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 2/05 p:/C|RC/DateDuerindd-p.1 ENDOTHELIN A (ETA) AND ETB RECEPTOR INTERACTION IN ARTERIES AND VEINS BY Keshari Maya Thakali A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Pharmacology and Toxicology 2006 ABSTRACT ENDOTHELIN A (ETA) AND ETB RECEPTOR INTERACTION IN ARTERIES AND VEINS By Keshari Maya Thakali Veins maintain responsiveness while arteries lose responsiveness to the vasoactive hormone endothelin-1 (ET-1) in situations of exposure to ET-1 (eg. hypertension and experimental protocols). Thus, receptors for ET-1 — the heptahelical ETA and El"l3 receptors - may function differently in arteries and veins. While heptahelical receptors canonically interact with G-proteins in a 1:1 ratio, recent evidence suggests that receptor dimerization (hetero- or homo-) may occur. Receptor dimerization can potentially affect a number of receptor characteristics such as agonist affinity, potency and efficacy, as well as receptor interaction. While ET A and ET 3 receptors are present on the smooth muscle of rat thoracic aorta and vena cava, only vena cava have contractile ETA and Era receptors as in aorta only ET A receptors couple to contraction. Previous studies suggest that in vessels with functional populations of both ETA and El}, receptors, ET8 receptors are capable of physically or pharmacologically altering ETA receptor function. I hypothesized that ET A and ETl3 receptors can heterodimerize in rat thoracic vena cava but not aorta and that there are functional consequences of venous ETA and ETl3 receptor heterodimerization. To test this hypothesis that ET A and ETI3 receptors heterodimerize in vena cava but not aorta, I examined functional receptor interaction (vascular contractility), biochemical receptor interaction (interaction via signal transducers) and physical receptor interaction (receptor heterodimerization) of ETA and ET 3 receptors in rat thoracic aorta and vena cava. Isolated contractility experiments demonstrated that functional ETA and ET 3 receptor interaction occurred in vena cava but not aorta as venous I':'I'A receptors were more sensitive to receptor blockade when E‘l'l3 receptors were rendered dysfunctional either from desensitization or receptor blockade. Biochemical signaling interactions via H202 signaling, p38 MAPK, Erk MAPK, rho kinase, src and phosphatidyl inositol 3-kinase (PIS-K) do not mediate functional venous ETA and El'.3 receptor interaction. Receptor co-localization of ETA and ETB receptors on dissociated aortic and venous vascular smooth muscle cells demonstrated that ET A and ET B receptors co-localized to the plasma membrane of both aortic and venous vascular smooth muscle cells. These results suggest that functional ETA and El'I3 receptor interaction is independent of receptor location and may be an intrinsic property of veins and not due to receptor heterodimerization. ACKNOWLEDGEMENTS I would first and foremost like to thank my parents, Renee and Aita Thakali, for their continuous love and support. Morn, thank you for fostering my interest in science. Dad, thank you for passing on your inquisitiveness and skepticism. From you I Ieamed that with some Duct tape, superglue and the right set of tools, anything can be fixed (even if it was not broken in the first place). Thank you both for traveling around the world with me, expanding my horizons and molding me into the independent individual I am today. I would like to acknowledge my advisor, Stephanie W. Watts, PhD, for being the most exceptional mentor a graduate student could ever dream of. Thank you for taking the time to teach me both in the laboratory and the classroom. Thank you for all of your countless hours spent reading my abstracts, manuscripts and dissertation. Thank you for taking me to meetings and building up my scientific network. Thank you (in advance) for all of the future opportunities I will have as a direct result of the excellent training l have received under you mentorship. Thank you for putting up with my practical jokes and my odd sense of humor. Most of all, thank you for welcoming me into your family. I am constantly humbled and amazed by everything you do (and have already accomplished) and would like you to know that you will always be my role model. Lastly, I would like to acknowledge my boyfriend, Zachary C. Taylor, MD. Thank you for moving to “miserable” Michigan with me. Thank you for understanding how important science is to me, but also thank you for reminding me that there are other fun things in life, and those fun things typically involve bicycles and puppy dogs. I love you and hope that you will always save a spot on the couch for me and Kali Bear. TABLE OF CONTENTS LIST OF TABLES ................................................................................ viii LIST OF FIGURES ............................................................................... ix LIST OF ABBREVIATIONS ................................................................... xiii I. INTRODUCTION .......................................................................... 1 A. The circulatory system .............................................................. 1 1. Blood vessel composition .................................................. 1 8. Function of arteries and veins ..................................................... 3 1. Determinants of blood pressure .......................................... 3 2. Mechanisms of blood pressure regulation ............................. 8 C. Role of veins in blood pressure regulation ................................... 10 D. Humoral control of blood pressure by circulating peptides 11 E. Endothelin ............................................................................ 12 1. ET-1 synthesis and degradation ....................................... 13 F. ET-1 signal transduction .......................................................... 17 1. ETA and ETB receptors are G-protein coupled receptors ......... 17 2. ETA and ETB receptor signal transduction ........................... 19 3. Reactive oxygen species signaling .................................... 24 G. Distribution of ETA and ETB receptors in blood vessels .................. 25 H. ETA and ETB receptor function in hypertension ............................. 30 I. ETA and ET; receptor “cross-talk? or interaction ............................. 31 J. Receptor dimerization .............................................................. 33 1. Physical means for receptor dimerization ........................... 33 2. Caveolae as cellular locations for receptor dimerization ........ 34 3. Functional consequences of receptor dimerization ............... 37 4. Can ETA and ETB receptor heterodimerization occur and can heterodimerization affect ET receptor function? ................. 38 5. Significance of ETA and ETB receptor dimerization ............... 39 K. Hypothesis ........................................................................... 41 II. METHODS ............................................................................... 44 A. Isolated smooth muscle contraction ........................................... 44 1. ET-1 desensitization ...................................................... 45 2. ETa receptor desensitization ............................................ 45 3. Receptor antagonism studies ........................................... 45 4. H202 concentration response curves ................................. 46 B. Measurement of arterial and venous H202 production. ..46 C. Whole tissue immunohistochemistry .......................................... 47 D. Whole tissue protein isolation ................................................... 48 1. Cross-linking ............................................................... .48 E. Western blot analysis .............................................................. 49 receptor function. I hypothesized that ETA and EI'la receptors can heterodimerize in rat thoracic vena cava but not aorta and that there are functional consequences of venous EI'A and ET 3 receptor heterodimerization. To test this hypothesis that ETA and ETB receptors heterodimerize in vena cava but not aorta, I examined functional receptor interaction (vascular contractility), biochemical receptor interaction (interaction via signal transducers) and physical receptor interaction (receptor heterodimerization) of ETA and ETI3 receptors in rat thoracic aorta and vena cava. Isolated contractility experiments demonstrated that functional ETA and ETB receptor interaction occurred in vena cava but not aorta as venous ETA receptors were more sensitive to receptor blockade when ETl3 receptors were rendered dysfunctional either from desensitization or receptor blockade. Biochemical signaling interactions via H202 signaling, p38 MAPK, Erk MAPK, rho kinase, src and phosphatidyl inositol 3-kinase (PIS-K) do not mediate functional venous ETA and EI'B receptor interaction. Receptor co-Iocalization of ET, and ETl3 receptors on dissociated aortic and venous vascular smooth muscle cells demonstrated that ETA and ETB receptors co-localized to the plasma membrane of both aortic and venous vascular smooth muscle cells. These results suggest that functional ETA and Er, receptor interaction is independent of receptor location and may be an intrinsic property of veins and not due to receptor heterodimerization. ACKNOWLEDGEMENTS I would first and foremost like to thank my parents, Renee and Aita Thakali, for their continuous love and support. Morn, thank you for fostering my interest in science. Dad, thank you for passing on your inquisitiveness and skepticism. From you I Ieamed that with some Duct tape, superglue and the right set of tools, anything can be fixed (even if it was not broken in the first place). Thank you both for traveling around the world with me, expanding my horizons and molding me into the independent individual I am today. I would like to acknowledge my advisor, Stephanie W. Watts, PhD, for being the most exceptional mentor a graduate student could ever dream of. Thank you for taking the time to teach me both in the laboratory and the classroom. Thank you for all of your countless hours spent reading my abstracts, manuscripts and dissertation. Thank you for taking me to meetings and building up my scientific network. Thank you (in advance) for all of the future opportunities I will have as a direct result of the excellent training I have received under you mentorship. Thank you for putting up with my practical jokes and my odd sense of humor. Most of all, thank you for welcoming me into your family. I am constantly humbled and amazed by everything you do (and have already accomplished) and would like you to know that you will always be my role model. Lastly, I would like to acknowledge my boyfriend, Zachary C. Taylor, MD. Thank You for moving to “miserable” Michigan with me. Thank you for understanding how important science is to me, but also thank you for reminding me that there are other fun things in life, and those fun things typically involve bicycles and PUPPY dogs. I love you and hope that you will always save a spot on the couch for me and Kali Bear. iv TABLE OF CONTENTS LIST OF TABLES ................................................................................ viii LIST OF FIGURES ............................................................................... ix LIST OF ABBREVIATIONS ................................................................... xiii I. INTRODUCTION .......................................................................... 1 A. The circulatory system .............................................................. 1 1. Blood vessel composition .................................................. 1 B. Function of arteries and veins ..................................................... 3 1. Determinants of blood pressure .......................................... 3 2. Mechanisms of blood pressure regulation ............................. 8 C. Role of veins in blood pressure regulation ................................... 10 D. Humoral control of blood pressure by circulating peptides 11 E. Endothelin ............................................................................ 12 1. ET-1 synthesis and degradation ....................................... 13 F. ET-1 signal transduction .......................................................... 17 1. ETA and ETa receptors are G-protein coupled receptors 17 2. ETA and ETB receptor signal transduction ........................... 19 3. Reactive oxygen species signaling .................................... 24 G. Distribution of ETA and ETB receptors in blood vessels .................. 25 H. ETA and ETB receptor function in hypertension ............................. 30 I. ETA and ETB receptor “cross-talk” or interaction ............................. 31 J. Receptor dimerization .............................................................. 33 1. Physical means for receptor dimerization ........................... 33 2. Caveolae as cellular locations for receptor dimerization ........ 34 3. Functional consequences of receptor dimerization ............... 37 4. Can ETA and ETa receptor heterodimerization occur and can heterodimerization affect ET receptor function? ................. 38 5. Significance of ETA and ETB receptor dimerization ............... 39 K. Hypothesis ........................................................................... 41 II. METHODS ............................................................................... 44 A. Isolated smooth muscle contraction ........................................... 44 1. ET-1 desensitization ...................................................... 45 2. ET a receptor desensitization ............................................ 45 3. Receptor antagonism studies ........................................... 45 4. H202 concentration response curves ................................. 46 B. Measurement of arterial and venous H202 production. ..46 C. Whole tissue immunohistochemistry .......................................... 47 D. Whole tissue protein isolation ................................................... 48 1. Cross-linking ............................................................... .48 E. Western blot analysis .............................................................. 49 1. SDS-polyacrylamide gel electrophoresis (PAGE) (denaturing PAGE) ............................................................ 49 2. SDS-urea PAGE ............................................................ 49 3. Non-denaturing PAGE (NATIVE PAGE) ............................. 50 F. Co-immunoprecipitation ........................................................... 50 1. Traditional co-immunoprecipitation .................................... 50 2. True Blot® co-immunoprecipitation kit ................................ 51 3. Pierce ProFound® mammalian Co-IP kit ............................ 51 4. ExactaCruz® co-immunoprecipitation kit ............................ 52 5. Co-immunoprecipitation using Dynal® magnetic beads 53 G. Dissociation of vascular smooth muscle cells ............................... 53 H. Immunocytochemistry in freshly dissociated vascular smooth muscle cells ..................................................................... 54 I. Data analysis and statistics ....................................................... 55 RESULTS ................................................................................. 57 A. Subhypothesis 1: Pharmacological receptor interaction in veins but not arteries ........................................................................... 57 1. Non-specific ET,.,/ETl3 receptor desensitization ..................... 57 2. ET 8 receptor desensitization ............................................. 65 3. Does ETl3 receptor desensitization alter ET A receptor function? ......................................................................... 65 4. Receptor interaction: contractility studies using receptor antagonists ...................................................................... 7O 5. Section summary ........................................................... 74 B. Subhypothesis 2: Biochemical signaling interactions ...................... 75 1. Reactive oxygen species and ET -1-induced contraction 80 2. Signal transduction pathways ........................................... 83 3. Section summary ........................................................... 91 C. Subhypothesis 3: Physical endothelin receptor interaction 91 1. Antibody testing: dot blots ................................................ 92 2. Receptor co-Iocalization .................................................. 93 3. Co-immunoprecipitation of ETA and ET B receptors .............. 103 4. Section summary ......................................................... 107 DISCUSSION .......................................................................... 109 A. Rationale ........................................................................... 109 B. Differences in ET A and ETB receptor desensitization ................... 111 C. Functional ETA and ETl3 receptor interaction occurs in veins but not arteries .................................................................... 1 14 D. Signaling pathway interactions may account for functional venous ET A and ETl3 receptor interaction ............................................. 116 1. Reactive oxygen species signaling .................................. 117 2. ET-1 signal transduction ................................................ 120 E. Can ETA and ET B receptors physically interact via vi VI. receptor heterodimerization? .................................................. 124 1. Co-immunoprecipitation of ETA and ET B receptors in rat thoracic aorta and vena cava ............................................. 128 2. Receptor co-Iocalization ................................................ 129 F. Conclusions ....................................................................... 134 G. Speculation ........................................................................ 135 REFERENCES ........................................................................ 138 CURRICULUM VITAE ............................................................... 148 vii Table 1. Table 2. Table 3. Table 4. LIST OF TABLES Estimated EC50 values for Er-1-induced contraction in aorta and vena cava in the presence of functional or SGc-desensitized ETB receptors with and without ETA receptor antagonism ................. 69 Estimated EC,o values for ET -1-induced contraction in aorta and vena cava and the presence and absence of functional ETl3 receptors with and without ETA receptor antagonism ................. 73 An abbreviated list of commercially available ETA and ETB receptor antibodies and their antigenic sequences used for dot blot analysis ........................................................................... 94 Different co-immunoprecipitation methods were performed in several attempts to co-immunoprecipitate ETA and E'I'l3 receptors from rat thoracic aorta and vena cava lysates ........................ 1 O8 viii Figure 1. Figure 2. Figure 3. Figure 4. Figure 5. Figure 6. Figure 7. Figure 8. Figure 9. Figure 10. Figure 11. Figure 12. Figure 13. Figure 14. Figure 15. LIST OF FIGURES Rendered picture of arteries and veins ................................... 2 Histological cross-sections of rat thoracic aorta and thoracic vena cava stained with a Masson trichrome stain 4 Alpha actin immunohistochemistry of rat thoracic aorta and vena cava ......................................................................... 5 Western blot analysis of smooth muscle a-actin expression in rat thoracic aorta and vena cava lysates ..................................... 7 Guyton-Coleman diagram of physiological factors that affect pressure natriuresis and control blood pressure ........................ 9 Diagram of endothelin-1 (ET-1) biosynthesis ........................... 14 Effect of phosphoramidon, an endothelin converting enyzme inhibitor, on big ET -1-induced contraction ............................... 16 Diagram of endothelin signal transduction .............................. 18 Diagram of ET A receptor signal transduction ........................... 20 Diagram of EI',3 receptor signal transduction ........................... 21 ET. and El’l3 receptor western blot analysis in rat thoracic vena cava ............................................................................... 26 ETA and ETa receptor immunohistochemistry in rat thoracic aorta and vena cava .................................................................. 27 ET-1 and 86c cumulative concentration response curves in rat thoracic aorta and vena cava .............................................. 28 Electron micrographs of caveolae present on the plasma membrane of aortic and venous vascular smooth muscle cells ............................................................................... 36 ET -1 concentration response curves in rat thoracic aorta and vena cava desensitized with ET-1 ............................................... 58 Figure 16. Figure 17. Figure 18. Figure 19. Figure 20. Figure 21. Figure 22. Figure 23. Figure 24. Figure 25. Figure 26. Figure 27. Figure 28. ET -1 [1-31] concentration response curves in rat thoracic aorta and vena cava desensitized with ET-1 ................................... 59 SSc-induced contraction of rat thoracic vena cava desensitized with ET-1 or $60 ................................................................ 61 Norepinephrine-induced contraction of rat thoracic aorta and vena cava was unaltered with ET-1 desensitization .................. 62 ET-t concentration response curves in the presence of ETA receptor blockade or ET 3 receptor blockade in rat thoracic vena cava desensitized with ET-t ......................................... 64 86c desensitization protocol and quantification of results in rat thoracic vena cava ........................................................ 66 Comparison of ETA receptor blockade of ET-1 cumulative concentration response curves in rat thoracic aorta with functional EI'.3 receptors and desensitized ET B receptors 67 Comparison of ETA receptor blockade of ET-1 cumulative concentration response curves in rat thoracic vena cava with functional ETB receptors and desensitized Iz'l'l3 receptors 68 Comparison of ETA receptor blockade of ET-1 cumulative concentration response curves in rat thoracic aorta with functional ETB receptors and without functional ET B receptors 71 Comparison of ETA receptor blockade of ET-1 cumulative concentration response curves in rat thoracic vena cava with functional EI'l3 receptors and without functional ET 3 receptors 72 Exogenously added H202 contracts both rat thoracic aorta and vena cava ........................................................................ 76 Basal H202 levels are significantly higher in rat thoracic vena cava compared to aorta ...................................................... 77 ET -1 stimulates H202 production in rat thoracic vena cava but not aorta .......................................................................... 78 ET -1-stimulated H202 production in vena cava is not concentration- dependent and is reduced by either ETA receptor blockade or ET a receptor blockade ............................................................. 79 Figure 29. Figure 30. Figure 31. Figure 32. Figure 33. Figure 34. Figure 35. Figure 36. Figure 37. Figure 38. Figure 39. Figure 40. Figure 41. H202 is not involved in venous ET-1-induced contraction as neither catalase nor PEG-catalase reduced venous ET-1-induced contraction .................................................... 81 The catalase inhibitor 3-AT had no effect on maximal venous ET -1 -induced contraction .................................................... 82 Effect of LY294002, a phosphatidyl inositol-3-kinase inhibitor, on ET-t -induced contraction of rat thoracic aorta and vena cava ........................................................................ 85 Effect of PD98059, an Erk MAPK, on ET-t -induced contraction of rat thoracic aorta and vena cava ....................................... 86 Effect of PP1 (10 IIM), a src tyrosine kinase inhbitor, on ET-t - induced contraction of rat thoracic aorta and vena cava 87 Effect of SB203580, a p38 MAPK inhbitor, on ET-1-induced contraction of rat thoracic aorta and vena cava ........................ 88 Effect of Y27632, a rho kinase inhbitor, on ET-1-induced contraction of rat thoracic aorta and vena cava ........................ 89 Effect of HA1077, a rho kinase inhbitor, on ET -1 -induced contraction of rat thoracic aorta and vena cava ........................ 90 Dot blots testing ETA receptor antibodies from Alomone Laboratories, Biodesign lntemational, Biogenesis and Fitzgerald Industries ........................................................................ 95 Dot blots testing ETl3 receptor antibodies from Alomone Laboratories, Biodesign lntemational and Biogenesis ............... 96 Projected confocal images of smooth muscle a-actin expression in freshly dissociated aortic and venous vascular smooth muscle cells ............................................................................... 98 Confocal images of ETA receptor, E'I'B receptor and cadherin expression in freshly dissociated aortic vascular smooth muscle cells ............................................................................... 99 Confocal images of ETA receptor, ETB receptor and cadherin expression in freshly dissociated venous vascular smooth xi Figure 42. Figure 43. Figure 44. Figure 45. muscle cells ................................................................... 100 Confocal images of ETA receptor, ET 3 receptor and cadherin expression in ET -1-stimulated freshly dissociated aortic vascular smooth muscle cells ............................................ 101 Confocal images of ET A receptor, ETl3 receptor and cadherin expression in ET-1-stimulated freshly dissociated venous vascular smooth muscle cells ............................................ 1 02 Western blot analysis of ETA receptor expression and ETB receptor expression in aorta and vena cava lysates ................................ 1 04 Example immunoprecipitation of ETl3 receptors from aortic and venous lysates ........................................................................... 1 05 xii AC CAMP CHO DAG DOCA DZ ET-1 ECL ERK FRET GABA GPCR HEK H202 IHC JNK MAP MAPK NEM NE 02' PE PI3-K PKA (or B or C) PLC ROS 860 LIST OF ABBREVIATIONS adenylate cyclase cyclic adenosine monophosphate Chinese Hamster Ovary diacyl glycerol deoxycorticosterone acetate dopamine 2 receptor endothelin-1 enhanced chemiluminescence extracellular regulated kinase (p42/44 MAPK) Fluorescence Resonance Energy Transfer y-aminobutyric acid G-protein coupled receptor human embryonic kidney hydrogen peroxide immunohistochemistry inositol 1,4,5-triphosphate c-jun N-terrninal kinase mean arterial pressure mitogen activated protein kinase N-ethyl maleimide norepinephrine superoxide anion phenylephrine phosphotidyl inositol 3-kinase protein kinase A (or B or C) phospholipase reactive oxygen species sarafotoxin 6c, ET 3 receptor specific agonist xiii SDS sodium dodecyl sulfate SEM standard error of the mean UNaV urinary sodium excretion xiv I. INTRODUCTION A. The circulatory system The circulatory system, consisting of arteries that distribute oxygenated blood to tissues, and veins that return oxygen-poor blood to the heart for re-oxygenation, is critical for providing tissues with nutrients and oxygen and removing waste byproducts of tissue metabolism. Arteries and veins have different physiological functions, and it is a goal of this dissertation to investigate how differences in endothelin receptors determine arterial and venous function. 