THESE :50] F'BPARY Michigan State , University This is to certify that the dissertation entitled Relationship Between Experimental Angiotensin II Induced Hypertension and Activation of the Enddhelin System in Male Rats: Effects of Salt Intake presented by Jennifer Rebecca Ballew has been accepted towards fulfillment of the requirements for Ph.D. degree“. Pharmacology and Toxicology 47%égrgi Date 8/5//';ZOCDO MS U is an Affirmative Action/Equal Opportunity Institution 0- 12771 PLACE IN REFURN BOX to remove this checkout from your record. TO AVOID FlNES return on or before date due. MAY BE RECALLED with earlier due date if requested. In A ‘VVY DATE DUE DATE DUE DATE DUE 11m mm.“ ‘5 I RELATIONSHIP BE HYDERTENSICN Mn. RELATIONSHIP BETWEEN EXPERIMENTAL ANGIOTENSIN II INDUCED HYPERTENSION AND ACTIVATION OF THE ENDOTHELIN SYSTEM IN MALE RATS: EFFECTS OF SALT INTAKE By Jennifer Rebecca Ballew 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 2000 RELATIONSHIP BE HYPERTENQI’I- ' ; V'V' ‘A “Mr. ‘h . . u 'U = - ‘5 m “V0 T5,: .5 ‘1’E’CEN |' by Digger-3e : A TIM 'I My): H‘Vfifien "~ ' Sisr‘ »- I ’1‘" MA. ”Eris: st " L' n _ 3‘” [1°55 15:. v, :5 ‘ uf “‘6 ”. age» VJ.,‘&N ABSTRACT RELATIONSHIP BETWEEN EXPERIMENTAL ANGIOTENSIN II INDUCED HYPERTENSION AND ACTIVATION OF THE ENDOTHELIN SYSTEM IN MALE RATS: EFFECTS OF SALT INTAKE By Jennifer Rebecca Ballew The overall objective of my thesis project was to determine the role of endothelin-1 in the salt-sensitivity of angiotensin II induced hypertension. The relationship between these two peptide hormones has been well documented, but the specific mechanisms through which they interact have yet to be fully elucidated. The main experimental strategy | employed in this thesis project was measurement of hemodynamic and renal responses to endothelin-l receptor antagonists in vivo. This approach was supplemented by use of in vitro studies of vascular contractility. The data presented in this thesis Show that selective ETA receptor blockade alone normalizes mean arterial pressure in angiotensin II induced hypertension, particularly under conditions of high salt intake. This presents a strong case that endothelin-1 acting at ETA receptors is an important mechanism of the salt-sensitivity of angiotensin II induced hypertension. Conversely, my data further demonstrate that selective ETB receptor activation decreases mean arterial pressure in angiotensin II induced hypertension, especially under high salt conditions. The potential clinical implications of my data are that combined blockade of both ETA and ETB receptors will be less effective at decreasing blood pressure than selective ETA receptor antagonism, especially in people with salt-sensitive hypertension. To my lax/WWW Marlo. Yaw WWW IF. the 51;“; Limersty as a gas: '94 15“.»! An.‘ -5”. v»; "VIII W a»... N“. “2:: work, 3:; F'I'S'f I WIS“ :: :‘fl'o b a» .IJ‘ far IT, 1,. W3“: flint: in v *w s _‘_ as: A m “Omrh't‘aa . 4’"——— —....-—a—r— Acknowledgments In the slightly more than five years I have spent here at Michigan State University as a graduate student in the Department of Pharmacology and Toxicology, it has become my considered opinion that the best scientific work results from collaborative efforts. This thesis project was the outcome of my own hard work and the input, advice, and assistance of many other people. First I wish to express my utmost love, appreciation, respect and admiration for my thesis advisor, Dr. Greg Fink. From the commencement of this project, Greg has encouraged me to think creatively and individually, even when my ideas and opinions were in contrast to his. He helped me retain my enthusiasm for science through the most trying times, and for this I am profoundly grateful. Greg, you have been a true mentor and friend to me. Thank you. I would like to thank and acknowledge the other members of my thesis guidance committee: Dr. Susan Barman, Dr. Jim Galligan, Dr. Stephanie Watts, and Dr. Donna Wang. It has been a privilege to work with such distinguished scientists. I thank Jim in particular for his humor, encouragement, discretion, and commitment to graduate education. Maize? See": E'ocsf’ated l hfi'f‘. ' Ar- I owe a 3 n.’ a J“ a D‘jieissi, ”‘7'“ I .. Name of c I am deeply grateful to Dr. Veronica Maher and Dr. Justin McCormick, co- directors of the Michigan State University College of Osteopathic Medicine’s Medical Scientist Training Program. Drs. Maher and McCormick have demonstrated, time and again, their faith in my abilities and their dedication to helping me receive the best possible scientific training. I owe a great debt of gratitude to my fellow co-workers, Barbara Grant, Dawna Dylewski, and Evan Brown, without whom this thesis project could not have been completed in such an efficient manner. It has been my sincere pleasure to work and laugh with all of you on a daily basis, and I am truly appreciative of our friendly working environment. Special thanks are due to Dr. John Thornburg, Dr. Bill Atchison, and Dr. Keith Lookingland. Dr. Thornburg allowed me the opportunity to continue seeing patients while working on this thesis and went above and beyond the call of duty to make sure I stayed up-to-date in my clinical training. Dr. Atchison helped me juggle medical school and graduate school requirements, and frequently and humorously reminded me how to keep scientific politics in perspective. Dr. Lookingland enthusiastically recruited me to this training program and helped me get started in my graduate school career. vi later lov fig ‘3 i..'.sR Ba“ew wt: Ia.“ and whose 'Ic re tat lar g'afe‘q; : . 333‘s? 399mg m3: -‘ 3* II" 3‘3 Bethe L; firm; and a: 1.7; £5“ . 3 SC Wine fUfi’ We . WI 390 mos? . . : Canaan, 8M ., 3‘: I offer loving thanks to my mother, Anne Marie Ballew, and my father, Julius R. Ballew, whose high expectations kept me on a challenging upward path, and whose love and encouragement convinced me to pursue my academic goals. I am grateful to my brother, Jamie Ballew, for believing in my aptitudes and for offering motivation whenever possible. Thanks also to my parents-in- law, Jim and Bethel Lanz, and my grandmother-in-law, Helen Jameson, for their continuing and abiding faith in my career goals and their countless prayers on my behalf. My thankful appreciation goes to my friends, Kaisa Johnson, Tiffany Lasky, Mike O’Neill, Coleen Warriner, and Fran Wolber, as well as to my uncle and aunt, Jim and Dianne McPharlin. The seven of you have formed my long- distance support group and I can always count on each of you to listen, offer advice, make me laugh, and comfort me during rough times. Thanks also for being so much fun! Lastly, and most importantly, I want to thank my husband, Mark Lanz. Mark has been a constant and unconditional source of love, support, encouragement, and energy to me. He has helped me keep my sense of humor throughout this ordeal and I am eternally grateful to him. Thank you, Mark, for gracing my life with your presence and your astonishing ability to keep long-term goals in sight. I love you. vii LIST OF TABLES LIST OF FIGURES LIST OF ABBREVIA Chapler1: INTROE I Hypertension E539" E‘. 31:3, Hews,- Me-d'far , Presser-:- Sai’. Se"! ' .NIA'. Tea. ,1;- Remn'AngIOIEr E‘Iects o‘ #935»: A“; 3196: Sat 86".: l EndoIIleIin B’Osy'itre S:tes OIC TABLE OF CONTENTS LISTOF FIGURES LIST OF ABBREVIATIONS.................................................. Chapter1: INTRODUCTION.............................................. I. Hypertension................................... . . Epidemiology...........................'.ffj.'.'."..'."..'.'Z.'."..'."..'.".327...".III... Etiology. Hemodynamics Mechanisms of HypertenSIon Pressure-Natriuresis... Salt Sensitivity... Treatment and Prevention II. Renin-Angiotensin System... Effects of Angiotensin II . Angiotensin II Induced Hypertensron Angiotensin II and Pressure-Natriuresis Salt Sensitivity of Angiotensin II Induced HypertenSIon........'.'......'.. .. Ill. Endothelin... Biosyntheosis... . . . Sites of Generation and Secretion Endothelin Receptors... Endothelin Signal Transduction Mechanisms Endothelin Receptor Antagonists... Endothelin and the Renin-Angiotensin System... V. Hypothesis... Specific Aim I Specific Aim II... viii .3 Z .xii .xiii .xix ooo'xi 12 .....13 ...14 15 .....17 ......17 ......18 .....18 ......20 ...21 Cardiovascular Physiology of EndothelIn ..23 Renal Physiology of Endothelin........................................... Endothelin and Essential Hypertension....................... .25 .27 .31 ......35 ...35 I Chapter); GENEIh Chapter 3 Acacia I Surge: Chrom: Hem: Metals-:I lscae: rags Statstc. Prelimin 9:353) H‘y':e".e Efficacy N HI’DE'TE" Ra: :1 Te Chapter 2: Chapter 3: Chapter 4: GENERAL METHODS37 Animals. . 37 Surgical Procedures .. 37 -Arterial and Venous Cathetenzatron 38 -Surgical Uninephrectomy” .. ... ...39 -Deoxycortisosterone Acetate Implantation . .. ... ... ..39 Chronic Rat Maintenance and Measurements. 40 HemodynamicMeasurements... 40 MetabolicMeasurements... 41 Isolated Tissue Bath Measurements... 42 StatIstIcalAnalysrs 43 Preliminary Studies... ...45 Efficacy of PD156707 Tested In ”ET- 1 Induced Model of” Hypertension... ............45 Efficacy of PD156707 TeStedm In DOCA-Salt Model 6r” Hypertension... . 49 Rapid Termination of Endothelin Infusion . ...52 Autonomic Blockade and PD156707... 57 Effect of PD156707 on Angiotensin II Induced Hypertension” ....59 Efficacy of ABT-627 TeS-ted. in Endothelin 1 Induced Model of Hypertension... . ..64 Eff cacres of ABT-627 and Minoxidil Tested in DOCA-Salt Model of Hypertension... 67 Effect of ABT-627 on Angrotensm WII Induced Hypertension... ... . .70 Effect of ABT-627 on Acute Sarafotoxrn 6c Dose ResponSe Curve... ..........73 Effect of ABT-627 on Acute Endothelin 1 Dose Response Curve... ......76 Effect of A-192621 on Acute Sarafotoxm 6c Dose Response Curve... .........79 Effect of A-192621 on Acute EndotheIIn 1 Dose Response Curve... .. ...........82 Efficacy of A-182086 Tested in ET-1 Induced Model of” Hypertension... 85 Role of ET. Receptors in Experimental Angiotensin II Induced Hypertension in RatsBB Introduction89 Methods91 Results94 DIscussmn101 Chapter 5; Chapter 6 Chapter 7- a“alters, Role 0 Induce Intros. Metro Res.fs 0 secs: Charac Diureti Hypene Infra/1 ,. t.“ vuu‘ M9123: Rescis 0.58955 Effects ReaCfi\ 1pm,. . R A.“ “L" fin V\, : m Chapter 5: Chapter 6: Chapter 7: Chapter 8: Role of Endothelin ETB Receptor Activation in Angiotensin II Induced Hypertension: Effects of Salt Intake... 105 IntroductIon 106 Methods108 Results111 DIscuSSIon123 Characterization of the Antihypertensive Effect of a Thiazide Diuretic in Angiotensin II Induced HypertenSIon ........130 lntroductron131 Methods133 Results136 DIscuSSIon144 Effects of Salt Intake and Angiotensin II on Vascular Reactivity to Endothelin-1... .....148 IntroductIon149 Methods............... .........150 Results ....154 DISCUSSIon 161 General Discussion and Conclusions...... .168 Ang II and ET-1 can each cause hypertension. ...169 Salt intake contributes to severity of Ang II and ET-1 induced hypertension... . ............169 ET- 1 mediates some of the cardiorenal effects of Ang II. .... .170 Ang II induced hypertensron is the beSt model to Study the” interaction between Ang II and ET—1.. ... .. .171. ET-1 receptor antagonists reveal ET—1 mediated effects In vivo and invitro... . ..171 ET—1, especially under conditions of increased salt intake is required for the maintenance of experimental Ang II induced hypertension... . . .. .. ..1 72 Ang II induced hypertenSIon depends primarily on the actions of ET—1 on ETA receptors, and this effect Is greater under conditions of high salt intake... 176 The IF Induce 699-: ETB re hy’Wte Cons;| REFERENCES The initial hypotensive effect of ETA receptor blockade in Ang II induced hypertension is not due to a diuretic effect............. 179 are receptor aetiia'Iiéfi 5555:5555 ii 153.1553 ' hypertensron ....180 Conclusions....................................... 184 xi Chapter 3 Tabe Z Tate 3 Chapter 3 Table 3.1: Table 3.2: Chapter 7 Table 7.1: Chapter 8 Table 8.1: LIST QF TABLES Comparison of the change in mean arterial pressure (mmHg) over time with endothelin-1 infusion cessation and injection of P0156707... .55 Comparison of the change in heart rate (bpm) over time with Endothelin-1 infusion cessation and injection of PD156707...... ”......56 Mean arterial pressure in rats given chronic Ang II infusion (5 ng/mIn)159 Novel findings from this thesis on the vascular and renal effects of Ang II and salt intake and the activation of ETA and ETB receptors. xii Chapter 3 F -g'.'e 3 1 nge 3 2 FigJe 3 3 F‘9‘».1334 Faveas “We 3 e. {-7 hfllna— 0’00 Chapter 3 Figure 3.1: Figure 3.2: Figure 3.3: Figure 3.4: Figure 3.5: Figure 3.6: Figure 3.7: Figure 3.8: Figure 3.9: Figure 3.10: List of Figures Effect of PD156707 on endothelin—1 induced hypertension in high saltrats” ..........47 Effect of PD156707 on endothelin-1 induced hypertension in normal saltrats ...48 Effect of PD156707 on DOCA-salt hypertension... .51 Effect of termination of endothelin-1 infusion on mean arterial pressure (mm Hg) in endothelin-1 induced hypertension... ......54 Daily mean arterial pressures (mmHg) in high and normal salt rats in the presence and absence of angiotensin II ..... 61 Changes in mean arterial pressure (mmHg) between daily control pressures and pressures averaged over 35-50 minutes post injection of PD156707 in high salt rats ......... 62 Changes in mean arterial pressure (mmHg) between daily control pressures and pressures averaged over 35-50 minutes post injection of PD156707 in normal salt rats ..... 63 Hemodynamic response to infusion of endothelin-1 (ET-1) at 8 pmol/kglmin over a 72 hour time period prior to, during, and after administration of the ETA selective antagonist, ABT-627, 2 mg/kg/day in drinking water... ...66 Hemodynamic responses of DOCA-salt rats, 4-5 weeks post DOCA-Salt treatment, to ABT-627 and Minoxidil... ..69 Changes in mean arterial pressure (mmHg) with increasing doses of ABT-627 in the presence and absence of angiotensin II in high salt rats... ....72 xiii F.g.rre 3 1‘ i3 ' _.931“ =4 Pg. ui63 ’9 Chalter 4 F's‘efe 4 1 : Figure 3.11: Figure 3.12: Figure 3.13: Figure 3.14: Figure 3.15: Chapter 4 Figure 4.1: Figure 4.2: Hemodynamic response to acute bolus i.v. injection of sarafotcxin 6c (1-500 pmollkg) in rats on high salt intake..75 Hemodynamic response to acute bolus i.v. injection of endothelin-1 (0—500 pmollkg) in rats on high salt intake... .78 Hemodynamic responses to acute bolus i.v. injections of sarafotcxin 6c (doses = 62.5 to 500 pmollkg) in rats on high salt intake at various time points after receiving the ETB selective receptor antagonist A-192621 at 12 mglkg/day I. v. injection... .. .. 8.1 Hemodynamic responses to acute bolus i.v. injections of endothelin-1 (doses = 62.5 to 500 pmollkg) in rats on high salt intake at various time points after receiving the ETB selective receptor antagcnist, A-192621 at 12 mglkg/day I. v injection... ...84 Hemodynamic response to infusion of endothelin-1 (8 pmollkglmin) over a 72 hour time period prior to, during, and after administration of the non-selective ET-1 receptor antagonist, A-182086 (12 mglkglday) i.v. injection... ..89 Line plots show hemodynamic responses to infusion of angiotensin II (Ang II) at 5.0 nglmin and administration of ABT-627 at 2 mglkglday in rats on either high sodium (top panel) or normal sodium (bottom panel) intake... ...95 Line plots show hemodynamic responses to bolus intravenous injection of ABT-627 at 2 mglkg in rats on high sodium (top panel) or normal sodium (bottom panel) intake... ...97 xiv nge43 Fgre44 Frg‘u'e 4 5 ”s .