmonary tumors. Endocrinology, 1995. 136(9): p. 4099-105. Kitamuro, T., et al., Induction of adrenomedullin during hypoxia in cultured human glioblastoma cells. J Neurochem, 2000. 75(5): p. 1826-33. Nakayama, M., eta1., Induction of adrenomedullin by hypoxia and cobalt chloride in human colorectal carcinoma cells. Biochem Biophys Res Commun, 1998. 243(2): p. 514-7. 214 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. Takahashi, K., et al., Production and secretion of two vasoactive peptides, adrenomedullin and endothelin-I, by cultured human adrenocortical carcinoma cells. Peptides, 2000. 21(2): p. 251-6. Guevara, M., et al., Increased adrenomedullin levels in cirrhosis: relationship with hemodynamic abnormalities and vasoconstrictor systems. Gastroenterology, 1998. 114(2): p. 336-43. Hjelmqvist, H., et al., Vasodilation and glomerular binding of adrenomedullin in rabbit kidney are not CGRP receptor mediated. Am J Physiol, 1997. 273(2 Pt 2): p. R716-24. Parkes, D.G. and C.N. May, Direct cardiac and vascular actions of adrenomedullin in conscious sheep. Br J Pharmacol, 1997. 120(6): p. 1179-85. Nakamura, M., et al., Potent and long-lasting vasodilatory effects of adrenomedullin in humans. Comparisons between normal subjects and patients with chronic heart failure. Circulation, 1997. 95(5): p. 1214-21. Entzeroth, M., et al., Adrenomedullin mediates vasodilation via C GRPI receptors. Life Sci, 1995. 56(1): p. PL19-25. Chang, C.P., et al., Identification of a seven transmembrane helix receptor for corticotropin-releasing factor and sauvagine in mammalian brain. Neuron, 1993. 11(6): p. 1187-95. Feng, C.J., et al., L-NAME modulates responses to adrenomedullin in the hindquarters vascular bed of the rat. Life Sci, 1994. 55(22): p. PL433-8. Hirata, Y., et al., Mechanisms of adrenomedullin-induced vasodilation in the rat kidney. Hypertension, 1995. 25(4 Pt 2): p. 790-5. Miura, K., et al., Attenuation of adrenomedullin-induced renal vasodilatation by NG-nitro L-arginine but not glibenclamide. Br J Pharmacol, 1995. 115(6): p. 917- 24. Nossaman, B.D., et al., Pulmonary vasodilator responses to adrenomedullin are reduced by NOS inhibitors in rats but not in cats. Am J Physiol, 1996. 270(5 Pt I): p. L782-9. Champion, H.C., et al., Comparison of responses to rat and human adrenomedullin in the hindlimb vascular bed of the cat. Regul Pept, 1997. 70(2- 3): p. 161-5. 215 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. Barber, D.A., et al., Adrenomedullin-mediated relaxations in veins are endothelium-dependent and distinct from arteries. J Cardiovasc Pharmacol, 1997. 30(5): p. 695-701. Majid, D.S., et al., Renal responses to intra-arterial administration of adrenomedullin in dogs. Am J Physiol, 1996. 270(1 Pt 2): p. F200-5. Miller, M.J., eta1., Adrenomedullin expression in human tumor cell lines. Its potential role as an autocrine growth factor. J Biol Chem, 1996. 271(38): p. 23345-51. Withers, D.J., et al., Adrenomedullin stimulates DNA synthesis and cell proliferation via elevation of cAMP in Swiss 3 T 3 cells. FEBS Lett, 1996. 378(1): p. 83-7. Kapas, S., et al., Adrenomedullin has mitogenic effects on human oral keratinocytes: involvement of cyclic AMP. FEBS Lett, 1997. 418(3): p. 287-90. Wang, H., et al., Sequential expression of adrenomedullin and its receptor during gastric ulcer healing in rats. Dig Dis Sci, 2000. 45(3): p. 591-8. Udono, T., et al., Adrenomedullin in cultured human retinal pigment epithelial cells. Invest Ophthalmol Vis Sci, 2000. 41(7): p. 1962-70. Tsuruda, T., et al., An autocrine or a paracrine role of adrenomedullin in modulating cardiac fibroblast growth. Cardiovasc Res, 1999. 43(4): p. 958-67. Tsuruda, T., et al., Adrenomedullin: a possible autocrine or paracrine inhibitor of hypertrophy of cardiomyocytes. Hypertension, 1998. 31(1 Pt 2): p. 505-10. Parameswaran, N., et al., Regulation of glomerular mesangial cell proliferation in culture by adrenomedullin. Eur J Pharmacol, 1999. 372(1): p. 85-95. Kano, H., et al., Adrenomedullin as a novel antiproliferative factor of vascular smooth muscle cells. J Hypertens, 1996. 14(2): p. 209-l3. Jensen, B.L., et al., Eflects of dietary salt on adrenomedullin and its receptor mRNAs in rat kidney. Am J Physiol, 1998. 275(1 Pt 2): p. F55-61. Ebara, T., eta1., Effect of adrenomedullin on renal hemodynamics and functions in dogs. Eur J Pharmacol, 1994. 263(1-2): p. 69-73. Vari, R.C., S.D. Adkins, and WK. Samson, Renal effects of adrenomedullin in the rat. Proc Soc Exp Biol Med, 1996. 211(2): p. 178-83. 216 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. Leclerc, M. and MG. Brunette, The paradoxical eflect of adrenomedullin on Na+ transport by the renal distal tubule luminal membrane. Mol Cell Endocrinol, 2000. 164(1-2): p. 159-67. Yang, B.C., et al., Adrenomedullin dilates rat pulmonary artery rings during hypoxia: role of nitric oxide and vasodilator prostaglandins. J Cardiovasc Pharmacol, 1996. 28(3): p. 458-62. Szokodi, I., et al., Evidence for cAMP-independent mechanisms mediating the eflects of adrenomedullin, a new inotropic peptide. Circulation, 1998. 97(11): p. 1062-70. He, H., et al., Eflects of a synthetic rat adrenomedullin on regional hemodynamics in rats. Eur J Pharmacol, 1995. 273(3): p. 209-14. Kureishi, Y., eta1., Adrenomedullin decreases both cytosolic Ca2+ concentration and Ca(2+)-sensitivity in pig coronary arterial smooth muscle. Biochem Biophys Res Commun, 1995. 212(2): p. 572-9. Elhawary, A.M., J. Poon, and CC. Pang, Effects of calcitonin gene-related peptide receptor antagonists on renal actions of adrenomedullin. Br J Pharmacol, 1995. 115(7): p. 1133-40. Nagaya, N., et al., Cardiovascular and renal effects of adrenomedullin in rats with heart failure. Am J Physiol, 1999. 276(1 Pt 2): p. R213-8. Togawa, M., et al., Beraprost sodium, an analogue of prostacyclin, induces the expression of mitogen-activated protein kinase phosphatase and inhibits the proliferation of cultured mesangial cells. Eur J Pharmacol, 1997. 336(2-3): p. 291-4. Segawa, K., et al., Inhibitory effect of adrenomedullin on rat mesangial cell mitogenesis. Nephron, 1996. 74(3): p. 577-9. Jensen, B.L., B.K. Kramer, and A. Kurtz, Adrenomedullin stimulates renin release and renin mRNA in mouse juxtaglomerular granular cells. Hypertension, 1997. 