1. Blood vessel composition Blood vessels are hollow organs comprised of three layers of cells (Figure 1). A single layer of endothelial cells line the lumen of blood vessels and define the intimal region of a blood vessel. The outermost layer of the blood vessel, the adventitia, comprised primarily of fibroblasts, is a highly collagenous layer providing structural integrity to vessels. Smooth muscle cells are present in the medial layer of a blood vessel, between the intima and adventitia. The smooth muscle cells of blood vessels are contractile cells that allow vessels to contract and relax to various neuro-humoral stimuli. Between the layers of smooth muscle are elastin fibers, which like the collagenous adventitia are critical for structural support, but also provide elastic recoil to accommodate stretching of the blood vessel. Recent studies suggest that in addition to medial smooth Tunica adventitia Carry blood away from Carry blood away from the heartto organs organs and tissues and tissues back to the heart Flgure 1. Rendered picture of arteries and veins. The tunica intima is the innermost layer of blood vessels. The tunica media, the middle layer, is where contractile smooth muscles cells are located. The tunica adventitia is the outermost layer, comprised primarily of collagen. muscle cells, the adventitia may contribute to changes in vascular tone (Laflamme ef al, 2006).Unique to veins are valves, which assist in returning blood to the heart. While the structure of blood vessels can be generalized by the presence of the intima, media and adventitia, a closer histological analysis of the structure of arteries and veins reveals several differences in arterial and venous structure. Masson trichrome staining of histological sections of rat thoracic aorta and thoracic vena cava, delineates the presence of smooth muscle (light pink), collagen (blue staining in the adventitia), nuclei (dark purple) and elastin fibers (bright red inbetween smooth muscle layers) (Figure 2). In aorta, multiple layers of smooth muscle cells are clearly defined between the collagenous adventitia and the single layer of endothelial cells lining the lumen of the blood vessel. The vena cava is highly collagenous, with a single layer of endothelial cells lining the intima. lmmunohistochemical analysis of smooth muscle a—actin expression reveals the presence of smooth muscle in between elastin fibers in aorta and a very thin layer of smooth muscle cells in vena cava (Figure 3). B. Function of arteries and veins 1. Determinants of blood pressure While these three layers of cells are common to both arteries and veins, how these components are organized to function together likely determines differences in arterial and venous function. Arteries typically have thicker layers I. INTRODUCTION A. The circulatory system The circulatory system, consisting of arteries that distribute oxygenated blood to tissues, and veins that return oxygen-poor blood to the heart for re-oxygenation, is critical for providing tissues with nutrients and oxygen and removing waste byproducts of tissue metabolism. Arteries and veins have different physiological functions, and it is a goal of this dissertation to investigate how differences in endothelin receptors determine arterial and venous function. 1. Blood vessel composition Blood vessels are hollow organs comprised of three layers of cells (Figure 1). A single layer of endothelial cells line the lumen of blood vessels and define the intimal region of a blood vessel. The outermost layer of the blood vessel, the adventitia, comprised primarily of fibroblasts, is a highly collagenous layer providing structural integrity to vessels. Smooth muscle cells are present in the medial layer of a blood vessel, between the intima and adventitia. The smooth muscle cells of blood vessels are contractile cells that allow vessels to contract and relax to various neuro-humoral stimuli. Between the layers of smooth muscle are elastin fibers, which like the collagenous adventitia are critical for structural support, but also provide elastic recoil to accommodate stretching of the blood vessel. Recent studies suggest that in addition to medial smooth Tunica intima Tunica media Tunica adventitia Carry blood away from Carry blood away from the heart to organs organs and tissues and tissues back to the heart Flgure 1. Rendered picture of arteries and veins. The tunica intima is the innermost layer of blood vessels. The tunica media, the middle layer, is where contractile smooth muscles cells are located. The tunica adventitia is the outermost layer, comprised primarily of collagen. muscle cells, the adventitia may contribute to changes in vascular tone (Laflamme ef al, 2006).Unique to veins are valves, which assist in returning blood to the heart. While the structure of blood vessels can be generalized by the presence of the intima, media and adventitia, a closer histological analysis of the structure of arteries and veins reveals several differences in arterial and venous structure. Masson trichrome staining of histological sections of rat thoracic aorta and thoracic vena cava, delineates the presence of smooth muscle (light pink), collagen (blue staining in the adventitia), nuclei (dark purple) and elastin fibers (bright red inbetween smooth muscle layers) (Figure 2). In aorta, multiple layers of smooth muscle cells are clearly defined between the collagenous adventitia and the single layer of endothelial cells lining the lumen of the blood vessel. The vena cava is highly collagenous, with a single layer of endothelial cells lining the intima. lmmunohistochemical analysis of smooth muscle a-actin expression reveals the presence of smooth muscle in between elastin fibers in aorta and a very thin layer of smooth muscle cells in vena cava (Figure 3). B. Function of arteries and veins 1. Determinants of blood pressure While these three layers of cells are common to both arteries and veins, how these components are organized to function together likely determines differences in arterial and venous function. Arteries typically have thicker layers Vena cava H AdventitialMedia? Flgure 2. Histological cross-sections of rat thoracic aorta and thoracic vena cava stained with a Masson trichrome stain. The three layers of a blood vessel are clearly defined in the aorta (as indicated by arrows) but are not obvious in vena cava. Adventitia Media mama-r:— Vena cava <——> Adventitia/Media? Flgure 3. Alpha actin immunohistochemistry of rat thoracic aorta and vena cava. Arrows indicate smooth muscle a-actin antibody binding (the darker purple areas in between elastin fibers in aorta, and the thin, subendothelial layer staining in vena cava), delineating where smooth muscle is located aorta and vena cava. of smooth muscle than veins and express more smooth muscle a-actin (a key contractile protein) per microgram of total protein (Figure 4). Arterial smooth muscle contraction or relaxation causes changes in arterial diameter, or tone. If the circulatory system is thought of as an electrical circuit, arteries determine resistance, while veins determine capacitance. Resistance is the impedance to flow, thus increases in arterial tone (or decreases in arterial lumen diameter) increase vascular resistance, and reductions in arterial tone (l.e. increases in arterial diameter) decrease resistance. Capacitance is the ability to store charge, and as functional capacitors, veins act as reservoirs for storing blood and contain approximately 60-70% of the total blood volume. Factors that determine arterial blood pressure include total peripheral resistance, determined by the tone or contraction of arteries, and cardiac output, the volume of blood pumped out of the heart per minute. Cardiac output is determined by heart rate, how fast the heart contracts, and stroke volume, the volume of blood ejected from the heart with each contraction. Elevated arterial blood pressure can lead to organ damage, such as renal failure, stroke, myocardial ischemia and myocardial infarction, while low blood pressure can cause insufficient organ perfusion and organ damage due to hypoxia. Thus, it is of critical importance for the body to maintain a homeostatic blood pressure that does not induce end- organ damage, but allows for adequate organ perfusion. Aorta Vena Cava 1234567891011. 1234567891011 40 ROM W40 kDA (N=11) a-actin den sitometry (arbitrary units) Vena Cava Figure 4. Western blot analysis of smooth muscle a-actin expression in rat thoracic aorta and vena cava lysates. Top: representative Western blots of a- actin expression; bottom: quantitation of a-actin expression. Fifty micrograms of protein (as measured by BCA protein assay) from aortic and venous lysates from 11 different rats (N=11) were loaded in each well, electrophoresed, transferred to PVDF membrane and a-actin detected using a primary antibody specific for a- actin and secondary antibody conjugated to horse radish peroxidase. 2. Mechanisms of blood pressure regulation Acutely (from seconds to minutes), blood pressure is regulated by the baroreceptor reflex. Changes in arterial pressure alter the firing rate of pressure- sensitive baroreceptors located in the aortic arch and carotid sinus, signaling regions in the brain (ie. rostral vental lateral medulla, RVLM) that control sympathetic outflow to control the force and rate of cardiac contraction and venous return to ultimately alter blood pressure. A more chronic mechanism to control blood pressure is through regulation of blood volume through alterations in natriuresis (Figure 5). Decreases in blood pressure activate the renin- angiotensin system to increase sodium and water retention and increase blood volume. Increased blood volume increases stroke volume and cardiac output, thus increasing blood pressure (Boron and Boulpaep, 2003). While our bodies have developed regulatory mechanisms for controlling blood pressure, there are times when these systems fail to keep blood pressure constant and high blood pressure can cause end-organ damage such as renal failure, stroke, and heart failure. There many clinically approved pharrnacotherapies for the treatment of hypertension, including diuretics, beta- blockers, calcium channel blockers, angiotensin (AT1) receptor blockers/angiotensin converting enzyme (ACE) inhibitors, etc., but typically monotherapy is not adequate to maintain blood pressure at levels recommended by The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure (JNC Vll). Since Fluid intake Non—renal fluid loss I I Rate of change of extracellular fluid volume I I... l_ ....... ular fluid volume Arterial —9 pressure L $ if MAP Blood Total G—- I' I i n volume peripheral resistance f $ ' Mean circulatory Cardiac (I T Venous T ' . T output return :I fllllng pressure i I Cardiac inotropy T [ Venoconstriction l ‘ I Vascular capacity lO—-—J Flgure 5. Guyton-Coleman diagram of physiological factors that affect pressure natriuresis and control blood pressure, demonstrating how increased venoconstriction can drive increases in cardiac output to ultimately increase arterial pressure. MAP, mean arterial pressure; UNaV, urinary sodium excretion. hypertension is a risk factor for cardiovascular disease and cardiovascular disease is the number one cause of death in the United States, clearly a better understanding of this disease and more effective treatments for hypertension are needed. C. Role of veins in blood pressure regulation The majority of the studies to date on how vascular smooth muscle tone and structure affect blood pressure have been performed in arteries, as they are primarily responsible for controlling total peripheral resistance. As mentioned previously, blood pressure can be increased by either increasing blood volume or by increasing vascular resistance by vasoconstriction (narrowing of the arterial lumen diameter) (Boron and Boulpaep, 2003). However, the mechanisms by which chronically elevated blood pressure, I.e. hypertension, develop are more complex than simply increasing blood volume or arterial vasoconstriction. During the development of human essential hypertension, cardiac output increases but not total blood volume. The increase in cardiac output is attributable to increased effective blood volume or the blood volume relative to vascular capacitance, a measure of venous compliance (or contractility) (Safar et al, 1975). Reduced vascular capacitance or increased venous tone (decreased venous compliance) is observed in some animal models of hypertension as well as human hypertension (Ricksten et al, 1981). Specifically, in mineralocorticoid hypertension, mean circulatory filling pressure (MCFP), an in vivo measure of venomotor tone, is elevated suggesting that venous capacitance is reduced 10 (venous contractility is increased) in hypertension (Johnson et al, 2001; Palacios et al, 1997). MCFP is measured when the heart is momentarily stopped and pressure throughout the circulatory system equilibrates. The two physiological determinants of MCFP are venous capacitance and blood volume, such that either decreases in venous capacitance or increases in blood volume lead to increases in MCFP. In human hypertension (Safar et al, 1975) and some animal models of hypertension (Ricksten et al, 1981), total blood volume is not increased, suggesting that the measured increase in MCFP is not driven by changes in blood volume, but changes in venous tone. In hypertension, the cause(s) of decreased vascular capacitance are still under investigation (Johnson et al, 2001; Palacios et al, 1997). I contend that veins play an important role in raising blood pressure by causing a shift of blood volume from the abdominal cavity into the thoracic cavity, increasing venous return to the heart and increasing cardiac output (Figure 5). D. Humoral control of blood pressure by circulating peptides The regulation of blood pressure is complex as multiple physiological processes are involved, including the autonomic nervous system, the kidneys and circulating hormones. These physiological processes do not operate independently, but instead modulate each other’s activity and act in concert to maintain blood pressure homeostasis. Circulating peptides involved in blood pressure regulation include vasopressin, neuropeptide Y, angiotensin II, bradykinin, the natriuretic peptides and endothelin-1 (ET-1). Vasopressin, 11 neuropeptide Y, angiotension II and ET-1 are all vasoconstrictors in mammals and elevate blood pressure by increasing total peripheral resistance, while bradykinin and the natriuretic peptides are vasodilators. However, these peptides can also influence sympathetic nervous system activity and sodium and water retention in the kidney. Synthesis and degradation of the various peptides involved in blood pressure regulation are tightly controlled as these systems are physiological compensatory mechanisms designed to maintain constant blood pressure (Boron and Boulpaep, 2003). Likewise, dysfunction in the biosynthetic pathways of any of these vasoactive peptides can cause significant fluctuations in blood pressure. A number of cardiovascular diseases, including hypertension, stroke and atherosclerosis involve alterations in the synthesis of vasoactive hormones, thus understanding the pharmacological and physiological function and regulation of these systems is paramount to understanding the pathophysiology and treatment of these diseases. E. Endothelin Endothelin -1 (ET-1) is a potent endogenous vasoconstrictor and is a peptide in the endothelin family of vasoconstrictor peptides discovered by Yanagisawa and co-workers in 1988. The three endothelin isoforrns, encoded by separate genes, - ET-1, ET-2 and ET-3 — vary in amino acid sequence and length. Endothelin exerts its physiological response via binding to receptors, of which there are two, the endothelin A (ETA) and the endothelin B (ETB) receptor; both ETA and El]; receptors are G-protein coupled receptors (GPCRs). The endothelin isoforrns 12 vary in their affinity for the endothelin receptors; ET-1 has higher affinity for ETA receptors than ET-3, while ET-1 and ET-3 have similar affinities for the ETB receptor (Kedzierski and Yanagisawa, 2001; Decker and Brok, 1998; Masaki, 2004). ET-1, ET-2 and ET-3 are present in plasma, but ET-1 is the only endothelin found in the vasculature (Kedzierski and Yanagisawa, 2001). ET-1 binds both ETA and ETB receptors with similar affinities (Kr approximately 20 and 15 pM, respectively) (Lee et al, 1998). Another family of peptides, the sarafotoxins 86b and 86c, isolated from asp (Atractaspis sp. of snake) venom are structurally similar to the endothelins and are agonists at endothelin receptors (Kloog and Sokolovsky, 1989; D'Orleans-Juste et al, 2003). 1. ET-1 synthesis and degradation Biosynthesis of ET-1 initiates with transcription of the ET-1 gene that encodes for preproET-1, a 212 amino acid peptide (Figure 6). PreproET-1 is cleaved to proET-1 by a furin-like peptidase, and then proET-1 is cleaved to big ET-1 by a subtilisin-like convertase. Big ET—1, the precursor to vasoactive ET-1, is a substrate for the protease endothelin converting enzyme (ECE) (D’Orleans-Juste et al, 2002). ECE, a metalloendoprotease that is structurally related to neutral endopeptidase (NEP), occurs in three isoforrns: ECE-1, ECE-2 and EOE-3, of which EOE-1 is predominantly physiologically relevant as EOE-3 specifically forms ET-3 and knockout of EOE-2 has no developmental effects (D’Orleans- Juste et al, 2003). There are four ECE—1 isoforrns: EOE-1a, ECE-1b, ECE-1c 13 ET-1 mRNA I Pre-proET-1 (212 amino acids) *MMET- 1* Big ET-1 (38 amino acids) are all: as (21 amino 1acids) *WETJU'M] ET-1[1-32] CONTRACTION Flgure 6. Diagram of endothelin-1 (ET-1) biosynthesis. Multiple enzymes are capable of processing big ET-1 into different vasoactive endothelin peptides, though ECE cleavage of big ET-1 is the classical pathway for ET-1 synthesis/big ET-1 metabolism. ECE, endothelin converting enzyme; MMP, matrix metalloprotease; NEP, neutral endopeptidase. 14 and ECE-1d due to four different promoters on a single gene (Yanagisawa ef al, 2000).The different ECE-1 isoforrn expression is due to alternative splicing and the isoforrns differ in their N-terminal sequences and subcellular locations; ECE- 1a is located in the cytosol, nucleus and plasma membrane, ECE-1 b is located in the cytoplasm, ECE-1c is located at the plasma membrane and ECE-1d is present in several subcellular locations (Jafri and Ergul, 2003). ECE cleaves big ET—1 (39 amino acids long) at the VaI21-Trp22 bond into active ET-1 (21 amino acids long). Big ET-1 is also a substrate for chymase, which cleaves big ET-1 at the Tyr31-Gly32 bond to form ET-1[1-31] (Nakano et al, 1997), which is vasoactive in rat thoracic aorta (Kishi et al, 1998; Watts of al, 2002) and vena cava (Watts et al, 2002). ET-1[1-31] can be proteolytically cleaved by NEP and/or ECE to form ET-1 in some tissues (Kloog and Sokolovsky, 1989). In rat aorta, phosphoramidon, a non-specific ECE/NEP inhibitor does not reduce ET-1[1-31]- induced contraction, suggesting that in this vessel, ET-1[1-31] is not cleaved to ET-1 to cause contraction (Kishi et al, 1998). Big ET-1 is also a substrate for matrix metalloproteinase-2 (MMP-2) (Fernandez-Patron ef al, 1999). MMP-2 is responsible for the digestion of extracellular matrix proteins including collagen, laminin, elastin and fibronectin and cleaves big ET—1 at the Gly"“’-Leu33 bond to form ET -1[1-32]. ET-1[1-32] is vasoactive and contracts rat mesenteric arteries (Fernandez-Patron et al, 1999). In some tissues, NEP degrades ET-1 (D’Orleans-Juste et al, 2003; Kedzierski and Yanagisawa, 2001), though in rat thoracic aorta and vena cava, the non-specific ECE/NEP inhibitor phosphoramidon reduced big ET—1-induced contraction (Figure 7) suggesting 15 A. 100 . Aorta +VehIcle -D- Phosphorarnidon (10 (M) c 75- ("=5 rte mg 5’: 5.5. 5°“ 22 d) :5. n_ * S 25‘ 0- . -12 -11 -1O -9 -8 -7 -l5 log big ET-1 [M] B. 700 Vena Cava -e-Vehicle eooe -o-Phosphoramldon (10 pM) c (N=5) “1% 5(1)! 2 m 8,13 4004 £38 300 §3 ‘ Q :1. 0'2 MT 100- o. -12 -11 -10 -9 -8 -7 -6 logbig ET-1[M] Flgure 7. Effect of phosphoramidon (10 IIM), an endothelin converting enyzme inhibitor, on big ET-t-induced contraction of rat thoracic aorta (A) and vena cava (B). Points represent means a: SEM for the number of animals (N) indicated in parenthesis. statistically significant differe nce from Vehicle (p<0.05). 16 NE, norepinephrine; PE, phenylephrine. Asterisks represent a that ECE and possible NEP are primarily involved in cleavage of big ET-1 into vasoactive ET-1. Thus, the synthesis of ET-1 relies on several proteolytic enzymes of which the activity and presence may differ between arteries and veins. F. ET-1 signal transduction 1. ETA and ETa receptors are G-protein coupled receptors The ETA and ETB receptors are G-protein coupled receptors (GPCRs). GPCRs couple to heterotrimeric G-proteins comprised of an a subunit and a By subunit that are tightly bound together. The a subunit of the heterotrimeric G-protein binds guanine nucleotides such as GDP and GTP and has intrinsic GTPase activity. Ligand binding to the GPCR induces a conformational change in the receptor and a subunit, allowing the active at subunit to hydrolyze GDP for GTP and dissociate from the By subunit. Conventionally, the GTP-bound a subunit is the primary mediator of GPCR signaling, but the dissociated By subunit is also capable of activating downstream signaling cascades and eliciting cellular responses. ET-1 activates multiple signaling pathways, the activation of which may account for the multitude of physiological responses mediated by ET-1 in vascular smooth muscle cells including, but not limited to vasoconstriction, mitogenesis, Na“/H+ exchange, and anti-apoptosis (Kitamura et al, 1999; Decker and Brok, 1998). The myriad of signaling pathways activated by ET-1 include, but again are not limited to, phospholipase C B, phospholipase A2, phospholipase 17 Ii A - Gun 6012/13 Gui/Gas (36‘! l I I \ PLCfi RhoA AC ERK, I N I I ”Sim IP3 DAG Rho Kinase PKA \ J V CONTRACTION Flgure 8. Diagram of endothelin signal transduction via ET, and ET B receptors. AC, adenylate cyclase; DAG, diacyl glycerol; Erk, extracellular regulated kinase; lPa, inositol 1,4,5-triphosphate; MAPK, mitogen activated kinase; Pl3-K, phosphatidyl inositol 3-kinase; PLC, phospholipase C; PKA, protein kinase A; PKC, protein kinase C. 18 D, the MAP kinases (ERK 1/2, JNK and p38), rho kinase, CAMP-mediated signaling and tyrosine kinase-mediated signaling (specifically Src signaling) (Kitamura ef al, 1999; Decker and Brok, 1998) (Figure 8). 2. ET, and ET; receptor signal transduction The ETA and ETB receptors are capable of coupling to several G-proteins to activate a number of signaling cascades (Figure 9, 10). The primary mechanism for vascular ET-1-induced contraction is most likely via ETA and ETB receptors coupling to Gag, a G-protein that activates phospholipase C B (PLCB). PLCB hydrolyzes phosphatidylinositol 4,5-biosphophate (Ple) into inositol 1,4,5- triphosphate (IP3) and sn1,2—diacylglycerol (DAG). IP3 binds to IP3 receptors in the sarcoplasmic reticulum to cause release of intracellular calcium stores, while DAG activates protein kinase C (PKC). ET-1 typically causes a biphasic increase in intracellular calcium; the initial transient rise in intracellular calcium is ng-mediated calcium release from intracellular stores, while the second sustained rise in intracellular calcium occurs via PKG-mediated influx of extracellular calcium (Kitamura et al, 1999). When ETA and ETB receptors are over-expressed in cell lines, the receptors can also couple to Go. and Gas, G-proteins that inactivate and active adenylate cyclase, respectively. When human ET, and ETB receptors are expressed in Chinese hamster ovary (CHO) cells, ET A receptors couple to Go... while ETB receptors couple to Get. (Aramori and Nakanishi, 1992; Takagi et al, 1995). Get,3 and Go. increase and inhibit adenylate cyclase, respectively, affecting cAMP 19 ET, receptor: NADPH Gus Gag Gatzm oxidase I f AC 1' \ RIP" I I CA1”? I \ Rho Kinase 1 IP DAG ' 3 (“i i PKA l l Myosin light H202 Caz. “fleas, “om PKG chaln phosphatase H Intracellular stores Influx of Vascular SI'I'IOOU'I Vascular smooth muscle growth muscle growth extracellular Cal" Vascular smooth muscle contraction< W Figure 9. Diagram of ET, receptor signal transduction. AC, adenylate cyclase; cAMP, cyclic adenosine monophosphate; H202, hydrogen peroxide; IP3, inositol triphosphate; 02’, superoxide anion; PLC, phospholipase C, PKA, protein kinase A; (-), inhibition; 1, activation. 20 ETl3 receptor: Got, 657 631;;13I I \ fa \. l I A? / \ chg RhoA Ia...» Ep'E’SZMi'I’é Pram / \ i Rho Kinase I I ii=3 DAG I P z l 1 H K\ PKC Cal: release from Myosin llght . l, Intracellular stores 1 chain phosphatase Vascular smooth Influx of muscle growth extracellular Ca” 1, / W T" Z > Vascular smooth muscle contraction < Flgure 10. Diagram of ET a receptor signal transduction. AC, adenylate cyclase; cAMP, cyclic adenosine monophosphate; DAG, diacylglycerol; ERK, extracellular regulated kinase; H202, hydrogen peroxide; lPa, inositol triphosphate; JNK, c-Jun N-terminal kinase; MAPK, mitogen activated kinase; 02‘, superoxide anion; PLC, phospholipase C, PKA, protein kinase A; PKB, protein kinase B, PKC, protein kinase c; (-), inhibition; 1. activation. 21 production and protein kinase A (PKA) activity. PKA activity in vascular smooth muscle cells inhibits mitogenesis via inhibition of the ERK pathway and induces vasodilation (Bornfeldt and Krebs, 1999). Thus, predicting PKA-mediated responses depends on tissue expression of ET, and ETB receptor and receptor- mediated G-protein coupling, factors which have not yet been determined in blood vessels or smooth muscle cells. Another class of G-proteins activated by ET—1 is the 612113 family. In human embryonic kidney (HEK)—293 cells, over-expressed ETB receptors couple to Gaia (Kitamura et al, 1999) while in embryonic mouse fibroblasts microinjected with plasmids encoding for the human ETA receptor, ETA receptors couple to 6012 (Gohla ef al, 1999). Intestinal smooth muscle cell ETA receptors couple to (3013 (Hersch et al, 2004), thus receptor-coupling to either Gan or Gaia may be cell- specific as different cell types may have different complements of G-proteins. 