‘t‘e 5 2 Figure 4.3: Figure 4.4: Figure 4.5: Chapter 5 Figure 5.1: Figure 5.2: Figure 5.3: Effects of acute intravenous injection of angiotensin II (Ang II) on mean arterial pressure in rats on high sodium intake, before and after administration of ABT-627 at 2 mglkg/day for 72 hours... ...98 Line plots Show water balance responses in milliliters per day (mls/day) to infusion of angiotensin II (Ang II) at 5.0 nglmin and administration of ABT—627 at 2 mglkg/day in rats on either high sodium (top panel) or normal sodium (bottom panel) intake... ......99 Line plots Show sodium balance responses in milliequivalents per day (mEq/day) to infusion of angiotensin II (Ang II) at 5. 0 nglmin and administration of ABT-627 at 2 mglkglday in rats on either high sodium ”(top panel) or normal sodium (bottom panel) intake... .. ..100 Line plots show hemodynamic responses to infusion of angiotensin II (Ang II) at 5. 0 nglmin and administration of A— 192621 at 24 mglkglday”. In rats on either high sodium or normalsodiumintake... ................112 Line plots Show water balance responses in milliliters per day (mls/day) to infusion of angiotensin II (Ang II) at 5.0 nglmin and administration of A-192621 at 24 mglkg/day in rats on either high sodium or normal sodium intake ........ 113 Line plots show sodium balance responses in milliequivalents per day (mEq/day) to infusion of angiotensin II (Ang II) at 5.0 nglmin and administration of A-192621 at 24 mglkg/day in rats on either high sodium or normal sodium intake... ......1 14 XV Fgcre 5 4 Fzgs'e 5 5 F'gu’e 5 g Figure 5.4: Figure 5.5: Figure 5.6: Figure 5.7: Figure 5.8: Chapter 6 Figure 6.1: Figure 6.2: Line plots Show hemodynamic responses to bolus intra- arterial injection of A-192621 at 24 mglkg in rats on high sodium or normal sodium intake... ....1 15 Line plots Show hemodynamic responses to infusion of angiotensin II (Ang II) at 5. 0 nglmin and administration of A- 182086 at 24 mglkg/day”. In rats on either high sodium or normalsodiumintake... . . . . ......118 Line plots show water balance responses in milliliters per day (mIS/day) to infusion of angiotensin II (Ang II) at 5.0 nglmin and administration of A-182086 at 24 mglkg/day in rats on either high sodium or normal sodium intake ........ 119 Line plots Show sodium balance responses in milliequivalents per day (mEq/day) to infusion of angiotensin II (Ang II) at 5.0 nglmin and administration of A-182086 at 24mglkg/day in rats on either high sodium or normal sodium intake... ....120 Line plots show hemodynamic responses to bolus intra- arterial injection cf A-182086 at 24 mglkg in rats on high sodium or normal sodium intake... .. .. ...121 Hemodynamic responses to infusion of angiotensin II (Ang II) at 5. 0 nglmin and administration of trichlormethiazide at 10 mglkg/day In rats on 8 mEq/day sodium intake or on 2 mEq/dayscdiumintake... .137 Water balance responses in milliliters per day (mls/day) in response to infusion of angiotensin II (Ang II) at 5.0 nglmin and administration of trichlormethiazide at 10 mglkg/day in rats on 8 mEq/day sodium intake or 2 mEq/day sodium intake... ....138 (A) Frgure 5 Chapter 7 F'Sc'e 7 1 Figure 6.3: Figure 6.4: Figure 6.5: Chapter 7 Figure 7.1: Figure 7.2: Figure 7.3: Sodium balance responses in milliequivalents per day (mEq/day) in response to infusion of angiotensin II (Ang II) at 5. 0 nglmin and administration of trichlormethiazide at 10 mglkg/day In rats on 8 mqulday sodium intake or 2 mEq/daysodiumintake... . 139 Hemodynamic responses to bolus intravenous injection of trichlormethiazide at 10 mglkg in rats on 8 mEq/day sodium intake or on 2 mEq/day sodium intake... ....140 Blood volumes in milliliters in rats on 8 mEq/day sodium intake or on 2 mEq/day sodium intake on control day 2, and Ang II infusion days 5, 10, and 14... ...142 Concentration dependent contraction to endothelin—1 (ET-1) in control, A-192621 (ETB selective receptor antagonist, 30nM) incubated, ABT-627 (ETA selective receptor antagonist, 30nM) incubated, and A-182086 (ETA/3 nonselective receptor antagonist, 30nM) incubated superior mesenteric artery of the rat... 155 Concentration dependent contraction to endothelin-1 (ET-1) in control and angiotensin II (Ang II) incubated superior mesenteric artery of the rat... 156 Line plots Show mean arterial pressure (MAP) responses to infusion of angiotensin II at 5.0 nglmin or phenlyephrine at 2uglmin for two hours in rats on either high (circles) or normal (triangles) sodium intake... ....158 xvii ." a: ', fitters V. «:3 CD (I) A V 3"" -4 A'- nv Figure 7.4: Chapter 8 Figure 8.1: Concentration dependent contraction to endothelin-1 (ET-1) and phenylephrine (PE) in superior mesenteric artery from normotensive control and hypertensive angiotensin II (Ang II) infused rats on normal and high salt intake... 160 A model representing the inter-relationships between salt- intake, Ang II, and ET-1 in the pathogenesis of hypertension... 174 xviii ACE ANS ACE ANS BP BV Ca++ cAMP cGMP CNS CO DAG DOCA-salt E050 ECE ET ET-1 ET-2 ET-3 HR IP3 [Ca”]i i.a. i.p. i.v. L-NAME L-NMMA MAP NE N0 NOS 1 K1 C PGI2 PKC PLA2 PLC RAS 86c s.c. SHR TPR 2K1 C VSMC ABBREVIATIONS angiotensin converting enzyme autonomic nervous system blood pressure blood volume calcium cyclic adenosine monophosphate cyclic guanlyl monophosphate central nervous system cardiac output diacylglycerol decxycorticostercne acetate-salt effective concentration 50 endothelin converting enzyme endothelin endothelin-1 endothelin-2 endothelin-3 heart rate inositol 1,4,5-triphosphate intracellular calcium intra-arterial intraperitoneal intravenous NG-nitro-L-arginine methyl ester NG-monomethyI-L-arginine mean arterial pressure norepinephrine nitric oxide nitric oxide synthase one kidney-one clip prostacyclin protein kinase C phospholipase A2 phospholipase C renin-angiotensin-system sarafotcxin 6c subcutaneous spontaneously hypertensive rat total peripheral resistance two-kidney-one-clip vascular smooth muscle cell xix I Hyperten A: Ire pres cases of death. r II‘ ‘35 30001.01 are Tiese cardzovasa gereratmg a firm- fNH‘LBI. 199.7) H. $323!: for came... "£3353: ve heart .‘I 155556 (Burt e: a Sn.“ R9900 of the Chapter 1 INTRODUCTION I. Hypertension At the present time, heart disease and stroke are the first and third leading causes of death, respectively, in the United States. Each year, 43% of all deaths in this country are attributed to cardiovascular disease (Whelton et al., 1994). These cardiovascular killers present an enormous public health problem by generating a financial burden of more than $259 billion in direct and indirect costs (NI-ILBI, 1997). High blood pressure is one of the most important modifiable risk factors for cardiovascular disorders such as myocardial infarction, stroke, congestive heart failure, end-stage renal disease, and peripheral vascular disease (Burt et al., 1995). Hypertension is somewhat arbitrarily defined by the Sixth Report of the Joint National Committee on Detection, Evaluation, and Treatment of High Blood Pressure as the finding of an average systolic blood pressure >140 mmHg, or a diastolic blood pressure >90mmHg, or current treatment with an antihypertensive medication. The objective of identifying and treating hypertension is to reduce the risk of cardiovascular disease and its associated morbidity and mortality (JNC-Vl, 1997). Epidemiology Hypertension affects nearly 50 million Americans (Burt et al., 1995). The prevalence of hypertension increases with age, so that an estimated 50 percent 0ij Unrted 335 D’BSSJG read I" _: peiatron I699; to tower her béo: tarantam good :Iahagenent Is ;, A8333 Mergers are reament of r‘ ma). ENE" , . WIDQREHSIOD III @311 e .955 of age I The Hispanrc 9ch 501'. n . orIHISpanIc t Wen and me.n 5* he I A fee preaching met: than n . Ii fork» :Imn . Amerca' en u; Siesta,- M“ 3"." i la. [er In WC- 5m, ”.3"ch ‘- of the United States population over 60 years of age have elevated blood pressure readings (Cushman, 1994). Two-thirds of the entire hypertensive population (69%) are aware of their diagnosis, half (53%) are taking medication to lower their blood pressure, and only a quarter of these people (27%) are able to maintain good blood pressure control (Burt et al., 1995). Hypertension management is particularly problematic in population subgroups, including African Americans, Hispanics, and Native Americans, in whom both awareness and treatment of high blood pressure continues to be suboptimal (Semchenko et al., 1998). Epidemiological studies have consistently shown a higher incidence of hypertension in African Americans compared with Caucasian Americans, regardless of age, although the difference is most pronounced at younger ages. The Hispanic population has lower hypertension prevalence rates than those of both non-Hispanic blacks and whites (Flack and Yunis, 1993). Women and men share many of the same risk factors for hypertension, despite the male predominance of this disorder. Hypertension is less common among women than men up to 65 years of age in Caucasians and up to 45 years of age for African Americans. In the older age groups, prevalence rates are actually slightly higher in women than men. In all age groups, prevalence rates are higher among black women than white women, and black women have greater reductions in morbidity and mortality with antihypertensive treatment than white women (LaCroix, 1993). Tne US Depa-"I" Wrgflmg blOOC year 2000 {DHH Etiology One great heterogeneous Ir mgr door: press. byix'fensI-on EJI 33.59 is Know a ciagnosls of esse h mafierrulln -'5 Us a?» n .ch aways d, '.v’I':‘-.-a7*.s. 1994' The US Department of Health and Human Services has established a goal of controlling blood pressure in at least 50% of the hypertensive population by the year 2000 (DHHS, 1991). Etiology One great difficulty in studying hypertension is that the disease process is heterogeneous in its etiology. In nearly 95% of affected people, the cause of high blood pressure is unknown, a condition referred to as essential hypertension. Elevated blood pressure that can be attributed to a definable cause is known as secondary hypertension (Deshmukh et al., 1998). The diagnosis of essential hypertension is one of exclusion in that it is the only option left after ruling out the potential causes of secondary hypertension, which are almost always due to an alteration in renal function and/or hormone secretion (Williams, 1994). Secondary hypertension is often curable with correction of its cause. Essential hypertension is more a description than a diagnosis since it indicates only one particular physical finding, elevated blood pressure, rather than a cause for the finding. The difficulty in identifying a cause lies in the fact that multiple systems play a role in regulating arterial pressure, including the peripheral and/or central nervous (Brody et al., 1987), renal (Guyton, 1991), endocrine (Ely et al., 1997), and circulatory systems (Safer and London, 1987). The complex interplay of these systems with one another further complicates the issue. There l5 5 5,159 s.‘gn:.‘." r'. metres of hype to he dIsorc'er ; S tims {Deshmkf‘ comes from the u: Nth-etch et al 13 It Is most I.- ‘essental hype'te deeds especal, p'es M e .atlon that l ’IQNI. 5,, ‘5 Value to l There is also a clear genetic component to hypertension, as demonstrated by the significantly higher rate of elevated blood pressure among first-degree relatives of hypertensives than the general population. Additionally, concordance for the disorder is much greater between monozygotic twins than for dizygotic twins (Deshmukh et al., 1998). Further support for a genetic role in hypertension comes from the uneven distribution of hypertension among racial groups (Whelton et al., 1994; Burt et al., 1995). It is most likely that several different underlying defects are responsible for “essential hypertension” in different subpopulations of patients and that these defects, especially when mixed with environmental stressors, result in the clinical presentation that is known as essential hypertension. It is therefore of great scientific value to understand the intricate relationships between the various systems that work to control blood pressure. Hemodynamics Blood pressure (BP) is the product of cardiac output (CO) and total peripheral resistance (TPR). Likewise, CO is the product of cardiac stroke volume (SV) and heart rate (HR). These five functional parameters depend on the heart to supply the pumping pressure, blood vessel tone to determine the systemic resistance, and the kidneys to regulate intravascular volume. It is important to keep the role of the kidneys in mind because renal fluid excretion alone can completely return blood pressure to normal levels by reducing intravascular volume. Therefore, the maintenance of chronic hypertension requres renal pa hype'tenspn a'e Hemodym m..-rd a filgl‘. CO HCWEI'ef. as the ' amai shdes tie Incease in TPR . mwyfiaf'llc pr: 99-31% VaSCgfar ”5559C (Cofema' Hechanisms of H) The nerve] In tIe hyperfine-or ‘5 lesrlbec abori- rless accompane :l 3‘-' :zcchL‘lQ TPR l5 3 requires renal participation even when the initial factors responsible for the hypertension are external to the kidney (Deshmukh et al., 1998). Hemodynamic studies performed in the initial stages of hypertension found a high CO and normal TPR during periods of rest (Lund-Johansen, 1994). However, as the hypertension progresses to a chronic state, both human and animal studies have demonstrated a reduction in CO accompanied by an increase in TPR (Lund-Johansen, 1989). Therefore the characteristic hemodynamic profile of a chronic hypertensive subject is normal blood flow with elevated vascular resistance, suggesting that regional blood flow is generally not impaired (Coleman and Hall, 1993). Mechanisms of Hypertension The peripheral vasculature is one of the most extensively studied systems in the hypertension field because of its obvious relationship to blood pressure. As described above, an increase in TPR is sufficient to increase blood pressure unless accompanied by a decrease in CO. The most common mechanism of increasing TPR is direct vasoconstriction. Several neural, endocrine, and paracrine factors function as vasoconstrictors, including norepinephrine (NE), angiotensin II (Ang II), endothelin-1 (ET-1), vasopressin, insulin, and various cytokines (Webb and Strachan, 1998). There is also evidence that TPR is increased by remodelling and growth of the vascular smooth muscle itself, resulting in decreased lumen size and therefore higher pressure through the vessel (Alexander and Griendling, 1993). The sympathetic nervous system (SNS) acts as both an initiator and a facilitator of chronic hypertension (lzzo, 1999). Acute stimulation of SNS control centers causes an increase in systemic blood pressure by producing arteriolar and venous constriction. The SNS acts to maintain blood pressure through a complex system of interactions with other systems. Activation of a-adrenergic receptors in the vasculature causes a rise in TPR, while stimulation of a- adrenergic receptors in the kidney produces volume expansion via renal vasoconstriction and sodium retention. Activation of BI-adrenergic receptors in the heart increase heart rate, and in the kidney stimulates release of renin, thereby activating the renin-angiotensin-system (RAS), as discussed in the next section. Experimental models have shown that lesions in the anteroventral third ventricle, posterior hypothalamus, and area postrema result in decreased blood pressure, thereby indicating the pressor effects of these brain regions (Wyss, 1999). Additional feedback neural control of blood pressure is from sensory nerve endings in the carotid sinuses and aortic arch, the baroreceptors, which relay blood pressure status to the brain (Chapleau, 1993). The kidney plays a role in most forms of hypertension and a defect in renal function is almost definitely involved in the pathogenesis of essential hypertension (Kaplan, 1998a). Furthermore, the course of hypertension often causes renal damage, leading to a circular relationship in which a kidney defect leads to hypertension, which in turn further damages the kidney, and thus causes more severe hypertension. An alteration in renal function which causes a change in sodium and water output in response to arterial pressure shifts is present is al: to natvureSIs is d s Pressure-Natl'iu Under nor' keney's excetor Icme and a retu frs‘. mmsed by ( ‘ 990 COWley and IOU cream 5X33;- F 4%“ Kaplan. 