29(5): p. 1148-55. Cornish, J., et al., Adrenomedullin is a potent stimulator of osteoblastic activity in vitro and in vivo. Am J Physiol, 1997. 273(6 Pt 1): p. E] 1 13-20. Cornish, J ., eta1., Systemic administration of adrenomedullin(2 7-52) increases bone volume and strength in male mice. J Endocrinol, 2001. 170(1): p. 251-7. 217 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. Samson, W.K., T. Murphy, and DA. Schell, A novel vasoactive peptide, adrenomedullin, inhibits pituitary adrenocorticotropin release. Endocrinology, 1995. 136(5): p. 2349-52. Parkes, D.G. and C.N. May, AC T H-suppressive and vasodilator actions of adrenomedullin in conscious sheep. J Neurocndocrinol, 1995. 7(12): p. 923-9. Yamaguchi, T., et al., Inhibition of aldosterone production by adrenomedullin, a hypotensive peptide, in the rat. Hypertension, 1996. 28(2): p. 308-14. Saita, M., et al., Central actions of adrenomedullin on cardiovascular parameters and sympathetic outflow in conscious rats. Am J Physiol, 1998. 274(4 Pt 2): p. R979-84. Samson, W.K. and TC. Murphy, Adrenomedullin inhibits salt appetite. Endocrinology, 1997. 138(2): p. 613-6. Murphy, TC. and WK. Samson, The novel vasoactive hormone, adrenomedullin, inhibits water drinking in the rat. Endocrinology, 1995. 136(6): p. 2459-63. Taylor, G.M., eta1., Adrenomedullin inhibits feeding in the rat by a mechanism involving calcitonin gene-related peptide receptors. Endocrinology, 1996. 137(8): p. 3260-4. Yokoi, H., et al., Intracerebroventricular injection of adrenomedullin inhibits vasopressin release in conscious rats. Neurosci Lett, 1996. 216(1): p. 65-7. Kohno, M., et al., Plasma adrenomedullin concentrations in essential hypertension. Hypertension, 1996. 27(1): p. 102-7. Shimokubo, T., et al., Adrenomedullin: changes in circulating and cardiac tissue concentration in Dahl salt-sensitive rats on a high-salt diet. Clin Exp Hypertens, 1996. 18(7): p. 949-6]. Nishikimi, T., et al., Role of increased circulating and renal adrenomedullin in rats with malignant hypertension. Am J Physiol Regul Integr Comp Physiol, 2001. 281(6): p. R2079-87. Tadokoro, K., et al., Altered gene expression of adrenomedullin and its receptor system and molecular forms of tissue adrenomedullin in left ventricular hypertrophy induced by malignant hypertension. Regul Pept, 2003. 112(1-3): p. 71-8. Wang, X., et al., Upregulation of ligand, receptor system, and amidating activity of adrenomedullin in left ventricular hypertrophy of severely hypertensive rats: 218 88. 89. 90. 9]. 92. 93. 94. 95. 96. 97. 98. 99. eflects of angiotensin-converting enzyme inhibitors and diuretic. J Hypertens, 2003. 21(6): p. 1171-8]. Hirayama, N., et al., Molecular forms of circulating adrenomedullin in patients with congestive heart failure. J Endocrinol, 1999. 160(2): p. 297-303. Yu, C.M., et al., Increase in plasma adrenomedullin in patients with heart failure characterised by diastolic dysfunction. Heart, 2001. 86(2): p. 155-60. Jougasaki, M., et al., Elevation of circulating and ventricular adrenomedullin in human congestive heart failure. Circulation, 1995. 92(3): p. 286-9. Cueille, C., et al., Increased myocardial expression of RAMP] and RAMP3 in rats with chronic heart failure. Biochem Biophys Res Commun, 2002. 294(2): p. 340- 6. Nishikimi, T., et al., Cardiac production and secretion of adrenomedullin are increased in heart failure. Hypertension, 1997. 30(6): p. 1369-75. Nishikimi, T., et al., Ventricular adrenomedullin system in the transition from LVH to heart failure in rats. Hypertension, 2003. 41(3): p. 512-8. Totsune, K., et al., Increased gene expression of adrenomedullin and adrenomedullin—receptor complexes, receptor-activity modifying protein (RAMP)2 and calcitonin-receptor-like receptor (CRLR) in the hearts of rats with congestive heart failure. Clin Sci (Lond), 2000. 99(6): p. 541-6. Yoshihara, F., et al., Ventricular adrenomedullin concentration is a sensitive biochemical marker for volume and pressure overload in rats. Am J Physiol Heart Circ Physiol, 2000. 278(2): p. H633-42. Nagaya, N., et al., Hemodynamic, renal, and hormonal effects of adrenomedullin infusion in patients with congestive heart failure. Circulation, 2000. 101(5): p. 498-503. J ougasaki, M., et al., Attenuated natriuretic response to adrenomedullin in experimental heart failure. J Card Fail, 2001. 7(1): p. 75-83. Shinomiya, K., et al., Association of plasma adrenomedullin with carotid atherosclerosis in chronic ischemic stroke. Peptides, 2001. 22(11): p. 1873-80. Nakayama, M., et al., Adrenomedullin in monocytes and macrophages: possible involvement of macrophage-derived adrenomedullin in atherogenesis. Clin Sci (Lond), 1999. 97(2): p. 247-5]. 219 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. Beltowski, J. and A. Jarnroz, Adrenomedullin--what do we know 10 years since its discovery? Pol J Pharmacol, 2004. 56(1): p. 5-27. Agata, J ., et al., Adrenomedullin gene delivery inhibits neointima formation in rat artery after balloon angioplasty. Regul Pept, 2003. 112(1-3): p. 115-20. Ando, K. and T. Fujita, Lessons from the adrenomedullin knockout mouse. Regul Pept, 2003. 112(1-3): p. 185-8. Yamasaki, M., et al., Adrenomedullin overexpression to inhibit cuff-induced arterial intimal formation. Hypertension, 2003. 41(2): p. 302-7. Kobayashi, K., et al., Increased plasma adrenomedullin in acute myocardial infarction. Am Heart J, 1996. 131(4): p. 676-80. Miyao, Y., et al., Increased plasma adrenomedullin levels in patients with acute myocardial infarction in proportion to the clinical severity. Heart, 1998. 79(1): p. 39-44. Nagaya, N., et al., Cardiac adrenomedullin gene expression and peptide accumulation after acute myocardial infarction in rats. Am J Physiol Regul Integr Comp Physiol, 2000. 278(4): p. R1019-26. Oie, E., et al., Induction of a myocardial adrenomedullin signaling system during ischemic heart failure in rats. Circulation, 2000. 101(4): p. 415-22. Yoshihara, F., et al., Possible involvement of oxidative stress in hypoxia-induced adrenomedullin secretion in cultured rat cardiomyocytes. Eur J Pharmacol, 2002. 436(1-2): p. 1-6. Nishikimi, T., et al., Molecular forms of adrenomedullin in pericardial fluid and plasma in patients with ischaemic heart disease. Clin Sci (Lond), 2002. 