60.12/13 signaling mediates a number of other physiological responses including RhoA activation, regulating Na‘IH“ exchanger (NHE) activity, extracellular regulated kinase (ERK) activity, c-Jun N-terminal kinase (JNK) activity and nuclear factor IcB (NFIcB) activity (Kitamura et al, 1999; Gohla et al, 2000). RhoA is a monomeric G-protein that activates rho kinase, a kinase that phosphorylates and inactivates myosin light chain phosphatase (MYPT), increasing calcium sensitivity and allowing smooth muscle contraction to occur at concentrations of intracellular calcium that are typically sub-threshold for eliciting contraction (Somlyo and Somlyc, 2003). RhoA can be activated by a number of 22 G-proteins including Gotq and Gan/13 (Gohla et al, 2000). Contractility studies using isolated blood vessels suggest that vascular smooth muscle contraction to ET-1 is partially mediated via RhoA and activation of the rho kinase pathway (Hersch et al, 2004; Miao ef al, 2002). It is likely that ET A and ETB receptor activation of the rho kinase pathway can occur via coupling to Getq and Gong/13. Interestingly, cAMP and cGMP appear to inhibit the rho kinase pathway (Gohla et al, 2000; Dong et al, 1998). Thus, ET-1 activation of rho kinase may occur through several mechanisms, including direct activation of Gag by both ETA and ETB receptors, direct activation of 6012/13 by ETA and ETB receptors, or indirectly through ETB receptors, which via coupling to Go. decrease cAMP production. However, in smooth muscle cells with functional ET, receptors, ETA receptor coupling to Gas may reduce ETB receptor-mediated increases in rho kinase activity. The a subunits of G-proteins are not the only subunit responsible for GPCR signaling; the dissociated By dimer is also involved in ET-1 signaling. The cytoplasmic tail of the ETB receptor is involved in ET-1-induced activation of the ERK1/2, JNK and p38 MAP kinases, likely through By signaling (Aquilla ef al, 1996). As mentioned previously, ETB receptors activate Goo, causing the a and By subunits to dissociate. While the Ga. subunit decreases adenylate cyclase activity, decreasing cAMP levels and PKA activity, the dissociated By subunit is also capable of acting as a signaling molecule and can activate the protein kinase B (PKB)IAkt pathway in sinusoidal endothelial cells (Liu ef al, 2003). ET-1 23 activation of the PKB/Akt pathway in rabbit basilar artery is coupled to ET-1- induced contraction likely via activation of a MAPK pathway (Zubkov et al, 2000). ETa receptor-mediated Got. signaling can indirectly activate PLCB via the By subunit dimer (Decker and Brok, 1998). There are multiple signaling pathways that ETA and ETB receptors activate, thus there is significant potential for biochemical interactions of these signaling pathways in tissue populations with both ETA and ETB receptors. 3. Reactive oxygen species signaling Reactive oxygen species (ROS) such as superoxide anion (02') and hydrogen peroxide (H202) are formed by EM activation of El', receptors in smooth muscle cells (Li ef al, 2003a; Wedgwood ef al, 2001) and ET, receptors in endothelial cells (Duerrschmidt at al, 2000). El', receptors on smooth muscle cells of arteries and veins activate NADPH oxidase, an enzyme that generates 02'. Superoxide is catalytically broken down into H202 via the action of superoxide dismutase (SOD) of which there are 3: Cu/ZnSOD (extracellular and cytosolic) and MnSOD (mitochondrial). H202 is broken down by the enzymes catalase and glutathione peroxidase to form H20 and 0,. Both O,‘ and H202 can function as signaling molecules and can induce vasoconstriction (02' and H202), vasodilation (H202), and mitogenesis (02' and H202) (Wolin at al, 2001 ). Superoxide and H202 activate a number of signaling pathways that ET-1 activates and these ROS may mediate a number of the physiological affects of ET-1. ET-1 administered chronically to rats increased blood pressure by 18-20 mm Hg and increased renal 24 vascular resistance; tempol, a superoxide dismutase mimetic, reversed the effects of ET-1 on both blood pressure and renal vascular resistance (Sedeek ef al, 2003). These results suggest that the overall function of ET-t-stimulated ROS is vasoconstrictive. Li et al and other groups (Li et al, 2003a; Li et al, 2003b; Callera at al, 2003) observed that ET-1 increases aortic and venous 0,; via NADPH oxidase and that the ET, receptor mediates activation of NADPH oxidase. In DOCA-salt hypertension, where aortic tissue ET-1 is elevated, O,‘ levels are elevated and decreasing oxidative stress reduces blood pressure and improves hypertension- related tissue damage such as endothelial cell damage. Guzik et al observed no differences in basal superoxide levels in human saphenous vein and internal mammary artery (Guzik at al, 2004). Interestingly, the source of 02' varied between human arteries and veins. In human saphenous vein NAPDH oxidase was the primary source for 02' generation, while the xanthinelxanthine oxidase system generated 02' in human internal mammary arteries (Guzik at al, 2004). It is possible that ET-1 may differentially affect the ROS production and downstream signaling events in arteries and veins. G. Distribution of ET A and ET; receptors in blood vessels Both ETA and ETB receptors are present on vascular smooth muscle cells of blood vessels (Figure 11, 12) and couple to smooth muscle contraction (Figure 13), while ETB receptors on endothelial cells couple to nitric oxide release. 25 ETA ET, #I 120 kDa —> l». y . dimer? dimer? ~ —- --—- --- 45 kDa _. [:53 30 kDa _. 1o 10 + Competing . a... peptide 10 10 + Competing peptide Flgure 1 1 . El', and ET, receptor Western blot analysis in rat thoracic vena cava. Proteins from venous lysates were separated using SDS-PAGE, and transferred to PVDF. Membranes were probed with primary antibodies (1°) directed to ET, (left lanes) and ET, receptors (right lanes) or primary antibody neutralized with competing peptide (1° + competing peptide). Bands that are lighter in the presence of competing peptide represent specific primary antibody binding. The expected molecular weight of both the ET, and ET, receptor is around 45 kDA. 26 Aorta Vena cava < .2. I— Z ET, Receptor ADVENTITIA \ i.“ .0 . I ire ’5 Flgure 12. ET, and ET, receptor immunohistochemistry in rat thoracic aorta (left) and vena cava (right). Tissues are oriented with adventitia on the left and intima on the right. Black precipitate, as indicated by arrows, represents specific primary antibody binding. 27 Aorta .3" § (N=5-6) is Percentage PE/NE (10 pM) Contraction .8 Q- -12 -11 -10 5 -i3 logET-t [M] -l U) Aorta F” A v (Nfi) Percentage PEINE (10 pM) Connection Ou—W -12 -11 -1o -9 -s -7 IOQSGCIMI 700 Vena Cava coo (N=5-6) 500. 400. 300. 200.. 100.. 0‘ I T U I -12 -11 -1o -9 -8 -7 -45 IogET-1[M] Vena Cava 7.5 (N=4) 501 25. o. , , -12 -11 -10 -9 -e -7 I09 360 [Ml Flgure 13. ET -1 (A) and 860 (B) cumulative concentration response curves in rat thoracic aorta and vena cava. Points represent mean :1: SEM for the number (N) of animals in parenthesis. ET-1, endothelin-1; NE, norepinephrine; PE, phenylephrine; S6c, sarafotoxin 6c. 28 Endothelial ETB receptors also function as a clearance mechanism to remove plasma ET-1 (Bohm et al, 2003). Endothelin receptors are present in a number of other non-vascular tissues including renal collecting duct cells, cardiomyocytes, hepatocytes, neurons, osteoblasts, keratinocytes and adipocytes (Kedzierksi and Yanagisawa, 2001). On blood vessels, receptor expression and coupling to contraction is dependent on the vascular bed and species studied. ET, and ETB receptors that both participate in contraction are present in rabbit jugular vein (Lodge ef al, 1995), rabbit saphenous vein (Lodge at al, 1995), rat thoracic vena cava (Watts et al, 2002), rat superior mesenteric vein (Claing et al, 2002), rat small mesenteric arteries (Adner ef al, 2001) and hamster thoracic aorta (Lodge at al, 1995), while in other blood vessels such as rat thoracic aorta (Watts of al, 2002; Lodge et al, 1995), rabbit carotid artery (Lodge et al, 1995) and rat superior mesenteric artery (Claing et al, 2002) only the ET, receptor mediates ET-1-induced contraction. A few of studies have suggested that contractile ETB receptors are preferentially localized to veins, while ET-1-induced contraction is mediated by ET, receptors on arteries (Ekelund et al, 1994; Moreland et al, 1994). A recent study performed in human forearm veins suggests that contractile ETB receptors on veins are an important overlooked mechanism for increasing venous return and increasing blood ‘ pressure (Mitchell et al, 2004). The presence of both ET, and ETB receptors on rat thoracic aorta and vena cava has been confirmed using Western blot analysis (Watts of al, 2002; Figure 11) 29 and immunohistochemistry (Thakali et al, 2004; Figure 12). Both rat thoracic aorta and vena cava contract to ET-1, a non-selective ET, and ETB receptor agonist, while vena cava but not aorta contract to 86c, an ETa receptor agonist (Figure 13). Why ETB receptors are present in rat thoracic aorta, but are not coupled to contraction is not understood, but it is also possible that smooth muscle contraction is not the correct endpoint for assessing ETB receptor activity in rat thoracic aorta. H. ET, and El'a receptor function in hypertension The endothelin receptors present on vascular smooth muscle cells of arteries and veins are important in blood pressure regulation. Systemic administration of endothelin receptor antagonists reduce blood pressure in healthy, normotensive people, suggesting that ET—1 is normally involved in control of vascular tone (Haynes et al, 1996; Verhaar et al, 2000). The endothelin system also plays a role in both human hypertension and several animal models of hypertension. African American hypertensive patients have significantly higher plasma ET-1 levels than their normotensive counterparts and Caucasian hypertensive patients (Grubbs et al, 2002). In DOCA-salt hypertension, aortic but not venous tissue levels of ET-1 are elevated, and aorta but not vena cava from DOCA-salt hypertensive rats exhibit reduced contraction to ET-1 (Watts of al, 2002). Recent studies suggest that the ET, receptor is down regulated in human and animal models of hypertension, leading to impaired ET-1 mediated arterial contraction, while ETB receptor expression is not reduced (Reinhart et al, 2002; Telemaque— 3O Potts et al, 2002). Giulumian et al reported that decreased Ca2+ influx, but not reduced ET-1 binding to its receptors, could explain impaired contraction to ET-1 in the small coronary arteries of DOCA-salt rats, suggesting an uncoupling of the ET-1 signaling mechanism (Giulumian ef al, 2002). Lodge et al (1995) and Moreland et al (1996) suggest that ET, receptors mediate contraction on most arteries and ET; receptors mediate contraction on most veins. Mitchell ef al report that hand vein contraction to ET-1 is not reduced by an orally administered ET, receptor antagonist (Mitchell ef al, 2004). Lack of arterial responsiveness to ET-1 under high ET-1 conditions suggests that vasoconstrictive ET; receptors present on veins may play an underestimated and underappreciated role in regulating blood pressure. I. ET, and ET; receptor “cross-talk” or interaction As mentioned previously, there are vascular beds where both ET, and ET; receptors mediate ET-1-induced contraction. Several reports also suggest “cross-talk” between ET, and ET; receptors occurs, meaning that activation of one receptor subtype alters the function of the other receptor subtype. In rabbit jugular and saphenous veins and hamster aorta (vessels that have contractile ET; receptors), ET-1-induced contraction is not reduced by ET, receptor antagonism. However, when the ET; receptors on these vessels were desensitized using S6c, an ET; selective agonist, ET-I-induced contraction was sensitive to ET, receptor blockade. Interestingly, in rat aorta and rabbit carotid artery (vessels that do not contract to ET; agonists), ET; receptor desensitization 31 using S6c did not alter ET, receptor blockade of ET-1-induced contraction (Lodge at al, 1995). Lodge et al suggest that in rabbit jugular and saphenous vein, ET-1 elicits contraction via both ET, and ET; receptors, but that under normal conditions, ET, receptors are under receptor blockade by ET; receptors. When ET; receptors are desensitized, ET, receptors are released from receptor blockade and are able to participate fully in ET-1-induced contraction. ET, and ET; receptor interaction has also been studied in the small mesenteric arteries from rats, in which both ET, and ET; receptors are contractile (Adner et al, 2001), unlike the superior mesenteric artery from rat in which only ET, receptors are responsible for ET-1-induced contraction (Lodge at al, 1995). In small mesenteric arteries, desensitization of ET; receptors using 86c increased the potency of ET-1 and the apparent affinity of FR139317, an ET, receptor antagonist (Adner et al, 2001). Endothelin receptor interaction has also been observed in mouse mesenteric veins but not arteries (Perez-Rivera et al, 2005), renal afferent but not efferent arterioles (lnscho ef al, 2005), and pulmonary arteries (Sauvageau et al, 2006). Thus, in tissues with mixed functional endothelin receptor populations, pharmacological data suggests that ET; receptors normally function to antagonize ET, receptors, but ET; receptor desensitization releases ET, receptors from receptor blockade. Whether this interaction between ET, and ET; receptors occurs at a physical or signaling level is not known. 32 One possible explanation for differences between aortic and venous responses to ET-1 is differences in G-protein complement in arteries and veins. To our knowledge, a study comparing G-protein complement in arteries and veins has not been performed to date. Adams et al compared gene expression of 4048 genes in macaque aorta and vena cava and observed that mRNA expression was elevated in 68 out of 4048 genes in aorta compared to vena cava, with the largest difference in mRNA expression occurring in a regulator of G-protein signaling (RGS) 5 (Adams et al, 2000). Northern blot analysis suggests that a similar pattern of increased RGS expression in aorta compared to vena cava occurs in the rat (Adams of al, 2000). RGS proteins are GTPase-activating proteins and function to inhibit GPCR signaling; specifically, RGSS binds and inhibits Ga. and Gaq-coupled signaling (Zhou et al, 2001), two G-proteins involved in ET-1 signaling. Over-expressed RGS5 inhibits ET-1-induced Ca2+ transients in HEK-293T cells, suggesting that RG85 can interfere with ET-1 signaling (Zhou et al, 2001). Higher RGSS expression in rat thoracic aorta compared to vena cava may inhibit ET-1 signal transduction and specifically may mask aortic ET; receptor-mediated signaling. J. Receptor dimerization 1. Physical means for receptor dimerization Conventionally, GPCR signaling is thought of as a ligand binding to its receptor and the receptor coupling to a G-protein in a 1:1 ratio, causing a specific response through effector molecules. However, recent investigations suggest 33 that a number of GPCRs are capable of homo— and heterodimerization (a receptor dimerizing with an identical receptor or dimerizing with a different receptor, respectively), altering the pharmacology of these receptors. Heptahelical GPCRs, such as glutamate (Romano et al, 2001), angiotensin (AbdAIla et al, 2001), B2 adrenergic (Hebert et al, 1996), and GABA; receptors can form homo— and heterodimers (White et al, 1998). Dimerization has been proposed to occur through N-terminal disulfide bonds such as in the case of the Ca2+ sensing receptor (Goldsmith et al, 1999; Pace et al, 1999) and the metabotropic glutamate receptor (Romano et al, 2001). Disulfide bonds in residues cys101 and cys263 allow Ca2+ sensing receptor homodimerization, and disruption of these disulflde bonds decreases agonist affinity for the receptors and slows the kinetics of receptor activation and inactivation (Goldsmith ef al, 1999; Pace et al, 1999). Dimerization can also occur through interactions in the C-terminus of receptors, as is the case for 6 opioid receptor homodimerization (Cvejic and Devi, 1997). Coiled-coil interactions through leucine-zipper motifs have also been proposed to facilitate receptor dimerization, such as in the case of the GABA; receptor (Kammerer et al, 1999). The final mechanism by which dimerization can occur is through transmembrane interactions as occurs with the DZ dopamine receptor. Using truncated DZ receptor mutants in COS-7 and Sf9 cells, Lee et a! determined that for DZ dopamine receptors, transmembrane 4 was critical for receptor dimerization (Lee et al, 2003). Z. Caveolae as cellular locations for receptor dimerization Caveolae play an important role in receptor trafficking as internalization of caveolae is inducible. The caveolin proteins (-1,2, and 3) function as scaffolding proteins in the vasculature, caveolin-3 is muscle specific, while caveolin-1 (the predominant caveolin) is required for caveolae formation and internalization. Tyrosine phosphorylation of caveolin-1 by c-src is the signal required for caveolin-1 to induce caveolae internalization (Stan, 2002). ET-1 activates a number of signaling molecules, including c-src (Kodama ef al, 2003), thus if ET receptors are localized to caveolae in aorta and vena cava, ET-1 could potentially mediate receptor internalization into caveolae. Caveolin-1 may also interact with filamin, an actin binding protein, suggesting that caveolae can potentially affect smooth muscle contractility (Dreja et al, 2002). Dreja et al have performed a number of experiments investigating the role of caveolae in 5-hydroxytryptamine (5-HT)—induced contraction in rat tail artery. They observed that 5-HT-induced contraction is dependent on caveolae as cholesterol depletion (which depletes smooth muscle cell membrane caveolae verified by electron microscopy) inhibits 5-HT-induced contraction. In the tail artery, cholesterol depletion also inhibits ET-1-induced contraction (Dreja et al, 2002). Thus, in rat thoracic aorta and vena cava, caveolae (Figure 14) may represent a cellular location for ET,/ET; receptor dimerization as well as a mechanism for 1 receptor internalization and desensitization under ET-1- stimulating conditions. 35 Flgure 14. Electron micrographs of caveolae (invaginations indicated by black arrows) present on the plasma membrane of aortic and venous vascular smooth muscle cells. 3. Functional consequences of receptor dimerization A number of studies suggest that several receptor characteristics such as agonist affinity, potency and efficacy as well as receptor internalization are affected by receptor dimerization. In the case of GABA; receptor dimerization, heterodimerization between the GABA;R1 and GABA;R2 receptors increases agonist affinity 10-fold, as was observed by inhibition of radiolabeled GABA; agonist binding by GABA and baclofen (Kaupmann et al, 1998). Agonist affinity is not always affected by receptor dimerization - heterodimerization between the B2 adrenergic receptor and the 6 and 1c opioid receptors does not affect ligand binding, as ligand binding affinity for each heterodimer pair was similar to ligand binding affinities in preparations with each receptor alone (Jordan et al, 2001), suggesting that functional consequences of receptor dimerization vary for each pairing. While ligand binding properties are unaffected by heterodimerization between the B2 adrenergic receptor and the 6 and 1c opioid receptors, receptor trafficking can vary, depending on the type of heterodimer formed. B2 adrenergic receptor heterodimerized with the 6 opioid receptor undergoes agonist-mediated internalization, while the B2/Ic receptor heterodimer does not undergo agonist- mediated internalization (Jordan at al, 2001). Other receptor characteristics that are potentially modifiable by receptor dimerization include agonist potency and efficacy. With heterodimerization of the 6 and 1c opioid receptors, a synergistic effect of dual agonist (DPDPE and U69593, 6 and 1c opioid receptor agonists, respectively) binding leading to increased effector function was observed (Jordan and Devi, 1999). When It and 6 heterodimers were expressed, 6 opioid receptor 37 antagonism increased potency and efficacy of II opioid receptor signaling, and vice versa (Gomes et al, 2000). To date, much of work published on receptor dimerization has occurred in cell lines in which receptors were artificially expressed. Whether receptor dimerization functionally affects tissues - in this case, ET receptors in arteries and veins - is an important question. 4. Can ET, and ET; receptor heterodimerization occur and can heterodimerization affect ET receptor function? Gregan et al demonstrated that ET, and ET; receptors heterodimerize in HEK- 293 cells over-expressing human ET, and ET; receptors, results that have since been confirmed by Del and Galligan (2006). Short or prolonged incubation with the non-specific agonist ET-1 had no effect on the ET,/ET; receptor heterodimer, suggesting that heterodimer formation was constituitive. Interestingly, prolonged incubation with an ET; receptor-selective agonist disrupted the ET,/ET; receptor heterodimer, suggesting that with selective ET; receptor activation, the heterodimer dissociates and ET; receptor internalization occurs (Gregan ef al, 2004). This study represents an excellent proof-of-principle study - ET, and ET; receptors can heterodimerize. However, heterodimerization was assessed in a highly artificial system and functional consequences of tissue ET, and ET; receptor heterodimerization cannot be deduced from these over-expression studies. Radioligand binding studies of ET, and ET; receptors in the rat anterior pituitary gland demonstrated that ET; receptor agonists and antagonists did not compete for K125’ET-1 binding unless ET, receptors were bound by an ET, 38 receptor antagonist. Thus, the authors suggest two models of ET, and ET; receptor heterodimerization that explain their data. In the first model ET-1 acts as a molecular bridge between the ET, and ET; receptors, inducing receptor heterodimerization and in the second model ET, and ET; receptors constituitively heterodimerize (Harada ef al, 2002). But like the proof-of-principle study performed in HEK-293 cells, data from these radioligand binding studies provide no information on the functional consequences of ET, and ET; receptor heterodimerization in arteries and veins. There is a wealth of data that highlights the importance of the endothelin system in several animal models of hypertension and hypertension in humans, and accordingly the endothelin system has been pursued as a potential drug target for lowering blood pressure. However, if the endothelin receptors dimerize, this could alter the pharmacology of each receptor and also alter the response to drugs designed to interfere with the system. Thus, by understanding how ET receptors interact, whether physically, pharmacologically or via biochemical signaling, and determining if this interaction occurs in a similar fashion in arteries and veins, rational drug therapies can be devised for treating endothelin- dependent hypertension. 5. Significance of ET, and ET; receptor dimerization Receptor dimerization is a fascinating field in pharmacology that could potentially change the way tissues with different receptor populations are typically 39 characterized. This dissertation represents a foray into a phenomenon that has not been highly described in whole tissue, as most of the work involving receptor dimerization has been performed in cell lines over-expressing receptors of interest. Thus, there are many questions as to how and if receptor dimerization affects tissue or system function. In a number of human diseases, such as systemic hypertension, pulmonary hypertension, coronary atherosclerosis, myocardial ischemia and reperfusion and myocardial infarction, ET-1 plays a pathogenic role. Understanding how the ET, and ET; receptors function normally in blood vessels and under conditions of elevated ET-1 may provide useful information on the treatment of such diseases and point here to an under- appreciated role of the venous system in blood pressure regulation. 40 K. HYPOTHESIS Veins maintain responsiveness while arteries lose responsiveness to the vasoactive hormone endothelin-1 (ET-1) in situations of exposure to ET -1 (eg. hypertension, experimental protocols). Thus, receptors for ET-1 - the heptahelical ET, and ET; receptors — may function differently in arteries and veins. While heptahelical receptors canonically interact with G-proteins in a 1:1 ratio, recent evidence suggests that receptor dimerization (hetero- or homo-) may occur. Receptor dimerization can potentially affect a number of receptor characteristics such as agonist affinity, potency and efficacy, as well as receptor internalization. Aligned sequence analysis suggests that the ET, and ET; can interact through N-terminal residues. ET, and ET; receptors are present on the smooth muscle of arteries and veins and previous studies suggest that activated ET; receptors are capable of physically or pharmacologically uncoupling ET, receptors from receptor blockade. I hypothesized that ET, and ET; receptors physically interact via recepter heterodimerization in veins but not arteries and this heterodimerization functionally affects ET, and ET; receptor pharmacology. To test this hypothesis I designed three subhypotheses: Subhypothesis 1: To demonstrate a pharmacological interaction between ET, and ET; receptors in rat thoracic vena cava but not aorta and to demonstrate that activated ET; receptors pharmacologically uncouple ET, receptors from receptor blockade in rat thoracic vena cava. 41 I expect that compared to arteries, veins will not desensitize to ET-1 because veins but not arteries have functional (i.e. contractile) ET; receptors. Because veins possess contractile ET; receptors to interact with ET, receptors, I expect that venous ET-1-induced contraction will be reduced by either ET, or ET; receptor antagonists and dual ET,/ET; receptor antagonism will cause a greater inhibition than ET, or ET; receptor antagonism alone. Subhypothesis 2: To characterize biochemical signaling interactions between ET, and ET; receptors in rat thoracic aorta and vena cava. I hypothesize that reactive oxygen species signaling will differ between arteries and veins because of the presence of contractile ET; receptors on veins but not arteries. I speculate that the presence of contractile ET; receptors on veins but not arteries will mean that the signal transduction pathways activated by ET-1 in veins will differ from those activated in arteries and contribute to functional venous endothelin receptor interaction. Subhypothesis 3: To demonstrate a physical interaction, Le. heterodimerization, between ET, and ET; receptors in rat thoracic vena cava but not aorta. I expect that single smooth muscle cell ET, and ET; receptor immunocytochemistry of rat thoracic aorta and vena cava will 42 demonstrate that ET, and ET; receptors co-localize on the membrane of venous vascular smooth muscle cells. 0 I expect that with ET-1 stimulation, venous ET, and ET; receptors will internalize to a lesser degree than aortic ET, receptors. l posit that ET, and ET; receptors will co-immunoprecipitate in rat thoracic vena cava but not aorta. 43 II. METHODS A. Isolated smooth muscle contraction Male Sprague Dawylet rats (225-250 9, Charles River Laboratories, Portage, MI) were deeply anesthetized (until loss of eyelid reflex and withdrawal from painful stimuli) with a pentobarbital injection (50 mg/kg, l.p). The thoracic aorta and vena cava were removed and placed in warmed (37°C), oxygenated (95% O2, 5% CO2) physiological salt solution (PSS) of the following composition: 103 mM NaCl; 4.7 mM KCI; 1.18 mM KH2PO4; 1.17 mM MgSO4-7H2O; 1.6 mM CaCl2- ZH2O; 14.9 mM NaHCO;; 5.5 mM dextrose and 0.03 mM CaNa2 EDTA. Arteries and veins were cleaned of fat and connective tissue and dissected into rings (34 mm) with the endothelial layer left intact. The rings were hung between wire hooks with one end attached to a stationary glass rod and the other end attached to a force transducer connected to a Grass polygraph for measurement of isometric contractile force. Vessels were placed under optimum passive tension (aorta: 4000 mg, vena cava: 1000 mg). Arteries and veins were equilibrated for one hour in PSS prior to a wake-up challenge with a maximal concentration of phenylephrine (PE) (10 (M) or norepinephrine (NE) (10 IIM), respectively. PE was used to wake-up aorta to compare results to historical data, but was not used to wake-up vena save as reproducible contractions to PE were unattainable in our hands. Endothelial integrity was assessed by acetylcholine (1 uM)- induced relaxation after contraction to PE (10 nM) or NE (10 (M) in aorta and vena cava, respectively. 