193 in m ~371th .aflia . ‘I. «we: range Sun ' a present is all forms of hypertension. This relationship, known as pressure- natriuresis, is discussed further below. Pressure-Natriuresls Under normal circulation conditions, when blood pressure rises the kidney’s excretion of sodium and water increases, resulting in reduced body fluid volume and a return of blood pressure to the normal range. This phenomenon, first proposed by Guyton et al. (1964), is known as pressure-natriuresis (Hall, 1990; Cowley and Roman, 1996; Kaplan, 1998b). A small increase in mean arterial pressure produces a large increase in the rate of sodium and water excretion, creating a steep slope of the curve relating these two variables (Hall, 1990; Kaplan, 1998b). In many hypertensive patients, the indices commonly used to evaluate renal function (glomerular filtration rate and renal plasma flow) are within the normal range, suggesting that hypertension can develop in the absence of kidney defects (Kaplan, 1998b). However, the relationship between renal sodium and water excretion and mean arterial pressure is abnormal in all types of clinical and experimental hypertension, indicating that some degree of renal dysfunction is necessary for the development and maintenance of hypertension (Hall, 1990). Under hypertensive conditions, the pressure-natriuresis curve is shifted to higher pressures so that sodium balance can only occur at higher than normal blood pressures. The mechanism of pressure-natriuresis is at least in part intrinsic to the kidney, because it occurs in isolated perfused kidney preparations (Cowley and Roman, 19'. preSSLlre-nat'lw nerwus System p'essures by a“: mdomne contra Regardless of he "at-areas theory Seiafice and bioo hyze'fensrcn to k. 1062‘ w.) 33” Sensitivity Of all of I: 5 n‘ and Roman, 1996). Yet, factors external to the kidney also can influence pressure-natriuresis. Release of vasoconstrictor factors or excess sympathetic nervous system activation can shift the pressure-natriuresis relationship to higher pressures by affecting glomerular filtration. Additionally, alterations in neural or endocrine control of renal tubular function can precipitate such a shift. Regardless of how systemic blood pressure is initially increased, the pressure- natriuresis theory predicts that the relationship between sodium and water balance and blood pressure must be shifted towards higher pressure for hypertension to be maintained (Hall, 1990; Cowley and Roman, 1996; Kaplan, 1998). Salt Sensitivity Of all of the environmental factors implicated in the development of hypertension, salt intake has generated the most attention and the most controversy (Taubes, 1998). Several epidemiological studies, most notably the lntersalt study of 1988, have shown a weak but significant relation between salt intake and the development of hypertension (INTERSALT, 1988; Kaplan, 1993). The available data suggests a threshold relationship between salt intake and hypertension to the effect that those who ingest less than 50 mmol per day of sodium do not develop hypertension and those who ingest more will develop hypertension if they have other predisposing factors (Kaplan, 1993; Kotchen, 1999). Some researchers have suggested that chloride may play an important role in hypertension development as well since over 95% of dietary salt is served Inrreformfsl showr that soc associated it)? Most rese exz'ahs Why the WT?“ elevated 58 Increase In blood he sane lndzwdt. West In cider t mt: lowrenm sta nsfiaency (GI, in the form of sodium chloride (Weinberger, 1993). Sato et al. (1991) have shown that sodium chloride loading in angiotensin II infused rats potentiates the associated hypertension, whereas sodium citrate loading does. Most researchers believe that a condition known as “salt sensitivity” explains why the blood pressure of some individuals, but not others, increases with elevated salt intake. Salt sensitivity is generally defined as a 10% or greater increase in blood pressure after salt loading as compared to low-sodium intake in the same individual (Weinberger, 1993). The prevalence of salt sensitivity is highest in older, black, and/or diabetic hypertensive patients, as well as those with low-renin status, increased sympathetic nervous activity, or renal insufficiency (Guyton et al., 1964; Weinberger, 1993). The exact etiology of salt-sensitive hypertension is not known. Guyton et al. (1964) were among the first to suggest that hypertension may be a necessary consequence of the kidney’s reduced ability to excrete a salt load. The resulting sodium retention and consequent volume expansion produce a transient increase in cardiac output (causing acute hypertension) that ultimately leads to chronically increased peripheral vascular resistance (and therefore chronic hypertension). This theory has been somewhat problematic to test in humans because the onset of the increase in blood pressure is often difficult to determine, but several experimental animal studies provide support for this variation over time in hemodynamic response to salt loading (Campese, 1994). Proposed renal causes of salt-sensitivity include defects in renal plasma flow and increased tubular reabsorption of sodium. Potential neural mechanisms Involve direct C' levels have ai'sc" m saft-resIstant barorempt res noreomephnne t r gored rena: d: l'lSu'llfl. the kale: tare 3i: Men pr. {Canoese 1994 Salt-sens : Ire development eq‘ ,, A. ah. maggots as. involve direct or indirect stimulation of the SNS by high salt intake. High salt levels have also been shown to sensitize cardiopulmonary baroreceptor reflexes in salt-resistant, but not salt-sensitive patients, suggesting a lack of appropriate baroreceptor response in salt-sensitive people. There is evidence that increased norepinephrine to dopamine ratios exist in salt-sensitive patients, suggesting that reduced renal dopamine levels may contribute to salt-sensitivity. Additionally, insulin, the kallikrein-kinin system, prostaglandins, and atrial natriuretic factor have all been proposed to play a role in the salt-sensitivity of hypertension (Campese, 1994). Salt-sensitive hypertensives frequently show a greater propensity towards the development of renal failure, left ventricular hypertrophy, microalbuminuria, and metabolic abnormalities that may predispose them to cardiovascular diseases (Campese, 1994). Ferri et al. (1998) have recently shown that salt- sensitive hypertensives have elevated plasma levels of the endothelium—derived substances endothelin-1, von Willebrand factor, and E-selectin. This finding supports the theory that salt sensitivity is correlated with increased risk for development of hypertension-related vascular damage. Treatment and Prevention Clinical management of hypertension is a challenge due to its multifactorial nature. Prevention is most easily achieved through patient education and lifestyle modifications, including weight loss, salt restriction, increased potassium intake, decreased alcohol consumption, smoking cessation, 1O and relaxation t Desnrnukn et a a firstllne treat unsafeleveis p acrenergfc rece; sy‘rpat‘tolytlcs. traders. and an Graham, 1993 F innitrtors are be {Jackson and G; ”9911)! bang l! ‘2‘: ‘Y ‘9 ~ eat'nent of and relaxation therapies (OMCC, 1996; JNC-V, 1997; Semchenko et al., 1998; Deshmukh et al., 1998). These patient-controlled activities may also be used as a first-line treatment of hypertension. If the blood pressure remains elevated to unsafe levels, pharmacological treatments may be used, including diuretics, B- adrenergic receptor antagonists, a-adrenergic receptor antagonists, sympatholytics, angiotensin converting enzyme inhibitors, calcium channel blockers, and angiotensin II receptor antagonists (Frishman, 1993; Gavras, 1993; Graham, 1993; Perry, 1993; Puschett, 1993; Weir, 1993; Oates, 1996). Renin inhibitors are being evaluated for possible use as antihypertensive agents (Jackson and Garrison, 1996). Endothelin-1 receptor antagonists are also currently being investigated as a promising class of novel therapeutic agents for the treatment of hypertension (Webb and Strachan, 1998). 11 IL Renin-A! l. lt has lor Integral rote In t' oa‘ance In both I experzmental by, enzyme of the R )uraglomerular ; orreduced set C $9,...” “ s i! 'Ifi“ av» S. 099". 35”” n ~ alvte;.S;n I (A:- ll. Renin-Angiotensin System It has long been known that the renin-angiotensin system (RAS) plays an integral role in the regulation of blood pressure as well as in electrolyte and fluid balance in both normotensive individuals, and in a variety of forms of clinical and experimental hypertension. Renin, an aspartyl protease, is the rate—limiting enzyme of the RAS. It is secreted into the blood by the granular cells of the juxtaglomerular apparatus of the kidney in response to decreased renal perfusion or reduced salt delivery. Circulating renin cleaves the circulating preprohormone, angiotensinogen, which is secreted by the liver, to produce a decapeptide called angiotensin I (Ang I). This Ang I is in turn cleaved by angiotensin converting enzyme (ACE) to produce an octapeptide called angiotensin II (Ang II). ACE is located on the endothelial surface of pulmonary capillaries. Ang II is the primary effector molecule of the RAS (Bader et al., 1994; Griendling and Alexander, 1994; Deshmukh, 1998). The rate of Ang II formation is inversely related to salt intake because high salt levels inhibit renal release of renin. Effects of Angiotensin II There are at least two subtypes of Ang II receptors, denoted AT1 and AT2, that have been cloned and characterized. These receptors belongs to the superfamily of G-protein coupled receptors that have seven transmembrane regions (Murphy et al., 1991; Sasaki et al., 1991). Ang II binding to the AT1 receptor results in a myriad of physiological events. In arterial smooth muscle, it 12 causes market: giand, It acts tc renal sodium re noreplnephrme :rrty mm a." mt'actrlty and causes growth a {Gelsterfer et al . dIre—c‘dy on the tu' Deshmukh et al Angiotensin ll In lthas long EVE-59p DVOgress asei causes marked vasoconstriction followed by a drop in blood flow. In the adrenal gland, it acts to stimulate release of aldosterone which in turn acts to increase renal sodium reabsorption. In the SNS, it facilitates the release of norepinephrine to increase CO and TPR. In the brain, it stimulates sympathetic activity, thirst and vasopressin secretion. It works in the heart to enhance contractility and ventricular hypertrophy. Ang II is also a growth factor that causes growth and remodelling of the vascular media, thereby increasing TPR (Geisterfer et al., 1985; Robertson and Nicholls, 1993). In the kidney, Ang II acts directly on the tubular cells to promote sodium and water reabsorption (Deshmukh et al., 1998). Angiotensin II Induced Hypertension It has long been known that animals receiving daily infusions of Ang II will develop progressive hypertension after a period of normotension (Koletsky et al., 1966). When Ang II is administered parenterally to either humans or experimental animals, the characteristics of the hypertension produced are dependent on the amount given. Large doses of Ang II (>30 nglkglmin) result in an acute pressor effect that occurs within seconds to minutes and returns to normal within minutes of infusion cessation (Brown et al., 1981 ). This acute pressor response is thought to be due to direct actions of Ang II on vascular smooth muscle (Li et al., 1996). Conversely, small doses of the peptide (2-20 nglkglmin) take hours to days to produce an increase in blood pressure. This slow pressor response persists for hours to days after stopping the infusion 13 (Brown et al . Increased DIG tone (Lult et a Sludes chronéc Infuse potassium. ace oontrols (Dowe pctentated ate the adrenergzc (Brown et al., 1981). This chronic increase in blood pressure is accompanied by increased production of aldosterone in some studies and increased sympathetic tone (Luft et al., 1989). Studies of vascular reactivity of mesenteric arteries from rats receiving chronic infusions of low-dose Ang II have shown that contractile responses to potassium, acetylcholine, and Ang II were not significantly changed compared to controls (Dowell et al., 1996). However, the response to phenylephrine was potentiated after chronic Ang II treatment, suggesting that Ang II may enhance the adrenergic response in vivo. Angiotensin II and Pressure-Natriuresis Angiotensin II is one of the strongest modulators of pressure-natriuresis. Long-term intravenous infusion of Ang II depresses the slope of the pressure- natn'uresis curve and shifts it to higher pressures (Cowley and Roman, 1996). Hall et al. (1990) have shown that dogs with intact renin-angiotensin systems, under conditions of chronic increased salt intake, can maintain sodium balance with only minor changes in blood pressure. However, in dogs under the same conditions that also received continuous low dose intravenous infusions of Ang II, large elevations in blood pressure were necessary to maintain sodium balance as sodium intake was increased. These findings suggest that the RAS plays an important role in the maintenance of normal blood pressure across a wide range of sodium intakes and that an inability to suppress the RAS in response to 14 excess sodrur and sense-qua These 30595 of Ang I‘COVl'fey anc deaease be; i815. Ang II E her excess sodium intake can lead to resetting of the pressure-natriuresis system and consequently produce a chronic rise in blood pressure (Hall 1990). These observations are softened by the report that, in rats, relatively large doses of Ang II are needed to produce a shift in the pressure-natriuresis curve (Cowley and Roman, 1996). Since these doses are still not sufficient to decrease blood flow or renal interstitial hydrostatic pressure, it seems that, in rats, Ang II acts to alter the pressure-natriuresis curve via enhanced renal tubular reabsorption of sodium. The role of Ang II in pressure-natriuresis has not yet been fully defined. Salt Sensitivity of Angiotensin II Induced Hypertension Blood pressure responses to Ang II are salt—dependent in that infusion of Ang II at low doses causes hypertension in subjects with high salt intake but not in subjects with normal salt intake (Muirhead et al., 1975; Robertson and Nicholls, 1993; Haynes and Webb, 1998; Simon et al., 1998). Additionally, the decrease in renal plasma flow seen in response to Ang II in subjects on a high salt diet is normalized when the same subjects are switched to a low salt diet (Sirvio et al., 1990; Robertson and Nicholls, 1993). Hall et al. (1980) that the RAS plays a major role in maintaining steady- state levels of arterial pressure and renal hemodynamics during chronic changes in sodium intake. The mechanism by which Ang II chronically conserves sodium is through increased renal sodium reabsorption. A decade later, Krieger et al. (1989) showed that Ang II salt-dependent hypertension is consistently related to 15 Signrfioant Increases and TPR. Ando et a and hypertenswe e.