102(6): p. 669-77. Nayeem, MA. and SJ. Mustafa, Mechanisms of delayed preconditioning with A1 adenosine receptor activation in porcine coronary smooth muscle cells. Pol J Pharmacol, 2002. 54(5): p. 443-53. Chao, J ., et al., Human adrenomedullin gene delivery protects against cardiovascular remodeling and renal injury. Peptides, 2001. 22(11): p. 1731-7. Nakamura, K, eta1., Beneficial effects of adrenomedullin on left ventricular remodeling after myocardial infarction in rats. Cardiovasc Res, 2002. 56(3): p. 373-80. 220 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. Kinoshita, H., et al., Increased plasma levels of mature adrenomedullin in chronic glomerulonephritis. Nephron, 2000. 86(3): p. 333-8. Kubo, A., et al., Plasma and urinary levels of adrenomedullin in chronic glomerulonephritis patients with proteinuria. Nephron, 1998. 80(2): p. 227-30. Nagata, D., et al., Hypoxia-induced adrenomedullin production in the kidney. Kidney Int, 1999. 55(4): p. 1259-67. Nishimatsu, H., et al., Role of endogenous adrenomedullin in the regulation of vascular tone and ischemic renal injury: studies on transgenic/knockout mice of adrenomedullin gene. Circ Res, 2002. 90(6): p. 657-63. Caron, KM. and O. Smithies, Extreme hydropsfetalis and cardiovascular abnormalities in mice lacking a functional Adrenomedullin gene. Proc Natl Acad Sci U s A, 2001. 98(2): p. 615-9. Shindo, T., et al., Vascular abnormalities and elevated blood pressure in mice lacking adrenomedullin gene. Circulation, 2001. 104(16): p. 1964-71. Shimosawa, T., et al., Adrenomedullin, an endogenous peptide, counteracts cardiovascular damage. Circulation, 2002. 105(1): p. 106-11. Nuki, C., et al., Vasodilator effect of adrenomedullin and calcitonin gene-related peptide receptors in rat mesenteric vascular beds. Biochem Biophys Res Commun, 1993. 196(1): p. 245-5]. Perret, M., et al., The eflect of adrenomedullin on the isolated heart. Life Sci, 1993. 53(22): p. PL377-9. Hall, J .M., eta1., Interaction of human adrenomedullin 13-52 with calcitonin gene-related peptide receptors in the microvasculature of the rat and hamster. Br J Pharmacol, 1995. 114(3): p. 592-7. Heaton, J ., et al., Pulmonary vasodilation to adrenomedullin: a novel peptide in humans. Am J Physiol, 1995. 268(6 Pt 2): p. H2211-5. Champion, H.C., et al., [MPR14]-rADM(14-50), a novel analog of adrenomedullin, possesses potent vasodilator activity in the hindlimb vascular bed of the cat. Life Sci, 1996. 59(1): p. PL1-7. Pinto, A., et al., Effects of adrenomedullin and calcitonin gene-related peptide on airway and pulmonary vascular smooth muscle in guinea-pigs. Br J Pharmacol, 1996. 119(7): p. 1477-83. 221 126. 127. 128. 129. 130. 131. 132. 133. 134. 135. 136. 137. I38. Takahashi, Y., et al., Mechanisms of adrenomedullin-induced increase of pulmonary blood flow in fetal sheep. Pediatr Res, 1999. 45(2): p. 276-81. Osajima, A., et al., Adrenomedullin-sensitive receptors are preferentially expressed in cultured rat mesangial cells. Eur J Pharmacol, 1996. 315(3): p. 319- 25. Kapas, S., K.J. Catt, and A.J. Clark, Cloning and expression of CDNA encoding a rat adrenomedullin receptor. J Biol Chem, 1995. 270(43): p. 25344-7. Harrison, J .K., C.M. Barber, and KR. Lynch, Molecular cloning of a novel rat G- protein-coupled receptor gene expressed prominently in lung, adrenal, and liver. FEBS Lett, 1993. 318(1): p. 17-22. Eva, C. and R. Sprengel, A novel putative G protein-coupled receptor highly expressed in lung and testis. DNA Cell Biol, 1993. 12(5): p. 393-9. Kapas, S. and A.J. Clark, Identification of an orphan receptor gene as a type I calcitonin gene-related peptide receptor. Biochem Biophys Res Commun, 1995. 217(3): p. 832-8. Chakravarty, P., et al., C GRP and adrenomedullin binding correlates with transcript levels for calcitonin receptor-like receptor (CRLR) and receptor activity modijying proteins (RAMPS) in rat tissues. Br J Pharmacol, 2000. 130(1): p. 189-95. Kennedy, S.P., et al., Expression of the rat adrenomedullin receptor or a putative human adrenomedullin receptor does not correlate with adrenomedullin binding or functional response. Biochem Biophys Res Commun, 1998. 244(3): p. 832-7. McLatchie, L.M., et al., RAMPS regulate the transport and ligand specificity of the calcitonin-receptor-like receptor. Nature, 1998. 393(6683): p. 333-9. Aiyar, N., et al., A CDNA encoding the calcitonin gene-related peptide type I receptor. J Biol Chem, 1996. 271(19): p. 11325-9. Elshourbagy, N.A., et al., Molecular cloning and characterization of the porcine calcitonin gene-related peptide receptor. Endocrinology, 1998. 139(4): p. 1678- 83. Han, Z.Q., et al., The interaction of CGRP and adrenomedullin with a receptor expressed in the rat pulmonary vascular endothelium. J Mol Endocrinol, 1997. 18(3): p. 267-72. Njuki, F., et al., A new calcitonin-receptor—like sequence in rat pulmonary blood vessels. Clin Sci (Lond), 1993. 85(4): p. 385-8. 222 139. 140. 141. 142. 143. 144. 145. 146. 147. 148. 149. 150. Fluhmann, B., et al., A human orphan calcitonin receptor-like structure. Biochem Biophys Res Commun, 1995. 206(1): p. 341-7. Zumpe, E.T., et al., Multiple ramp domains are required for generation of amylin receptor phenotype from the calcitonin receptor gene product. Biochem Biophys Res Commun, 2000. 267(1): p. 368-72. Aldecoa, A., et al., Mammalian calcitonin receptor-like receptor/receptor activity modifying protein complexes define calcitonin gene-related peptide and adrenomedullin receptors in Drosophila Schneider 2 cells. FEBS Lett, 2000. 471(2-3): p. 156-60. Buhlmann, N., et al., A receptor activity modijying protein (RAMP)2—dependent adrenomedullin receptor is a calcitonin gene-related peptide receptor when coexpressed with human RAMP]. Endocrinology, 1999. 140(6): p. 2883-90. Kamitani, S., eta1., The RAMP2/CRLR complex is a functional adrenomedullin receptor in human endothelial and vascular smooth muscle cells. FEBS Lett, 1999. 448(1): p. 111-4. Fraser, N.J., et al., The amino terminus of receptor activity modifying proteins is a critical determinant of glycosylation state and ligand binding of calcitonin receptor-like receptor. Mol Pharmacol, 1999. 