1. ET-1 desensitization Aorta and vena cava were challenged with ET-1 (100 nM) for 15 minutes (when aortic ET—1-induced contraction plauteaued). Then vessels were washed with PSS for 90 minutes (5 minutes between washes), during which tone returned to baseline. Vessels were allowed to rest (no washing) for 30 minutes before performing either an ET-1 cumulative concentration response curve (10 pM — 100 nM), a norepinephrine concentration response curve (1 nM — 10 nM), or challenged with a bolus of $60 (100 nM). If antagonists were used (atrasentan, 10 nM; 30-788, 100 nM), they were added during the 30-minute rest period before performing ET-1 cumulative concentration response curves (10 pM — 100 nM). 2. ET; receptor desensitization Vena cava were challenged with a bolus of the ET; receptor agonist 86c (100 nM, 15 minutes). Vena cava were washed with PSS for 15 or 30 minutes (5 minutes between washes) and then vena cava challenged again with S6c (100 nM). To investigate ET, receptor function in ET; receptor desensitized aorta and vena cava, ET; receptors were desensitized with 86c (100 nM) and ET, receptors blocked with atrasentan (10 nM) for one hour and then cumulative ET- 1 concentration response curves (10 pM - 100 nM) were performed. 3. Receptor antagonism studies Aorta and vena cava were incubated with vehicle (0.0001% DMSO), ET, receptor antagonist (atrasentan, 10 nM), ET; receptor antagonist (BO-788, 100 45 nM) or dual ET, and ET; receptor blockade (atrasentan + BQ-788) for one hour and then ET-1 concentration response curves (10 pM — 100 nM) were performed. 4. H202 concentration response curves Cumulative concentration response curves to exogenous H202 (1 uM- 1 mM) were performed in rat thoracic aorta and vena cava. To determine if H202 mediated ET-1-induced contraction, aorta and vena cava were incubated with catalase (2000 UlmL), PEG-catalase (163 and 326 UlmL), or 3-aminotriazole (50 mM) for one hour before performing cumulative concentration response curves to ET-1 (10 pM — 100 nM). B. Measurement of arterlal and venous H202 productlon H202 production from rat aorta and vena cava was assessed using an Amplex Red® Hydrogen Peroxide Assay kit (Molecular Probes, Eugene, OR). Three to four millimeter segments of thoracic aorta and thoracic vena cava were dissected from rats, cleaned of fat, connective tissue and blood and then incubated in 500 LII. of modified Kreb’s buffer containing (in mM): HEPES, 20; NaCl, 119; KCI, 4.6; MgSO4 7H20, 1.0; Na,HPO,, 0.15; KH,P0,, 0.4; NaHCO,, 5, CaCl, 1.2; dextrose, 5.5) at 37°C for one hour. Antagonists or vehicle were incubated during the one hour equilibration. Buffer was carefully removed and 200 uL of Amplex Red® reaction solution was added to the tissues. Tissues were incubated with ET -1 (1, 10 and 100 nM) or ET-1 (100 nM) plus antagonists [atrasentan (10 nM): ET, receptor antagonist, BQ-788 (100 nM): ET, receptor antagonist or DDC (10 mM): SOD inhibitor] for 4 hours at 37°C. Li er al 46 previously determined that maximal ET-1-stimulated superoxide production occurred after a four-hour incubation (Li et al, 2003a; Li et al, 2003b), thus I chose to measure ET-1-stimulated H202 production after a four-hour incubation. Then the Amplex Red® solution was transferred to a 96-well plate and fluorescence emission was measured (excitation = 544 nm, emission = 584 nm) on a SpectraMax Gemini plate reader (Molecular Devices, Sunnyvale CA) using SoftMax®Pro software. A standard curve using known H202 concentrations was performed with each experiment and was used to determine H202 concentration. After each experiment, aortic and venous segments were homogenized and protein concentrations determined using the bicinchoninic assay (BCA) for protein determination. H202 production from rat aorta and vena cava is represented as nmol/mg protein or as a percentage of control response. C. Whole tissue immunohistochemistry Cleaned rings of thoracic aorta and vena cava were formalin-fixed (10%) overnight and paraffin-embedded at the histology lab at Michigan State University. Sections (5 micron) were cut and placed on glass slides. Slides were dewaxed (Z dips in xylenes; 3 minutes each), rehydrated (2 dips in 100% ethanol, 2 dips in 95% ethanol, 2 dips in 90% ethanol, 2 dips in deionized water; 3 minutes each) and unmasked (boiled for 5 minutes in unmasking solution in the microwave). Endogenous peroxidases were blocked by incubating sections for 30 minutes in 0.3% H202 (dissolved in phosphate buffered saline) at room temperature and non-specific secondary binding blocked by incubating sections 47 for 1 hour in 1.5% blocking serum (for the species the secondary antibody was raised in) at room temperature. Sections were incubated overnight (humidified chamber, 4°C) with primary antibody [ET, antibody (5 ug/mL, Alomone Laboratories), ET; antibody (5 uglmL, Alomone Laboratories)]. Sections were also incubated with primary antibody quenched with five times the concentration of competing peptide (solution made 2 hours before application). Sections were incubated with secondary antibody for 30 minutes (humidified chamber, room temperature) and with ABC reagent for 30 minutes (humidified chamber, room temperature, Vector Laboratories). Staining was detected using the diaminobenzidine peroxidase reagent (Vector Laboratories) and slides were counterstained with hematoxylin. Slides were allowed to air dry, coverslips mounted with Vectamount® and pictures taken with a Spot 2 digital camera on a Leica light microscope. D. Whole tissue protein isolation Thoracic aorta and vena cava were snap frozen in liquid nitrogen, pulverized in a liquid nitrogen-cooled mortar and pestle and solubilized in buffer (62.5 mM Tris, pH 6.8, 2% SDS wlv, 10% glycerol vlv) with protease inhibitors (1 mM PMSF, 100 uglml aprotinin and 100 jig/ml leupeptin). Homogenates were centrifuged (11,000 rpm for 10 minutes, 4°C) and supernatant collected. Total protein in the supernatant was measured using the BCA method of protein measurement (Sigma, St. Louis, MO). Samples were stored at -80 °C until use. 1. Cross-linking: Rat thoracic aorta and vena cava were isolated, dissected and 48 cleaned. Vessels were incubated with 5 mM disuccinimidyl tartrate (DST) (dissolved in 20 mM HEPES pH 8.0, 1 hour, room temperature, end-over-end mixing). The cross-linking reaction was stopped with 20 mM Tris (pH 7.5, 15 minutes, room temperature, with end-over-end mixing). Aorta and vena cava were snap frozen in liquid nitrogen and homogenized with a HEPES lysis buffer (20 mM HEPES pH 8.0, 150 mM NaCl, 1% SDS) according to the protein isolation protocol mentioned above. E. Western blot analysis 1. SDS-polyacrylamide gel electrophoresis (PAGE) (denaturing PAGE) Fifty micrograms of protein (either aorta or vena cava lysates) was diluted in 4:1 denaturing sample buffer (250 mM Tris pH 6.8, 5% SDS wlv, 0.005% bromophenol blue wlv, 45% glycerol v/v, 9.4% B-mercaptoethanol vlv), incubated for 10 minutes at room temperature, separated on 10% SDS-polyacrylamide gels and transferred (1 hour, 100 V) to lmmobilon P (PVDF) membranes. Membranes were blocked overnight (Tris-buffered saline + 0.5% Tween (T EST), 5% nonfat dry milk (Biorad), 0.025% sodium azide). Blots were probed overnight with primary antibody [ET,: 1:200 dilution (raised in rabbit, Alomone Laboratories), ET;: 1:200 dilution (raised in rabbit, Alomone Laboratories)] at 4 °C. rinsed in TBS-T, with a final rinse in TBS, and incubated with secondary antibody (anti- rabbit lgG, 1:1000 dilution) for 1 hour at 4 °C. After secondary incubation, blots were rinsed in TBS and then incubated with ECL® reagents to visualize bands. 2. SDS-urea PAGE 49 A standard SDS-PAGE was performed except for the addition of 8M urea to the standard 4:1 denaturing sample buffer, 10% SDS-acrylamide gels and the Laemmli running buffer. 3. Non-denaturing PAGE (NATIVE PAGE) A standard SDS-PAGE was performed except for the omission of SDS in 10% acrylamide gels and the standard 4:1 denaturing buffer. B-mercaptoethanol was also omitted from standard 4:1 denaturing buffer. F. Co-immunoprecipitation 1 . Traditional co-immunoprecipitation Cleaned thoracic aorta and vena cava were snap frozen in liquid nitrogen and ground in a mild homogenization buffer (62.5 mM Tris pH 6.8, 2% SDS wlv, 10% glycerol WM and freshly added peptidase inhibitors including 100 jig/ml aprotinin, 100 uglml leupeptin, 1 mM PMSF). Five hundred micrograms of protein (either aorta or vena cava lysates) was incubated with 2 ug ET, or ET; receptor antibody (Alomone Laboratories) for 2 hours at room temperature. Thirty microliters of protein AIG beads (Santa Cruz) were incubated with antibody/antigen complexes overnight at 4 °C with rocking. The beads were washed 3 times with phosphate buffered saline (pH 7.4) buffer, with centrifugation (1 minute, 2500 rpm) in between washes. Beads were boiled (96°C) for five minutes in 2:1 sample buffer (plus 9.4% B-mercaptoethanol) and centrifuged (1 minute, 2500 rpm). The supernatant containing the immunoprecipitates were loaded and separated with 10% SDS-PAGE. Samples 50 were electrically transferred (1 hour, 100 V) to lmmobilon P (PVDF), blots blocked overnight (T ris—buffered saline + 0.5% Tween (TBS—T), 5% nonfat dry milk (Biorad), 0.025% sodium azide) and then incubated overnight (4°C) with ET, and ET; receptor antibodies (1:200 dilution, Alomone Laboratories). Blots were developed according to standard western blot protocols. 2. True Blot® co-immunoprecipitation kit Rat thoracic aorta and vena cava were isolated, dissected and cleaned. Protein was isolated with a modified lysis buffer (50 mM Tris pH 8.0, 150 mM NaCl, 1% nonidet P-40 substitute) following the protein isolation protocol mentioned above. Five hundred micrograms of aortic and venous lysates were pre-cleared with fifty uL of TrueBIot rabbit lgG beads (3 min, 10,000 x 9). Five micrograms of antibody (ET,, Alomone Laboratories; ET;, Alomone Laboratories; caveolin-1, Transduction Laboratories) and pre-cleared lysates were incubated (2 hours, room temperature). Fifty microliters of TrueBIot rabbit lgG beads was added to the antibody-lysate complex and incubated overnight (4°C with end-cver-end mixing). Beads were washed three times with lysis buffer and immunoprecipitates recovered by boiling (10 minutes) in 2X reducing sample buffer. Samples were then run through standard SDS-PAGE protocol and developed using the TnieBlot anti-rabbit secondary antibody (1 :1000). 3. Pierce ProFound® mammalian Co-IP kit One hundred microliters of antibody coupling gel was added to a spin cup column and washed twice with phosphate buffered saline. Two micrograms of ET; receptor antibody (Alomone Laboratories) was diluted in 100 uL of 51 phosphate buffered saline + sodium cyanoborohydride (5 mM) and incubated overnight (4°C) with end-over-end mixing. The spin cup column was centrifuged (1 min, 3000 x g) and quenching buffer plus sodium cyanoborohydride added to the column for 30 minutes at room temperature with end-over-end mixing. The antibody-coupled gel was washed 4 times with wash solution (with centrifugation: 1 min, 3000 x g). Lysates of rat thoracic aorta and vena cava were made using the M-Per lysis buffer. Protein (1500 micrograms) from aortic and venous lysates were added to the spin columns and diluted to a final volume of 300 (IL with phosphate buffered saline. Columns were incubated overnight (4°C) with end- over—end mixing. Columns were then washed 3 times with phosphate buffered saline and immunoprecipitated proteins eluted off the column with three 50 BL aliquots of elution buffer. The pH of samples was neutralized with Tris (1 M, pH 9.5) and samples ran through standard SDS-PAGE as mentioned above. 4. ExactaCruzO co-immunoprecipitation kit The ExactaCruz® kit for homologous immunoprecipitation and western blotting for antibodies raised in rabbit (Santa Cruz, sc-45043). Two micrograms of ET; receptor antibody (Alomone Laboratories) was added to 50 BL of IP matrix (beads) and incubated with end-over-end mixing (1 hour, 4°C). Beads were washed twice with phosphate buffered saline (30 sec, 3000 rpm centrifugation). Five hundred micrograms of aortic lysate was added to the antibody-bead complex and incubated with end-over—end mixing (overnight, 4°C). Beads were washed three times with phosphate buffered saline. lmmunoprecipitated proteins were separated from beads by boiling (5 minutes, 96°C) in 2X denaturing sample 52 buffer and samples run through the standard SDS-PAGE protocol as described above. lmmunoprecipitated proteins were detected (after primary antibody binding) with a secondary antibody that came with the kit (1 :2000 dilution). 5. Co-immunoprecipitation using DynalO magnetic beads Dynal magnetic rabbit IgG beads or protein A beads (60 ILL, Invitrogen) were washed two times with phosphate buffered saline and then incubated with ET, or ET; receptor antibody (2 pg, Alomone Laboratories; overnight, 4°C with end- over-end mixing). Antibody bound beads were washed three times with phosphate buffered saline and then cross-linked with DMP (100 mM, 30 minutes at room temperature with end-over-end mixing). The cross-linking reaction was stopped with the addition of Tris (50 mM, pH 7.5; 15 minutes with end-over-end mixing). Cross-linked beads were washed three times with phosphate buffered saline and then 500 pg of aortic or venous lysate was added to the beads (1 hour with end-over-end mixing). Beads were washed three times with phosphate buffered saline and then immunoprecipitated proteins were eluted off of the bead-antibody complex by boiling for 5 minutes in 2X denaturing sample buffer. Samples were then run through the standard SDS-PAGE protocol as described above. G. Dissociation of vascular smooth muscle cells Rat thoracic aorta and vena cava were isolated, dissected and cleaned in chilled dissociation solution containing (in mM): NaCl, 136; KCI, 5.6; M902, 1; Na2HPO4, 0.42; NaH2P04, 0.43; NaHCOa, 4.2; HEPES, 10; sodium nitroprusside, 8.72 and 53 bovine serum albumin, 1mg/mL (pH 7.4 with NaOH). After dissection, the whole vena cava or 4-5 millimeters of aorta were cut into thin rings (as thin as possible) and equilibrated at room temperature for 10 minutes in one milliliter of fresh dissociation solution. Vessel pieces were incubated in one milliliter of an enzymatic solution containing papain (26 UnitslmL) and dithiothreitol (1mglmL) dissolved in dissociation solution for 45 minutes (at 37°C, with shaking). Then vessel pieces were incubated in 1-2 mL of a second enzymatic solution containing collagenase (1.95 Units/mL), elastase (0.15 mglmL) and soybean trypsin inhibitor (1 mglmL) (aorta: 35 minutes, vena cava: 45 minutes with shaking, 37°C) dissolved in dissociation solution. After the second enzymatic digestion, the digestion solution was pulled off (leaving the tissue and cells in the tube) and 4 mL of fresh, cold dissociation solution was added. Cells were placed on ice for five minutes, the dissociation solution was discarded (carefully leaving cells at the bottom of tube) and cells were rinsed again with 4 mL of fresh, cold dissociation solution. The second wash of dissociation solution was gently pipetted off, and cells were suspended and triturated (forcefully pipetted approximately 10 times) in 0ptiMEM (plus sodium nitroprusside, 872 nM) to dissociate vascular smooth muscle cells from the blood vessel matrix. H. Immunocytochemistry in freshly dissociated vascular smooth muscle cells Two hundred microliters of freshly dissociated vascular smooth muscle cells were placed on poly-lysine (50 ug/mL) coated coverslips (12 mm) and allowed to 54 adhere to the coverslips for 45 minutes (37°C, 4% CO2). Cells were fixed in Zamboni’s fixative (4% paraforrnaldehyde, 15% picric acid, 0.1M phosphate buffer pH 7.4, 20 minutes). Cells were permeabilized with Triton-X 100 (0.5%, 20 minutes) and incubated cells with lmageiT® signal enhancer (30 minutes, 37°C, Invitrogen). Coverslips with cells were incubated with primary antibodies (ET,: anti-sheep, Fitzgerald Industries; ET;: anti-rabbit, Alomone Laboratories; pan- cadherin: anti-mouse, Sigma; 1:200 dilution in phosphate buffered saline, 0.5% Triton-X 100) for 2 hours (37°C). Cells were then incubated with secondary antibodies (Alexa555 anti-rabbit, 1:200; Alexa488 anti-sheep, 1:200; Alexa633 anti-mouse, 1:200; Invitrogen) for 1 hour (37°C). Coverslips were then mounted on slides with ProFound® anti-fade mounting media (Invitrogen). Confocal images were captured at the Center for Advanced Microscopy at Michigan State University on a Zeiss confocal microscope. I. Data analysis and statistics Data was derived from 4-10 animals per group. Western blots densitometry was performed using NIH image and normalized to smooth muscle ot-actin expression. Contractility data is presented as mean :I: SEM, as a percentage of the initial response to phenylephrine (10 IIM for arteries) or norepinephrine (10 BM for veins) for the number of animals indicated in parentheses. Agonist E050 values were calculated using a nonlinear regression analysis using the algorithm [effect = maximum response I1 +(E050Iagonist concentration” (GraphPad Prism, San Diego, CA). When clear maximal responses were not obtained, EC50 values 55 were considered estimates with the true EC50 value being equal to or greater than the calculated value. When comparing concentration response curves, two-way ANOVA with Bonferroni’s post hoc was performed. When comparing two groups of estimated EC50 values, the appropriate Student’s f-test was used, and when comparing EC50 values between three or more groups, one-way ANOVA with Bonferroni’s post hoc test was performed. In all cases, a P value s 0.05 was considered statistically significant. Some images in this dissertation are presented in color. 56 III. RESULTS Hypothesls: ET, and ET, receptors physically interact via receptor heterodimerization in veins but not arteries and this heterodimerization functionally affects ET, and ET, receptor pharmacology. A. Subhypothesls 1: To demonstrate a pharmacological interaction between ET, and ET, receptors in rat thoracic vena cava but not aorta and to demonstrate that activated ET, receptors pharmacologically uncouple ET, receptors from receptor blockade in rat thoracic vena cava. 1. Non-speclflc ET,/ET, receptor desensltlzatlon Since veins but not arteries have functional, contractile ET, receptors, I investigated the role of ET, receptor in ET-1 desensitization. Flgure 15 displays lack of arterial contraction but partial venous contraction to ET-1 (10 pM — 100 nM) after desensitization with ET-1 (100 nM). Contraction to ET-1, given cumulatively, after exposure to a concentration of ET-1 causing maximal contraction (15 minutes, 100 nM, followed by 2 hours of washing) was abolished in aorta (Flgure 15A), while in veins 36.3 a: 0.2% of maximal contraction to ET-1 remained (Flgure 158). Figure 16 displays ET, receptor-mediated contraction of rat thoracic aorta and vena cava after ET-1 desensitization (100 nM, ). Aorta were unresponsive to the ET, receptor agonist ET -1[1-31] 57 A. Aorta +Control -D- ET-1 (100 nM) desens. (N=5) 5 Percentage PE (10 I‘M) CODUBCIIOI’I .8 i9 -12 -11 -10 -9 -8 -7 -l3 IogET-1 [M] Vena Cava -e- Control .. -o- ET-1 (100 nM) desens. (N=9) 1 Percentage NE (10 pM) Contraction a a a a a a s -12 -11 -10 -9 36 -7 ~15 IogET-1[M] Flgure 15. ET -1 (10 pM - 100 nM) concentration response curves in rat thoracic aorta (A) and vena cava (B) desensitized with ET -1 (100 nM). Points represent means a; SEM for the number of animals (N) indicated in parentheses. Asterisks (*) indicate statistically significant differences from control responses (p<0.05). ET-1, endothelin-1; NE, norepinephrine; PE, phenylephrine. 58 Aorta 75 +Control -D- ET-1 (100 nM) desens. g (N=4) m. a- m 50. is :0 6s 33 g 25. o, * -12 -11 ~10 -9 -8 -7 -6 log ET-1[1-31] [M] ' Vena Cava 500 -e-Control -o- ET-I (100 nM) desens. g 400. (N=4) a) a 3m- .88 go 93 2m: 8 3. g a 100.. o. -12 -11 -10 -9 -8 -7 -6 log ET-1[1-31] [M] Flgure 16. ET -1[1 -31] (10 pM — 100 nM) concentration response curves in rat thoracic aorta (A) and vena cava (B) desensitized with ET-1 (100 nM). Points represent means a: SEM for the number of animals (N) indicated in parentheses. Asterisks (*) indicate statistically significant differences from control responses (p<0.05). ET-1, endothelin-1; NE, norepinephrine; PE, phenylephrine. 59 after ET-1 exposure (Flgure 16A), while veins remained responsive to ET-1[1- 31] (though the response was significantly diminished) after ET-1 exposure (Flgure 16B) (21.9 :I: 0.6% of maximum venous contraction to ET -1 ). While the ET, receptor on arteries and veins desensitized in a qualitatively similar manner, the ET, receptor on veins did not desensitize to ET-1 (Flgure 17A) or the ET, agonist, 86c (Flgure 178) when a similar desensitization paradigm was used (15 minute agonist challenge followed by 2 hour agonist washout). A bolus concentration of 86c (100 nM) was administered because cumulative concentration response curves to S6c were not reproducible. Contraction to a maximal concentration of ET, receptor agonist S6c (100 nM) after EM (100 nM) was not altered in veins (117 :I: 7% of norepinephrine contraction in control tissue vs 118 a: 6% in ET -1 desensitized tissues, P> 0.05). Moreover, maximal venous contraction to S6c after 86c pre-exposure (100 nM) was not altered (80 :I: 15% of norepinephrine contraction in control tissues vs 87 :I: 12% in 86c desensitized tissues, P > 0.05). Contraction to SSC (10 pM - 100 nM) was not observed in aorta. These data suggest that the ET, receptor may account for maintained venous responsiveness to additional ET challenges after desensitization, as the ET, receptor remains functional (i.e. contractile) and responsive to ET-1 in the desensitization time frame used. Desensitization of the ET receptors after tissue exposure to ET-1 was specific to 60 A Vena Cava 175 -Control 150.. IZIET-1 (100 nM)desens. c (N=4) .9 125. E g —r— §§ 100. C: 5 g 75-: a) :1. °- 2 50. 25. 0. Contraction to 86c (100 nM) Vena Cava 175 -Control 150- DSGC (100 nM)desens. c (N=4) .9125- % ii an r. . C 63 75' (I) 1 m e 50. 25. 0. Contraction to S6c (100 nM) Flgure 17. 86c (100 nM) -induced contraction of rat thoracic vena cava desensitized with ET-1 (100 nM) (A) or 86c (100 nM) (B). Points represent means a: SEM for the number of animals (N) indicated in parentheses. Asterisks (*) indicate statistically significant differences from control responses (p<0.05). ET-1, endothelin-1; NE, norepinephrine; PE, phenylephrine; 86c, sarafotoxin 6c. 61 A Aorta 150 -l-Control 125. -I:I-ET-1 (100 nM)desens. g (N=3) $8100- CB 0 DIE as 75- 6s a g 50- $- 0- I I -10 -9 -8 -7 -6 -5 -4 '09 NE [W B Vena Cava 125 -e-Control -o-ET-1 (100 nM) desens. c:100- (N=3) .9 g g 751 b a a a 8 {is 501 d) :1. O- o z: 25, -10 -9 - -7 -i3 -' -4 '09 NE [W Flgure 18. Norepinephrine-induced contraction (1 nM - 10 BM) of rat thoracic aorta (A) and vena cava (B) was unaltered with ET -1 (100 nM) desensitization. Points represent means a: SEM for the number of animals (N) indicated in parentheses. ET-1, endothelin-1; NE, norepinephrine; PE, phenylephrine. 62 the ET system (I.e. displayed homologous desensitization). Contraction to the ot- adrenergic agonist norepinephrine was not reduced or rightward-shifted in control or ET-t -desensitized aorta (Flgure 18A) or vena cava (Flgure 18B). To determine which receptor subtype accounted for maintained venous ET -1- induced contraction after desensitization, veins were desensitized to ET-1 as described above and incubated with either vehicle, an ET, receptor antagonist (atrasentan, 100 nM) or an ET, receptor antagonist (BO-788, 100 nM) for 30 minutes prior to performing ET -1 concentration response curves (10 pM — 10 nM). The ET, receptor antagonist 80-788 was unable to shift the remaining contraction to ET-1 (EC50 (-log M) = 8110.1 in control tissues versus 81:01 in 80-788 incubated tissues), but did reduce maximal contraction to ET -1, though this reduction was not statistically significant (Flgure 19B). However, the ET, receptor antagonist atrasentan significantly reduced the remaining venous contraction to ET-1 after desensitization (E05, (-log M) = 81:01 in control tissues vs unmeasurable in atrasentan-incubated tissues, Flgure 19A). These data suggest that the ET, receptor, along with small contributions by the ET, receptor, is primarily responsible for continued venous responsiveness to ET-1. These data also suggest that the presence of contractile ET, receptors in vena cava but not aorta may alter ET, receptor pharmacology such that the ET, receptor is less susceptible to receptor desensitizationfintemalization. A. 175 Vena Cava -e-Control 1504 -o-Atrasentm(100nM) r: (N=5-6) “1% 1254 2 (U 3.3 100. g o :0 75 £95 ' o s. as w. v t 25. * * 0. -12 -11 -10 -9 -8 -7 -6 log ET-1[M] Vena Cava B 175 ' -e-Control 1501 -o-BQ~788(100nM) (N=5-6) 125. 5 Percentage NE (10 pM) Contraction .3 -12 -11 -10 -9 -'8 -7 -6 logET-1 [M] Flgure 19. ET -1 (10 pM - 100 nM) concentration response curves in the presence of ET, receptor blockade (atrasentan, 100 nM) (A) or ET, receptor blockade (BO-788, 100 nM) (B) in rat thoracic vena cava desensitized with ET-t (100 nM). Points represent means a: SEM for the number of animals (N) indicated in parentheses. Asterisks (*) indicate statistically significant differences from control responses (p<0.05). ET -1, endothelin-1; NE, norepinephrine; PE, phenylephrine. 2. ET, receptor desensltlzatlon Since there are clear differences in desensitization between the ET, and ET, receptors in vena cava, l next determined if the ET, receptor in vena cava did in fact desensitize. The desensitization protocol was changed to alter the amount of re-sensitization time (Is. change the number of washes, Flgure 20A). The data in Flgure 208 demonstrates that S6c (100 nM, 15 minute) desensitization followed by 15 minutes of re-sensitization (washing every 5 minutes) completely abolished venous contraction to a subsequent challenge of 86c (100 nM). When the re-sensitization time (and number of washes) was increased to 30 minutes (6 washes), venous contraction to subsequent challenges of 86c after 86c desensitization were not different from the control contraction to $60 (100 nM, Flgure 20B), suggesting that while venous ET, receptors do desensitize, they re- sensitize in a significantly shorter time frame than desensitized ET, receptors. 3. Does ET, receptor desensltlzatlon alter ET, mceptor function? I then investigated whether contractile ET, receptors could alter ET, receptor function in both rat thoracic aorta and vena cava by desensitizing ET, receptors with 86c and assessing ET, receptor function using the ET, receptor antagonist, atrasentan. To ensure maximum ET, receptor desensitization, 36c (100 nM) was incubated for one hour concurrent with ET, receptor antagonism (atrasentan, 10 nM) and cumulative concentration response curves to ET-1 (10 65 A. . _ - Vena cava Example DOIygraph recordings. 1 86c (100 nM, 1 hr) k 3 washes (15 min re-sensitization) l 360(100 nM,1hr) ,2 x 6 washes (30 min re-sensitization) I 860(100 nM,1hr) k Contraction to 86¢ (100 nM) l 9 Percentage NE (10 pM) contraction .8 .8 .3 * # Control 15 3'0 Re-sensitlzatlon time (minutes) Flgure 20. 86c desensitization protocol (A) and quantification of results (B) in rat thoracic vena cava. NE, norepinephrine. Asterisks represents a statistically significant difference from control (p<0.05). # represents a statistically significant difference from 30 minutes re-sensitization (p<0.05). 