“ parallel one another In addition to I reoent‘y reported the ngrtglmin for two we [Lombardi et al . 199 fiat/l and functiona: ' .4 "vi-9‘3) that favor so: significant increases in fluid retention, blood volume expansion, cardiac output, and TPR. Ando et al. (1990) have suggested, however, that the sodium retention and hypertensive effects of Ang II salt-dependent hypertension do not always parallel one another. In addition to chronic infusion of low doses of Ang II, Lombardi et al. have recently reported that transient exposure to high pressor doses of Ang II (435 nglkglmin for two weeks) results in the development of sustained hypertension (Lombardi et al., 1999). This is purportedly due to structural (microvascular injury) and functional (loss in intrarenal nitric oxide formation) changes in the kidney that favor sodium retention. 16 m Endothelin By the Mid: endothelium-denve otnrric oxide as a: HICKEY et al. repor was partied. sect.- at In 1988 (Yanag the most potent va BiC’syrlthesis The family R3}, the snake I MC). EEC—h ET IS Seedfic precursor Poélocig 1998. w"2 r Ill. Endothelin By the mid-1980s, cardiovascular researchers were searching for an endothelium-derived constricting factor (EDCF) to counterbalance the discovery of nitric oxide as an endothelium-derived relaxation factor (EDRF). In 1985, Hickey et al. reported release of such an EDCF (Hickey et al., 1985). This factor was purified, sequenced, and cloned in the monumental work of Yanagisawa et al. in 1988 (Yanagisawa et al., 1988). The factor was named endothelin and is the most potent vasoconstrictor yet known. Biosynthesis The family of endothelins (ET) consists of three isoforms (ET-1, ET-2, and ET-3), the snake venom sarafotcxin (86c), and vasoactive intestinal contractor (VIC). Each ET isopeptide is represented by a distinct gene that encodes a specific precursor for the mature isoform (lnoue et al., 1989; lnagami et al., 1993; Pollock, 1998; Webb et al., 1998). Each ET gene is transcribed and translated to produce a 212 amino acid preproendothelin. A short secretory sequence is then removed to form proendothelin, which is in turn acted on by a furin-like enzyme to produce the inactive 38 residue peptide known as big ET. Endothelin converting enzyme-1 (ECE-1 ), a neutral metalloendopeptidase, cleaves 17 amino acid residues off the C-terminal of big ET to form the final 21 amino acid peptide, ET (lnagami et al., 1993; Webb et al., 1998; Takayanagi et al., 1998). Each ET isoform contains two intrachain disulphide bridges linking paired 17 qsteine reSId. Webb, 1998). Sites of Gene PM or expression of generation of ”MUM by? SiStem. and (Gray, 19335; tier (How; Expressed m A . . no: veer: Icer Endothelin j The GI ha fife been Ctr or , | . , shawls; s t W. . 3-3 has cysteine residues, thereby producing a semi-conical structure (Haynes and Webb, 1998). Sites of Generation and Secretion ET-1 comprises 80% of total endothelin formation. Studies assessing the expression of mRNA for preproendothelin—1 have shown that the major site of generation of ET-1 is in endothelial cells (Webb et al., 1998). ET-1 is also produced by the heart, kidney, posterior pituitary gland, the central nervous system, and, to a very limited extent, human aortic vascular smooth muscle cells (Gray, 1995). ET-2 is found in small amounts in endothelial cells, heart and kidney (Howard et al., 1992; Plumpton et al., 1993). ET-3 is selectively expressed in the endocrine, gastro-intestinal, and nervous systems. ET—3 has not been identified in endothelial cells (Gray, 1995). Endothelin Receptors The endothelins act on at least two receptor subtypes, ETA and ETB, that have been cloned, sequenced and characterized on the basis of their pharmacology. The ETA receptor is preferentially activated by ET-1 and ET-2, but ET-3 has very low affinity for this receptor subtype. ETB is a nonselective receptor with practically equal affinity for all three isoforms of endothelin, as well as for sarafotcxin 6c. ETA receptor mRNA is expressed most highly in the vascular smooth muscle, aorta, heart, and kidney, but not in endothelial cells. Conversely, mRNA for the ETB receptor is most highly expressed in cultured 18 endothelial cells an; al.1993; Haynes 3' The ET. and snanng only a 70% 1 Structural features Ir mooted receptors, II and a relatively long phospholipase C an vasoconstrictors, ET Night to be clue I: at. 1993). and Mlnlfjaj EXP-'ess Subc;aSS ”my 0n Vasmlar yeSGCoT‘IStnqunl a was responsible to endothelial cells and in smaller amounts in vascular smooth muscle (lnagami et al., 1993; Haynes and Webb, 1998). The ETA and ETB receptor subtypes appear to be functionally distinct, sharing only a 70% sequence homology. However, both receptors have structural features in common with the superfamily of rhodopsin-like G-protein- coupled receptors, including seven hydrophobic membrane-spanning domains and a relatively long extracellular N terminal. Both subtypes activate phospholipase C and L-type calcium channels. In comparison to other vasoconstrictors, ET is known to produce a prolonged contractile response that is thought to be due to tenacious binding of the peptide to the receptor (lnagami et al., 1993). Expression of ET receptors is increased by ischemia and cyclosporin, and decreased by ET-1, Angll, and phorbol esters (Haynes and Webb, 1998). The initial subclassiflcation of ET receptors held that the ETA receptor, located mainly on vascular smooth muscle, was generally responsible for vasoconstriction, and that the ETB receptor, located mainly on endothelial cells, was responsible for vasodilation via nitric oxide and prostacyclin release (Pollock, 1998). Webb et al. showed that endogenous ET—1 confers basal constrictor tone on vascular smooth muscle in humans through ETA receptor activation which is modulated by endothelial cell NO-dependent ETB-mediated dilator tone (Webb et al., 1998). However, it has recently been observed that some non-ETA receptors mediate at least part of the vasoconstrictor actions of ET. Binding studies and other in vitro experiments have shown that some vasculature contain ETB receptor mediated constrictor elements (Bigaud et al., 1992; Clozel et al., 1992; 19 Cristal et a of ET 5 ago Icon). It IS Waugh ET l‘I’eno. 193 U Internatze; Cristol et al., 1993). Additionally, other studies have demonstrated that infusion of ETB agonists will elicit a hypertensive response in most species (Sakurai et al., 1990). It is believed that a substantial proportion of clearance of ET-1 occurs through ETa receptor binding followed by receptor internalization (Haynes and Webb, 1998; Webb and Strachan, 1998; Miyauchi and Masaki, 1999). The internalized ET-1/ET3 receptor complex is sequestered into lysosomes where the acidic environment facilitates ligand dissociation (Miyauchi and Masaki, 1999). Antagonism of the ETB receptor subtype results in increased plasma concentrations of ET-1, whereas blockade of ETA receptors does not. The clearance function of ETB receptors contributes to the short half-life (~1 minute) of ET-1 in the blood (Webb and Strachan, 1998). Endothelin Signal Transduction Mechanisms Binding of ET-1 to ETA or ETB receptors results in G-protein dependent action of phospholipase C, leading to the rapid hydrolysis of the membrane inositol phospholipid, phosphatidylinositol 4,5-bisphosphate, yielding cytosolic inositol 1,4,5-triphosphate (IP3) and membrane-bound sn1,2-diacylglycerol (DAG). IP3 causes a rapid increase in the intracellular concentration of calcium through its release from intracellular stores, thereby activating Ca++ dependent processes and leading to cell membrane depolarization and sustained extracellular Ca+ influx. DAG activates protein kinase C, which in turn sensitizes the contractile apparatus to the elevated levels of intracellular calcium. DAG also 20 aciiaes nudea’ 5 regulation Of 09”“[a were 1998) Endothelin Recep Many pharrr patwlany drugs t adotional benefits renal Impairment t' orsuit. Several e development. Sir utinately prove tc wt? be dissussed P0156707 ”O'I‘Depme Com; rare shown this < activates nuclear signalling mechanism that may have long-term effects on the regulation of cellular growth and function (lnagami et al., 1993; Haynes and Webb, 1998). Endothelin Receptor Antagonists Many pharmacological treatments for hypertension currently exist, particularly drugs that function to counteract vasoconstriction. It is the potential additional benefits of ET-1 receptor antagonists on vascular hypertrophy and renal impairment that make the further refinement of these drugs a worthy pursuit. Several endothelin receptor antagonists are currently under development. Since it remains to be seen which of these antagonists will ultimately prove to be of the most scientific value, only those used in this thesis will be discussed herein. PD156707 (Parke-Davis Pharmaceutical Research, Ann Arbor, MI) is a non-peptide competitive antagonist for both ET receptor subtypes. Maguire et al. have shown this compound to possess up to a 15000-fold greater selectivity for the ETA receptor (subnanomolar affinity) compared with the ETB receptor (micromolar affinity) in in vitro pharmacological experiments. This group also demonstrated in functional experiments that PD156707 potently antagonized the vasoconstrictor responses to ET-1 in isolated vascular preparations. An additional interesting finding is that PD156707 proved to be a more effective antagonist at lower concentrations than higher ones (Maguire et al., 1997). 21 Abbott Lab: selecjjve receptor é Porseror. A-127‘ (Opgenorth et al., ‘ high polenCy, craft-y I Laboratories, 1998 ETA receptors {K = 3 ET t. receptors Col”)? tare shown ART-62 In Isolated rat aorta ( rt. “1 Aoastonal studies he antagonism of ET-1 I ill-192621 (Ab! bIC-a‘ralia'ole nonpe~ ptl Abbott Laboratories (Abbott Park, IL) has developed a nonpeptide ETA selective receptor antagonist, A-127722, that is structurally distinct from PD156707. A-127722 is the most potent ETA receptor antagonist yet reported (Opgenorth et al., 1996). The active enantiomer of this racemate, ABT-627, is a high potency, orally bioavailable and efficacious ETA receptor antagonist (Abbott Laboratories, 1998). In vitro, ABT-627 has displayed very high affinity binding to ETA receptors (K; = 34 pM), and a more than 1000-fold greater binding affinity to ETA receptors compared with ETB receptors (K; = 63nM). Functional studies have shown ABT-627 to dose-dependently inhibit ETA mediated vasoconstriction in isolated rat aorta (Opgenorth et al., 1996; Abbott Laboratories, 1998). Additional studies have shown this compound to be orally efficacious for in vivo antagonism of ET-1 induced increases in MAP (Abbott Laboratories, 1998). A-192621 (Abbott Laboratories, Abbott Park, IL) is a highly potent, orally bioavailable nonpeptide selective antagonist of ETB receptors. A-192621 has been shown to competitively antagonize the specific binding of [mu-labeled ET-1 to CHO cells transfected with human ETB receptors (KI = 8.8 nM) but is far less potent in inhibiting binding to CHO cells transfected with human ETA receptors (K; = 5.6 uM; Abbott Laboratories, 1998). In vivo, A-192621 inhibits the ETB- receptor-mediated initial depressor response to intravenous sarafotcxin 60 (S6c, an ETB receptor agonist; Abbot Laboratories, 1998). This blockade occurred when A-192621 was administered either orally or intravenously. A-192621 did not block ET-1 induced contractions in the rat aorta. 22 A482 bioavaia’ce 182386 has labeted ET. on CHO cej 7998} {n v: Diessor res Intrarenogs Cardiovas. Stud m he Cara. meaSE-d ‘4 W the RA A-182086 (Abbott Laboratories, Abbott Park, IL) is a highly potent, orally bioavailable nonpeptide mixed antagonist of both ETA and ETB receptors. A- 182086 has been shown to competitively antagonize the specific binding of [ml]- labeled ET-1 to ETA receptors (K. of 0.2 nM) and to ETB receptors (KI of 1.23 nM) on CHO cells transfected with the ET-1 receptor subtypes (Abbott Laboratories, 1998). In vivo, A—182086 inhibits both the ETA-receptor-mediated prolonged pressor response and the ETB-receptor-mediated initial depressor response to intravenous big ET-1 (Abbot Laboratories, 1998). Cardiovascular Physiology of Endothelin Studies with ET receptor antagonists have helped to define the role of ET in the cardiovascular system. The cardiovascular effects of ET-1 include increased vascular tone, increased mitogenesis, and activation of both the SNS and the RAS. The marked pressor effect of ET-1 occurs mainly through an increase in peripheral vascular resistance. Haynes and Webb have shown that endogenous ET-1 generation and binding to ETA receptors contributes to the maintenance of basal vascular tone in forearm resistance vessels of healthy human subjects (Haynes and Webb, 1998). ET-1 produces a prolonged vasoconstriction in the peripheral vasculature that lasts for up to 2 hours after the infusion is stopped. This vasoconstriction leads to a slowly developing dose- dependent decrease in blood flow through resistance vessels (Brunner, 1998; Webb and Strachan, 1998). ET-1 has also been shown to produce venoconstriction, most notably in the human dorsal hand veins in vivo (Webb and 23 St'acnan, ‘ vasodratio 1998). ind.