55(6): p. 1054-9. Nowak, W., Regulation of Receptor Activity Modifying Protein Expression as a Novel Mechanism for Modulation of Adrenomedullin Activity in Rat Mesangial Cells. Doctoral Dissertation, 2003. Nowak, W., et al., Novel regulation of adrenomedullin receptor by PDGF: role of receptor activity modifi/ing protein-3. Am J Physiol Cell Physiol, 2002. 282(6): p. C1322-31. Hay, UL. and D.M. Smith, Knockouts and transgenics confirm the importance of adrenomedullin in the vasculature. Trends Pharmacol Sci, 2001. 22(2): p. 57-9. Sexton, P.M., et al., Receptor activity modifying proteins. Cell Signal, 2001. 13(2): p. 73-83. Husmann, K., et al., Mouse receptor-activity—modijying proteins 1, -2 and -3: amino acid sequence, expression and function. Mol Cell Endocrinol, 2000. 162(1- 2): p. 35-43. Nagae, T., et al., Rat receptor-activity-modijying proteins (RAMPS) for adrenomedullin/CGRP receptor: cloning and upregulation in obstructive nephropathy. Biochem Biophys Res Commun, 2000. 270(1): p. 89-93. 223 151. 152. 153. 154. 155. 156. 157. 158. 159. 160. I61. 162. Muff, R., W. Born, and J .A. Fischer, Adrenomedullin and related peptides: receptors and accessory proteins. Peptides, 2001. 22(11): p. 1765-72. Hall, J.M. and D.M. Smith, Calcitonin gene-relatedpeptide--a new concept in receptor-ligand specificity? Trends Pharmacol Sci, 1998. 19(8): p. 303-5. Bomberger, J .M., et al., Novel function for receptor activity modifiring proteins (RAMPS) in post-endocytic receptor traflicking. J Biol Chem, 2004. Hall, R.A., eta1., A C-terminal motif found in the beta2-adrenergic receptor, P2 Y1 receptor and cystic fibrosis transmembrane conductance regulator determines binding to the Na +/H+ exchanger regulatory factor family of PDZ proteins. Proc Natl Acad Sci U S A, 1998. 95(15): p. 8496-501. Ueda, T., et al., Expression of receptor-activity modifying protein (RAMP) mRNAs in the mouse brain. Brain Res Mol Brain Res, 2001. 93(1): p. 36-45. Ono, Y., et al., Decreased gene expression of adrenomedullin receptor in mouse lungs during sepsis. Biochem Biophys Res Commun, 2000. 271(1): p. 197-202. Muff, R., et al., Receptor activity modijying proteins regulate the activity of a calcitonin gene-related peptide receptor in rabbit aortic endothelial cells. FEBS Lett, 1998. 441(3): p. 366-8. Oliver, K.R., et al., Cloning, characterization and central nervous system distribution of receptor activity modifying proteins in the rat. Eur J Neurosci, 2001. 14(4): p. 618-28. Yoshihara, F., eta1., Alterations of intrarenal adrenomedullin and its receptor system in heart failure rats. Hypertension, 2001. 37(2): p. 216-22. Totsune, K., eta1., Adrenomedullin and its receptor complexes in remnant kidneys of rats with renal mass ablation: decreased expression of calcitonin receptor-like receptor and receptor-activity modifying protein-3. Peptides, 2001. 22(1 1): p. 1933-7. Christopoulos, G., et al., Multiple amylin receptors arise from receptor activity- modijying protein interaction with the calcitonin receptor gene product. Mol Pharmacol, 1999. 56(1): p. 235-42. Kuwasako, K., et al., Visualization of the calcitonin receptor-like receptor and its receptor activity-modifying proteins during internalization and recycling. J Biol Chem, 2000. 275(38): p. 29602-9. 224 163. 164. 165. 166. 167. 168. 169. 170. 171. 172. 173. Hilairet, S., et al., Protein-protein interaction and not glycosylation determines the binding selectivity of heterodimers between the calcitonin receptor-like receptor and the receptor activity-modifying proteins. J Biol Chem, 2001. 276(31): p. 29575-81. Hilairet, S., et al., Agonist-promoted internalization of a ternary complex between calcitonin receptor-like receptor, receptor activity-modifying protein 1 (RAMPI), and beta-arrestin. J Biol Chem, 2001. 276(45): p. 42182-90. F itzsimmons, T.J., X. Zhao, and SA. Wank, The extracellular domain of receptor activity-modifying protein 1 is sufficient for calcitonin receptor-like receptor function. J Biol Chem, 2003. 278(16): p. 14313-20. Steiner, S., et al., The transmembrane domain of receptor-activity-modijying protein 1 is essential for the functional expression of a calcitonin gene-related peptide receptor. Biochemistry, 2002. 41(38): p. 11398-404. Kuwasako, K., eta1., Identification of the human receptor activity-modifiing protein 1 domains responsible for agonist binding specificity. J Biol Chem, 2003. 278(25): p. 22623-30. Udawela, M., D.L. Hay, and PM. Sexton, The receptor activity modifiting protein family of G protein coupled receptor accessory proteins. Semin Cell Dev Biol, 2004. 15(3): p. 299-308. Kuwasako, K., et al., The seven amino acids of human RAMP2 (86) and RAMP3 (59) are critical for agonist binding to human adrenomedullin receptors. J Biol Chem, 2001. 276(52): p. 49459-65. Flahaut, M., et al., N-Glycosylation and conserved cysteine residues in RAMP3 play a critical role for the functional expression of CRLR/RAMP3 adrenomedullin receptor. Biochemistry, 2003. 42(34): p. 10333-41. Sexton, P.M., D.M. Findlay, and T]. Martin, Calcitonin. Curr Med Chem, 1999. 6(11): p. 1067-93. Muff, R., et al., An amylin receptor is revealed following co-transfection of a calcitonin receptor with receptor activity modifying proteins-I or -3. Endocrinology, 1999. 140(6): p. 2924-7. Tilakaratne, N., et al., Amylin receptor phenotypes derived from human calcitonin receptor/RAMP coexpression exhibit pharmacological differences dependent on receptor isoform and host cell environment. J Pharmacol Exp Ther, 2000. 294(1): p. 61-72. 225 174. 175. 176. I77. 178. 179. 180. 181. 182. 183. 184. 185. Christopoulos, A., et al., Novel receptor partners and function of receptor activity-modifying proteins. J Biol Chem, 2003. 278(5): p. 3293-7. Qi, Y.F., et al., Changes of adrenomedullin and receptor activity modifying protein 2 (RAMP2) in myocardium and aorta in rats with isoproterenol—induced myocardial ischemia. Peptides, 2003. 24(3): p. 463-8. Stachniak, T.J. and TL. Krukoff, Receptor activity modifying protein 2 distribution in the rat central nervous system and regulation by changes in blood pressure. J Neurocndocrinol, 2003. 15(9): p. 840-50. Cao, Y.N., et al., Chronic salt loading upregulates expression of adrenomedullin and its receptors in adrenal glands and kidneys of the rat. Hypertension, 2003. 