66 A. 175 Aorta +Control 150- -I:I-Atrasentan(10nM) N=4 mi 125. ( ) “-3 3.3 100. $8 75 55:, ‘12 so. 251! O. -12 -11 .10... 3 -7 -6 logET-1[M] B_ Aorta -I- 86c desens. -I:I- SGc desens. «I- Atrasentan (10 nM) (N=4) Percentage PE (10 pM) Contraction .8 .8 .8 -12 -11 -10 -9 is 5! -6 logET-1[M] Figure 21. Comparison of ET, receptor blockade (Atrasentan, 10 nM) of ET -1 cumulative concentration response curves in rat thoracic aorta with unbound ET, receptors (A) and agonist-bound ET, receptors (B) (86c desens). Points represent means a: SEM for the number of animals (N) indicated in parenthesis. NE, norepinephrine; PE, phenylephrine. 67 A 700 Vena Cava -e- Control 600, -o-Atrasentan (10 nM) (N=4) Percentage NE (10 pM) Contraction -12 -11 -10 59 -8 -7 -l IOQET-IIMI U) B Vena Cava -e- 86c desens. 500,, -o-SGC desens. + Atrasentan (10 nM) (Nd) Percentage NE (10 pM) Contraction -12 -11 -10 -9 -8 37 -6 logET-HMI Flgure 22. Comparison of ET, receptor blockade (Atrasentan, 10 nM) of ET-1 cumulative concentration response curves in rat thoracic vena cava with unbound ET, receptors (A) and agonist-bound ET, receptors (8) (86¢ desens). Points represent means a: SEM for the number of animals (N) indicated in parenthesis. NE, norepinephrine; PE, phenylephrine. 68 Aorta A. Condition Aorta EC50 Aorta (dog M) Fold shift Control + ET, 8.2 3 2 Atrasentan (10 nM) receptors 7.7 ' 86c (100 nM) - ET, 8.2 5 0 Atrasentan + 86c receptors 7.5 ' B. Vena cava . . Vena cava Vena cava 0°"d't'°" EC, (4% M) Fold shift Control + ET, 8 2 1 3 Atrasentan (10 nM) receptors 8.1 ' S6c (100 nM) - ET, 8.5 6 3,, Atrasentan + 86c receptors 7.7 ' Table 1. Estimated E05, (-Iog M) values for ET-1-induced contraction in aorta (A) and vena cava (B) in the presence of unbound or agonist-bound ET, receptors with and without ET, receptor antagonism (atrasentan, 10 nM). Asterisks (*) represents a statistically significant difference from + ET, receptors (p<0.05). 86c, sarafotoxin 6c. 69 pM — 100 nM) were performed without washing out 86c and the ET, receptor antagonist. In rat thoracic aorta with unbound ET, receptors (control, not S6c- desensitized), atrasentan (10 nM) caused a 3.2-fold rightward shift in ET-1- induced contraction (Flgure 21A, Table 1). In aorta with agonist-bound ET, receptors (SGc-desensitized aorta), atrasentan (10 nM) caused a 5.0-fold rightward shift in ET-1-induced contraction (Figure 21 B). In vena cava with unbound ET, receptors (control, not S6c-desensitized), atrasentan caused a 1.3- fold rightward shift in ET-1-induced contraction (Flgure 22A, Table 1). In vena cava with agonist-bound ET, receptors (SSc-desensitized), atrasentan caused a 63-fold rightward shift in ET-1-induced contraction (Flgure 228). These data suggest that in vena cava, the presence of agonist-bound contractile ET, receptors alters ET, receptor function such that ET, receptors are less susceptible to receptor blockade. 4. Receptor Interactlon: contractlllty studies uslng receptor antagonists Receptor antagonism studies were also performed to investigate howfif ET, receptors alter ET, receptor function. Cumulative ET-1 concentration response curves were performed in the presence of vehicle, ET, receptor blockade (atrasentan, 10 nM), ET, receptor blockade (BO-788, 100 nM) or ET, plus ET, receptor blockade (atrasentan, 10 nM + BQ-788, 100 nM) and estimated 70 A. Aorta -I- Vehicle ~0- Atrasentan (10 nM) (N=4-5) a Percentage PE (10 BM) Contraction .8 .8 .3 o I V -12 -11 -10 -9 43 -7 -6 log ET-t [M] Aorta 8' 200 -I-BQ-788 (100 nM) ~D-Atrasentan (10 nM) :150‘ +BQ-788(100nM) w .9 (N=4-5) .. a E 0 100- § 3 a a c 50-1 0' . -12 -11 -10 -9 -8 -7 -6 log ET-1 [M] Flgure 23. Comparison of ET, receptor blockade (Atrasentan, 10 nM) of ET-1 cumulative concentration response curves in rat thoracic aorta without ET, receptor blockade (A) and with ET, receptor blockade (B) (BO-788, 100 nM). Points represent means a SEM for the number of animals (N) indicated in parenthesis. NE, norepinephrine; PE, phenylephrine. 71 A 800 Vena Cava -e-Vehicle 700- -o-Atrasentan(10"M) 600. ("‘45) Percentage NE (10 pM) Contraction 100, o. , -12 -11 -10 -9 -e -7 -6 log ET-1 [M] B- Vena Cava -e-BQ-788 (100 nM) " -o-Atrasentan (10 nM) + 80-788 (100 nM) (N=4—5) l 1 Percentage NE (10 pM) Contraction assesses ‘P I _A 2 -11 -10 -9 -'8 -7 -l IogET-1 [M] U) Figure 24. Comparison of ET, receptor blockade (Atrasentan, 10 nM) of ET-1 cumulative concentration response curves in rat thoracic vena cava without ET, receptor blockade (A) and with ET, receptor blockade (B) (80-788, 100 nM). Points represent means a; SEM for the number of animals (N) indicated in parenthesis. NE, norepinephrine; PE, phenylephrine. 72 Aorta A. Condition Aorta EC,o Aorta Hog M) Fold shift Vehicle + ET, 8.2 6 7 Atrasentan (10 nM) receptors 7.3 ' BQ-788 (10 pM) Er 8.3 Atrasentan + BQ- ' 9 6.4 788 receptors 7.5 B. Vena cava . . Vena cava Vena cava C°"""'°" _ ECin (499 M) Fold shift _ Vehicle + ET, 8.1 2 3 Atrasentan (10 nM) receptors 7.8 ' 80-788 (10 (IM) ET 8.5 - - B * Atrasegteag + BO receptors 7.6 8.1 Table 2. Estimated EC50 (-log M) values for ET-1-induced contraction in aorta (A) and vena cava (B) and the presence and absence of ET, receptor blockade, with and without ET, receptor antagonism. Asterisks (*) represents a statistically significant difference from + ET, receptors (p<0.05). 73 ECso values were calculated and compared between groups. In aorta without ET, receptor blockade, atrasentan caused a 67-fold rightward shift in ET-1 - induced contraction (vehicle vs atrasentan) (Flgure 23A, Table 2). In aorta with ET, receptor blockade, atrasentan caused a 6.4-fold rightward shift in ET-1- induced contraction (BO-788 vs 80-788 + atrasentan) (Flgure 238), which was not different from the shift in the presence of functional ET, receptors. In vena cava without ET, receptor blockade, atrasentan caused a 2.3-fold rightward shift in ET -1-induced contraction (vehicle vs atrasentan) (Flgure 24A, Table 2). When ET, receptors in vena cava were rendered non-functional with ET, receptor blockade, atrasentan caused a 8.1-fold rightward shift in ET-1-induced contraction (Flgure 24B), which was significantly greater than when ET, receptors were not blocked, suggesting that functional ET, and ET, receptor interaction occurs in vena cava, our model vein, but not aorta, our model artery. 5. Section summary Through the use of contractility studies, I have demonstrated that functional, contractile ET, receptors (present in vena cava but not aorta) are capable of altering ET, receptor pharmacology. Desensitization experiments and receptor antagonist experiments provide evidence for functional ET,/ET, receptor interaction in veins but not arteries, suggesting that the rat thoracic vena cava is an appropriate model to investigate ET,/ET, receptor heterodimerization. 74 B. Subhypothesls 2: To characterize biochemical signaling interactions between ET, and ET, receptors in rat thoracic aorta and vena cava. One possible explanation for functional endothelin receptor interaction in vena cava but not aorta could be differences in endothelin receptor signal transduction in arteries and veins. ET-I can also potentially signal through the production of reactive oxygen species (ROS). ET-1 via ET, receptors stimulates NADPH oxidase to increase superoxide production (Li et al, 2003; Loomis ef al, 2005). Superoxide is a substrate for superoxide dismutase, which rapidly converts superoxide into H202. Both superoxide and H202 have been reported to alter vascular tone, causing both contraction and vascular bed depending on experimental conditions and vascular beds studied (Clempus and Griendling, 2006). It is possible that differences in ROS production and signaling in arteries and veins may explain why endothelin receptor interaction occurs in veins but not arteries. Contractility studies were performed to investigate the signal transduction pathways mediating ET-t-induced contraction in aorta and vena cava and investigated the role of ROS in aortic and venous ET -1-induced contraction. l determined if H202 was vasoactive in rat thoracic aorta and vena cava, if ET-1 stimulated H202 production and if H202 mediated aortic and venous ET-1-induced contraction. Also, to determine if signaling interactions can explain functional venous ET ,IET , receptor interaction, cumulative concentration response curves to ET -1 were performed in the presence of specific inhibitors of 75 150 +Aorta(+ E) -l:I-Aorta(- E) -e- Vena Cava (+ E) w 8 (iv-10> fi 8 100- s s Q) C a e c A 2 d) 1 q es 5° 0- S 0:7 -6 -5 -4 -3 -2 I09 l"I202 [MI Figure 25. Exogenously added H202 (1 uM — 1mM) contracts both rat thoracic aorta and vena cava. Points represent means :1: SEM for the number of animals (N) indicated in parenthesis. +E, endothelium intact; -E, endothelium denuded; NE, norepinephrine; PE, phenylephrine. 76 (N=9-14) If 1.504 I 1.25- [H 202] (nmol/mg protein) Flgure 26. Basal H202 levels (nmol/mg protein) are significantly higher in rat thoracic vena cava compared to aorta. Basal H202 production is significantly reduced in aorta and vena cava by DDC (diethyldithiocarbamate, 10 mM), an inhibitor of superoxide dismustase. Data are represented as nmol H202/mg of total protein for the number (N) of animals indicated in parentheses. Asterisks (*) represent a statistically significant difference from basal H202 levels (p<0.05), pound signs (if) represent a statistically significant difference between aorta and vena cava (p<0.05). 77 (N=9-14) 6 Tissue H202 (as a % control response) 3'3 $ C Aorta ET-1 (100 nM): . Flgure 27. ET-1 (100 nM, 4 hour incubation) stimulates H202 production in rat thoracic vena cava but not aorta. Data are represented as tissue H202 production as a percent of control H202 levels for the number (N) of animals indicated in parentheses. Asterisks (*) represent a statistically significant difference from -ET-1 (p<0.05). 78 Tissue H202 (as a % control response) Figure 28. ET -1-stimulated H202 production in vena cava is not concentration- dependent and is reduced by either ET, receptor blockade (atrasentan, 30 nM) or ET, receptor blockade (100 nM). Data are represented as tissue H202 production as a percent of control H202 levels for the number (N) of animals indicated in parentheses. Asterisks (*) represent a statistically significant difference from basal H202 levels. 79 several signaling pathways, including rho kinase, p38 MAPK, Erk MAPK, src and Pl3-K. 1. Reactive oxygen species and ET-1-lnduced contraction Exogenously added H202 (1 IIM - 1 mM) contracted both rat thoracic aorta and vena cava, but contracted veins much more robustly than arteries (Figure 25). H202 levels were significantly higher in vena cava compared to aorta (vena cava: 1.26 a: 0.33 nmol/mg H202; aorta: 0.27 a: 0.07 nmol/mg H202) (Figure 26). ET-1 incubation (100 nM, 4 hour) increased venous but not aortic H202 levels (vena cava ET -1: 154:17% control H202; aorta ET-1 94:13 % H202) (Figure 27). Both ET, receptor blockade with atrasentan (10 nM) and ET, receptor blockade with BQ-788 (100 nM) significantly reduced ET-1-stimulated H202 production in vena cava (atrasentan: 95:30% control H202; BQ-788: 104:21% H202) (Figure 28). To determine if H202 partially mediated ET-1-induced contraction, ET-1 cumulative concentration response curves were performed in the presence of catalase or polyethyleneglwa (PEG) -catalase (which is supposedly more membrane permeable because of the PEG moieties) to break down any ET-1- stimulated H202 production. Catalase and PEG-catalase potentiated maximal venous ET -1-induced contraction (Figure 29), suggesting that in veins, ET-1- stimulated H202 may actually be vasodilatory to counter the vasoconstriction elicited by ET-1. Cumulative ET-1 concentration response curves were then performed in the presence of 3-aminotriazole (3-AT) (50 mM), a catalase 80 A 600 Vena Cava +Control Sm, -D-Catalase (2000 U/ITII) (N=4) as 23; 400- 8e a s 300‘ Is: as 2002 100‘ 0- U I V -12 -11 -10 -9 -8 -7 -6 Log ET-1 [M] B- 800 Vena Cava +Control v: e 7001 -o-PEG-Catalase 4 (1a; Ulml) I: 600 “1.0 -I:I-PEG-Catalase z§ 50,. (326U/ml) 0 or? d (N=4) g g 400 a a 3001 0.0 5 200' 100' 912 -11 -10 -'9 is -'7 -6 Log ET-1 [M] Figure 29. H202 is not involved in venous ET -1-induced contraction as neither catalase (2000 U/ml) (A) nor PEG-catalase (163 and 326 U/ml) (B) reduced venous ET-1-induced contraction. NE = norepinephrine, PEG-catalase = polyethylene glycol catalase. Points represent means :1: SEM for the number (N) of animals indicated in parentheses. Asterisks (*) represent a statistically significant difference from control (p<0.05). 81 Vena Cava -e- Control -o-3-AT (50 mM) (N=4) l I 1 Percentage NE (10 pM) contraction assesses L :9 2 -11 -10 -9 T8 -7 -l LogET-i [M] U) Figure 30. The catalase inhibitor 3-AT (50 mM) had no effect on maximal venous ET -1-induced contraction. 3-AT = 3-aminotriazole, NE = norepinephrine. Points represent means :I: SEM for the number (N) of animals indicated in parentheses. 82 inhibitor, in an attempt to potentiate/enhance ET-1-stimulated H202 levels. Catalase inhibition with 3-AT did not alter aortic or venous ET -1-induced contraction (Figure 30). These data suggest that in veins, ET -1 increases H202 production, but this H202 does not play a significant role in mediating venous ET - 1-induced contraction, but may actually be vasodilatory. In arteries, ET-1 does not produce H202, thus it is unlikely that H202 mediates aortic ET-1-induced contraction. Thus, I conclude that because reduction of venous H202 with catalase did not inhibit ET -1-induced contraction, ET-t signaling via H202 production is not a likely mechanism responsible for venous endothelin receptor interaction, but cannot extend my conclusions to ET -1 signaling through the production of other ROS such as superoxide or hydroxyl radical. 2. Signal transduction pathways Since ET -1 signal transduction through H202 production was not a likely mechanism for explaining why venous ET, and ET, receptors functionally interact, I examined the possibility that ET -1 activates differential signaling in aorta and vena cava through other signaling pathways known to mediate vascular contraction. I asked if inhibitors of p38 MAPK, Erk MAPK, src, rho kinase and Pl3—K reduced aortic and venous ET-1-induced contraction. The choice and concentration of inhibitors used was based on previous studies in our laboratory that determined inhibitor specificity in cultured smooth muscle cells (Watts, 1996; Banes et al, 1999; Florian and Watts, 1999; Northcott et al, 2002). 83 Also, except for the rho kinase inhibitor Y27632, none of the inhibitors used significantly reduced KCl-induced contraction (data not shown), a measure of receptor-independent contraction, suggesting that these inhibitors (LY294002, P098059, PP1, and 88203580) do not inhibit vascular contraction per se. Inhibition of Pl3—K with LY294002 (20 pM) did not reduce aortic or venous ET-1- induced contraction (Figure 31). Inhibition of src with PP1 (10 pM) did not reduce aortic or venous ET-1-induced contraction (Figure 32). PD98059 (10 am, an inhibitor of Erk MAPK, also did not inhibit aortic or venous ET -1-induced contraction (Figure 33). 88203580 (10 (M), an inhibitor of the p38 MAPK pathway did not reduce venous ET -1-induced contraction, but significantly reduced maximum aortic ET -1-induced contraction [vehiclez 153:8% PE (10 pM) contraction; SB203580: 106:11%] (Figure 34). Inhibition of rho kinase with Y27632 (10 pM) significantly reduced both maximum aortic and venous ET-1- induced contraction [aorta vehicle: 106:5% (10 nM) PE contraction; aorta Y27632: 59:7%; vena cava vehicle: 469:50% NE (10 (M) contraction; vena cava Y27632: 272:39%] (Figure 35). While Y27632 has as higher affinity for rho kinase, it is also capable of inhibiting PKC in some biochemical assays. Thus ET -1 cumulative concentration response curves were performed in the presence of another rho kinase inhibitor, HA1077. HA1077 (10 (1M) significantly reduced aortic but not venous ET-1-induced contraction [aorta vehicle: 131 :1 1% (10 pM) PE contraction; aortic HA1077: 92:11%; vena cava vehicle: 571:87% NE (10 pM) contraction; vena cava HA1077: 471 :67%] (Figure 36). 84 if Aorta Percentage PE (10 pM) Contraction a is I 5? | _L o. 2 ~11 + Vehicle -D- LY294002 (20 pM) (N =3) 33 log ET-1 [M] -10 -9 Vena Cava L U) ...§§ assess Percentage NE (10 pM) Contraction .5 a? Figure 31. 2 -e- Vehicle -o- LY294002 (20 pM) (N=3) -10 -'9 -'8 logET-I [M] -11 Effect of LY294002 (20 (M), a phosphatidyl inositol-3-kinase Inhibitor, on ET-1-induced contraction of rat thoracic aorta (A) and vena cava (8). Points represent means : SEM for the number of animals (N) indicated in parenthesis. NE, norepinephrine; PE, phenylephrine. 85 A Aorta +Vehicle -I:I- P098059 (10 pM) (N=3) Percentage PE (10 pM) Contraction $ 3.9 I? -12 -11 -10 -9 -e -7 -6 IOQET-IIMI B- Vena Cava aoo -e-Vehicle 700- -o-P098059(10 nM) (N=3) m g 600- Z to 500. SE as 400. 8A a 5:. 300- 0.0 I: 200- 100- -12 -11 -1o -9 -§ 37 -6 log ET-1 [M] Figure 32. Effect of P098059 (10 (M), an Erk MAPK, on ET -1-induced contraction of rat thoracic aorta (A) and vena cava (8). Points represent means : SEM for the number of animals (N) indicated in parenthesis. NE, norepinephrine; PE, phenylephrine. 86 A. Aorta 200 +Vehicle -I:l-PP1 (10 pM) 150.. (Nfi) Percentage PE (10 PM) CORITBCUOII .8 .12 -11 -10 -9 13 -7 -l IogET-1 [M] U) B. Vena Cava -e-Vehicle .. -o- PP1 (10 BM) (N=4) Percentage NE (10 pM) Contraction -12 -11 -10 -§ 13 -7 -6 log ET-1[M] Figure 33. Effect of PP1 (10 (M), a src tyrosine kinase inhbitor, on ET-1-induced contraction of rat thoracic aorta (A) and vena cava (8). Points represent means : SEM for the number of animals (N) indicated in parenthesis. NE, norepinephrine; PE, phenylephrine. 87 A Aorta +Vehicle -D-S8203580 (10 pM) (N=6) €93 Percentage PE (10 pM) Contraction 3 39 -12 -11 -1o 39 -8 5 -6 logET-t [M] B. VenaCava 800 -e-Vehicle 700- -o-SBZOBSBO(10 pM) mg 600- (N=4) Z to 500. 6%: E0 400- ESE-.3001 (Lo 5200- 100- 0. i V r -12 -11 -10 -9 -8 -7 -6 log ET-1 [M] Figure 34. Effect of 88203580 (10 (M), a p38 MAPK inhbitor, on ET-1-induced contraction of rat thoracic aorta (A) and vena cava (8). Points represent means : SEM for the number of animals (N) indicated in parenthesis. NE, norepinephrine; PE, phenylephrine. Asterisks (*) represent a statistically significant difference from Vehicle (p<0.05). 88 .> Aorta -I- Vehicle -D- Y27632 (10 pM) (N=10) if. Percentage PE (10 pM) Contraction 5.9 a $ 93 0' I t ~12 ~11 ~10 -9 ~8 ~7 ~6 log ET~1 [M] B- Vena Cava § -0- Vehicle -o- Y27632 (10 pM) (N=8) . é Percentage NE (10 pM) Contraction a a a -12 -11 -10 -9 -8 -7 -1 IOQET-IIMI U) Figure 35. Effect of Y27632 (10 (M), a rho kinase inhbitor, on ET -1 -induced contraction of rat thoracic aorta (A) and vena cava (8). Points represent means : SEM for the number of animals (N) indicated in parenthesis. NE, norepinephrine; PE, phenylephrine. Asterisks (*) represent a statistically significant difference from Vehicle (p<0.05). 89 Aorta .> § -I- Vehicle -D-HA1077(10 uM) (N=6) Percentage PE (10 pM) Contraction :3 .8 I9 from Vehicle (p<0.05). 90 0- . r -12 -11 -10 -9 ~8 - -15 logET-t [M] B- VenaCava 800 -e-Vehicle 700" -o-HA1077(10IIM) c 600- (N=4-6) as g 83g 500a ] _ . C a! $8 400, a?” c 200- i 100- . ‘. _ I" 0- ‘ I I I ~12 ~11 ~10 ~9 ~8 ~7 ~6 IogET-1 [M] Figure 36. Effect of HA1077 (10 BM), a rho kinase inhbitor, on ET -1-induced contraction of rat thoracic aorta (A) and vena cava (8). Points represent means : SEM for the number of animals (N) indicated in parenthesis. NE, norepinephrine; PE, phenylephrine. Asterisks (*) represent a statistically significant difference 3. Section summary l hypothesized that ET, and ET, receptor signaling pathway interactions might explain functional ET, and ET, receptor interaction observed in veins but not arteries. l determined that ET-1 stimulated the production of the vasoactive reactive oxygen species H202 in veins but not arteries, but this H202 does not mediate venous ET-1-induced contraction. Through the use of signal transduction inhibitors, I also observed that rho kinase and p38 MAPK mediated aortic ET-t-induced contraction and rho kinase mediated venous ET-1-induced contraction. Within the context of these specific signaling pathways mediating vascular contraction, ET-1 did not activate any clearly divergent pathways in vena cava compared to aorta, suggesting that functional venous ET, and ET, receptor interaction likely does not occur via activation of the specific signaling pathways investigated or H202-mediated signaling. C. Subhypothesis 3: To demonstrate a physical interaction, I.e. heterodimerization, between ET, and ET, receptors in rat thoracic vena cava but not aorta. Recently, several G-protein coupled receptors have been reported to interact via receptor dimerization, altering receptor function, trafficking and pharmacology (Angers et al, 2002; Milligan, 2004; Maggio at al, 2005; Prinster at al, 2005). Though ET, and ET, receptors can heterodimerize when they are co-transfected 91 in HEK293 cells (Gregan er al, 2004), it is not known whether ET, and ET, receptors heterodimerize under physiological conditions, like when they are normally expressed in blood vessels. Common approaches for determining if receptors dimerize include receoptor co-immunoprecipitation, fluorescence resonance energy transfer (FRET) between fluorescently labeled receptors, and functional measures of receptor interaction (Milligan and Bouvier, 2005). l have investigated functional ET, and ET, receptor interaction in Subhypotheses 1 and 2, and in Subhypothesis 3 I proposed to perform receptor co- immunoprecipitation experiments and FRET analysis to determine if ET, and ET, receptors physically interact via receptor heterodimerization in vena cava but not aorta. 1 . Antibody testing: dot blots The experiments in this section required the use of antibodies specific for either ET, or ET, receptors. Because the ET, and ET, receptor are approximately 70% homologous, it was necessary to verify antibody specificity. Dot blots were performed using different antigenic sequences of ET, and ET, receptors and various commercially available antibodies for ET, and ET, receptors (Figure 37 and 38, Table 3). I concluded that the most selective and sensitive antibody was the Alomone ET, primary (raised in rabbit). The Alomone ET, primary antibody (also raised in rabbit) was the next most sensitive primary antibody, but unfortunately did recognize higher concentrations of ET, receptor epitopes. The 92 next most sensitive and selective antibody was the Fitzgerald ET, receptor primary antibody (raised in sheep). For the Chemicon and Biogenesis ET, and ET, receptor primary antibodies l was unable to detect any primary antibody binding to any of the concentrations of antigenic peptides, as all of these primary antibodies gave the same pattern of binding, suggesting nonspecific binding of the anti-sheep secondary antibody used to detect primary antibody binding. I chose to use the Alomone ET, and ET, receptor primary antibodies for co- immunoprecipitation experiments and the Fitzgerald ET, and Alomone ET, receptor primary antibodies for co-localization experiments. 2. Receptor co-locallzatlon Western blot analysis and immunohistochemistry demonstrate that ET, and ET, receptors are present in both rat thoracic aorta and vena cava. Because the endothelium was not removed prior to vessel homogenization, I could not determine from Western blots which cell types (smooth muscle cells, fibroblasts or endothelial cells) contain ET, and ET, receptors. lmmunohistochemistry was performed on 5-micron thick sections of aorta and vena cava but because I was unable to distinguish single cells, I was unable to determine specifically which cell types express these receptors. Thus, smooth muscle cells were freshly dissociated from rat thoracic aorta and vena cava and adhered to coverslips. Immunocytochemistry using primary antibodies directed to ET, and ET, 93 Antibody source Antigenic sequence Host species Alomone Laboratories ET, receptor NHNT ERSSH KDSMN rabbit ET, receptor CEMLR KKSGM QIALN D rabbit Biodesign International ET, receptor Ac-SSHVE DFTPFP GTEFC- rabbit Amide ET, receptor KANDH GYDNF RSSNN sheep Biogenesis ET, receptor QEQNH NTERS SHK sheep ET, receptor C-terminal peptide (proprietary sheep sequence) Fitzgerald Industries ET, receptor QEQNH NTERS SHK sheep Table 3. An abbreviated list of commercially available ET, and ET, receptor antibodies and their antigenic sequences used for dot blot analysis. 94 ,2 ng 20 ng_ 299 ng 2 ug COIIEIIPETING . . PPTIDE- . ,. _. Alomone ET, AlomoneEJTo,a ' . 'I'. } _. '* Alomone ET, Prima anti . . '. ry y ‘ ' .- L ‘- ' ,1 . . Biodesign ET , g,- , Biodesign er, Biodesign ET Primary antibody Biogenesis ET, Primary antibody Fitzgerald ET Primary antibody Flgure 37. Dot blots testing ET, receptor antibodies from Alomone Laboratories, Biodesign International, Biogenesis and Fitzgerald Industries. Competing peptides (2 ng - 2 ug) were spotted on nitrocellulose membranes and blots were developed with different ET, receptor primary antibodies. The same pattern of peptide dotting was used for the Biodesign ET,, Biogenesis ET, and Fitzgerald ET, primaries as was used for the Alomone ET, primary antibody. 95 COMPETING PEPTIDE: 2 2 2 2 .ng 0 ng 00.an ug Alomone ET, PAlomone EbTo% AI ET rimary anti y omone o 0 fl " Biodesign ET, Biodesign ET, Biodesign ET, Primary antibody Biogenesis ET, Primary antibody Flgure 38. Dot blots testing ET, receptor antibodies from Alomone Laboratories, Biodesign International and Biogenesis. Competing peptides (2 ng - 2 ug) were spotted on nitrocellulose membranes and blots were developed with different ET, receptor primary antibodies. The same pattern of peptide dotting was used for the Biodesign ET, and Biogenesis ET, primaries as was used for the Alomone ET, primary antibody. 96 receptors and fluorescent secondary antibodies was performed to determine if both ET, and ET, receptors are expressed by aortic and venous vascular smooth muscle cells and if both ET, and ET, receptors co-Iocalize on venous vascular smooth muscle cells. No primary antibody control experiments (i.e. cells just stained with secondary antibodies) verified that the fluorescent images captured represent binding of secondary antibodies to primary antibodies and no background fluorescence from non-specific secondary antibody binding. Flgure 39 displays projected confocal images (I.e. 3-D images comprised of 6 micron stacks captured from the top to the bottom of cells) of smooth muscle a-actin expression on aortic and venous vascular smooth muscles cells. The high a- actin expression present in the cells and elongated shape of the cells verifies that the isolated cells are indeed vascular smooth muscle cells. To perform receptor co-Iocalization experiments, cells were double-labeled with Fitzgerald .ET, and Alomone ET, receptor antibodies (raised in sheep and rabbit, respectively). Initially, confocal images were captured with singly (either ET, or ET, antibody) labeled cells to confirm that there was no bleed-through or background emission of one fluorescent secondary antibody in bandwidth of the second fluorescent secondary antibody (data not shown). Cells were also labeled with a pan- cadherin antibody to highlight the plasma membrane of the vascular smooth muscle cells. 97 Aorta: Vena cava: a-actin a-actin Figure 39. Projected confocal images of smooth muscle a-actin expression in freshly dissociated aortic (left) and venous (right) vascular smooth muscle cells. 98 Aortic vascular smooth muscle cells ET, receptor ET, receptor .