- pnysologr As I endogeno, restated \ 1938; We: “are Silo“ tone Sofie Elms-r31; Strachan, 1998). Blockade of ET receptors in normotensive subjects produces vasodilation accompanied by a drop in blood pressure (Haynes and Webb, 1998), indicating again that endogenous endothelin is involved in the physiological maintenance of blood pressure. As previously mentioned, recent studies suggest that overall effects of endogenous ET-1 are due to a complex interplay between ETA receptor mediated vasoconstriction and ETB receptor mediated vasodilation (Brunner, 1998; Webb and Strachan, 1998; Haynes and Webb, 1998). Webb and Strachan have shown in human forearm vessels in vivo that the basal vascular constrictor tone conferred by endogenous ET-1 via the ETA receptors is modulated by the ETa mediated vascular dilator tone via NO release (Webb and Strachan, 1998). This proposed interplay is of course further complicated by the reported vasoconstrictor effects of ETB receptors (Sakurai et al., 1990). ET-1 is also a potent mitogen that acts to produce hypertrophy of vascular smooth muscle cells, as well as cardiac myocytes and fibroblasts (Webb and Strachan, 1998; Haynes and Webb, 1998). ET-1 causes enhanced expression of mRNA for the growth-promoting oncogenes, c-fos and c-myc, that play key roles in the control of cell growth and proliferation (Force, 1998; Haynes and Webb, 1998). The mitogenic effects of ET-1 are mediated by the ETA receptor and may serve to amplify the vasoconstrictor effects of ET—1 (Force, 1998). 24 Renal Physiology. ET-1 is syn'. nesangzal and Inte Immunonistocherr ltégrer levels of ET 1998). Specnlcaity greatest amount of t Kidneys from reeeptors. ET), rece recta. artuate alteI'II etal, 1995). ETB re Multan collecting IQtrey (Rubanyi ant Mentor subtype ca the malo’lty or the E Renal Physiology of Endothelin ET—1 is synthesized and released in the kidney by cells of renal tubular, mesangial and interstitial origin, as well as in the renal vascular endothelium. Immunohistochemical and gene expression studies have consistently found higher levels of ET-1 in the renal medulla compared to the renal cortex (Pollock, 1998). Specifically, the inner medullary collecting duct cells are the sites of the greatest amount of ET-1 gene expression. Kidneys from all investigated species included both ETA and ETB receptors. ETA receptors have been localized to the renal glomerulus, vasa recta, arcuate arteries, and mesangial cells (Rubanyi and Polokoff, 1994; Clavell et al., 1995). ETB receptors have been localized to the initial and terminal inner medullary collecting duct, as well as the glomerulus and mesangial cells of the kidney (Rubanyi and Polokoff, 1994; Clavell et al., 1995). Activation of either receptor subtype causes renal vasoconstriction. In the rat, unlike dog or human, the majority of the ET-1 mediated renal vasoconstrictor response is due to activation of ETB receptors (Gurbanov et al., 1996). However, ET-1 shifts the pressure-natriuresis relationship to higher pressure levels predominately via activation of ETA receptors (Kassab et al., 1998). ET-1 constricts both afferent and efferent arterioles equally, thereby causing reductions of renal plasma flow and glomerular filtration rate (Haynes and Webb, 1998; Baylis, 1999). Several lines of evidence suggest ET-1 plays a role in the pathogenesis of acute renal failure following renal ischemia. In rats, administration of an ET-1 antibody prior 25 to clamping of thei reduction ol renal Masaki, 1999) A bilateral OCduSIon (Miyauchi and Mas administration of Bt renal failure (Brook! In spite of ET Suggests that ET-1 by direct actions on vasopressm-stimuia ETs receptors (Ede 50dlum by Inhibiting asoenoing limb, an Banks et al 1 998 and natriuretic effe- §l3iltemlar filtratior MET‘1 has a Cl; to clamping of the renal artery prevents the increase of arteriolar resistance and reduction of renal plasma flow and glomerular filtration rate (Miyauchi and Masaki, 1999). Administration of the ETA receptor antagonist, BQ-123, prior to bilateral occlusion of the renal arteries slows renal functional impairment (Miyauchi and Masaki, 1999). Furthermore, at least two studies have shown that administration of BQ-123 24-48 hours following renal ischemia can reverse acute renal failure (Brooks et al., 1998; Miyauchi and Masaki, 1999). In spite of ET-1’s potent vasoconstrictor effects, accumulating evidence suggests that ET-1 has an important role in renal excretion of sodium and water by direct actions on the renal tubules. ET-1 has been reported to inhibit arginine vasopressin-stimulated water and chloride reabsorption in the collecting duct via ETB receptors (Edwards et al., 1993). Additionally, ET-1 blocks reabsorption of sodium by inhibiting the sodium-potassium ATPase in the proximal tubule, thick ascending limb, and inner medullary-collecting duct (Haynes and Webb, 1998; Banks et al., 1998; Plato and Garvin, 1999). ET-1 produces these potent diuretic and natriuretic effects at doses too low to produce significant reduction of glomerular filtration rate (Banks et al., 1998). Plato and Garvin have suggested that ET-1 has a biphasic effect on sodium/water transport in the proximal tubule by stimulating reabsorption at low doses of ET-1 and inhibiting reabsorption at high doses of ET-1 (Plato and Garvin, 1999). These tubular effects of ET-1 also occur with ETB selective receptor agonists and are not blocked by BQ-123, indiwting that they are mediated by ETB receptors (Haynes and Webb, 1998). These findings are supported by the report that ETB knockout mice develop 26 hypertenSlon seC€ studies have Sho‘ generation 01 ET' reabsorption of 5‘ (Haynes and We. Endothelin and The multll vascular hypertrr activation of the pathogenesis of 3-1 alone will r: humans and ex: hypertension secondary to renal sodium retention (Ohuchi et al., 1999). Other studies have shown that under experimental hypertensive conditions, renal generation of ET-1 is decreased, resulting in decreased tonic inhibition of tubular reabsorption of sodium and water, and thereby leading to sodium retention (Haynes and Webb, 1998). Endothelin and Essential Hypertension The multiple cardiovascular actions of ET-1 (increased vascular tone, vascular hypertrophy, stimulation of the sympathetic nervous system, and activation of the renin-angiotensin system) suggest a role for this peptide in the pathogenesis of hypertension. It is well documented that short-term infusion of ET-1 alone will produce a significant increase in mean arterial pressure in both humans and experimental animals (Mortensen and Fink, 1990; Brunner, 1998). Mortensen and Fink have further demonstrated that hypertension induced by long-term ET-1 infusion in conscious rats is salt-dependent. In these studies, only animals receiving 6 mEq per day of sodium experienced a significant increase in MAP in response to ET-1 infusion at 5 pmollkglmin, whereas animals receiving 2 mEq/day of sodium remained normotensive (Mortensen and Fink, 1992a). Cardiac output, stroke volume, water balance, and urinary sodium and potassium excretion remained unchanged. Termination of the ET-1 infusion resulted in rapid normalization of MAP. Therefore, ET-1 induced hypertension is a salt-dependent model of hypertension. A connection between salt-sensitive 27 ’T hwenenSlon and levels 0i ET" 'n s tudles 9V6 nave prOduced var only In caseS 0" 56 nought to be 5900' 1993) However. Ci since ET-l Is thou; decxycorticosteronr correlation beMeer and the level of sys In vitro studies of IC levels In spontanec levels In nonnotens However, polymer; W bl00d presetirr hypertension. Thzs hypertension and ET-1 is further supported by the finding of increased plasma levels of ET-1 in salt-sensitive human hypertensives (Ferri et al., 1998). Studies evaluating plasma concentrations of ET-1 in human hypertension have produced variable results. Circulating levels of ET-1 tend to be increased only in cases of severe hypertension, and even then the increased levels are thought to be secondary to impaired renal clearance of ET-1 (Haynes and Webb, 1998). However, circulating ET-1 may not reflect the role of ET-1 in hypertension since ET -1 is thought to primarily act at the local tissue level. In rats with decxycorticostercne acetate (DOCA)-salt induced hypertension, a significant correlation between the concentration of immunoreactive ET-1 in vascular tissue and the level of systemic blood pressure has been reported (Clozel et al., 1994). In vitro studies of local mesenteric generation of ET-1 have shown increased levels in spontaneously hypertensive rats (SHR) as compared to vascular tissue levels in normotensive control Wistar-Kyoto (WKY) rats (Miyamori et al., 1991 ). However, polymorphisms of the preproendothelin-1 gene are not co-segregated with blood pressure for inbred salt-sensitive Dahl rats, a low-renin model of hypertension. This finding indicates that changes in the levels of ET-1 expression per se may not affect the pathophysiology of this model of hypertension (Haynes and Webb, 1998). Interestingly, human patients with ET- secreting haemangioendotheliomas are generally hypertensive (Ruschitzka et al., 1998) and a specific polymorphism (Lys198Asn) in the ET-1 gene is associated with increased blood pressure in obese human patients (Tiret et al., 1999). Other studies in hypertensive rats have shown reduced numbers of 28 ETJETa "3C9 res/113mm“l The mi amplify V8500 ETjreoeDlOr 5 model of hype oells {Shanta a' It IS cffil oonfounorng e.‘ ofresstance vr conduit vessefs 1998). Expenme drierent forms c Ilbenension. 5L ”IDIIKIC. a me ETA/ET; receptors in mesenteric vessels, but increased numbers of ETA receptors in the cortex of the kidney (Brooks et al., 1998). The mitogenic effects of ET-1 resulting in vascular hypertrophy may act to amplify vasoconstrictor effects of ET-1. Sharifi and Schiffrin have shown that ETA receptor antagonists act to normalize vascular structure in the DOCA—salt model of hypertension, possibly through apoptosis of vascular smooth muscle cells (Sharifi and Schiffrin, 1997). It is difficult to interpret sensitivity to ET-1 in hypertension because of the confounding effects of ET-1 induced vascular hypertrophy. In general, sensitivity of resistance vessels to ET-1 is reduced and sensitivity of capacitance and conduit vessels is increased under hypertensive conditions (Haynes and Webb, 1 998). Experimental evidence suggests that the role of ET-1 may differ in different forms of hypertension. In severe, salt-sensitive, low-renin models of hypertension, such as the DOCA-salt, Dahl salt-sensitive and one-kidney one- clip (1 K1 C, a model of renovascular hypertension in which one kidney is removed and the contralateral renal artery is clipped) models, circulating and tissue levels of ET-1 are increased (Haynes and Webb, 1998; Ruschitzka et al., 1998; Schiffrin, 1998). Similarly, these models appear to be particularly sensitive to the hypotensive effects of ET-1 receptor antagonists (Haynes and Webb, 1998; Ruschitzka et al., 1998; Schiffrin, 1998). In contrast, the endothelin system does not appear to be augmented in spontaneously hypertensive rats (SHR), a normal-renin model of hypertension. Furthermore, in two-kidney one-clip (2K1C, 29 a model of renc contralateral kic system Is not 3: al., 1996) The: receptor antago ”mnenson. Two recs Ir.- humans with I blOOd flow respc $816-ng regeptc ”Made of ET~‘ esma‘ hl’perte lrteresnngly, the We efiedlve at Sim‘ga’ly. 1 a model of renovascular hypertension in which one renal artery is clipped and the contralateral kidney is left intact) hypertension, a renin-dependent model, the ET system is not activated and bosentan does not lower blood pressure (Sventek et al., 1996). These strong experimental findings would appear to indicate that ET receptor antagonism might be most effective in RAS-independent models of hypertension. Two recent studies have reported finding increased vascular ET-1 activity in humans with essential hypertension. Cardillo et al. (1999) compared forearm blood flow responses to an ETA selective receptor antagonist and an ETB selective receptor antagonist, alone and in combination. They found that blockade of ET-1 receptors produced a vasodilator response in patients with essential hypertension, but not in normotensive controls. These results indicate enhanced ET-1 mediated vasoconstrictor activity in the hypertensive subjects. Interestingly, this study found that combined ETA and ETB receptor blockade was more effective at producing vasodilation than ETA receptor antagonism alone. Similarly, Taddei et al., ( 1999) also showed that combined ETA and ETB receptor blockade caused a larger vasodilation in hypertensive patients compared to normotensive controls. This group associated the difference between groups with a decrease in tonic nitric oxide (NO) release. Taken together, these studies in human hypertensive patients strongly support a role for ET-1 in the pathogenesis of essential hypertension. 30 In addition ET -I may contrib. left venlncular hyp In addition to its effects on the pathogenesis of essential hypertension, ET-t may contribute to the complications of secondary hypertension, including left ventricular hypertrophy, pre-eclampsia, and renal failure. 31 w. Endotheli There app I gulatton of bloc ET-1 and Ang I l I etal..1992. Brur i losing Subpres: neaitny rats (ch sgnifirant nse In predated an Incr t=‘r.!>e.".ensive e‘fe urine volume, or I ‘0 renal actions 0 Several gr re; IV. Endothelin and the Renin-Angiotensin System There appears to be a connection between ET-1 and the RAS in the regulation of blood pressure. It has been established that coadministration of ET-1 and Ang II in rats will produce a synergistic rise in blood pressure (Yoshida et al., 1992; Brunner, 1998). Yoshida et al. first showed this by continuously infusing subpressor doses of ET-1 and Ang II, separately and in combination, in healthy rats (Yoshida et al., 1992). Neither ET-1 nor Ang II alone produced a significant rise in blood pressure, but the combination of these two agents produced an increase of 32% compared to controls (lmai et al., 1992). This hypertensive effect occurred without any changes in body weight, fluid retention, urine volume, or urinary sodium excretion, indicating that this effect was not due to renal actions of ET-1. Several groups have reported that Ang II stimulates the generation and release of ET-1 from endothelial cells (Emori et al., 1991; lmai et al., 1992; Webb and Strachan, 1998; Brunner, 1998). lmai et al. were the first to show that Ang II and arginine vasopressin immediately and dose-dependently induce expression of preproendothelin-1 mRNA in multiple tissues, including endothelial cells (lmai et al., 1992). More recently, Moreau and colleagues demonstrated that long-term treatment with Ang II caused both elevated tissue levels of ET-1 and vascular hypertrophy that was inhibited by ETA receptor blockade (Moreau et al., 1997). Likewise, d’Uscio et al. showed that losartan, an AT1 receptor antagonist, inhibits Ang II induced tissue increases of ET-1 (D’Uscio et al., 1995). Verapamil, a 32 mum channel {I hatATI receptor trough an actor Angll -ET-1 Inte' producton of ET-‘- "$583118th 338085 ET-t production me The flip Side modulate the actlvl Ireeased 2 Stem l Illawaguchi et al.. moment admInIs development of ET All “ Concentrate calcium channel blocker, did not prevent the increased ET-1 levels, suggesting that AT1 receptor antagonists directly modulate tissue levels of ET-1, rather than . through an action on systemic arterial pressure. A functional response of the Ang II — ET-1 interaction was reported by Dohi et al (1992). Ang II stimulated production of ET-1 potentiates norepinephrine-induced contractions in mesenteric arteries of spontaneously hypertensive rats, indicating that vascular ET-1 production may act to amplify the pressor effects of the RAS. The flip side of this Ang II — ET-l relationship is that ET-1 is able to modulate the activity of the RAS. Kawaguchi et al. reported that ACE activity is increased 25-fold in the presence of ET-1 in cultured endothelial cells (Kawaguchi et al., 1991). Similarly, Mortensen and Fink (1992b) showed that concurrent administration of ET—1 and the ACE-inhibitor captopril prevented the development of ET-1-induced hypertension. Surprisingly, an increase in plasma Ang II concentration was not observed in rats receiving the same concentration of ET-1 alone. These findings suggest the potential role of Ang II in ET-1- induced hypertension is played out at a local tissue level, or that one peptide increases the sensitivity of cardiovascular tissues to the other. As discussed earlier, experimental evidence suggests that the role of ET-1 may differ in different forms of hypertension. In severe, salt-sensitive, low-renin models of hypertension, circulating and tissue levels of ET—1 are increased and these models appear to be particularly sensitive to the hypotensive effects of ET- 1 receptor antagonists (Schiffrin, 1998; Haynes and Webb, 1998; Ruschitzka et al., 1999). In contrast, the endothelin system does not appear to be augmented 33 in normalrenin o receptor antagon hypertensron. However, a | l tat are hard to re: administration of i4 d‘Uso'o et al that c Increase of systolic “ Induced hypertenl fled the possum n..