42(3): p. 369-72. Mishima, K., et al., Angiotensin II modulates gene expression of adrenomedullin receptor components in rat cardiomyocytes. Life Sci, 2003. 73(13): p. 1629-35. Mishima, K., et al., Effects of endothelin on adrenomedullin secretion and expression of adrenomedullin receptors in rat cardiomyocytes. Biochem Biophys Res Commun, 2001. 287(1): p. 264-9. Qing, X., J. Svaren, and I.M. Keith, mRNA expression of novel CGRP] receptors and their activity-modifying proteins in hypoxic rat lung. Am J Physiol Lung Cell Mol Physiol, 2001. 280(3): p. L547-54. Kitamuro, T., et al., Differential expression of adrenomedullin and its receptor component, receptor activity modijying protein (RAMP) 2 during hypoxia in cultured human neuroblastoma cells. Peptides, 2001. 22(11): p. 1795-801. Mukoyama, M., et al., Role of adrenomedullin and its receptor system in renal pathophysiology. Peptides, 2001. 22(1 1): p. 1925-31. Luebke, A.E., eta1., Identification of a protein that confers calcitonin gene- related peptide responsiveness to oocytes by using a cystic fibrosis transmembrane conductance regulator assay. Proc Natl Acad Sci U S A, 1996. 93(8): p. 3455-60. Rosenblatt, M.I., G.P. Dahl, and I.M. Dickerson, Characterization and localization of the rabbit ocular calcitonin gene-related peptide (C GRP)-receptor component protein (RCP). Invest Ophthalmol Vis Sci, 2000. 41(5): p. 1159-67. Naghashpour, M., et al., Inhibitory eflect of calcitonin gene-related peptide on myometrial contractility is diminished at parturition. Endocrinology, 1997. 138(10): p. 4207-14. 226 186. 187. 188. 189. 190. 191. 192. 193. 194. 195. 196. 197. Evans, B.N., et al., C GRP-RCP, a novel protein required for signal transduction at calcitonin gene-related peptide and adrenomedullin receptors. J Biol Chem, 2000. 275(40): p. 31438-43. Prado, M.A., et al., The role of the CGRP-receptor component protein (RCP) in adrenomedullin receptor signal transduction. Peptides, 2001. 22(11): p. 1773-81. Yu, Y., T. Lundeberg, and LC. Yu, Role of calcitonin gene-related peptide and its antagonist on the evoked discharge frequency of wide dynamic range neurons in the dorsal horn of the spinal cord in rats. Regul Pept, 2002. 103(1): p. 23-7. Pokabla, M.I., I.M. Dickerson, and RE. Papka, Calcitonin gene-relatedpeptide- receptor component protein expression in the uterine cervix, lumbosacral spinal cord, and dorsal root ganglia. Peptides, 2002. 23(3): p. 507-14. Dickerson, I.M. and SC. Loiseau, CGRP-Receptor: A malt-protein complex required for G protein-coupled signal transduction. Fifth Annual Great Lakes GPCR Retreat and, 2004. Leflcowitz, R.J., The superfamily of heptahelical receptors. Nature Cell Biology, 2000. 2: p. E133-E136. Pierce, K.L., L.M. Lutrell, and R.J. Lefltowitz, New mechanisms in heptahelical receptor signaling to mitogen activated protein kinase cascades. Oncogene, 2001. 20: p. 1532-1539. Pierce, K.L., R.T. Premont, and R]. Leflcowitz, Seven-transmembrane receptors. Nat Rev Mol Cell Biol, 2002. 3(9): p. 639-50. Krupnick, J .G. and J .L. Benovic, The role of receptor kinases and arrestins in G protein-coupled receptor regulation. Annu Rev Pharmacol Toxicol, 1998. 38: p. 289-319. Ferguson, S.S., et al., Molecular mechanisms of G protein-coupled receptor desensitization and resensitization. Life Sci, 1998. 62(17-18): p. 1561-5. Vazquez-Prado, J., P. Casas-Gonzalez, and J .A. Garcia-Sainz, G protein-coupled receptor cross-talk: pivotal roles of protein phosphorylation and protein-protein interactions. Cell Signal, 2003. 15(6): p. 549-57. Bohm, S.K., E.F. Grady, and NW. Bunnett, Regulatory mechanisms that modulate signalling by G protein-coupled receptors. Biochem J, 1997. 322: p. 1- 18. 227 198. 199. 200. 201. 202. 203. 204. 205. 206. 207. 208. 209. 210. Parameswaran, N., et al., Desensitization and resensitization of adrenomedullin- sensitive receptor in rat mesangial cells. European Journal of Pharmacology, 2000. 407: p. 205-210. Qi, Y.F., eta1., Desensitization of adrenomedullin and calcitonin gene-related peptide receptors in vascular smooth muscle cells-effects of receptor activity- modifying protein. Sheng Li Xue Bao, 2001. 53(5): p. 396-400. Drake, W.M., et al., Desensitization of C GRP and adrenomedullin receptors in SK-N-MC cells: implications for the RAMP hypothesis. Endocrinology, 1999. 140(1): p. 533-7. Aiyar, N., et al., Involvement of G protein-coupled receptor kinase-6 in desensitization of CGRP receptors. European Journal of Pharmacology, 2000. 403(1-2): p. 1-7. Hilairet, S., eta1., Agonist-promoted Intemalization of a Ternary Complex between Calcitonin Receptor-like Receptor, Receptor Activity Modifying Proteins (RAMPI), and Beta-Arrestin. J Biol Chem, 2001. 276(45): p. 42182-42190. Iaccarino, G., R]. Lefltowitz, and W.J. Koch, Myocardial G protein-coupled receptor kinases: implications for heart failure therapy. Proc Assoc Am Physicians, 1999. 111(5): p. 399-405. Koch, W.J., et al., Cardiac function in mice overexpressing the beta-adrenergic receptor kinase or a beta ARK inhibitor. Science, 1995. 268(5215): p. 1350-1353. Bohn, L.M., et al., Mu-opioid receptor desensitization by beta-arrestin-Z determines morphine tolerance but not dependence. Nature, 2000. 408(6813): p. 720-3. Whistler, J .L., et al., Functional dissociation of mu opioid receptor signaling and endocytosis: implications for the biology of opiate tolerance and addiction. Neuron, 1999. 23(4): p. 737-46. Maxfield, F.R. and TE. McGraw, Endocytic recycling. Nat Rev Mol Cell Biol, 2004. 5(2): p. 121-32. Kurten, R.C., Sorting motifs in receptor traflicking. Adv Drug Deliv Rev, 2003. 55(11): p. 1405-19. Hunyady, L., et al., Mechanisms and functions of A T(1) angiotensin receptor internalization. Regul Pept, 2000. 91(1-3): p. 29-44. Thomas, W.G. and H. Qian, Arresting angiotensin type 1 receptors. Trends Endocrinol Metab, 2003. 14(3): p. 130-6. 228 211. 212. 213. 214. 215. 216. 217. 218. 219. 220. 221. 222. 223. Ferguson, S.S., Evolving concepts in G protein-coupled receptor endocytosis: the role in receptor desensitization and signaling. Pharmacol Rev, 2001. 53(1): p. 1- 24. Conner, SD. and S.L. Schmid, Regulated portals of entry into the cell. Nature, 2003. 422(6927): p. 37-44. Johannes, L. and C. Lamaze, Clathrin-dependent or not: is it still the question? Traffic, 2002. 