____ 10mm 10mm cadherin overlay 10mm Figure 40. Confocal images (6 um thick sections) of ET, receptor, ET, receptor and cadherin expression in freshly dissociated aortic vascular smooth muscle cells. Images are representative of aortic vascular smooth muscle cells from five different rats. Yellow on the overlay image represents co-Iocalization of cadherin, ET, receptors and ET, receptors. 99 Venous vascular smooth muscle cells ETA receptor ET, receptor * '- D IOnm 'h f: 3 10um x; cadherin overlay 10mm 10pm Flgure 41. Confocal images (6 um thick sections) of ET, receptor, ET, receptor and cadherin expression in freshly dissociated venous vascular smooth muscle cells. Images are representative of venous vascular smooth muscle cells from four different rats. Yellow on the overlay image represents co-Iocalization of cadherin, ET, receptors and ET, receptors. 100 Aortic vascular smooth muscle cells ET-1 stimulated ET, receptor ET, receptor 10mm "———‘ IOllm cadhefln oveflay 10ml) 10mm Flgure 42. Confocal images (6 pm thick sections) of ET, receptor, ET, receptor and cadherin expression in ET-1-stimulated (100 nM) freshly dissociated aortic vascular smooth muscle cells. Images are representative of aortic vascular smooth muscle cells from five different rats. Yellow on the overlay image represents co-localization of cadherin, ET, receptors and ET, receptors. 101 Venous vascular smooth muscle cells ET-1 stimulated ET, receptor ET, receptor IOum cadherin overlay 10mm 'IOIIITI Figure 43. Confocal images (6 pm thick sections) of ET, receptor, ET, receptor and cadherin expression in ET-1-stimulated (100 nM) freshly dissociated venous vascular smooth muscle cells. Images are representative of venous vascular smooth muscle cells from four different rats. Yellow on the overlay image represents co-localization of cadherin, ET, receptors and ET, receptors. 102 Flgure 40 shows that ET, and ET, receptors are expressed at the membrane of aortic vascular smooth muscle cells. Flgure 41 shows that ET, and ET, (third panel) receptors were expressed at the membrane of venous vascular smooth muscle cells, with the white arrows indicating locations where ET, receptors and ET, receptors co-localized in vena cava. In vena cava, there was significant intracellular and membrane ET, receptor expression (Flgure 41). Aortic and venous vascular smooth muscle cells were stimulated with EM (100 nM) during the 45 minutes they were adhering to coverslips. ET-1 stimulation did not alter the location of aortic or venous ET, or ET, receptors (Flgure 42, aorta and Flgure 43, vena cava). ln vena cava, similar to basal conditions, significant ET, receptor staining was present inside vascular smooth muscle cells. 3. Co-Immunopreclpltatlon of ET, and ET, receptors Co-immunoprecipitation is another technique used to evaluate endothelin receptor dimerization (Le. physical receptor interaction). If ET, and ET, receptors dimerize in vena cava but not aorta, ET, and ET, receptors in venous but not aortic lysates should co-immunoprecipitate. Traditional co- immunoprecipitations were initially performed using protein AIG beads to immunoprecipitate the ET, receptor using the Alomone ET, receptor antibody. However upon immunoblotting with the Alomone ET, receptor antibody to determine if the immunoprecipitation was successful or with the Alomone ET, receptor antibody to determine if the ET, receptor co-immunoprecipitates with the 103 Aorta lysates Vena cava lysates _. ..___._ h... . ._.. _.._..- .-.r__-z._.. __.___._ . . _ - . -. I“ .‘ c.“ E: 5 E . fl ‘ i_,qu . ‘ . 1 3f- ' " "" " w b -' <—50kba 8 an -—--—-—-., q.. _-._.—L Aorta lysates Vena cava lysates hf“. m .. omw+5°k°a ‘. -.V + ‘4._, ,,., Flgure 44. Western blot analysis of ET, receptor expression (A) and ET, receptor expression (8) in aorta (left) and vena cava lysates (right) from eleven (11) different rats. The Alomone ET, receptor primary antibody (1 :200 dilution) was used to detect ET, receptors and the Alomone ET, receptor primary antibody (1 :200 dilution) was used to detect ET, receptors. 104 Aorta Vena cava IP lys IP lys IP lys IP lys Flgure 45. Example immunoprecipitation of ET, receptors from aortic and venous lysates. ET, receptors were both immunoprecipitated and detected (immunoblotted) with the Alomone ET, receptor antibody. IP, immunoprecipitate; lys, lysate. ET, receptor, immunoreactive bands for both the ET, and ET, receptor, which typically resolve at 50 kDa (Flgure 44) were obscured by a 50 kDA heavy chain antibody band from the immunoprecipitating antibody (Flgure 45). To avoid detection of the contaminating band from the immunoprecipitating antibody, a light chain-specific rabbit secondary antibody was then used for immunoblotting after immunoprecipitation, but this secondary antibody did not recognize either the Alomone ET, or ET, receptor antibodies. Several co-immunoprecipitation kits were tried, including the Pierce Mammalian colP kit, eBioscience True Blot, Santa Cruz ExactaBlot, and Dynal magnetic protein A beads (Table 4) in a effort to prevent loading/detecting of the 50 kDA heavy chain antibody band, however none of these kits successfully detected ET, receptor immunoprecipitation or ET, and ET, receptor co- immunoprecipitation. Cross-linking experiments were also performed to cross- link ET, and ET, receptors in aortic and venous lysates with the hope that cross- linked ET, and ET, receptors would migrate at a higher molecular weight separating them from the immunoprecipitating antibody, however the degree of protein degradation from aortic and venous lysates was too great to perform Western blot analysis. Also, caveolar fraction were isolated by sucrose gradient centrifugation, in an attempt to localize ET, and ET, receptors to caveolae, but these fractions were too dilute to quantify ET, and ET, receptors. 106 4. Section summary l hypothesized that ET, and ET, receptors would co-localize to the plasma membrane in venous but not aortic vascular smooth muscle cells and that ET, and ET, receptors would co-immunoprecipitate in venous but not aortic lysates. Immunocytochemistry analysis demonstrated that ET, and ET, receptors co- localized to the plasma membrane in both aortic and venous vascular smooth muscle cells and that ET-1 stimulation did not qualitatively alter ET, and ET, receptor localization in both aortic and venous vascular smooth muscle cells. Receptor co-immunoprecipitation experiments were inconclusive as in every immunoprecipitation technique employed, the immunoprecipitating antibody masked detection of either the ET, or ET, receptor antibody. Based on our receptor co-Iocalization experiments I concluded that ET, and ET, receptors co- localize to the membrane in both aorta and vena cava, but I was unable to determine if the receptors are within close enough proximity for receptor heterodimerization to occur. 107 ColP Method: Successful? Problem with technique . . Heavy chain IP antibody band Traditional IF NO contamination . Secondary antibody not True Blot colP kit NO specific Profound Mammalian NO Sample too diluted — colP kit No signal detected ExactaCruz homologous . colP kit NO Did not work Poor resolution of protein NATIVE PAGE NO bands . . Significant protein degradation Receptor cross-linking NO during cross-linking Dynal magnetic beads NO Table 4. Different co-immunoprecipitation methods were performed in several attempts to co-immunoprecipitate ET, and ET, receptors from rat thoracic aorta and vena cava lysates. 108 IV. DISCUSSION A. Ratlonale In the United States, approximately 30% of Americans have hypertension. A small percentage of hypertensive patients have primary hypertension, in which a single known gene is responsible for increasing blood pressure. However, most patients are diagnosed with essential hypertension and there is no known etiology for their elevated blood pressure. Anti-hypertensive drugs such as calcium channel blockers, beta adrenergic receptor blockers, diuretics and angiotension converting enzyme inhibitors and receptor blockers are available but typically a combination of two or more of the above mentioned anti- hypertensive drugs are required to achieve adequate decreases in blood pressure as recommended by the Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. Hypertension is a risk factor for many diseases including heart failure, stroke, renal failure, etc. Thus, inadequate control of blood pressure leaves patients at significantly higher risks of negative outcomes and increased risk of death. Clearly, more effective treatments for controlling high blood pressure are needed. The endothelin system represents a potential target for anti-hypertensive therapy that has not yet been aggressively pursued in the clinical arena. While 109 endothelin receptor antagonists are currently approved for the treatment of pulmonary hypertension, they are not available for the treatment of systemic hypertension. The mechanism of action of current anti-hypertensive drugs is primarily to reduce arterial tone to reduce total peripheral resistance and blood pressure. I posit that veins, specifically the endothelin receptors on veins, play an important role in regulating and elevating blood pressure. I hypothesize that the endothelin receptors, the ET, and ET, receptors, on veins interact via receptor heterodimerization to alter the pharmacology of these receptors. I also hypothesize that ET, and ET, receptors do not heterodimerize on arteries, making the venous endothelin receptor system a unique target for anti- hypertensive therapies. I began this study by choosing a model artery and a model vein to compare endothelin receptor heterodimerization. I chose the rat thoracic aorta as a model artery and the rat thoracic vena cava as a model vein. Though both of these vessels are considered to be conduit vessels and do not directly determine arterial resistance or venous compliance, both vessels (rat thoracic aorta and vena cava) were large enough to provide adequate protein yields for biochemical and functional assays to assess receptor heterodimerization. Western blot analysis and immunohistochemical analysis demonstrated that rat thoracic aorta and vena cava both express ET, and ET, receptors, suggesting that these vessels were appropriate models for studying endothelin receptors (Flgures 11 110 and 12). Preliminary contractility experiments suggested that while rat thoracic aorta express both ET, and ET, receptors, only ET, receptors couple to contraction as ET, receptor agonists do not alter tone in aorta under basal tone (Watts et al, 2002). Differently from rat thoracic aorta, in rat thoracic vena cava both ET, and ET, receptors couple to contraction. This initial observation of a difference in function of ET, and ET, receptors in arteries compared to veins prompted the idea that perhaps the two receptor subtypes interact in veins differently than in arteries. B. leferences In ET, and ET, receptor desensltlzatlon To enhance our understanding of the nature of a possible functional interaction between ET, and ET, receptors in vena cava but not aorta, I performed contractility studies examining ET, and ET, receptor-mediated contraction. Thoracic aorta from DOCA-salt hypertensive rats display significantly reduced contraction to ET-t when compared to their normotensive counterparts, while ET- 1-induced contraction was not different between vena cava from DOCA-salt hypertensive rats and their normotensive counterparts (Watts 9! al, 2002). I hypothesized that the difference in response to ET-1 in arteries and veins from DOCA-salt hypertensive rats could be due to the presence of functional ET, receptors that couple to contraction on veins but not on arteries. The first contractility experiments to assess receptor desensitization were designed to mimic the high levels of plasma ET-1 in DOCA-salt hypertension. Aorta and 111 vena cava were desensitized with a concentration of ET-1 that induced maximal contraction, the ET-1 was washed out until tone returned to baseline, vessels were challenged again with ET-1 and the contraction to ET-1 after desensitization quantified. Aorta desensitized with ET-1 did not contract to an additional ET-1 challenge, while vena cava contracted with ET-1 were able to contract to additional ET-1 stimulation, though the magnitude of contraction was significantly reduced (Flgure 15). l hypothesized that functional (contractile) ET, receptors on vena cava might mediate the remaining venous contraction to ET -1 after ET-1 desensitization since aorta (where only ET, receptors couple to contraction) completely desensitize to ET -1. Through the use of specific receptor antagonists I determined that the remaining ET -1 contraction in veins after ET-1 desensitization was mediated by ET, receptors (Flgure 19), suggesting that the contractile ET, receptors on vena cava were not responsible for the remaining contraction to ET-1 after desensitization. ET, receptor desensitization experiments with the ET, receptor agonist sarafotoxin 60 (S60) demonstrated that the venous ET, receptor does in fact desensitize (Flgure 20). However, the resensitization time frame is much shorter than that of the ET, receptor, highlighting an important difference between ET, and ET, receptor desensitization and resensitization. 112 Classical G-protein receptor desensitization occurs through receptor internalization, initiating with G-protein kinase (GRK) phosphorylation of the cytoplasmic tail of the receptor, allowing B-arrestin binding and receptor internalization via dynamin/clathrin-coated pits. Sequence analysis of the cytoplasmic tails of the ET, and ET, receptors suggest that because of low sequence homology in this region, ET, and ET, receptors may be phosphorylated by different GRK isoforms and internalized differently. Freedman et al demonstrated that when ET, and ET, receptors were overexpressed in HEK293 cells, both receptors were primarily phosphorylated by GRK2, suggesting that despite sequence differences, ET, and ET, receptor desensitization is initiated by the same GRK (Freedman et al, 1997). However, Bremnes at al observed that when ET, and ET, receptors were overexpressed in CHO or COS cells, intracellular trafficking of the ET, and ET, receptors differed. While both receptors where similarly internalized via an arrestin/dynamin- dependent pathway, ET, receptors were recycled and reinserted into the plasma membrane and ET, receptors targeted to Iysosomal compartments (Bremnes et al, 2000), demonstrating that following internalization the fates of the ET, and ET, receptor significantly differed. Since most of the experiments investigating ET, and ET, receptor desensitization and internalization have been performed in cells lines overexpressing ET, and ET, receptors, it is unknown if the conclusions from these experiments correlate to ET, and ET, receptor trafficking in tissues such as the rat thoracic aorta and vena cava. It is possible that ET, receptors in 113 veins are recycled and reinserted in the plasma membrane or there may be a large pool of intracellular ET, receptors ready to be inserted in the plasma membrane after desensitization and lysosomal degradation of ET, receptors. Our desensitization experiments highlight differences between aortic and venous ET, and ET, receptor function and suggest that the presence of a functional ET, receptor (as is present on the vena cava) might alter the function of the ET, receptor. C. Functlonal ET, and ET, receptor Interactlon occurs In velns but not arteries To further test the hypothesis that functional ET, and ET, receptor interaction occurs in veins but not arteries to alter the pharmacology of the receptors, I designed contractility experiments utilizing selective ET, and ET, receptor antagonists. In these experiments, the ability of an ET, receptor antagonist, atrasentan, to shift ET -1-induced contraction was compared in aorta and vena cava with unbound and agonist-bound ET, receptors (accomplished by desensitization with S6c). I observed that in aorta, ET, receptor blockade (determined by comparing EC50 values) was not significantly different whether ET, receptors were unbound or agonist-bound (desensitized with $60) (Flgure 21). Differently from the aorta, in vena cava ET, receptor blockade was quantitatively greater when ET, receptors were agonist-bound (S6c desensitized) 114 compared to when ET, receptors were unbound (Flgure 22), suggesting that ET, and ET, receptors functionally interact in vena cava but not aorta. As another way of assessing ET, receptor function (via atrasentan inhibition of ET-1-induced contraction) in the presence and absence of ET, receptors, ET, receptor blockade with the selective ET, receptor antagonist 80-788. In aorta, the degree of ET, receptor-inhibited ET-1-induced contraction was not different whether ET, were not blocked (vehicle incubated) or blocked (BO-788 incubated) (Figure 23). However, in vena cava, ET, receptor antagonism inhibited ET-1- induced contraction to a significantly greater extent when ET, receptors were rendered blocked with BQ-788 (100 nM), compared to vehicle-incubated vena cava without ET, receptor blockade (Figure 24). These data support the idea that functional ET receptor interaction occurs in vena cava but not aorta. It was a goal of this dissertation to understand why and how this receptor interaction occurs in veins but not arteries. Our studies are not the first to describe functional ET, and ET, receptor interaction. Lodge at a/ initially described in rabbit saphenous vein and jugular vein (vessels that have both contractile ET, and ET, receptors) a population of ET, receptors that were only sensitive to ET, receptor blockade when ET, receptors were either desensitized or antagonized (Lodge at al, 1995). In vessels with only contractile ET, receptors, such as the rat aorta and rabbit 115 carotid artery, ET, receptor desensitization or inhibition had no effect on ET, receptor function (Lodge at al, 1995). Adner at al observed in small mesenteric arteries that ET, receptor desensitization increased ET -1 potency and the apparent affinity of an ET, receptor antagonist (Adner at al, 2001). Receptor “cross-talk" between the ET, and ET, receptors has also been described to occur in rat trachea where ET, receptor or ET, receptor antagonism alone was not sufficient to inhibit ET -1 -induced contraction, but dual ET, and ET, receptor blockade significantly inhibited ET-1-induced contraction (CIozeI and Gray, 1995). Similar results were observed in human isolated bronchi (Fukuroda et al, 1996), rat mesenteric veins but not arteries (Claing at al, 2002), mouse mesenteric veins but not arteries (Perez-Rivera et al, 2005), rat renal afferent but not efferent arterioles (lnscho at al, 2005) and pulmonary arteries (Sauvageau at al, 2006), suggesting that ET, and ET, receptor “cross-talk" or interaction may occur in vessels that have both functional ET, and ET, receptors. Why some vessels have both contractile ET, and ET, receptors and others only have contractile ET, receptors (I would hypothesize veins and arteries, respectively) is still unknown. D. Signaling pathway Interactions may account for functlonal venous ET, and ET, receptor lnteractlon One possible explanation for how ET, and ET, receptors functionally interact in vena cava but not aorta is that cross-talk or interaction between ET, and ET, 116 receptor-induced signal transducers could occur. The ET, and ET, receptors activate multiple signal transduction pathways such as phospholipases (A, B and D), MAPK kinases (Erk, JNK and p38), PKC, rho kinase and tyrosine kinases such as src (Figures 8-10) via coupling to several G-proteins, including G, 6,, G, and (5,2,3. ET-1 also increases intracellular calcium through opening of non- selective cation channels, L-type voltage gated Ca2+ channels, and Ca2+ release from intracellular stores (both lP,-sensitive Caz” channels and ryanodine- sensitive Ca2+ channels) (Neylon, 1999). ET-1 also stimulates production of reactive oxygen species (Li et al, 2003a; Thakali at al, 2005; Wedgwood et al, 2001), which can potentially induce signaling in addition to what is activated by ET-1 itself. I envisioned in vena cava, which have both functional ET, and ET, receptors, ET-1-activated signal transduction pathways coupling to contraction would differ from aorta that only have functional ET, receptors. I also hypothesized that differences in ET -1 signaling between aorta and vena cava could explain how functional ET, and ET, receptor interaction occurs in vena cava but not aorta. 1. Reactlve oxygen specles slgnallng Several groups have reported that ET-1 via ET, receptors activates the superoxide-producing enzyme NADPH oxidase (Banes-Berceli er al, 2005; Li et al, 2003a; Loomis at al, 2005; Wedgwood ef al, 2001). Superoxide, the product of NADPH oxidase activity, H202 generated by the catalytic reduction of 117 superoxide by superoxide dismutase, and hydroxyl radical are examples of ROS that can alter vascular tone (Ardanaz and Pagano, 2006; Lyle and Griendling, 2006). Superoxide can directly constrict smooth muscle as superoxide generating systems constrict endothelium-denuded arteries (Jin et al, 2004), and can potentiate constriction by quenching nitric oxide, the endothelium derived relaxing factor. H202 has been reported to induce both contraction and relaxation, depending on experimental conditions, species studied and vascular bed studied (Lucchesi er al, 2005; Gil-Longo and Gonzalez-Vazquez, 2005). | hypothesized that ROS production would differ between aorta and vena cava because of the different complement of functional ET receptors in these two vessels. I chose to focus on the ability of ET-1 in aorta and vena cava to stimulate production of H202 instead of superoxide for the following reasons: 1) H202 is not a free radical like superoxide and is inherently more stable than superoxide; 2) H20, has a larger diffusion radius compared to superoxide; and 3) H202 can cross membranes. Thus, in aorta and vena cava I compared basal levels of H202 production, ET-1-stimulated H202 production, the ability of exogenously added H202 to alter vascular tone, and whether H202 mediates ET- 1-induced contraction. When basal H202 levels were measured using the Amplex Red H202 assay, I observed that basal levels of H20, production were significantly higher in vena cava compared to aorta (Flgure 26). Measurable H202 levels were nearly 118 A completely reduced when vessels were incubated with diethyldithiocarbate (DDC or DETC), an inhibitor of superoxide dismutase (the enzyme that catalyzes the hydrolysis of superoxide to H202), suggesting that the primary source of H202 in both aorta and vena cava was via the reduction of superoxide. In isolated tissue bath experiments, exogenously added H20, contracted both rat thoracic aorta and vena cava and the contraction in vena cava was much more robust than that in aorta (when the contraction to H202 was normalized to maximal contraction to an adrenergic agonist) (Flgure 25). I observed that ET -1 increased H20, production in vena cava but not aorta (Flgure 27). While the ET-l-stimulated H202 production in vena cava was not concentration-dependent (for the concentrations investigated), inhibitors of ET, and ET, receptors significantly reduced ET-1-induced H20, production, suggesting that ET-1-stimulated H202 production was not a non-specific effect of ET-1 (Flgure 28). Since ET-1 stimulated H202 production in vena cava and exogenous H20, constricted vena cava l hypothesized that venous ET-1-induced contraction might be partially mediated by ET -1-stimulated H202. Thus, removing or reducing endogenous H202 with catalase should have reduced venous ET -1-induced contraction. Contrary to what I expected, when vena cava were incubated with catalase or PEG-catalase (the PEG moiety supposedly increases cell permeability), maximum ET -1 -induced contraction was enhanced, suggesting that endogenous H20, may mediate vasodilation in these vessels (Figure 29). In the presence of 3-aminotriazole, a catalase inhibitor, venous ET -1-induced contraction was not 119 altered from vehicle-incubated vena cava (Flgure 30). Collectively, these data suggest that though ET-1 stimulates H202 production in vena cava and exogenous H202 constricts vena cava, it is unlikely that ET-1-stimulated H20, mediates venous ET -1 -induced contraction. From these experiments I concluded that ET-1 signaling via H202 production was not a likely mediator of ET, and ET, receptor functional interaction in vena cava. It is possible that ET -1 activates signaling pathways through the production of other reactive oxygen species such as superoxide or hydroxyl radical thus I may have limited our conclusions by only investigating the actions of H202. 2. ET-1 slgnal transduction Since ET -1-stimulated H202 signaling does not explain why functional ET, and ET, receptor interaction occurs in vena cava, I chose to examine which specific signal transducers were activated by ET, and ET, receptors that couple to contraction in aorta and vena cava. I chose to investigate the involvement of p38 MAPK, Erk MAPK, src, rho kinase and PI3-K in mediating ET-1-induced contraction in aorta and vena cava because these pathways are known to couple to smooth muscle contraction (Somlyo and Somlyc, 2000; Hilgers and Webb, 2005), have been reported to mediate ET-t-induced contraction (Yamboliev at al, 2000; Zubkov et al, 2000; Miao ef al, 2002; Kawanabe at al, 2004, Kodama er al, 2003), and are important in arterial hyperresponsiveness in hypertension 120 (Northcott er al, 2002; Touyz, 2003; Kwon at al, 2004; Wehrwein at al, 2004). While there are many reports investigating ET-1 signal transduction in the literature, most of these reports recount ET-1 signaling in cells over-expressing ET, and ET, receptors and cultured smooth muscle cell lines, I.e. they have investigated ET-1 signal transduction under non-physiological conditions. Of the reports regarding ET-1 signaling, different vascular beds have been studied, different species of animals have been studied, and experimental protocols vary enough such that a generalized statement summarizing ET receptor signaling in the vasculature would likely be unfair to make. To determine which signaling pathways mediated aortic and venous ET-1- induced contraction, I compared the ability of specific signaling inhibitors to reduce ET -1-induced contraction. I observed that pharmacological inhibition of rho kinase significantly inhibited both aortic and venous ET-1-induced contraction (Figure 35) and p38 MAPK inhibition marginally reduced maximal aortic ET-1- induced contraction (Flgure 34). Inhibitors of Erk MAPK, src and Pl3-K did not alter maximal aortic or venous ET-t-induced contraction (Flgures 31-33), suggesting that activation of these pathways is not necessary for aortic or venous ET-1-induced contraction. Using cultured aortic smooth muscle cells, our group previously verified that the concentrations of inhibitors used caused selective inhibition of the corresponding signaling molecule (Watts, 1996; Banes et al, 1999; Florian and Watts, 1999; Northcott et al, 2002) and except for the rho 121 kinase inhibitor Y27632, did not reduce non-receptor mediated contraction (KCI- induced contraction, data not shown). Since the rho kinase pathway was similarly involved in mediating ET-1-induced contraction in both aorta and vena cava and p38 MAPK played only a minor role in aortic ET-1-induced contraction, I chose not to further investigate specific ET, and ET, receptor signaling. I concluded that ET -1-mediated signal transduction via Erk MAPK, src and Pl3-K were not required for aortic and venous ET -1-induced contraction, thus these pathways are likely not responsible for ET, and ET, receptor interaction in vena cava. I expected that since vena cava have both contractile ET, and ET, receptors and aorta only have contractile ET, receptors, ET -1 -induced signaling and contraction in vena cava would be mediated by several signaling transduction pathways that would be distinctly different from ET, receptor mediated signaling and contraction in aorta. However, of the signaling pathways investigated, both aortic and venous ET-t-induced contraction was similarly dependent on rho kinase activity, suggesting that signaling interactions through the rho kinase, p38 MAPK, Erk MAPK, src and Pl3-K pathways are not likely responsible for mediating functional venous ET receptor interaction. There are other mechanisms/signaling pathways involved in smooth muscle contraction that I did not investigate. It is possible that by only investigating a select number of signaling pathways, l have overlooked other pathways that contribute to ET, and ET, receptor interaction in vena cava 122 w——~ or that there yet undiscovered pathways mediating ET, and ET, receptor contraction in both aorta and vena cava. An important mechanism of smooth muscle contraction that I chose not to investigate was the increase in intracellular calcium. ET -1-induced contraction and increases in intracellular calcium in dissociated rat mesenteric arteries and veins was insensitive to L-type calcium channel blockade with nifedipine (Claing et al, 2002), Supporting previous studies by Guilumian at al who demonstrated that ET -1-induced contraction in small mesenteric arteries was dependent on sarcoplasmic reticular ryanodine receptor activation and intracellular calcium release (Giulumian at al, 2000). However, in porcine coronary arteries, extracellular calcium influx mediated ET-1-induced contraction as voltage-gated calcium channel blockers reduced ET -1-induced contraction (Kasuya at al, 1989), suggesting that cellular mechanisms regulating ET -1-induced increases in intracellular calcium vary depending on species and vascular bed studied. Claing et al observed that ET, receptor activation increased intracellular calcium in mesenteric vein smooth muscle cells but not in mesenteric artery smooth muscle cells (Claing at al, 2002), suggesting that there may be differences in ET -1- induced calcium signaling in arteries and veins because of the presence of both contractile ET, and ET, receptors on veins. 