} h . I'YI‘IEIIEDSIOTI may 3 Stud yshowed that A vasoconstriCtlon we' antagonists (Rajago leveiopment of Ang anéxed ETA/ETB T95 that endogenous ET in normal-renin or renin-dependent models of hypertension, indicating that ET receptor antagonism might be most effective in RAS-independent models of hypertension. However, a handful of studies have investigated the effects of concomitant administration of Ang II and ET receptor antagonists and have reported results that are hard to reconcile with the above findings. It has been reported by d’Uscio et al. that chronic ETA receptor antagonist therapy attenuates the increase of systolic blood pressure and alterations of endothelial function in Ang II induced hypertension (D’Uscio et al., 1997). Rajagopalan and colleagues raised the possibility that many of the vascular effects seen in Ang II induced hypertension may actually be mediated by endogenously expressed ET-1. Their study showed that Ang II induced hypertension and its associated vasoconstriction were prevented by concomitant adminstration of ETA receptor antagonists (Rajagopalan et al., 1997). Herizi et al. reported that the development of Ang II induced hypertension was totally prevented by bosentan, a mixed ETA/ETB receptor antagonist, thereby further supporting the possibility that endogenous ET-1 contributes to the cardiovascular effects of Ang II (Herizi et al., 1998). In vitro studies have reported that acute exposure to subcontractile concentrations of Ang II potentiates vascular contractile responses to other agonists (Henrion et al., 1992a). This action of Ang II has not been tested using ET-1 as the second agonist, but the strong similarity in signaling mechanisms used by Ang II and ET-1 in vascular smooth muscle (Tsuda et al., 1993) makes it a likely possi on It, have reported the aortic strips from I 1998). A recent st In humans with es s“Oiled that AT, I 95.891 013 corny)”. hYDertensIon 3,, vamnstrlctjgn | 1999), ms ,5 em .eouoe the V3800 This tiWests ’96th anta’aon; a likely possibility. Some studies using Ang II induced models of hypertension have reported markedly attenuated vascular contractile responses to ET-1 in aortic strips from Ang II infused rats (D’Uscio et al., 1997; Rajagopalan et al., 1998). A recent study highlights a potential critical link between Ang II and ET-1 in humans with essential hypertension (Ghiadoni et al., 2000),. This group showed that AT1 receptor blockade with candesartan eliminates the vasodilating effect of a combined ETA/ETB receptor antagonist in patients with essential hypertension. Since it is known that endogenous ET-1 increases vasoconstriction in essential hypertension (Cardillo et al., 1999; Taddei et al., 1999), this is evidence that inhibiting the actions of Ang II at ATI receptors will reduce the vasoconstrictor effect of ET-1 in humans with essential hypertension. This thesis is the first body of work investigating the effect of ET-1 receptor antagonists on the salt-sensitivity of Ang II induced hypertension. 35 V. Hy Tn ET-l Sys‘ Intake Is linemen. 9900gen the 3"“A vtf 'V V. Hypotheses The overall objective of this research project is to examine the role of the ET-1 system in experimental Ang II induced hypertension. Our working hypothesis is that the presence of ET-1, under conditions of increased salt intake, is required for the maintenance of experimental Ang II induced hypertension. This may occur either by Ang II stimulated production of endogenous ET—1, or by an as yet unknown synergistic response resulting from the actions of the two peptides. The following specific hypotheses will be addressed: I. In experimental Ang II induced hypertension, especially under conditions of high salt intake: A. ET-1 activates ETA receptors to increase blood pressure. 8. ET-1 activates ETg receptors to decrease blood pressure. C. ET-1 affects blood pressure by modifying renal sodium and water excretion. D. ET-1 receptor antagonists will cause changes in blood pressure and renal fluid excretion qualitatively similar to those produced by the diuretic drug trichlormethiazide. II. In experimental Ang II induced hypertension, ET-1 affects blood pressure: A. As a result of changes in vascular reactivity to ET-1 produced by angiotensin II, in vitro. B. By altering ET-1 induced vascular contractility in a salt-dependent manner. 37 a?“ i Chapter 2 GENERAL EXPERIMENTAL METHODS I. Animals Male Sprague-Dawley rats (Charles River Laboratories, Portage, MI) weighing 325-450 grams are used in these experiments. Upon arrival at our facility, rats are maintained according to standards approved by the Michigan State University All-University Committee on Animal Use and Care. All experimental procedures are carried out in accordance with the “Guiding Principles in the Care and Use of Animals” of the American Physiological Society. Rats are acclimatized for at least two days prior to any surgical procedures in clear plastic boxes with woodchip bedding. Rats are allowed access to standard rodent chow (Teklad 22/5 Rodent Diet W 8640, Madison, WI) and tap water ad libitum. II. Surgical Procedures All surgical procedures are performed after administration of pentobarbital sodium (Nembutal’, Abbott Laboratories, N, Chicago, IL), 50 mglkg i.p., and atropine sulfate (Sigma, St. Louis, M0), 0.2 mg, i.p. If necessary, anesthesia is supplemented using methohexital sodium (Brevital®, Eli Lilly, Indianapolis, IN), 5- ‘IO mglkg, i.v. Before surgery is initiated, areas to be incised are shaved free of fur and cleaned with disinfectant (Betadine®, Purdue Frederick Co., Norwlk, CT). 38 thong CSTID Inner it 3.933;. aC‘Ia "I{ sang edema "e..“, Normal body temperature is maintained during anesthesia by a water-heated pad (Gorham-Rupp, Inc.). A. Arterial and Venous Catheten'zation The cannulae used in these procedures are constructed of polyvinyl chloride (Tygon® Microbore) tubing with 4.5 cm silicone rubber tips (Dow Corning” Silastic). The femoral vessels are exposed via a 2.0 cm incision in the inner left hind leg of the rat. Small incisions are made in each vessel and approximately 4.5 cm of the arterial and 7.0 cm of the venous catheters are advanced through the vessels into the aorta and vena cava, respectively. The larger arterial catheter is sutured to nearby leg muscle in a way that protects against occlusion of the catheter during normal rat movement. Some rats are additionally catheterized at the facial vein. This procedure involves a 1.0 cm incision in the right side of the animal’s neck, just superior to the jugular pulse point. The facial vein is cleared and catheterized using the same procedure described above for the femoral vessels. The free ends of the catheters are tunneled subcutaneously to the rat’s head, where they are exteriorized by threading them through a stainless steel spring tether. This tether is then anchored to the skull using jeweler's screws and dental acrylic. After the rats have recovered from the anesthesia, they are individually housed in stainless steel metabolism cages for the remainder of the experimental protocol. A minimum of three recovery days is allowed before any experiments are started. 39 ,n’. ‘uuG ureter Is ere BIS-313‘ 35am Some: 8. Surgical Uninephrectom y The skin over the left lateral abdomenal wall is shaved and prepared with an iodine antiseptic cleanser (Betadine‘a, Purdue Frederick 00., Nonivalk, CT). A 1.5 cm vertical incision is made through the skin and underlying muscle just caudal to the rib age with scissors and tissue forceps. The renal vessels and ureter are ligated with 4-0 suture silk (Ethicon‘m, Somerville, NJ). The left kidney is exteriorized and excised with a number 10 scalpel blade (Bard-Parker“, Becton-Dickinson, Franklin Lakes, NJ). The muscle layers and skin are separately closed with 4-0 silk and 4-0 monofilament nylon suture (Ethicon®, Somerville, NJ), respectively. C. Deoxycorticosterone Acetate Implantation A 3 x 1.5 cm rectangular area between the scapulae is shaved and disinfected. Following a 1 cm lengthwise incision, a decxycorticostercne acetate (DOCA) patch is implanted subcutaneously. DOCA implants are prepared by mixing 1 part DOCA (Sigma, St. Louis, MO) with 2 parts silicone rubber (Dow Corning”, Midland, MI) by weight, then curing overnight at room temperature. Surgical placement of implants at 600 mglkg results in a dose of 200 mglkg DOCA. Skin is closed with 40 nylon suture. Butorphanol tartate (Stadol‘m, Bristol Laboratories, Princeton, NJ), 2 mglkg subcutaneous, is given post-operatively for analgesia. Enrofloxacin (Baytril®, Bayer Corp., Shawnee Mission, KA), 5 mglkg s.c., is administered daily for 3 days beginning the day of surgery for bacterial prophylaxis. 4o room wrtl hater ant Ih’lstlfig It meaS-urer OIEtreasm Iterrain ; Ill. Chronic Rat Maintenance and Measurements The metabolism cages housing the rats are located in a climate-controlled room with a 12-hour light-dark cycle. The rats are allowed access to distilled water and either standard rodent chow (Teklad 22l5 Rodent Diet W 8640, Madison, WI) or sodium-deficient rat chow (Teklad TD 170950, Madison, WI) ad libitum. depending on the experimental protocol. The free end of the stainless steel spring tether anchored to the rat’s skull is attached to a plastic hydraulic swivel that will allow the instrumented rat to move freely in its cage without twisting the catheters. This tethering spring also allows for cardiovascular measurements and manipulations to be made without handling the rat, thereby decreasing the amount of animal distress. Catheters are flushed daily to maintain patency and the arterial catheters are filled with a heparinized sucrose solution and occluded when not in use. IV. Hemodynamic Measurements Mean arterial pressure (MAP) and heart rate (HR) are measured via the arterial catheter each morning between 8:00 and 11:00 am. The arterial catheters are connected to external pressure transducers that have first been zeroed at the level of the rat’s heart. The transducers are connected to digital pressure monitors (Digi-Med Blood Pressure Analyzer, Micro-Med, Louisville, KY, USA ) that output directly to a computerized data acquisition and storage system (Digimed System Integrator, Micro-Med, Louisville, KY, USA). Data are collected once every second for 20—30 minutes. The daily value is determined by 41 heav trod the average of the one-second recordings taken over the last five minutes of the recording session. V. Metabolic Measurements Rats received drinking water in graduated cylinders that were refilled each day, allowing for daily measurements of water intake. Rats were housed in metabolism cages designed for urine collection, allowing for daily measurements of urine output. Additionally, daily urine samples were collected and analyzed by ion selective electrodes (Nova Electrolyte 16+ Analyzer, Nova Biomedical, Waltham, MA, USA) for sodium, potassium, chloride, and creatinine levels as indices of renal function. On control day 2 and experimental days 5, 10, and 14, whole blood samples (1 cc) were collected by syringe via the arterial catheters, placed on ice and analyzed by ion selective electrodes ((Nova Electrolyte 16+ Analyzer, Nova Biomedical, Waltham, MA, USA) for sodium, potassium, chloride, BUN, and creatinine levels. Sodium and potassium excretion were calculated by multiplying electrolyte concentration by daily urinary volume. Plasma volumes were determined, via Evan’s Blue dye dilution, approximately every fifth day of the experimental protocols. Evan’s Blue dye binds to albumin in the plasma. The plasma volume measurement involved collecting a 0.5 cc sample of blood by syringe from the rat’s arterial catheter before injection of 0.2 cc Evan’s Blue dye into the catheter and collecting a second 0.5 cc sample 10 minutes after injection of the dye. The samples were then centrifuged and the plasma layer was collected. The plasma samples were 42 read PW sari; resui of the and r “a II-"g-“Ij’l- read on a fluorescent plate reader at 610 nm. The color detected was directly proportional to the concentration of Evan’s Blue in the sample. The control sample reading was subtracted from the post-injection sample reading and the resulting number was compared to a standard curve, allowing for determination of the total plasma volume. Blood volume was calculated from plasma volume and hematocrit using the standard formula. VI. Isolated Tissue Bath Measurements Rats were killed (80 mglkg pentobarbital i.p.) and the superior mesenteric arteries were dissected into helical strips. The endothelium was left intact. Tissues were placed in physiological salt solution containing (mmol/l) 130 NaCl, 4.7 KCl, 1.18 KH2PO4, 1.17 MgSOa~7H20, 1.6 CaCL2-2H20, 14.9 NaHCOa, 5.5 dextrose, and 0.03 CaNa—EDTA. One end of the preparation was attached to a glass rod and the other to a force transducer (model FT03, Grass Instruments, Quincy, MA), and the strip was placed under optimum resting tension (600 mg, as previously determined) and allowed to equilibrate for one hour. Muscle baths were filled with warmed (37°C), aerated (95% 02 -5% C02) physiological salt solution. Changes in isometric force were recorded on a polygraph (Grass Instruments). After the hour of equilibration, arteries were challenged with a maximal concentration of the dI-adrenergic receptor agonist phenylephrine (PE, 10 umoI/I). Tissues were then washed, and the status of the endothelium was examined by observing arterial relaxation to the endothelium-dependent agonist ACh (1 umol/l) in tissues contracted by a half-maximal concentration of PE (~10 nmolll). 