3(7): p. 443-51. Roettger, B.F., et al., Dual pathways of internalization of the Cholecystokinin receptor. J Cell Biol, 1995. 128(6): p. 1029-4]. Rapacciuolo, A., et al., Protein kinase A and G protein-coupled receptor kinase phosphorylation mediates beta-I adrenergic receptor endocytosis through difl’erent pathways. J Biol Chem, 2003. 278(37): p. 35403-11. Di Guglielmo, G.M., et al., Distinct endocytic pathways regulate T GF -beta receptor signalling and turnover. Nat Cell Biol, 2003. 5(5): p. 410-2]. Lazar, C.S., eta1., The Na +/H+ exchanger regulatory factor stabilizes epidermal growth factor receptors at the cell surface. Mol Biol Cell, 2004. 15(12): p. 5470- 80. Sneddon, W.B., et al., Activation-independent parathyroid hormone receptor internalization is regulated by NHERF1 (EBP50). J Biol Chem, 2003. 278(44): p. 43787-96. Marchese, A. and J .L. Benovic, Agonist-promoted ubiquitination of the G protein- coupled receptor CXCR4 mediates lysosomal sorting. J Biol Chem, 2001. 276(49): p. 45509-45512. Shenoy, S.K., et al., Regulation of Receptor Fate by Ubiquitination of Activated BetaZ-Adrenergic Receptor and Beta-Arrestin. Science, 2001. 294: p. 1307-1313. Whistler, J .L., et al., Modulation of Postendocytic Sorting of G Protein-Coupled Receptors. Science, 2002. 297(5581): p. 615-620. Martin, N.P., R.J. Lefkowitz, and SK. Shenoy, Regulation of V2 vasopressin receptor degradation by agonist-promoted ubiquitination. J Biol Chem, 2003. 278(46): p. 45954-9. Marchese, A., et al., The E3 ubiquitin ligase AIP4 mediates ubiquitination and sorting of the G protein-coupled receptor CXCR4. Dev Cell, 2003. 5(5): p. 709- 22. 229 224. 225. 226. 227. 228. 229. 230. 231. 232. 233. 234. 235. Cao, T.T., et al., A kinase-regulated PDZ-domain interaction controls endocytic sorting of the betaZ-adrenergic receptor. Nature, 1999. 401(6750): p. 286-90. Cong, M., et al., Binding of the betaZ-Adrenergic Receptor to N-Ethylmaleimide- sensitive Factor Regulates Receptor Recycling. J Biol Chem, 2001. 276(48): p. 45145-45152. Li, J .-G., C. Chen, and L.-Y. Lin-Chen, Ezrin-Radt'xin-Moesin-binding Phosphoprotein-50/Na +/H+ Exchanger Regulatory Factor (EBP50/NI-IERF) Blocks U50, 488H-induced Down-regulation of the Human Kappa Opoid Receptor by Enhancing Its Recycling Rate. J Biol Chem, 2002. 277(30): p. 27545-27552. Gage, R.M., et al., A transplantable sorting signal that is sufl‘icient to mediate rapid recycling of G protein-coupled receptors. J Biol Chem, 2001. 276(48): p. 4471 2-20. Parameswaran, N., et al., Desensitization and resensitization of adrenomedullin- sensitive receptor in rat mesangial cells. Eur J Pharmacol, 2000. 407(3): p. 205- 10. Sawyer, T.K., Src homology-2 domains: structure, mechanisms, and drug discovery. Biopolymers, 1998. 47(3): p. 243-61. Grucza, R.A., et al., SH2 domains: from structure to energetics, a dual approach to the study of structure-function relationships. Med Res Rev, 1999. 19(4): p. 273-93. Fan, G., et al., c-Src tyrosine kinase binds the beta 2-adrenergic receptor via phospho- Tyr-3 50, phosphorylates G-protein-linked receptor kinase 2, and mediates agonist-induced receptor desensitization. J Biol Chem, 2001. 276(16): p. 13240-7. Gmeiner, W.H. and DA. Horita, Implications of SH3 domain structure and dynamics for protein regulation and drug design. Cell Biochem Biophys, 2001. 35(2): p. 127-40. Zarrinpar, A., R.P. Bhattacharyya, and WA. Lim, The structure and function of proline recognition domains. Sci STKE, 2003. 2003(179): p. RE8. Liu, J ., et al., Src homology 3 binding sites in the P2 Y2 nucleotide receptor interact with Src and regulate activities of Src, proline-rich tyrosine kinase 2, and growth factor receptors. J Biol Chem, 2004. 279(9): p. 8212-8. Ilsley, J .L., M. Sudol, and SJ. Winder, The W domain: linking cell signalling to the membrane cytoskeleton. Cell Signal, 2002. 14(3): p. 183-9. 230 236. 237. 238. 239. 240. 241. 242. 243. 244. 245. 246. 247. 248. Ball, L.J., et al., E VH1 domains: structure, function and interactions. FEBS Lett, 2002. 513(1): p. 45-52. Kato, A., et al., Novel members of the Vesl/Homer family of PDZ proteins that bind metabotropic glutamate receptors. J Biol Chem, 1998. 273(37): p. 23969-75. Bezprozvanny, I. and A. Maximov, Classification of PDZ domains. F EBS Lett, 2001. 509(3): p. 457-62. Laura, R.P., et al., The Erbin PDZ domain binds with high affinity and specificity to the carboxyl termini of delta-catenin and AR VCF. J Biol Chem, 2002. 277(15): p. 12906-14. Hall, R.A., et al., The beta2-adrenergic receptor interacts with the Na +/H+- exchanger regulatory factor to control Na +/H+ exchange. Nature, 1998. 392(6676): p. 626-30. Komau, H.C., et al., Domain interaction between NMDA receptor subunits and the postsynaptic density protein PSD-95. Science, 1995. 269(5231): p. 1737-40. Grootjans, J .J ., et al., Syntenin, a PDZ protein that binds syndecan cytoplasmic domains. Proc Natl Acad Sci U S A, 1997. 94(25): p. 13683-8. Hata, Y., S. Butz, and TC. Sudhof, CASK: a novel dlg/PSD95 homolog with an N-terminal calmodulin-dependent protein kinase domain identified by interaction with neurexins. J Neurosci, 1996. 16(8): p. 2488-94. Hsueh, Y.P. and M. Sheng, Regulated expression and subcellular localization of syndecan heparan sulfate proteoglycans and the syndecan-binding protein CASK/LIN-Z during rat brain development. J Neurosci, 1999. 19(17): p. 7415-25. Torres, R., eta1., PDZ proteins bind, cluster, and synaptically colocalize with Eph receptors and their ephrin ligands. Neuron, 1998. 21(6): p. 1453-63. Stricker, N.L., et al., PDZ domain of neuronal nitric oxide synthase recognizes novel C—terminal peptide sequences. Nat Biotechnol, 1997. 15(4): p. 336-42. Cohen, N.A., et al., Binding of the inward rectifier K + channel Kir 2.3 to PSD-95 is regulated by protein kinase A phosphorylation. Neuron, 1996. 17(4): p. 759-67. Doyle, D.A., et al., Crystal structures of a complexed and peptide-free membrane protein-binding domain: molecular basis of peptide recognition by PDZ. Cell, 1996. 85(7): p. 1067-76. 231 249. 250. 251. 252. 253. 254. 255. 256. 257. 258. 259. 260. 261. Nourry, C., S.G. Grant, and J .P. Borg, PDZ domain proteins: plug and play! Sci STKE, 2003. 2003(179): p. RE7. Maudsley, S., et al., Platelet-derived growth factor receptor association with Na(+)/H(+) exchanger regulatory factor potentiates receptor activity. Mol Cell Biol, 2000. 