123 l have preliminary evidence suggesting that H202-induced contraction in aorta and vena cava differ in their dependence on extracellular Ca2+ influx. Vena cava contract to H202 in the presence of L-type Ca2+ blockers and when Ca2+ is omitted from PSS, while aortic contraction to H202 (after KCI contraction) is completely inhibited when Ca'“ is removed from PSS (data not shown), suggesting that aorta require extracellular Ca2+ influx for H202-induced contraction, while vena cava do not. While it is likely the conclusions I can make regarding differences in Ca2+ handling in aorta and vena cava may only be limited to H202-induced contraction, the source intracellular Ca“ (extracellular influx vs. release of intracellular stores) may be an important difference between contraction of aorta and vena cava and may provide an explanation to why functional ET, and ET, receptor interaction is observed in vena cava but not aorta. E. Can ET, and ET, receptors physlcally Interact vla' mceptor heterodlmerlzatlon? Another possible explanation for why I observed functional ET, and ET, receptor interaction in vena cava but not aorta is that ET, and ET, receptors might physically interact in vena cava via receptor heterodimerization. G-protein receptor dimerization, originally proposed in the early 1980s, has recently emerged as an important phenomenon that regulates the function and pharmacology of G-protein coupled receptors. Several G-protein coupled receptors have been reported to homodimerize (two receptors of the same 124 subtype physically interacting), heterodimerize (two receptors of different subtypes physically interacting), and oligomerize (multiple receptors physically interacting with each other). The best characterized example of receptor homodimerization is that of B, adrenergic receptors (reviewed by Prinster et al, 2005). Receptor heterodimerization of GABA,1 and GABA,, receptors, (3, adrenergic receptors and angiotensin (AT)1 receptors, taste receptors (T1 R2” 1 R3; T1R1/T 1 R3), 6, adrenergic receptors and opioid (5 and x) receptors, opioid receptors (M: and 6m), somatostatin receptors, purinergic receptors and olfactory receptors have been investigated in depth (reviewed by Prinster et al, 2005). Receptor dimerization can alter how receptors function by altering downstream signal transduction events, receptor internalization, receptor localization (specifically endoplasmic reticulum retention and membrane trafficking), and receptor pharmacology (agonist affinity and efficacy) (Angers et al, 2002; Milligan, 2004; Maggio et al, 2005; Prinster at al, 2005). Currently, reports of G-protein coupled receptor dimerization are primarily limited to cells systems overexpressing receptors. Heterologous overexpression systems are excellent tools for assessing receptor dimerization because these systems can be easily modified and manipulated to determine how GPCRs can physically interact with each other. One caveat to using heterologous overexpression systems is that these are immortalized cell lines, with significantly different properties from the native cells or tissues where GPCRs would normally exist. In the few studies investigating the physiological significance of receptor 125 dimerization, conclusions are typically based on indirect observations of receptor function, thus the physiological significance of receptor dimerization has not yet been fully explored and heterologous overexpression systems are currently the best available tools for assessing GPCR dimerization. It is important to note that of all available prescription drugs, approximately half of these rely on G-protein coupled receptors to achieve their therapeutic effects, thus receptor dimerization may be of clinical significance. Like many other G-protein coupled receptors, the ET, and ET, receptors, when over-expressed in HEK293 cells, can heterodimerize (Gregan et al, 2004). Gregan er al demonstrated that while ET, and ET, receptor heterodimerization was constitutive, prolonged ET, receptor activation caused the ET, and ET, receptor heterodimer to dissociate, likely due to ET, receptor internalization (Gregan et al, 2004). Dai at al confirmed that the ET, and ET, receptors co- localize (suggesting that the ET, and ET, receptors heterodimerize) in an independent study over-expressing human ET, and ET, receptors in HEK293 cells and that co-transfection of ET, and ET, receptors altered receptor desensitization and trafficking (Dai and Galligan, 2006). While these studies by Gregan er al and Dai at al demonstrate that in fact ET, and ET, receptors can heterodimerize and colocalize, respectively, whether ET, and ET, receptors heterodimerize in tissues, such as rat thoracic aorta or vena cava, and whether receptor heterodimerization alters receptor or tissue function is still unknown. 126 The following approaches are generally accepted for assessing G-protein coupled receptors dimerization: 1) there should be some measure of functional receptor interaction (though personal interpretation of functional responses can be highly variable); 2) the receptors should co-immunoprecipitate/co-localize; and 3) fluorescence resonance energy transfer (FRET) or another measure of receptor proximity (such as BRET) should demonstrate that the receptors are within close proximity to each other (Milligan and Bouvier, 2005). Taking these guidelines in to consideration, I initially proposed to perform co- immunoprecipitation experiments with the hypothesis that ET, and ET, receptors would co-immunoprecipitate in venous but not aortic lysates. I also proposed to perform FRET experiments with fluorescently labeled ET, and ET, receptors on aortic and venous vascular smooth muscle cells to demonstrate that ET, and ET, receptors were in closer proximity to each other in venous compared to aortic vascular smooth muscle cells because of receptor heterodimerization. Both the co-immunoprecipitation and FRET experiments relied on the use of antibodies to selectively detect ET, and ET, receptors. Antibody selectivity can often be questionable and since the sequences of the human ET, and ET, receptor are approximately 60% homologous, I compared the selectivity of several commercially available ET, and ET, receptor antibodies. Also, FRET experiments required ET, and ET, receptor antibodies that were raised in different species. I performed a series of dot blots on which increasing amounts 127 of antigenic peptides (which the different antibodies were raised against) were spotted on a nitrocellulose membrane. Based on the dot blot results (Figures 37 and 38) I decided to use the Alomone ET, and ET, receptor primary antibodies for co-immunoprecipitation experiments and the Fitzgerald ET, and Alomone ET, receptor primary antibodies for co-Iocalization experiments. 1. Co-lmmunopreclpltatlon of ET, and ET, receptors In rat thoracic aorta and vena cava Since I hypothesized that ET, and ET, receptors would heterodimerize in veins but not arteries, I expected that ET, and ET, receptors would co- immunoprecipitate in vena cava but not aorta. I initially immunoprecipitated the ET, receptor using the Alomone ET, receptor antibody and asked if the ET, receptor (detected with Alomone ET, receptor primary antibody) co- immunoprecipitated with the ET, receptor. Unfortunately, contamination with the 50 kDa heavy chain of the immunoprecipitating antibody prevented detection of either the ET, and ET, receptors as both of these proteins also resolve at approximately 50 kDa. Since the traditional immunoprecipitation method was not successful for determining if ET, and ET, receptors co-immunoprecipitate, I tested several commercially available co-immunoprecipitation kits to attempt to avoid detection of the immunoprecipitating antibody (Table 4). Unfortunately, none of these kits/techniques were successful in detecting co- 128 immunoprecipitated ET, and ET, receptors, and in fact none of these kits were successful in confirming/detecting a successful immunoprecipitation. I also attempted to crosslink ET, and ET, receptors with the idea that if the receptors were in close proximity of each other, cross-linking them would slow their migration through a polyacrylamide gel. This would allow us to detect cross- Iinked ET, and ET, receptors that should theoretically migrate at a higher molecular weight than uncross-Iinked ET, and ET, receptors, resolving immunoreactive bands for the ET, and ET, receptors separately from the immunoprecipitating antibody band. However, after incubating both vessels and lysates with cross-linkers, the degree of protein degradation was too high for Western blot analysis. It is probable that I chose the inappropriate cross-linker for cross-linking ET, and ET, receptors, as how ET, and ET, receptors physically associate is still unknown. It is also possible that ET, and ET, receptor heterodimerization may be an artifact of receptor overexpression in a cell line and does not occur in blood vessels. 2. Receptor co-locallzatlon In conclusion, our receptor co-immunoprecipitation experiments were inconclusive as l was unable to either confirm or deny if ET, and ET, receptors co-immunoprecipitate in vena cava but not aorta. Since the co- immunoprecipitation experiments were inconclusive I next attempted receptor co- 129 localization and FRET experiments to determine if ET, and ET, receptors physically interact via receptor heterodimerization in vena cava but not aorta. Ideally I would have performed receptor co-localization and FRET analysis on paraffin-embedded sections of rat thoracic aorta and vena cava to determine where, in whole vessels, ET, and ET, receptors were located. However, the collagen and elastin fibers of blood vessels autofluoresce at the wavelengths of commonly used fluorescent secondary antibodies. l was primarily concerned with the location of ET, and ET, receptors on the smooth muscle cells of aorta and vena cava, as l have assumed that vascular smooth muscle cells are the cell types where ET, and ET, receptors would heterodimerize and affect receptor function. I enzymatically dissociated vascular smooth muscle cells from rat thoracic aorta and vena cava, adhered the cells to coverslips and performed immunocytochemistry experiments on fixed vascular smooth muscle cells. I labeled the ET, and ET, receptors with the Fitzgerald ET, and Alomone ET, receptor primary antibodies, respectively and labeled the plasma membrane with an antibody for pan-cadherin (a membrane marker). Confocal images of 6 micron-thick sections of the whole smooth muscle cell (from the top to bottom) were captured for comparison of the cellular location of ET, and ET, receptors from aortic and venous vascular smooth muscle cells. Surprisingly, I observed that in both aortic and venous vascular smooth muscle cells, both ET, and ET, receptors co-localized at the plasma membrane (Flgures 130 40 and 41). I observed that ET, receptors in both aorta and vena cava were primarily located at the membrane (co-localized with pan-cadherin staining), while ET, receptors were found both at the plasma membrane and intracellularly. The membrane marker, pan-cadherin, was present at the plasma membrane of vascular smooth muscle cells, but was also present intracellularly. It is possible that the intracellular presence of ET, receptors and pan-cadherin may be an artifact of our immunocytochemistry protocol (perhaps resulting from fixation or cell permeabilization) or may be due to antibody non-specificity (though our dot blots suggest that the Alomone ET, receptor antibody is selective for its antigenic sequence). Human ET, receptors overexpressed in HEK293 cells were primarily expressed on the plasma membrane, while ET, receptors overexpressed in HEK293 cells were present equally expressed on the plasma membrane and intracellular compartments (Dai and Galligan, 2006). When both ET, and ET, receptors were co-transfected in HEK293 cells, both receptors were present at the plasma membrane, suggesting that ET, receptor expression was necessary for membrane targeting of the ET, receptor (Dai and Galligan, 2006). l was also interested in determining if ET, and ET, receptors in aorta and vena cava internalize differently after ET-1 stimulation. I observed that stimulation of aortic and venous vascular smooth muscle cells with ET-1 did not qualitatively alter ET, or ET, receptor location, I.e. ET, and ET, receptors co-localized at the plasma membrane in both aortic and venous vascular smooth muscle cells even 131 after ET-1 stimulation (Figures 42 and 43). Our observations are in agreement with Gregan et al who observed that ET, and ET, receptors remained at the plasma membrane even after 60 minutes of ET-1 stimulation in HEK293 cells co- transfected with ET, and ET, receptors (Gregan at al, 2004). Interestingly, Gregan et al observed that selective ET, receptor activation induced ET, receptor internalization in co-transfected cells, suggesting that selective ET, receptor activation disrupted the constitutive ET,/ET, receptor heterodimer. There are other possible explanations for why I did not observe ET, and ET, receptor internalization after ET-1 stimulation, the simplest but not necessarily the correct explanation being that ET, and ET, receptors on vascular smooth muscle cells do not internalize with ET -1 stimulation, though the contractile response to ET-1 desensitizes. Immunocytochemistry experiments in freshly dissociated vascular smooth muscle cells were performed in the presence of sodium nitroprusside, a nitric oxide donor, and in the absence of Ca2+ to optimize imaging conditions of the vascular smooth muscle cells. To mt knowledge, it is unknown if the presence of nitric oxide or the absence of Ca2+ acutely affects ET, and ET, receptor signaling, localization and internalization. While immunocytochemistry results suggested that in both aorta and vena cava ET, and ET, receptors co-localize to the plasma membrane of vascular smooth muscle cells, the resolution (4000 — 7000 A) of conventional confocal microscopy 132 does not give any information about molecular receptor proximity. I proposed to perform FRET experiments where one receptor subtype (for example the ET, receptor) was labeled with a donor fluorophore and the other receptor subtype (the ET, receptor) was labeled with an acceptor fluorophore and the donor and acceptor fluorophores have overlapping excitation/emission spectra. If the two receptors are within close enough proximity (within a nanometer range) (Wallrabe and Periasamy, 2005), specific excitation of the donor fluorophore will cause the donor fluorophore to emit energy that will in turn increase acceptor fluorescence while quenching donor emission. Successful FRET experiments depend on several factors including primary antibody binding efficiency, fluorophore dipole alignment, distance between donor and acceptor fluorophores (which becomes an issue when fluorescent secondary antibodies are used to detect primary antibody binding) and spectral bleed through due to spectral overlap of donor and acceptor fluorophores. In immunocytochemistry experiments of dissociated aortic and venous vascular smooth muscle cells I labeled the ET, receptor with the Alexa488® secondary antibody and the ET, receptor with the Alexa555® secondary antibody. The Alexa fluorophores are brighter than other fluorophores that emit at similar wavelengths, are more photostable (resistant to bleaching) (Panchuk—Voloshina ef al, 1999), and this pair of donor/acceptor fluorophores (Alexa488®/Alexa555®) has been successfully used in FRET experiments 133 (Wallrabe and Periasamy, 2005). In our experiments with fluorescently labeled ET, and ET, receptors on aortic and venous vascular smooth muscle cells, I did not observe significant bleed through between the two fluorescent secondary antibodies (Alexa488® and Alexa555®) or background fluorescence, but the intensity of fluorescence was very low. Because of the low fluorescence intensity from labeled ET, and ET, receptors, I concluded that FRET experiments were not likely to be successful. F. Conclusions I hypothesized that ET, and ET, receptors heterodimerize in rat thoracic vena cava but not aorta, and that receptor heterodimerization would alter ET, and ET, receptor function in vena cava. l generated contractility data that suggests that ET, and ET, receptors functionally interact in vena cava but not aorta. In vena cava, when ET, receptors were rendered nonfunctional, either through receptor desensitization or receptor blockade, ET, receptors were more susceptible to receptor blockade, suggesting that in vena cava, ET, receptors modulate ET, receptor function. I investigated whether signal transduction interactions could explain why ET, and ET, receptors functionally interact in vena cava, but concluded that signaling via the following pathways — H202, p38 MAPK, Erk MAPK, src, PI3-K and rho kinase - were not responsible for ET, and ET, receptor cross-talk in vena cava. Receptor co-localization experiments suggested that ET, and ET, receptors co-Iocalize at the plasma membrane of 134 both aortic and venous vascular smooth muscle cells, suggesting that receptor location does not govern functional receptor interaction. Unfortunately, other methods of assessing physical ET, and ET, receptor interaction, such as oo- immunoprecipitation experiments and FRET experiments did not provide any conclusive results regarding the physical association and proximity of ET, and ET, receptors in aortic and venous vascular smooth muscle cells. G. Speculatlon It is possible that ET, and ET, receptors, when over-expressed at artificially high levels heterodimerize with alterations in receptor pharmacology, but when these receptors are normally expressed in tissues, like vascular smooth muscle cells, they do not heterodimerize or their physical location does not determine receptor function. It is also possible that I relied too heavily on the current tools available to assess receptor heterodimerization, specifically commercially available ET, and ET, receptor antibodies. I expect that future development of better antibodies for the rat ET, and ET, receptors and novel co-immunoprecipitation protocols will allow FRET experiments and receptor co-immunoprecipitation experiments to be performed to determine if ET, and ET, receptors do in fact heterodimerize in tissues such as the rat thoracic vena cava. Future experiments investigating how ET, and ET, receptors increase intracellular calcium in arteries and veins are likely important for understanding 135 how functional ET, and ET, receptor interaction occurs in rat thoracic vena cava. In general, there is a lack of data surrounding venous vascular biology and to our knowledge, there has yet been a comprehensive comparison of calcium handling in arteries and veins. The use of ET, receptor deficient rats may be useful in determining if ET, and ET, receptors heterodimerize in vena cava but not aorta. I have preliminary contractility data that suggests that homozygous ET, receptor deficient rats do not exhibit functional ET, and ET, receptor interaction, unlike their homozygous wild type counterparts, confirming the idea that a contractile ET, receptor is required for ET,/ET, receptor interaction. Western blot analysis suggests that both homozygous ET, receptor deficient and ET, receptor wild type rats express the ET, receptor In vena cava, but in the homozygous ET, receptor deficient rats, the ET, receptor is nonfunctional and does not induce contraction. In the future, immunocytochemistry experiments should be performed to determine cellular localization of ET, and ET, receptors in vena cava and aorta of homozygous ET, receptor deficient and ET, receptor wild type rats. Receptor dimerization is a nascent field in receptor characterization and has more recently been recognized as a phenomenon with potentially clinical ramifications as many therapeutic drugs target G-protein coupled receptors. Most accounts of receptor dimerization have been reported in cell lines 136 overexpressing tagged receptors. Only a few reports question whether receptor dimerization is a physiological phenomenon with functional consequences. While our data suggest that functional ET, and ET, receptor interaction in vena cava does not appear to be dependent on receptor co-localization, only with improved tools to assess receptor dimerization, like better ET, and ET, receptor antibodies and novel co-immunoprecipitation approaches, can I actually determine if ET, and ET, receptors heterodimerize when natively expressed in tissues. 137 V. REFERENCES AbdAlla S, Lother H, Abdel-Tawab AM, Quitterer U. The angiotensin II AT2 receptor is an AT1 receptor antagonist. J Biol Chem. 2001;276:39721-39726. Adams LD, Geary RL, McManus B, Schwartz SM. A comparison of aorta and vena cava medial message expression by cDNA array analysis identifies a set of 68 consistently differentially expressed genes, all in aortic media. Circ Res. 2000;87(7):623-631 . Adner M, Shankley N, Edvinsson L. Evidence that ET-1, but not ET-3 and 86b, ET(A)-receptor mediated contractions in isolated rat mesenteric are modulated by co-activation of ET(B) receptors. Br J Pharmacol. 2001;133(6):927-935. Angers S, Salahpour A, Joly E, Hilairet S, Chelsky D, Dennis M, Bouvier M. Detection of beta 2-adrenergic receptor dimerization in living cells using bioluminescence resonance energy transfer (BRET). Proc Natl Acad Sci U S A. 2000;97(7):3684-3689. Aquilla E, Whelchel A, Knot HJ, Nelson M, Posada J. Activation of multiple mitogen-activated protein kinase signal transduction pathways by the endothelin B receptor requires the cytoplasmic tail. J Biol Chem. 1996;271(49):31572- 31579. Aramori I, Nakanishi S. Coupling of two endothelin receptors subtypes to differing signal transduction in transfected Chinese hamster ovary cells. J Biol Chem. 1 992;267(1 8): 1 2468-1 2474. Ardanaz N, Pagano PJ. Hydrogen peroxide as a paracrine vascular mediator: regulation and signaling leading to dysfunction. Exp Biol Med (Maywood). 2006;231:237-251 . Banes A, Florian JA, Watts SW. Mechanisms of 5-hydroxytryptamine(2A) receptor activation of the mitogen-activated protein kinase pathway in vascular smooth muscle. J Pharmacol Exp Ther. 1999;291:1179-1187. Banes-Berceli AK, Ogobi S, Tawfik A, Patel B, Shirley A, Pollock DM, Fulton D, Marrero MB. Endothelin-1 activation of JAK2 in vascular smooth muscle cells involves NAD(P)H oxidase-derived reactive oxygen species. Vascul Pharmacol. 2005;43:310-319. Bohm F, Pemow J, Lindstrom J, Ahlborg G. ET, receptors mediate vasoconstriction whereas ETB receptors clear endothelin-1 in the splanchnic and renal circulation of healthy men. Clin Sci (Lond). 2003;104(2):143-151. 138 Bomfeldt KE, Krebs EG. Crosstalk between protein kinase A and growth factor receptor signaling pathways in arterial smooth muscle. Cell Signal. 1999;1 1 (7)2465-477. Boron WF, Boulpaep EL, eds. Medical Physiology. Elsevier Science (USA), 2003: 447-462. Bremnes T, Paasche JD, Mehlum A, Sandberg C, Bremnes B, Attramadal H. Regulation and intracellular trafficking pathways of the endothelin receptors. J Biol Chem. 2000;275:17596-17604. Callera GE, Touyz RM, Teixeira SA, Muscara MN, Carvalho MH, Fortes ZB, Nigro D, Schiffrin EL, Tostes RC. ETA receptor blockade decreases vascular superoxide generation in DOCA-salt hypertension. Hypertension. 2003;42(4):81 1-817. Claing A, Shbaklo H, Plante M, Bkaily G, D’Orleans-Juste P. Comparison of the contractile and calcium-increasing properties of platelet-activating factor and endothelin-1 in the rat mesenteric artery and vein. Br J Pharmacol. 2002;135(2):433-443. Clozel M, Gray GA. Are there different ET, receptors mediating constriction and relaxation? J Cardiovasc Pharmacol. 1995;26 Suppl 3:8262-S264. Cocks TM, Faulkner NL, Sudhir K, Angus J. Reactivity of endothelin-1 on human and canine large veins compared with larger arteries in vivo. Eur J Pharmacol. 1989;171:17-24. Cvejic S, Devi LA. Dimerization of the delta opioid receptor: implication for a role in receptor internalization. J Biol Chem. 1997;272(43):26959-26964. Dai X, Galligan JJ. Differential trafficking and desensitization of human ET (A) and ET(B) receptors expressed in HEK 293 cells. Exp Biol Med (Maywood). 2006;231 :746-751 . Decker ER, Brok TA. Endothelin receptor-signaling mechanisms in vascular smooth muscle. In: Highsmith RF, ed. Endothelin: Molecular biology, physiology and pathology. New Jersey: Humana Press Inc 1998: 93-120. Dong J-M, Leung T, Manser E. Lim L. cAMP-induced morphological changes are counteracted by activated RhoA small GTPase and the Rho Kinase ROKa. J Biol Chem. 1998;273(35):22554»22562. 139 D’Orleans-Juste P, Labonte J, Bkaily G, Choufani S, Plante M, Honore JC. Function of the endothelinB receptor in cardiovascular physiology and pathophysiology. Pharmacol Ther. 2002;95(3):221-238. D’Orleans-Juste P, Plante M, Honore JC, Carrier E, Labonte J. Synthesis and degradation of endothelin-1. Can J Physiol Pharmacol. 2003;81:503-510. Dreja K, Voldstedlund M, Vinten J, Tranum-Jensen J, Hellstrand P, Sward K. Cholesterol depletion disrupts caveolae and differentially impairs agonist-induced arterial contraction. Arterioscler Thromb Vasc Biol. 2002;22(8):1267-1272. Duerrschmidt N, Wippich N, Goettsch W, Broemme H—J, Morawietz. Endothelin-1 induces NAD(P)H Oxidase in human endothelial cells. Biochem Biophys Res Comm. 2000;269:713-717. Ekelund U, Adner M, Edvinsson L, Mellander S. Effects of selective ETB-receptor stimulation on arterial, venous and capillary functions in cat skeletal muscle. Br J Pharmacol. 