43 toll each I VII. The vessels then were incubated with increasing concentrations of PE (10‘ 9 to 10*" M) followed by ET-1 (10'11 to 10‘7 M). The tissues were incubated with each concentration for ~5 minutes before the next concentration was added. In some preparations, superior mesenteric arteries were incubated with Ang II (10'10 M), A-192621 (an ETB selective receptor antagonist, 30nM, Abbott Laboratories, Abbott Park, IL, USA), ABT-627 (an ETA selective receptor antagonist, 30 nM, Abbott Laboratories, Abbott Park, IL, USA), or A-182086 (an ET NB non-selective receptor antagonist, 30 nM, Abbott Laboratories, Abbott Park, IL, USA), for one hour prior to the production of ET-1 dose response curves. VII. Drugs The ETA receptor antagonist, PD156707, was a generous gift from Parke- Davis Pharmaceutical Research (Ann Arbor, MI). The ETA receptor antagonist, ABT-627, the ETB receptor antagonist, A192621, and the non-selective ETA/3 receptor antagonist, A-182086, were generous gifts from Abbott Laboratories (Abbott Park, IL). The Ang II and the thiazide diuretic, trichlormethiazide, were obtained from Sigma Chemical Company (St. Louis, MO, USA). VIII. Statistical Analysis Results are expressed as means i SEM. Mean values were compared statistically using a one-way ANOVA followed by the protected least significant difference test for post hoc comparisons. For in vivo data, within- and between- group differences were analyzed using mixed-design ANOVA. Post hoc 44 oonpansc Within grc Inerence Iaues we Delanete Results c Slghlficar comparisons between groups were performed by testing for simple main effects. Within group comparisons were made using the protected least significant difference test. For in vitro data, estimates of maximum response and E050 values were obtained from each concentration response curve using a four- parameter logistic function. Other statistical procedures are described with the Results of the individual studies. For all analyses, criterion for statistical significance was a probability level of less than 0.05. 45 The efen‘ve 2 Planned lI P'G‘IIOusly C . Il- tartan Chapter 3 DOSE-FINDING STUDIES The purpose of the studies described in this chapter was to establish effective doses and in vivo time courses of the ET—1 receptor antagonists that I planned to use in the main part of my thesis project. These drugs had not been previously well—characterized in vivo, especially in chronic dosing studies. The first antagonist that was available to me for use in these studies was PD156707, a potent but short-acting ETA selective receptor antagonist. I chose to study an ETA selective receptor antagonist first because these receptors had been better characterized than the ETB receptors, arterial constriction was known to be regulated by ETA receptors, and at the onset of this project no selective ETB receptor antagonists had been described in the literature. Subsequent development by Abbott Laboratories of a long-acting and selective ETA antagonist, ABT-627, and the first long-acting and selective ETa antagonist, A- 192621, allowed me to further investigate the potential long-term effects of ET-1 in Ang II induced hypertension. I chose to use A-182086, Abbott Laboratories mixed ETA/ETB receptor antagonist, for my studies rather than the better characterized mixed antagonist, bosentan, in order to study drugs as chemically similar as possible. Unless otherwise noted, all studies were in normal conscious male Sprague-Dawley rats chronically instnrmented for direct, daily measurements of blood pressure and heart rate via catheterization of the femoral artery, femoral vein, and facial vein, and housed as previously described in the 46 general met DOCA-salt 2). general methods (chapter 2). In two of the protocols, drugs were tested in DOCA-salt rats prepared as described in the general methods section (chapter 2). 47 Ellertiven Hypertenl Rationale P015670 {Maguire We needs enOugh tr ”flatten: 0639mm a“: 90m: aterial p - ,J‘Ixoif Effectiveness of PD156707 Tested in Endothelin-1 Induced Model of Hypertension Rationale: At the time this study was conducted, the ETA receptor antagonist, PD156707, had been characterized mainly on the basis of in vitro pharmacology (Maguire et al., 1997). Since our studies all involve whole animal experiments, we needed to demonstrate that this drug would be both potent and efficacious enough to suit our purpose. PD156707 was tested in an ET-l induced model of hypertension. This model is both ET-1 and salt-dependent. We sought to determine the efficacy, time course and dose range of the ETA receptor antagonist that would produce an acute, dose-dependent decrease in the mean arterial pressure in rats receiving ET-1 infusions and high salt intake. Protocol: Some of the rats were maintained on high salt intake (n=8, ~8 mEq/day) and some were maintained on normal salt intake (n-8, ~2 mEq/day). All rats received 8 pmollkglmin ET-1 over a 24 hour time period prior to administration of the ETA receptor antagonist. Three doses of P0156707 (0.3, 0.03, and 0.003 mglkg) were then tested by injecting the drug into the facial vein catheters of the rats (n = 4-6 rats per experimental group). BP and HR were recorded from 10 minutes prior to until 60 minutes post-injection of P0156707. Results: Infusion of ET-1 raised BP only in the high salt intake rats, producing a control mean arterial pressure (MAP) of 144 mmHg in these rats compared to a 48 mntrol M were no 1 (0 003 In lowered by repee Dear. all ”Ilene: he did; allege-r interest. all Ol tl‘r ‘"9 $3?qu control MAP of 106 mmHg in the normal salt intake rats (figures 3.1 & 3.2). There were no signifimnt changes in BP with either the high (0.3 mglkg) or the low (0.003 mglkg) dose of P0156707. However, the 0.03 mglkg dose of PD156707 lowered BP in high salt intake rats by approximately 25 mmHg, as demonstrated by repeated measures ANOVA and Dunnetts multiple comparisons test, with peak effect occurring between 30-50 minutes post-injection. Since the hypertension in these high salt animals was induced by administration of ET-1, the drop in MAP after injection of PD156707 clearly indicates that this dose of the antagonist will reverse the actions of ET—1 mediated by ETA receptors in vivo. Interestingly, the injection of PD156707 also produced a significant tachycardia in all of the rats (data not shown). This response was seen with all three doses of the drug that were tested. 49 ad‘s. e1 2 1 1r . 9 C023). «OI-hut 3.1 figll'e —e— 0.3 NGKG, n=5 —O—- 0.03 WEI/KG, n=5 —A—- 0.003 NEIKG, n=5 10 5 CamuNMP=1441e41 0 ‘Kl l- \l l A fl: Ii \\ .5 4 ‘A A A % \ A MAP (mint-lg) 8 / >/ 451 k . l“ . , - " / C x> 20 lip—l NM” * .25 * * * * * m A __ .IL 5' 1s 30' 45' 60' 11NE(Mnures) Figure 3.1 Effect of PD156707 on endothelin-1 induced hypertension in high salt rats. 50 «WISE» p.332 9 + 0.3 MGIKG, n=5 —0— 0.03 MGIKG, n=5 —A- 0.003 MGIKG, n=5 20 Control MAP = 107.4 i 2.9 15 I _ l A 10 ~ E 5 l A A: E 0 " ‘ ‘/A ”A V O . / ’ ‘ (3;! ‘ <' '10‘ ' \b -15 ‘. -20 . . . . . . . e I . . . I 5' 15' 30' 45' 60' TIME (Minutes) F'QUI‘O 3.2 Effect of PD156707 on endothelin-1 induced hypertension in normal salt rats. 51 Effectii Rahm repent 95:83. ., k by ea: Effectiveness of P0156707 Tested in DOCA-Salt Model of Hypertension Rationale: After demonstrating that PD156707 was efficacious in an acute, ET- dependent model (see above), it was next desirable to test this drug in a well- established chronic model of hypertension. Therefore, PD156707 was tested in the decxycorticostercne acetate salt (DOCA-salt) model of hypertension. DOCA is a mineralocorticoid that acts intracellularly in cells of the renal tubule to cause sodium retention. This model is a chronic volume-expansion model and is RAS- independent (Kenyon et al., 1994). DOCA-salt is considered to be the best established model for studying the role of ET-1 in hypertension, as demonstrated by upregulation of ET-1 in DOCA-salt rats and decrease in BP in response to ET- 1 antagonists in these animals (Brooks et al., 1998). We sought to determine the dose of the ETA receptor antagonist that would produce an acute, dose- dependent decrease in mean arterial pressure in DOCA-salt rats. Protocol: Three doses of PD156707 (0.3, 0.03, and 0.003 mglkg) were tested by injecting the drug into the femoral vein catheters of the rats (n = 4-6 rats per experimental group). BP and HR were recorded from 10 minutes prior to until 60 minutes post-injection of PD156707. Results: In the DOCA-salt model, PD156707 produced a dose-dependent d9(3l‘ease in blood pressure (~12 mmHg) with a peak effect between 30-50 52 m, mutes r epea‘e 0" k ’ ays a \n m r . r T ecen pt: minutes (figure 3.3). This was a significant drop in BP, as demonstrated by repeated measures ANOVA and Dunnetts multiple comparisons test. Since ET-1 plays an established role in DOCA-salt hypertension, and PD156707 lowered BP in this model, we conclude that the antagonist is efficacious at blocking ETA receptors over the dose range used here. 53 «DIEEVn‘d‘S—Q i 9% 3.3 in + 0.3 MGIKG, n=5 —0— 0.03 MGIKG, n=5 —A— 0.003 MGIKG, n=5 :2 Control MAP = 183.6 i 13,6 T 10 ., T T T A AK 3 5 ‘ NM»). i A“ “H V '5 _ \. j; “ e w ‘5 a .. , 4 -10 ~ ‘.» s‘ Q -15 ‘ * * * .20 .4 * * * * -25 5' 15' 30' 45' 60' TIME (Minutes) Figure 3.3 Effect of PD156707 on DOCA-salt hypertension. Rapid ' Rationa P0156] 1 indUCi an: gor ntagor VESSUFE lamina: 91' ESSuFe We cops (6033” p : l en'eptOfs Rapid Termination of Endothelin-1 Infusion Rationale: In our first study, we showed that a moderate dose (0.03 mglkg) of PD156707 was sufficient to produce a significant decrease in BP in rats with ET- 1 induced hypertension, but that this decrease was delayed 30-40 minutes after antagonist injection. To demonstrate that this drop in BP was truly due to antagonism of ET receptor activity, we examined the time course of blood pressure fall after simply stopping the EM infusion. We hypothesized that termination of the ET-1 infusion would produce an identical time course of blood pressure fall to the administration of a moderate dose of PD156707 (0.03 mglkg). We considered cessation of ET-1 infusion to be roughly equivalent to ET—1 receptor antagonism because of the known tenacious binding of ET-1 to its receptors. Protocol: All of the rats were maintained on high salt intake (n=8, ~8 mEq/day). All rats received 8 pmollkglmin ET-1 over a 24 hour time period prior to administration of the ETA receptor antagonist. At the onset of the timed experiment, the ET—1 infusion was abruptly terminated. BP and HR were recorded from 10 minutes prior to until 60 minutes after stopping the ET—1 infusion. 55 RESuh ef‘m ~th iecree (zabie Soppe {22:39 1 antago ET; rec Results: Terminating ET-1 infusion resulted in a decrease in BP with a peak effect between 30-50 minutes after stopping the ET-1 infusion (figure 3.4). This decrease in BP closely paralleled the decrease seen after injection of PD156707 (table 3.1). An increase in HR was observed soon after the ET—1 infusion was stopped, similar to the tachycardia observed after administration of PD156707 (table 3.2). These results support the hypothesis that the ETA receptor antagonist, PD156707, acts primarily by preventing the actions of ET-1 at the ETA receptor. I I I I 1 1 :Ev GLJWQQLD. ..2LQHL4‘ :50: HQUre ; ‘in‘. H» +n= .3 \l O .x G O 150 - 140 - 130 - 120 “ 110 Mean Arterial Pressure (mm Hg) OI 10' j 20' j 30' 40' 50' 60' Time (minutes) Figure 3.4 Effect of cessation of endothelin-1 infusion on mean arterial pressure (mm Hg) in endothelin-1 induced hypertension. 57 O) C? ; /&T/S/c%/3_/c°5/&>:’/8§/e>f/za/’ot log/C3 I . 145. 156. 143. 151. 140. 143. 1 140. 136. 136. 135. 134. 1 142. 132. 1431 131. 139. 133. 1371 1 132. 136. 135. 136. 137. Table 1: Comparison of the change in mean arterial pressure (mmHg) over time with endothelin-1 infusion cessation and injection of PD156707 58 344. 353. 362.1 383. 363.1 355. 397. 382. 37 366.1 389. rate over endothelin-1 infusion cessation and injection of PD156707 59 Autonomic Blockade and PD156707 Rationale: The unexpected tachycardia observed with both administration of PD156707 and removal of exogenous ET—1 posed the question of whether or not this response was a function of blocking ET-1 activity or baroreflex-mediated sympathetic activation and parasympathetic withdrawal. To further investigate this phenomenon, we utilized two drugs known to antagonize actions of the autonomic nervous system (ANS) on the heart. Atropine is a competitive antagonist for muscarinic receptors and thereby blocks parasympathetic nervous system-mediated bradycardia (Brown and Taylor, 1996). Propranolol is a nonselective competitive is-adrenergic receptor antagonist whose actions include blockade of cardiac [31 receptors, resulting in decreased sympathetic actions on HR (Hoffman and Lefi—— lliglt saiH:+ llrlg IL n:-6 -—<>—— lligltlsait CCInthIL n==6 + Norm alsalt+ Ang II, n=6 c o -—£>—— hlorun al:salt ccintrtih n==5 :3 3(30 0 “ .25l) - ‘- ‘- c 200 ~ 0 e c o o ‘15ll a ‘ 0 ‘ ‘A—A I“ 100 « a ' A i O :2 5° Z. 0 .. In as ’A "'3" -11 -10 -9 -8 -7 I: 0 log ET-1(M) :3 2l)0 0 u 3 15¢) 4 C o 0 10¢) n ur a 50 " $ 0 .. -9 -8 -7 -6 -5 Hag FIE (M ) Figure 7.4 Concentration dependent contraction to endothelin-1 (ET-1) and phenylephrine (PE) in superior mesenteric artery from normotensive control and hypertensive angiotensin II (Ang II) infused rats on normal and high salt intake. Values are mean SE; n= number of animals. For ET-1, there were no differences in ECso values between groups. In normal salt rats, maximal responses to ET-1 were significantly greater in the group that received chronic angiotensin II (Ang II) infusion. For PE, there were no significant differences in ECso values or maximal responses between the groups. 165 infused rats on high salt intake had significantly decreased maximum response to ET-1 (123.4 i 35.4 % PE contraction) compared to controls (245.6 i 41.7 % PE contraction; p=0.0263). There was no significant difference in maximal contraction to ET-1 between high salt and normal salt control groups (p=0.141). There were no significant differences in the E050 values between the four groups in response to ET-1. 166 DISCUSSION The overall goal of my research is to define the mechanisms responsible for the salt-sensitivity of Ang II induced hypertension. Recent work by others implicates ET-1 in the acute and chronic pressor effects of Ang II. Specifically, it has been proposed that Ang II can interact with ET—1 in at least two ways. First, Ang II has been shown to increase endothelial cell synthesis and release of ET-1 both in vitro (Emori et al., 1991; Dohi et al., 1992; Imai et al., 1992; Kohno et al., 1992) and in vivo (Barton et al., 1997; Moreau et al., 1997; Lariviere et al., 1998; Ferri et al., 1999). Second, Ang II administered in vitro potentiates the vascular contractile response to other agonists (Henrion et al.,1992). Endogenous or infused Ang II administered in vivo also augments contractile responses to other agonists (Qui et al., 1994; Dowell et al.,1996). I recently demonstrated that blockade of ETA receptors in rats receiving chronic intravenous infusions of Ang II causes a larger and more sustained fall in arterial pressure when the rats were on a high versus a normal salt intake (Chapter 4). This result led me to conclude that ET-1 could participate in the mechanism of salt-sensitivity in Ang II induced hypertension. The goal of the current studies was to test the hypothesis that the salt-sensitivity of Ang II induced hypertension is due in part to amplification of the vascular contractile effects of ET-1. The main new finding from this work is that chronic infusion of Ang II alters vascular reactivity to ET-1 in a salt-dependent fashion: in rats on normal salt intake maximum contractile responses to ET-1 are increased, while in rats on high salt intake they are decreased. 