20(22): p. 8352-63. Weinman, E.J., D. Steplock, and S. Shenolikar, CAMP-mediated inhibition of the renal brush border membrane Na+-H+ exchanger requires a dissociable phosphoprotein cofactor. J Clin Invest, 1993. 92(4): p. 1781-6. Weinman, E.J., et al., Ezrin binding domain-deficient NHERF attenuates cAMP- mediated inhibition of Na( +)/H( +) exchange in OK cells. Am J Physiol Renal Physiol, 2001. 281(2): p. F374-80. Cunningham, R., et al., Defective parathyroid hormone regulation of NHE3 activity and phosphate adaptation in cultured NHERF -1 -/- renal proximal tubule cells. J Biol Chem, 2004. 279(36): p. 37815-21. Wade, J .B., et al., Diflerential renal distribution of NHERF isoforms and their colocalization with NHE3, ezrin, and ROMK. Am J Physiol Cell Physiol, 2001. 280(1): p. C192-8. Weinman, E.J. and S. Shenolikar, The Na-H exchanger regulatory factor. Exp Nephrol, 1997. 5(6): p. 449-52. Moyer, B.D., et al., A PDZ-interacting domain in CF T R is an apical membrane polarization signal. J Clin Invest, 1999. 104(10): p. 1353-61. Voltz, J .W., E.J. Weinman, and S. Shenolikar, Expanding the role of NHERF, a PDZ-domain containing protein adapter, to growth regulation. Oncogene, 2001. 20(44): p. 6309-14. Shenolikar, S. and E.J. Weinman, NHERF: targeting and trafficking membrane proteins. Am J Physiol Renal Physiol, 2001. 280(3): p. F389-95. Li, J .G., Chen, C., Liu-Chen, L.Y., Ezrin-radixin-moesin-binding phosphoprotein- 50/Na +/H+ exchanger regulatory factor (EBP5 0/NHERF ) blocks U50, 488H- induced down-regulation of the human kappa opioid receptor by enhancing its recycling rate. J Biol Chem, 2002. 277(30): p. 27545-52. Patel, S. and M. Latterich, The AAA team: related AT Pases with diverse functions. Trends Cell Biol, 1998. 8(2): p. 65-71. Brunger, A.T., Structural insights into the molecular mechanism of Ca(2+ )- dependent exocytosis. Curr Opin Neurobiol, 2000. 10: p. 293-302. 232 262. 263. 264. 265. 266. 267. 268. 269. 270. 271. 272. 273. Lin, R.C., Scheller, R.H., Mechanisms of synaptic vesicle exocytosis. Annu Rev Cell Dev Biol, 2000. 16: p. 19-49. Neuwald, A.F., The hexamerization domain of N-ethylmaleimide-sensitive factor: structural clues to chaperone function. Structure Fold Des, 1999. 7: p. Rl9-23. Morgan, A., R. Dimaline, and RD. Burgoyne, The ATPase activity of N- ethylmaleimide-sensitive fusion protein (NSF) is regulated by soluble NSF attachment proteins. J Biol Chem, 1994. 269(47): p. 29347-50. Barnard, R.J., A. Morgan, and RD. Burgoyne, Stimulation of NSF ATPase activity by alpha-SNAP is required for SNARE complex disassembly and exocytosis. J Cell Biol, 1997. 139(4): p. 875-83. Barnard, R.J., A. Morgan, and RD. Burgoyne, Domains of alpha-SNAP required for the stimulation of exocytosis and for N-ethylmalemide-sensitive fusion protein (NSF) binding and activation. Mol Biol Cell, 1996. 7(5): p. 693-701. Xia, 2., Gray, J .A., Compton-Toth, B.A., Roth B.L., A direct interaction of PSD- 95 with 5-H T 2A serotonin receptors regulates receptor trafficking and signal transduction. J Biol Chem, 2003. 278(24): p. 21901-8. Cong, M., Perry, S.J., Hu, L.A., Hanson, P.I., Claing, A., Leflcowitz, R.J., Binding of the beta 2 adrenergic receptor to N-ethylmaleimide sensitive factor regulates receptor recycling. J Biol Chem, 2001. 276(48): p. 45145-52. Song, 1., Kamboj, S., Xia, J ., Dong, H., Liao, D., Huganir, R.L., NSF binding to GluR2 regulates synaptic transmission. Neuron, 1998. 21: p. 393-400. Nishimune, A., Isaac, J.T., Molnar, E., Noel, J ., Nash, S.R., Tagaya, M., Collingridge, G.L., Nakanishi, S., Henley, J .M., NSF binding to GluR2 regulates synaptic transmission. Neuron, 1998. 21: p. 87-97. Hanley, J .G., et al., NSF AT Pase and alpha-/beta-SNAPs disassemble the AMPA receptor-PICK] complex. Neuron, 2002. 34(1): p. 53-67. Miller, W.E. and R.J. Lefltowitz, Expanding roles for beta-arrestins as scafl'olds and adapters in GPCR signaling and trafficking. Curr Opin Cell Biol, 2001. 13(2): p. 139-45. McDonald, RH. and R.J. Leflrowitz, Beta-Arrestins: new roles in regulating heptahelical receptors ' functions. Cell Signal, 2001. 13(10): p. 683-9. 233 274. 275. 276. 277. 278. 279. 280. 281. 282. 283. 284. Matveeva, E.A., et al., Phosphorylation of the N-ethylmaleimide-sensitive factor is associated with depolarization-dependent neurotransmitter release from synaptosomes. J Biol Chem, 2001. 276(15): p. 12174-81. McLatchie, L.M., Fraser, N.J., Main, M.J., Wise, A., Brown, J., Thompson, N., Solari, R., Lee, M.G., Foord, S.M., RAMPS regulate the transport and ligand specificity of the calcitonin-receptor-like receptor. Nature, 1998. 393: p. 333-9. Kuwasako, K., Shimekake, Y., Masuda, M., Nakahara, K., Yoshida, T., Kitaura, M., Kitamura, K., Eto T., Sakata, T., Visualization of the Calcitonin Receptor-like Receptor and Its Receptor Activity-modifying Proteins during Intemalization and Recycling. J Biol Chem, 2000. 275(38): p. 29602-9. Kamitani, S., Asakawa, M., Shimekake, Y., Kuwasako, K., Nakahara, K., Sakata, T., The RAMP2/CRLR complex is a functional adrenomedullin receptor in human endothelial and vascular smooth muscle cells. FEBS Lett, 1999. 448: p. 111-14. Buhlmann, N., Leuthauser, K., Muff, R., Fischer, J.A., Born, W., A receptor activity modifying protein (RAMP)2-dependent adrenomedullin receptor is a calcitonin gene-related peptide receptor when co-expressed with human RAMP]. Endocrinology, 1999. 140: p. 2883-90. Hinson, J.P., Kapas, S., Smith, D.M., Adrenomedullin, a multifunctional regulatory peptide. Endocrine Reviews, 2000. 21(2): p. 138-67. Hilairet, S., Belanger, C., Bertrand, J., Laperriere, A., Foord, S.M., Bouvier M., Agonist-promoted Intemalization of a Ternary Complex between Calcitonin Receptor-like Receptor, Receptor Activity-modifying Protein 1 (RAMPI), and ,6- Arrestin. J Biol Chem, 2001. 276(45): p. 42182-190. Aiyar, N., Disa, J., Ao, Z., Xu, D., Surya, A., Pillarisetti, K., Parameswaran, N., Gupta, S.K., Douglas, S.A., Nambi, P., Molecular cloning and pharmacological characterization of bovine calcitonin receptor-like receptor from bovine aortic endothelial cells. Biochem Pharmacol, 2002. 