1994;1 12(3):887—894. Fernandez-Patron C, Radomski MW, Davidge ST. Vascular matrix metalloproteinase-2 cleaves big endothelin-1 yielding a novel vasoconstrictor. Circ Res. 1999;85(10):906-911. Florian JA, Watts SW. Epidermal growth factor: a potent vasoconstrictor in experimental hypertension. Am J Physiol. 1999 ;276:H976-H983. Freedman NJ, Ament AS, Oppermann M, Stoffel RH, Exum ST, Lefkowitz RJ. Phosphorylation and desensitization of human endothelin A and B receptors. Evidence for G protein-coupled receptor kinase specificity. J Biol Chem. 1997 1 1 ;272:17734-17743. Fukuroda T, Ozaki S, lhara M, lshikawa K, Yano M, Miyauchi T, lshikawa S, Onizuka M, Goto K, Nishikibe M. Necessity of dual blockade of endothelin ET, and ET, receptor subtypes for antagonism of endothelin-1-induced contraction in human bronchi. BrJ Pharmacol. 1996;1 17:995-999. Gil-Longo J, Gonzalez-Vazquez C. Characterization of four different effects elicited by H202 in rat aorta. Vascul Pharmacol. 2005;43:128-138. Giulumian AD, Molero MM, Reddy VB, , Pollock JS, Pollock DM, Fuchs LC. Role of ET-1 receptor binding and [Cay]. in contraction of coronary arteries from DOCA-salt hypertensive rats. Am J Physiol Heart Circ Physiol.2002;282:H1944- H1949. 140 Gohla A, Offermanns S, Wilkie TM, Schultz G. Differential involvement of Galpha12 and Galpha13 in receptor-mediated stress fiber formation. J Biol Chem. 1999;274(25):17901-17907. Gohla A, Schultz G. Offermanns 8. Role for G(12)/G(13) in agonist-induced vascular smooth muscle cell contraction. Circ Res. 2000;87(3):221-227. Goldsmith PK, Fan GF, Ray K, Shiloach J, McPhie P, Rogers KV, Speigel AM. Expression, purification, and biochemical characterization of the amino-terminal extracellular domain of the human calcium receptor. J Biol Chem. 1999;274(16):11303-11309. Gomes I, Jordan BA, Gupta A, Trapaidze N, Nagy V, Devi LA. Heterodimerization of mu and delta opoioid receptors: A role in opiate synergy. J Neurosci. 2000;20(22):RCI 10. Gregan B, Jurgensen J, Papsdorf G, Furkert J, Schaefer M, Beyerrnann M, Rosenthal W, Oksche A. Ligand-dependent differences in the internalization of endothelin A and endothelin B receptor heterodimers. J Biol Chem. 2004;279(26):27679-27687. Grubbs AL, Anstadt MP, Ergul A. Saphenous vein endothelin system expression and activity in African American patients. Arterioscler Thromb Vasc Biol. 2002;22(7):1122-1 127. Guzik TJ, Sadowski J, Kapelak B, Jopek A, Rudzinski P, Pillai R, Korbut R, Channon KM. Systemic regulation of vascular NAD(P)H oxdiase activity and nox isoform expression in human arteries and veins. Arterioscler Thromb Vasc Biol. 2004;24:1614-1620. Harada N, Himeno A, Shigematsu K, Sumikawa K, Niwa M. Endothelin-1 binding to endothelin receptors in the rat anterior pituitary gland: possible formation of an ETA-ET, receptor heterodimer. Cell Mol Neurobiol. 2002;22(2):207-226. Haynes WG, Ferro CJ, O’Kane KP, Somerville D, Lomax CC, Webb DJ. Sytemic endothelin receptor blockade decreases peripheral vascular resistance and blood pressure in humans. Circulation. 1996;93(10):1860-1870. Hebert TE, Moffett S, Morello JP, Loisel TP, Bichet DG, Barret C, Bouvier M. A peptide derived from a beta2-adrenergic transmembrane domain inhibits both receptor dimerization and activation. J Biol Chem. 1996;271:16384—16392. Hersch E, Huang J, Grider JR, Murthy KS. G,/G13 signaling by ET-1 in smooth muscle: MYPT1 phosphorylation via ETA and CPI-17 dephosphorylation via ETg. Am J Physiol Cell Physiol. 2004;287:C1 209-01 21 8. 141 Hilgers RH, Webb RC. Molecular aspects of arterial smooth muscle contraction: focus on Rho. Exp Biol Med (Maywood). 2005;230:829-835. lnscho EW, Imig JD, Cook AK, Pollock DM. ET, and ET, receptors differentially modulate afferent and efferent arteriolar responses to endothelin. Br J Pharmacol. 2005;146:1 01 9-1 026. Jafri F, Ergul A. Nuclear lowlization of endothelin-converting enzyme-1: subisoform specificity. Arterioscler Thromb Vasc Biol. 2003;12:2192—2196. Jin L, Ying Z, Webb RC. Activation of Rho/Rho kinase signaling pathway by reactive oxygen species in rat aorta. Am J Physiol Heart Circ Physiol. 2004 ;287:H1 495-5H1 00. Johnson RJ, Galligan JJ, Fink GD. Effect of an ET(B)-selective and a mixed ET(AIB) endothelin receptor antagonist on venomotor tone in deoxycorticosterone-salt hypertension. J Hypertens. 2001;19(3):431-440. Jordan BA, Devi LA. G-protein-coupled receptor heterodimerization modulates receptor function. Nature. 1999;399(6737):697-700. Jordan BA, Trapaidze N, Gomes l, Nivarthi R, Devi LA. Oligomerization of opioid receptors with beta 2-adrenergic receptors: a role in trafficking and mitogen- activated protein kinase activation. Proc Natl Acad Sci U S A. 2001;98(1):343- 348. Kammerer RA, Frank S, Schulthess T, Landwehr R, Lustig A, Engel J. Heterodimerization of a functional GABAB receptor is mediated by parallel coiled-coil alpha-helices. Biochemistry. 1 999;38(40):1 3263-1 3269. Kasuya Y, lshikawa T, Yanagisawa M, Kimura S, Goto K, Masaki T. Mechanism of contraction to endothelin in isolated porcine coronary artery. Am J Physiol. 1989;257zH1828-H1835. Kaupmann K, Malitschek B, Schuler V, Heid J, Froestl W, Beck P, Mosbacher J, Bischoff S, Kulik A, Shigemoto R, Karschin A, Bettler B. GABA(B)-receptor subtypes assemble into functional heterotrimeric complexes. Nature. 1 998;396(671 2):683-687. Kawanabe Y, Hashimoto N, Masaki T. Effects of phosphoinositide 3-kinase on endothelin-1-induced activation of voltage-independent Ca2+ channels and vasoconstriction. Biochem Pharmacol. 2004;68:215-221. Kedzierski RM, Yanagisawa M. Endothelin system: The double-edged sword in health and disease. Annu Rev Pharmacol Toxicol. 2001;41:851-876. 142 Kishi F, Minami K, Okishima N, Murakami M, Mori S, Yano M, Niwa Y, Nakaya Y, Kido H. Novel 31-amino-acid-length endothelins cause constriction of vascular smooth muscle. Biochem Biophys Res Commun. 1998;248(2):387-390. Kitamura K, Shiraishi N, Singer WD, Handlogten ME, Tomita K, Miller RT. Endothelin-B receptors activate Galpha13. Am J Physiol. 1999;276(Pt 1)20930- C937. Kloog Y, Sokolovsky M. Similarities in mode and sites of action of sarafotoxins and endothelins. TRENDS Pharmacol Sci. 1989;10:12—14. Kodama H, Fukuda K, Takahashi E, Tahara S, Tomita Y, leda M, Kimura K, Owada KM, Vuori K, Ogawa S. Selective involvement of p13OCas/Crklek2/c- Src in endothelin-1-induced JNK activation. Hypertension. 2003;41(6):1372- 1379. Kwon S, Fang LH, Kim B, Ha TS, Lee SJ, Ahn HY. p38 Mitogen-activated protein kinase regulates vasoconstriction in spontaneously hypertensive rats. J Pharmacol Sci. 2004;95:267-272. Lee JA, Ohlstein EH, Peishoff CE, Elliott JD. Endothelin receptor-signaling mechanisms in vascular smooth muscle. In: Highsmith RF, ed. Endothelin: Molecular biology, physiology and pathology. New Jersey: Humana Press Inc 1998: 31-73. Lee SP, O’Dowd BF, Rajaram RD, Nguyen T, George SR. D2 dopamine receptor homodimerization is mediated by multiple sites of interaction, including an intermolecular interaction involving transmembrane domain 4. Biochemistry. 2003;42(37):1 1023-1 1031 . Li L, Fink GD, Watts SW, Northcott CA, Galligan JJ, Pagano PJ, Chen AF. Endothelin-1 increases vascular superoxide via endothelin(A)-NADPH oxidase pathway in low-renin hypertension. Circulation. 2003a;107(7):1053—1058. Li L, Watts SW, Banes AK, Galligan JJ, Fink GD, Chen AF. NADPH oxidase- derived superoxide augments endothelin-1-induced venoconstriction in mineralocorticoid hypertension. Hypertension. 2003b, 42(3):316-231. Liu S, Premont RT, Kontos CD, Huang J, Rockey DC. Endothelin-1 activates endothelial cell nitric-oxide synthase via heterotrimeric G-protein betagamma subunit signaling to protein kinase BlAkt. J Biol Chem. 2003;278(50):49929- 49935. 143 Lodge NJ, Zhang R, Halaka NN, Moreland S. Functional role of endothelin ET, and ET, receptors in venous and arterial smooth muscle. Eur J Pharmacol. 1995;287:297—285. Loomis ED, Sullivan JC, Osmond DA, Pollock DM, Pollock JS. Endothelin mediates superoxide production and vasoconstriction through activation of NADPH oxidase and uncoupled nitric-oxide synthase in the rat aorta. J Pharmacol Exp Ther. 2005;31 5:1058-1 064. Lucchesi PA, Belmadani S, Matrougui K. Hydrogen peroxide acts as both vasodilator and vasoconstrictor in the control of perfused mouse mesenteric resistance arteries. J Hypertens. 2005;23:571-579. Lyle AN, Griendling KK. Modulation of vascular smooth muscle signaling by reactive oxygen species. Physiology (Bethesda). 2006;21:269-280. Maggio R, Novi F, Scarselli M, Corsini GU. The impact of G-protein-coupled receptor hetero-Oligomerization on function and pharmacology. FEBS J. 2005;272:2939-2946. Masaki T. Historical review: Endothelin. TRENDS Pharmacol Sci. 2004;25:221- 224. Miao L, Dai Y, Zhang J. Mechanism of RhoA/Rho kinase activation in endothelin- 1-induced contraction in rabbit basilar artery. Am J Physiol Heart Circ Physiol. 2002;283(3):H983-H989. Milligan G. G protein-coupled receptor dimerization: function and ligand pharmacology. Mol Pharmacol. 2004;66:1-7. Milligan G, Bouvier M. Methods to monitor the quaternary structure of G protein- coupled receptors. FEBS J. 2005;272:2914-2925. Mitchell A, Luckebergfeld B, Buhrrnann S, Rushentsova U, Numberger J, Siffert W, Schafers RF, Philipp T, Wenzel RR. Effects of systemic endothelin A receptor antagonism in various vascular beds in men: In vivo interactions of the major blood pressure-regulating systems and association with the GNB3 0825T polymorphism. Clin Pharmacol Ther. 2004;76(5):396-408. Moreland S, McMullen D, Abboa-Offei B, Seymour A. Evidence for a differential location of vasoconstrictor endothelin receptors in the vasculature. Br J Pharmacol. 1994;1 12(2):?04-708. Nakano A, Kishi F, Minami K, Wakabayashi H, Nakaya Y, Kido H. Selective conversion of big endothelins to tracheal smooth muscle-constricting 31-amino 144 acid-length endothelins by chymase from human mast cells. J Immunol. 1 997:1 59(4): 1 987-1 992. Neylon CB. Vascular biology of endothelin signal transduction. Clin Exp Pharmacol Physiol. 1 999 ;26:1 49-1 53. Northcott CA, Poy MN, Najjar SM, Watts SW. Phosphoinositide 3-kinase mediates enhanced spontaneous and agonist-induced contraction in aorta of deoxycorticosterone acetate-salt hypertensive rats. Circ Res. 2002;91(4):360- 369. Pace AJ, Gama L, Breitwiesser GE. Dimerization of the calcium-sensing receptor occurs within the extracellular domain and is eliminated by Cys -) Ser mutations at Cys101 and Cys 236. J Biol Chem. 1999;274(17):11629—11634. Palacios B, Lim SL, Pang CC. Subtypes of endothelin receptors that mediate venous effects of endothelin-1 in anaesthetized rats. Br J Pharmacol. 1 997;122(6):993-998. Panchuk-Voloshina N, Haugland RP, Bishop-Stewart J, Bhalgat MK, Millard PJ, Mao F, Leung WY, Haugland RP. Alexa dyes, a series of new fluorescent dyes that yield exceptionally bright, photostable conjugates. J Histochem Cytochem. 1999;47(9):1 179-1 188. Perez-Rivera AA, Fink GD, Galligan JJ. Vascular reactivity of mesenteric arteries and veins to endothelin-1 in a murine model of high blood pressure. Vascul Pharmacol. 2005 ;43:1 -1 0. Prinster SC, Hague C, Hall RA. Heterodimerization of g protein-coupled receptors: specificity and functional significance. Pharmacol Rev. 2005;57:289- 298. Reinhart GA, Preusser LC, Burke SE, Wessale JL, Wegner CD, Opgenorth TJ, Cox BF. Hypertension induced by blockade of ET,, receptors in conscious nonhuman primates: role of ET(A) receptors. Am J Physiol Heart Circ Physiol.2002;283(4):H1555-1 561 . Ricksten SE, Yao T, Thoren P. Peripheral and central vascular compliance in conscious normotensive and spontaneously hypertensive rats. Acta Physiol Scand. 1981;112:169-177. Romano C, Miller JK, Hyrc K, Dikranian S, Mennerick S, Takeuchi Y, Goldberg MP, O’Malley KL. Covalent and noncovalent interactions mediate metabotropic glutamate receptor mGlu5 dimerization. Mol Pharmacol. 2001;59(1):46-53. 145 Safar ME, London GM, Weiss YA, Milliez PL. Altered blood volume regulation in sustained hypertension: A hemodynamic study. Kidney Intemat. 1975;8z42—47. Sauvageau S, Thorin E, Caron A, Dupuis J. Evaluation of endothelin-1-induced pulmonary vasoconstriction following myocardial infarction. Exp Biol Med (Maywood). 2006;231:840-846. Sedeek MH, Llinas MT, Drummond H, Fortepiani L, Abram SR, Alexander BT, Reckelhoff JF, Granger JP. Role of reactive oxygen species in endothelin- induced hypertension. Hypertension. 2003;42(4):806-810. Somlyo AP, Somlyo AV. Ca2+ sensitivity of smooth muscle and nonmuscle myosin ll: modulated by G proteins, kinases and myosin phosphatase. Physiol Rev. 2003;83:1325-1358. Stan RV. Structure and function of endothelial caveolae. Microsc Res and Tech. 2002;57:350-364. Takagi Y, Ninomiya H, Sakamoto A, Masaki T. Structural basis of G protein specificity of human endothelin receptors. A study with endothelin AIB chimeras. J Biol Chem. 1 995;270(17):10072—10078. Telemaque-Potts S, Kuc RE, Maguire JJ, Ohlstein E, Yanagisawa M, Davenport AP. Elevated systemic levels of endothelin-1 and blood pressure correlate with blunted constrictor responses and downregulation of endothelin(A), but not endothelin(B), receptors in an animal model of hypertension. Clin Sci (Lond).2002;48:357S-36ZS. Thakali K, Fink GD, Watts SW. Arteries and veins desensitize differently to endothelin. J Cardiovasc Pharmacol. 2004;43(3):387-393. Thakali K, Demel SL, Fink GD, Watts SW. Endothelin-t-induced contraction in veins is independent of hydrogen peroxide. Am J Physiol Heart Circ Physiol. 2005;289:H1115-H1122. Touyz RM. Recent advances in intracellular signalling in hypertension. Curr Opin Nephrol Hypertens. 2003 ;1 2:1 65-1 74. Verhaar MC, Grahn AY, Van Weerdt AW, Honing ML, Morrison PJ, Yang YP, Padley RJ, Rabelink TJ. Pharrnacokinetics and pharrnacodynamic effects of ABT-627, an oral ETA selective endothelin antagonist, in humans. Br J Clin Pharmacol. 2000;49(6):562-573. Wallrabe H, Periasamy A. Imaging protein molecules using FRET and FLlM microscopy. Curr Opin Biotechnol. 2005;16:19-27. 146 Watts SW. Serotonin activates the mitogen-activated protein kinase pathway in vascular smooth muscle: use of the mitogen-activated protein kinase kinase inhibitor PD098059. J Pharmacol Exp Ther. 1996;279:1541-1550. Watts SW, Fink GD, Northcott CA, Galligan JJ. Endothelin-I-induced venous contraction is maintained in DOCA-salt hypertension; studies with receptor agonists. Br J Pharmacol. 2002;137(1):69-79. Wedgwood S, McMullan DM, Bekker JM, Flneman JR, Black SM. Role for Endothelin-1-induced superoxide and peroxynitrite production in rebound pulmonary hypertension associated with inhaled nitric oxide therapy. Circ Res. 2001 ;89:357-364. Wehnivein EA, Northcott CA, Loberg RD, Watts SW. Rho/Rho kinase and phosphoinositide 3-kinase are parallel pathways in the development of spontaneous arterial tone in deoxycorticosterone acetate-salt hypertension. J Pharmacol Exp Ther. 2004;309:101 1-1019. White JH, Wise A, Main MG, Green A, Fraser NJ, Disney GH, Barnes AA, Emson P, Foords SM, Marshall FH. Heterodimerization is required for the formation of a functional GABA(B) receptor. Nature. 1998;396:679—682. Wolin MS, Gupte SA, Oeckler RA. Superoxide in the vascular system. J Vasc Res. 2001;39:191-207. Zubkov AY, Rollins KS, Parent AD, Zhang J, Bryan RM Jr. Mechanism of endothelin-1-induced contraction in rabbit basilar artery. Stroke. 2000;31(2):526- 33. Zhou J, Moroi K, Nishiyama M, Usui H, Seki N, Ishida J, Fukamizu A, Kimura S. Characterization of RG85 in regulation of G protein-coupled receptor signaling. Life Sci. 2001;68(13):1457-69. Yamboliev IA, Hedges JC, Mutnick JL, Adam LP, Gerthoffer WT. Evidence for modulation of smooth muscle force by the p38 MAP kinase/HSP27 pathway. Am J Physiol Heart Circ Physiol. 2000 ;278:H1 899-H1 907. Yanagisawa H, Hammer RE, Richardson JA, Emoto N, Williams SC, Takeda S-l, Clouthier DE, Yanagisawa M. Disruption of ECE-1 and ECE-2 reveals a role for endothelin-converting enzyme-2 in murine mrdiac development. J Clin Invest. 2000;105:1373-1382. Yang Z, Bauer E, con Segesser L, Stulz P, Turina M, Luscher TF. Different mobilization of calcium in endothelin-1-induced contractions in human arteries and veins: effects of calcium antagonists. J Cardiovasc Pharmacol. 1990;16:654- 660. 147 VI. CURRICULUM VITAE Keshari M. Thakali PERSONAL DATA Name: Keshari Maya Thakali B445 Life Sciences Bldg Born: 8/4/81 Dept of Pharmacology and Toxicology Michigan State University East Lansing, MI 48824 Phone: (517) 353-3900 Fax: (517) 353-8915 E-mail: thakalik@msu.edu EDUCATIONAL BACKGROUND 1998-2002 University of Arizona, Tucson, A2 8.8. (Environmental Science, Minor: Chemistry) 2002-present Michigan State University Doctoral candidate (Pharmacology and Toxicology) Project Title: “Endothelin A (E TA) and E T3 receptor interaction in arteries and veins" TEACHING ACTIVITIES 2004-2005 Tutor for Medical Pharmacology (PHM 563) Michigan State University 2005 Tutor for Veterinary Pharmacology (PHM 556) Michigan State University 2005-2006 Lecturer, Introduction to Chemical Toxicology (PHM 450) 148 Reproductive Toxicology Section Michigan State University RESEARCH TRAINING 2000—2002 Lab Assistant: Environmental Microbiology Laboratory, University of Arizona. Isolation and characterization of multi- copper oxidases in Escherichia coli. Supervisor: Dr. Christopher Rensing. 2002 Research Rotation: Cardiovascular Pharmacology 2002-present Laboratory, Michigan State University. Endothelin receptor desensitization in aorta and vena cava. Supervisor: Stephanie W. Watts, PhD Doctoral Dissertation Research: Cardiovascular Pharmacology Laboratory, Michigan State University. Endothelin A (ETA) and ET, receptor interaction in arteries and veins. Mentor: Stephanie W. Watts, PhD PROFESSIONAL ACTIVITIES American Heart Association American Physiological Society: Student member of the Women in Physiology Committee American Society for Pharmacology and Experimental Therapeutics: Student member of the ASPET Centennial Committee Reviewer for American Journal of Physiology Heart and Circulatory Physiology Reviewer for Journal of Pharmacology and Experimental Therapeutics Reviewer for Naunyn-Schmiedeberg's Archives of Pharmacology 149 ACADEMIC AND PROFESSIONAL HONORS 2006 NIDDK Minority Travel Award 2005 Caroline tum Suden/Francis Hellebrandt Professional Opportunity Award 2005 NIDDK Minority Travel Award 2004-present Ford Foundation Predoctoral Diversity Fellowship 2004 NIDDK Minority Travel Award 2004 Michigan State University Chronic Disease Initiative Fellowship 2003 Merck New Investigator Travel Award 2002-2003 Michigan State University Competitive Doctoral Enrichment Fellowship PRESENTATIONS ERA Thakali K Rondelli C, Fink GD, Watts SW. Chymase—dependent processing of big ET-1 in arteries but not veins. 60‘“ Annual Fall Conference of the Council for High Blood Pressure Research in Association with the Council on Kidney in Cardiovascular Disease, 2006 (San Antonio, TX). Thakali K, Lauren Davenport, Fink GD, Watts SW. Pleiotropic effects of hydrogen peroxide in arteries and veins from normotensive and deoxycorticosterone acetate-salt hypertensive rats. Michigan Hypertension Workshop; 2005 (Kellogg Biological Institute, Michigan State University). Thakali K, Fink GD, Galligan JJ, Watts SW. Arteries and veins desensitize differently to endothelin. Michigan Colloquium on Pharmacology, 2003 (University of Michigan). 150 Thakali K, Fink GD, Galligan JJ, Watts SW. Arteries and veins desensitize differently to endothelin. Michigan Hypertension Workshop; 2003 (Kellogg Biological Institute, Michigan State University). POSTERS Thakali K Galligan JJ, Fink GD, Watts SW. Pharmacological endothelin receptor interaction occurs in veins but not arteries. 60‘” Annual Fall Conference of the Council for High Blood Pressure Research in Association with the Council on Kidney in Cardiovascular Disease, 2006 (San Antonio, TX). Thakali K, Davenport L, Fink GD, Watts SW. Multiple signal transduction pathways are involved in hydrogen peroxide-induced contraction in rat thoracic aorta and vena cava. Experimental Biology 2006 (San Francisco, CA). Thakali K, Fink GD, Watts SW. K” channels differentially modulate contraction to hydrogen peroxide (H202) in arteries and veins; effects in deoxycorticosterone acetate (DOCA)-salt hypertension. 59‘" Annual Fall Conference of the Council for High Blood Pressure Research in Association with the Council on Kidney in Cardiovascular Disease, 2005 (Washington, DC). Thakali K, Rondelli C, Fink GD, Watts SW. Endothelin converting enzyme (ECE) is present and functional in rat thoracic aorta and vena mva. The Ninth lntemational Conference on Endothelin (E T-9), 2005 (Park City, UT). Thakali K, Fink GD, Watts SW. Endothelin-1 (ET-1) increases hydrogen peroxide (H202) in veins, but not arteries. Experimental Biology 2005 (San Diego, CA). Thakali K, Smark CT, Rondelli CM, Fink GD, Watts SW. Reduced aortic but not venous contraction to big Endothelin-1 (ET-1) in deoxycorticosterone acetate (DOCA)-salt hypertension: the role of endothelin converting enzyme (ECE). 58‘" Annual Fall Conference of the Council for High Blood Pressure Research in 151 Association with the Council on Kidney in Cardiovascular Disease, 2004 (Chicago, IL). Thakali K, Fink GD, Watts SW. Increased blood pressure causes changes in aortic endothelin-1 (ET-1) and norepinephrine (NE) contractility in deoxycorticosterone acetate (DOCA) salt hypertension. 14‘" Annual Phi Zeta Research Day, 2004 (College of Veterinary Medicine, Michigan State University). Thakali K, Fink GD, Watts SW. Increased blood pressure causes changes in aortic endothelin-1 (ET-1) and norepinephrine (NE) contractility in deoxycorticosterone acetate (DOCA) salt hypertension. Experimental Biology 2004 (Washington, DC). Thakali K, Watts SW. ET-1 contraction signaling in veins is independent of extracellular hydrogen peroxide (H202). 13‘" Annual Phi Zeta Research Day, 2003 (College of Veterinary Medicine, Michigan State University). Thakali K, Ni W, Li M, Fink GD, Watts SW. The anorexigen (+) norfenfluramine as a pressor; enhanced response in hypertension. 57‘” Annual Fall Conference of the Council for High Blood Pressure Research in Association with the Council on Kidney in Cardiovascular Disease; 2003 (Washington, DC). Thakali K, Watts SW. ET-1 contraction signaling in veins is independent of extracellular hydrogen peroxide (H202). 57‘" Annual Fall Conference of the Council for High Blood Pressure Research in Association with the Council on Kidney in Cardiovascular Disease; 2003 (Washington, DC). Thakali K, Fink GD, Galligan JJ, Watts SW. Arteries and veins desensitize differently to endothelin. XVth Scientific Meeting of the Inter-American Society of Hypertension; 2003 (San Antonio, TX). 152 PAPERS Thakali K, Davenport L, Fink GD, Watts SW. Cyclooxygenase, p38 MAPK, Erk MAPK, rho kinase and src mediate hydrogen peroxide-induced contraction of rat thoracic aorta and vena cava. J Pharmacol Exp Ther. 2006 Sep 26; [Epub ahead ofpnnfl. Thakali K, Lau Y, Fink GD, Galligan JJ, Chen AF, Watts SW. Mechanisms of hypertension induced by nitric oxide (NO) deficiency: focus on venous function. J Cardiovasc Pharmacol 2006, 47(6):742-750. Thakali K, Davenport L, Fink GD, Watts SW. Pleiotropic effects of hydrogen peroxide in arteries and veins from normotensive and deoxycorticosterone acetate-salt hypertensive rats. Hypertension. 2006, 47(3):482—487. Thakali K, Demel SL, Fink GD, Watts SW. Endothelin-1 (ET-1) —induced contraction in veins is independent of hydrogen peroxide (H202). Am J Physiol Heart Circ Physiol. 2005, 289(3):H1115-1 1 12. Ni W, Li W, Thakali K, Fink GD, Watts SW. The fenfluramine metabolite (+)- norfenfluramine is vasoactive. J Pharmacol Exp Ther. 2004, 309(2):845-852. Thakali K, Fink GD, Galligan JJ, Watts SW. Arteries and veins desensitize differently to endothelin. J Cardiovasc Pharmacol. 2004, 43(3):387-393. Grass G, Thakali K, Klebba PE, Thieme D, Muller A, Wildner GF, Rensing C. Linkage between catecholate siderophores and the multicopper oxidase CueO in Escherichia coli. J Bacteriol. 2004, 186(17):5826-5833. Roberts SA, Weichsel A, Grass G, Thakali K, Hazzard JT, Tollin G, Rensing C, Montfort WR. Crystal structure and electron transfer kinetics of CueO, a 153 multicopper oxidase required for copper homeostasis in Escherichia coli. Proc Natl Acad Sci U S A. 2002, 5;99(5):2766—2771. BOOK CHAPTERS Thakali K, Galligan JJ, Fink GD, Watts SW. Arterial and venous function in hypertension; a vascular focus. Comprehensive Hypertension. (in press). ABSTRACTS Thakali K Galligan JJ, Fink GD, Watts SW. Pharmacological endothelin receptor interaction occurs in veins but not arteries. [Abstract] Hypertens. 48:2006. Thakali K, Rondelli C, Fink GD, Watts SW. Chymase-dependent processing of big ET-1 in arteries but not veins. [Abstract] Hypertens. 48:2006. Rondelli C, Pervaiz MH, Watson RE, Thakali K Fink GD, Watts SW. Veins are not arteries: a story of remodeling in hypertension. [Abstract] Hypertens. 48:2006. Thakali K, Davenport L, Fink GD, Watts SW. Multiple signal transduction pathways are involved in hydrogen peroxide-induced contraction in rat thoracic aorta and vena wva. The FASEB Journal, Abstract #9043, 2006. Thakali K, Fink GD, Watts SW. K“ channels differentially modulate contraction to hydrogen peroxide (H202) in arteries and veins; effects in deoxycorticosterone acetate (DOCA)-salt hypertension. [Abstract] Hypertens. 46: 842, 2005. Thakali K, Rondelli C, Fink GD, Watts SW. Endothelin converting enzyme (ECE) is present and functional in rat thoracic aorta and vena cava. The Ninth lntemational Conference on Endothelin (E T-9), 2005 (Park City, UT). 154 Thakali K, Fink GD, Watts SW. Endothelin-1 (ET-1) increases hydrogen peroxide (H202) in veins, but not arteries. The FASEB Journal, Abstract #6862, 2005. Kayal A, Rondelli CM, Thakali K, Watson RE, Rovner AS, Fink GD, Watts SW. Myosin heavy chain B isoform is more predominant in rat veins than arteries. The FASEB Journal, Abstract #1135, 2005. Thakali K, Smark CT, Rondelli CM, Fink GD, Watts SW. Reduced aortic but not venous contraction to big Endothelin-1 (ET-1) in deoxycorticosterone acetate (DOCA)-salt hypertension: the role of endothelin converting enzyme (ECE). [Abstract]. Hypertens. 44: 562, 2004. Ni W, Li M, Thakali K, FinK, GD, and_Watts SW.: The fenfiuramine metabolite (+) norfenfluramine is vasoactive. Fund. Clin. Pharmacol. 18(S1):p.139, A38, 2004. Thakali K, Fink GD, Watts SW. Increased blood pressure causes changes in aortic endothelin-1 (ET-1) and norepinephrine (NE) contractility in deoxycorticosterone acetate (DOCA) salt hypertension. The FASEB Journal, 19, Abstract #2018, 2004. Smark CT, Thakali K, Watts SW. Role of caveolae in endothelin-1 (ET-1)- induced contraction in arteries and veins. Experimental Biology meeting abstracts [accessed at http://www.biosis-select.orglfaseb/index.htrnl]. The FASEB Journal, 19, Abstract #2044, 2004. ' Thakali K, Watts SW. ET-1 contraction signaling in veins is independent of extracellular hydrogen peroxide. [Abstract]. Hypertens. 43: 1352, 2003. Thakali K, Ni W, Li M, Fink GD, Watts SW. The anorexigen (+)-norfenfluramine as a pressor; enhanced response in hypertension. [Abstract]. Hypertens. 42: 387, 2003. 155 Thakali K, Fink GD, Galligan JJ, Watts SW. Arteries and veins desensitize differently to endothelin. XVth Scientific Meeting of the Inter-American Society of Hypertension. 2003 (San Antonio, TX). 156 MICHIGAN STATE UNIVERSITY LIBRARI IIIIflIlIlIIIQLIIIILIIIIIIQIIILIIIJI