63(11): p. 1949-59. Nowak, W., Parameswaran, N., Hall, C.S., Aiyar, N., Sparks, H.V., Spielman, W.S., Novel regulation of adrenomedullin receptor by PDGF: role of receptor activity modifying protein-3. Am J Physiol Cell Physiol, 2002. 282: p. C1322-3 l. Aiyar, N., Nambi, P., Whitman, M., Stassen, F .L., Crooke, S.T., Phorbol ester- mediated inhibition of vasopressin and beta-adrenergic responses in a vascular smooth muscle cell line. Mol Pharmacol, 1987. 31(2): p. 180-4. Salomon, Y., C. Londos, and M. Rodbell, A highly sensitive adenylate cyclase assay. Anal Biochem, 1974. 58(2): p. 541-8. 234 285. 286. 287. 288. 289. 290. 291. 292. 293. 294. 295. Parameswaran, N., Aiyar, N., Wu, H., Brooks, D.P., Nambi, P., Spielman, W.S., Desensitization and resensitization of adrenomedullin-sensitive receptor in rat mesangial cells. Eur J Pharmacol, 2000. 407(3): p. 205-10. Christopoulos, A., Christopoulos, G., Morfis, M., Udawela, M., Laburthe, M., Couvineau, A., Kuwasako, K., Tilakaratne, N., Sexton, P.M., Novel Receptor Partners and Function of Receptor Activity-modifidng Proteins. J Biol Chem, 2003. 278(5): p. 3293-97. Hay, D.L., D.R. Poyner, and D.M. Smith, Desensitisation of adrenomedullin and CGRP receptors. Regul Pept, 2003. 112(1-3): p. 139-45. Namkung, Y. and DR. Sibley, Protein kinase C mediates phosphorylation, desensitization, and trafficking of the D2 dopamine receptor. J Biol Chem, 2004. 279(47): p. 49533-41. Kawakami, N., et al., Direct phosphorylation of histamine H I receptor by various protein kinases in vitro. Methods Find Exp Clin Pharmacol, 2003. 25(9): p. 685- 93. Bhattacharyya, S., et al., Intemalization and recycling of 5-H T 2A receptors activated by serotonin and protein kinase C-mediated mechanisms. Proc Natl Acad Sci U S A, 2002. 99(22): p. 14470-5. Yan, F.X., et al., Serine 331 is major site of phosphorylation and desensitization induced by protein kinase C in thromboxane receptor alpha. Acta Pharmacol Sin, 2002. 23(10): p. 952-60. Liang, M., et al., Serine 232 of the alpha(2A)-adrenergic receptor is a protein kinase C-sensitive eflector coupling switch. Biochemistry, 2001. 40(49): p. 1503 1-7. Garcia-Unzueta, M.T., Montalban, C., Pesquera, C., Berrazueta, J .R., Amado, J .A., Plasma adrenomedullin levels in type 1 diabetes. Relationship with clinical parameters. Diabetes Care, 1998. 21(6): p. 999-1003. Kubo, A., Kurioka, H., Minamino, N., Nishitani, Y., Sato, H., Nishino, T., Iwano, M., Shiiki, H., Kangawa, K., Matsuo, H., Dohi, K., Plasma and urinary levels of adrenomedullin in chronic glomerulonephritis patients with proteinuria. Nephron, 1998. 80(2): p. 227-30. Nakamura, T., Honda, K., Ishikawa, S., Kitamura, K., Eto, T., Saito, T., Plasma adrenomedullin levels in patients with non-insulin dependent diabetes mellitus: close relationships with diabetic complications. Endocr J, 1998. 45(2): p. 241-6. 235 296. 297. 298. 299. 300. 301. 302. 303. 304. 305. 306. 307. Wang, C., Dobrzynski, E., Chao, J ., Chao, L., Adrenomedullin gene delivery attenuates renal damage and cardiac hypertrophy in Goldblatt hypertensive rats. Am J Physiol Renal Physiol, 2001. 280(6): p. F964-71. Zhang, J.J., Yoshida, H., Chao, L., Chao, J ., Human adrenomedullin gene delivery protects against cardiac hypertrophy, fibrosis, and renal damage in hypertensive dahl salt-sensitive rats. Hum Gene Ther, 2000. 11(13): p. 1817-27. Chao, J ., Jin, L., Lin, K.F., Chao, L., Adrenomedullin gene delivery reduces blood pressure in spontaneously hypertensive rats. Hypertens Res, 1997. 20(4): p. 269- 77. Dobrzynski, E., Wang, C., Chao, J ., Chao, L., Adrenomedullin gene delivery attenuates hypertension, cardiac remodeling, and renal injury in deoxycorticosterone acetate-salt hypertensive rats. Hypertension, 2000. 36(6): p. 995- l 001 . Dobrzynski, E., Montanari, D., Agata, J ., Zhu, J., Chao, J., Chao, L., Adrenomedullin improves cardiac function and prevents renal damage in streptozotocin-induced diabetic rats. Am J Physiol Endocrinol Metab, 2002. 283(6): p. E1291-8. Mahon, MJ. and G.V. Segre, Stimulation by parathyroid hormone of a NHERF- I-assembled complex consisting of the parathyroid hormone I receptor, phospholipase Cbeta, and actin increases intracellular calcium in opossum kidney cells. J Biol Chem, 2004. 279(22): p. 23550-8. Mahon, M.J., et al., Na(+)/H(+ ) exchanger regulatory factor 2 directs parathyroid hormone I receptor signalling. Nature, 2002. 417(6891): p. 858-6]. Bomberger, J .M., et al., RAMP isoform-specific regulation of adrenomedullin receptor traflicking by NHERF-I. J Biol Chem, 2005. Murthy, A., eta1., NHE-RF, a regulatory cofactor for Na(+)-H+ exchange, is a common interactor for merlin and ERM (MERM) proteins. J Biol Chem, 1998. 273(3): p. 1273-6. Brain, SD. and AD. Grant, Vascular actions of calcitonin gene-related peptide and adrenomedullin. Physiol Rev, 2004. 84(3): p. 903-34. Kurihara, H., et al., Targeted disruption of adrenomedullin and alphaCGRP genes reveals their distinct biological roles. Hypertens Res, 2003. 26 Suppl: p. 8105-8. Roh, J., Chang, C.L., Bhalla, A., Klein, C., Hsu, S.Y., Intermedin is a calcitonin/calcitonin gene-related peptide family peptide acting through the 236 308. 309. 310. 311. calcitonin receptor-like receptor/receptor activity-modifying protein receptor complexes. J Biol Chem, 2004. 279(8): p. 7264-74. Choksi, T., Hay, D.L., Legon, S., Poyner, D.R., Hagner, S., Bloom, S.R., Smith, D.M., Comparison of the expression of calcitonin receptor-like receptor (CRLR) and receptor activity modifying proteins (RAMPS) with C GRP and adrenomedullin binding in cell lines. Br J Pharmacol, 2002. 136: p. 784-792. Tadokoro, K., Nishikimi, T., Mori, Y., Wang, X., Akimoto, K., Matsuoka, H., Altered gene expression of adrenomedullin and its receptor system and molecular forms of tissue adrenomedullin in left ventricular hypertrophy induced by malignant hypertension. Regul Pept, 2003. 112(1-3): p. 71-8. Nishikimi, T., Tadokoro, K., Mori, Y., Wang, X., Akimoto, K., Yoshihara, F., Minamino, N., Kangawa, K., Matsuoka, H., Ventricular adrenomedullin system in the transition from L VH to heart failure in rats. Hypertension, 2003. 41(3): p. 512-8. Cueille, C., Pidoux, E., de Vemejoul, M.C., Ventura-Clapier, R., Garel, J .M., Increased myocardial expression of RAMP] and RAMP3 in rats with chronic heart failure. Biochem Biophys Res Commun, 2002. 294(2): p. 340-6. 237