167 In vitro studies have reported that short-term exposure (30-60 minutes) to very low (subthreshold for direct contraction) concentrations of Ang II potentiate vascular contractile responses to other agonists (Henrion et al., 1992a). The mechanism of this effect is not fully elucidated, but probably involves activation of protein kinase C in the vascular smooth muscle cell (Henrion et al., 1992b). This action of Ang II has not been tested with ET-1 as the second agonist, but the strong similarity in signaling mechanisms used by Ang II and ET-1 in vascular smooth muscle (Tsuda et al., 1993) makes it a likely possibility. Furthermore, there is evidence from chronic (6 day) in vivo experiments in rats that the pressor actions of ET-1 are potentiated by concomitant exposure to Ang II (Yoshida et al., 1992). Another study, though, failed to find such an effect during rapid, bolus injections of the two agonists in the canine coronary circulation (Kiss et al., 1998). My data do not support the idea that contractile effects of ET-1 are amplified after short-term exposure to Ang II in vitro, at least in the superior mesenteric artery of the rat. I observed no significant change in the concentration-response curve to ET-1 in arteries pre-exposed for one hour to Ang II. Pressor responses, and changes in hindlimb vascular resistance, to acute bolus injections of Ang II in vivo are reported to be reduced by prior blockade of ET-1 receptors, especially when low doses of Ang II are administered (Balakrishnan et al., 1996; Champion et al., 1998). This suggests that physiological amounts of ET-1 may amplify the vascular contractile response to Ang II in vivo. I performed experiments to investigate this phenomenon in rats on 168 normal and high salt intake. My results confirm that pressor responses to acute (2 hour) exposure to low amounts of Ang II in vivo are significantly reduced by prior blockade of ETA receptors. This effect appears to be specific for Ang II since no change in the pressor response to phenylephrine infusion was seen after administration of the ETA antagonist. One interpretation of these results is that Ang II infusion for 2 hours stimulated the release of ET-1 from arterial endothelial cells, and that this ET—1 accounted of the accompanying rise in arterial pressure. Most studies, however, have failed to find evidence for ET-1 release by Ang II in short-term infusion protocols (Klein et al., 1995; Delemarre et al., 1998), including an investigation in humans on differing levels of salt intake (Ferri et al., 1999). Thus, I interpret the results to indicate that physiological amounts of ET-1 acting at ETA receptors amplify the pressor actions of exogenous Ang II. My data, however, do not provide any insight into the mechanism of this interaction. It is notable though that this short-term effect was observed in rats on both normal and high salt intake, and therefore is not likely to alone explain the salt-sensitivity of chronic Ang II induced hypertension. In a final experiment, I evaluated vascular reactivity to ET-1 in vitro in superior mesenteric arteries from rats that received chronic (7 day) infusions of Ang II. Interestingly, I found divergent results depending on whether the rats were on normal or high salt intake. Rats on high salt intake became significantly more hypertensive than rats on normal salt intake, as has been described many times previously. Furthermore, maximal response of mesenteric arteries to ET-1 from these rats were significantly suppressed compared to responses in control 169 rats on high salt intake. Similar results have been reported by other investigators using a model of chronic Ang II induced hypertension involving infusion of higher doses of Ang II (200 nglkglmin, subcutaneously) in rats on normal salt intake (Rajagopalan et al., 1997; D’Uscio et al., 1997). Arteries from those rats exhibited a marked increase in preproET-1 gene expression and ET-1 peptide content (Rajagopalan et al., 1997; D’Uscio et al., 1997), and there was a strong negative correlation between reactivity to ET-1 and arterial peptide concentrations (D’Uscio et al., 1997). It has been suggested that a decrease in mesenteric response to ET-1 in vitro is a consequence of ETA receptor down- regulation caused by chronic increase in ET—1 release (Nguyen et al., 1992). Although I did not measure arterial content of ET-1 in my studies, I propose that a similar mechanism could account for depressed maximal responses to ET-1 in superior mesenteric arteries from rats in my study on high salt intake and Ang II infusion. Mesenteric arteries from rats receiving an infusion of Ang II for 7 days but maintained on a fixed normal salt intake exhibited a significantly increased maximum response to ET-1 in vitro. To my knowledge, this is the first report of potentiation by exogenous Ang II of in vitro contractile responses to ET—1 in vascular smooth muscle, although others have reported that Ang II amplifies the bronchoconstrictor actions of ET-1 via a leukotriene-dependent pathway (Pitt and Nally, 1999). As shown in figure 6.1, contraction of superior mesenteric arteries to ET-1 is mediated exclusively through ETA receptors, and Ang II has been reported to increase ETA receptor expression (Hatakeyama et al., 1994). Thus, 170 one potential explanation for my results is that chronic exposure to Ang II increased ETA receptor number in superior mesenteric arteries in rats on normal salt intake. This still does not explain, however, why the effects of chronic Ang II infusion on contraction of superior mesenteric arteries to ET-1 in vitro were different in rats on high versus normal salt intake. My experiments do not provide a definitive answer. I speculate that long-term exposure to Ang II can upregulate both ETA receptor number and preproET-1 gene expression. Though high salt intake alone does not stimulate vascular ET-1 formation (D’Uscio et al., 1997; Barton et al., 1998; Ikeda et al., 1999), my data are consistent with the idea that high salt intake plus Ang II may be a more effective stimulus to preproET-1 gene expression and ET-1 synthesis than Ang II alone. One possible mechanism could involve the effects of Ang II and high salt intake on endothelial nitric oxide (NO) action. Long-term exposure to Ang II in vivo (D’Uscio et al., 1997), and to high salt intake (Boegehold, 1995), are reported to impair NO activity in resistance arteries. There is also evidence that increased NO activity suppresses ET-1 formation in blood vessels (Boulanger and Liischer, 1990). The combination of Ang II and high salt could produce a larger increase in vascular ET-1 synthesis than Ang II alone because of less NO-mediated inhibition. In support of this idea, it has been shown that administration of the NO synthase inhibitor, L-nitro—arginine methyl ester, to rats on normal salt intake caused a highly significant potentiation of the chronic pressor responses to Ang II infusion in rats (Melaragno and Fink, 1996). Finally, there is evidence that the 171 pressor (Mortensen and Fink, 1992) and vascular resistance (Grossman et al., 1990) effects of ET-1 are increased by high salt intake. Therefore, enhanced synthesis of ET-1 in blood vessels in rats on high salt intake receiving Ang II, combined with some amplification of the pressor effect of ET-1 by high salt alone, could explain the larger contribution of ET-1 to Ang II induced hypertension under high salt conditions. Direct evidence for this theory needs to be obtained in future experiments. In summary, Ang II was shown to cause changes in pressor and vascular contractile effects of ET-1 that are dependent on time of exposure to Ang II, and on salt intake. During long-term infusion Ang II can increase both vascular reactivity to ET-1 and ET-1 formation. Each of these mechanisms may contribute to the dependence of Ang II induced hypertension on ETA receptor activation, but high salt intake apparently shifts the balance toward the latter. 172 Chapter 8 General Discussion A large proportion of the population of the modem westemized countries will develop hypertension during their lifetimes (Kaplan, 1998c; Mohrman and Heller, 1991). High blood pressure is now second only to upper respiratory infections as an indication for office visits to physicians in the United States (Woodwell, 1997). Hypertension is the most pervasive modifiable risk factor for cardiovascular disease (Kannel and Wilson, 1999) and heart disease is the major killer of people living in modernized societies. The high prevalence of hypertension, it’s robust impact on the incidence of heart disease, and it’s potential to be treated justify the continued quest of physicians, biomedical researchers, and health officials to develop a better understanding of this deceivingly complex medical disorder. Hypertension is defined simply as a chronic elevation of arterial blood pressure above 140/90 mmHg (JNC-V, 1997). Approximately 95% of diagnosed cases of hypertension are attributed to essential hypertension, or increased blood pressure without an obvious cause (Deshmukh et al., 1998). There are some universally accepted contributing factors to essential hypertension (Mohrman and Heller, 1991). Among these are environmental factors (stress, smoking, high salt/ high fat diet, sedentary lifestyle), genetic influences, structural changes in the heart and peripheral blood vessels, alterations in neural control of blood pressure, and renal defects. 173 There are also several known hormonal regulators of blood pressure, which include Ang II and ET-1. These two peptide hormones affect blood pressure both individually and in tandem, under both acute and chronic conditions. Ang II and ET-1 can each cause hypertension As was discussed in chapter 1, Ang II affects blood pressure by a myriad of mechanisms, including direct vasoconstriction, activation of the sympathetic nervous system, release of aldosterone from the adrenal gland, direct stimulation of renal sodium and water retention, and cardiac and vascular hypertrophy. The importance of Ang II as an etiologic factor in human hypertension is undisputed. ET-1 modulates local vascular tone and structure by both ETB receptor mediated release of vasodilating substances (namely NO and prostacyclin) from the endothelial cells, and by ETA receptor mediated strong, long-lasting constriction of the vascular smooth muscle cells. The renal actions of ET-1 are more controversial and could serve to either increase or decrease arterial pressure. Recent evidence strongly supports an important role for ET-1 in the pathogenesis of human hypertension. Salt intake contributes to severity of Ang II and ET-1 induced hypertension It has long been established that infusion of Ang II at low doses causes a more severe hypertension in subjects on high salt intake than in subjects on normal salt intake (Muirhead et al., 1975; Kanagy et al., 1990; Simon, 1998). 174 The magnitude of the hypertension in this model is dependent on the rate of Ang II infusion, the salt intake, and the duration of the infusion. In some studies, an initially subpressor infusion rate caused a gradual rise in arterial blood pressure after several days (refs). This salt sensitive model of hypertension was used in most experiments in this thesis project. Although my protocols involved identical rates and duration of Ang II infusion, the differences in blood pressure response between rats on normal and high salt intake were not identical between studies, even though the high salt rats always showed a larger pressor response. I cannot fully account for this variable effect of salt intake, but possibilities include variation between litters or seasonal effects. It also has been clearly demonstrated that hypertension induced by chronic ET—1 infusion is salt-dependent (Mortenson and Fink, 1990). While in many other forms of salt-sensitive hypertension the role of endogenous Ang II in blood pressure control is reduced, the contribution of ET-1 is enhanced. Increased ET-1 plasma levels have been reported in human patients with salt- sensitive hypertension, particularly African Americans and the elderly (Ferri et al., 1998; Ergul et al., 1996). Furthermore, ET-1 is now an accepted etiologic factor in several common salt-sensitive experimental models of hypertension, including DOCA-salt, Dahl salt-sensitive, one-kidney-one—clip Goldblatt (1 K1 C) and chronic renal failure (Schiffrin, 1999). 175 ET-1 mediates some of the cardiorenal effects of Ang II As was discussed in chapter 1, Ang II stimulates the synthesis and release of ET-1 from the endothelial cells and increases renal ET-1 formation. Moreover, ET-1 amplifies the pressor effects of Ang II and partially mediates Ang II induced vascular hypertrophy. ET-1 receptor blockade begun prior to the start of chronic Ang II infusion prevents the development of Ang II induced hypertension. Thus, Ang II and ET-1 may interact to increase blood pressure by renal as well as vascular mechanisms. Ang II induced hypertension is the best model to study the interaction between Ang II and ET-1 The overall objective of my thesis project was to examine the interaction between Ang II and ET—1 in salt-sensitive hypertension. Several experimental models exist to study the role of the RAS in hypertension. The two-kidney-one- clip Goldblatt model (2K1 C) is the best characterized renin-dependent model of hypertension. Most studies show that ET-1 antagonists do not signifiwntly affect blood pressure in this model (Sventek, 1996). A problem with using this model for studying possible ET-1 mediated effects of Ang II on blood pressure is that ET-1 can affect other components of the RAS besides Ang II. For example, ET—1 activates ACE (Kawaguchi et al., 1991) and can decrease renin secretion (Berthold et al., 1999). Additionally, I was interested in studying salt-sensitive hypertension and blood pressure in this model is not affected by salt intake. 176 Therefore, I chose to use an experimental model that is both salt-dependent and unequivocally caused by the actions of Ang II. ET-1 receptor antagonists reveal ET-1 mediated effects in vivo and in vitro The main experimental strategy I chose to study the interaction between Ang II and ET-1 in salt-sensitive hypertension was measurement of hemodynamic and renal responses to pharmacological antagonists in vivo. This strategy was supplemented by use of in vitro studies of vascular contractility. Other possible experimental approaches to addressing my overall hypothesis could have included measurements of ET-1 peptide concentrations, preproET-1 gene expression, ECE activity, or ET-1 receptor expression in vasculature and other target tissues in vivo and in vitro. Genetically manipulated strains of rodents lacking key components of the ET-1 system also could have been employed for in vivo and in vitro studies. A potential weakness of my pharmacological strategy would be a lack of either efficacy or selectivity of the ET-1 antagonists. I attempted to minimize these possibilities by extensively characterizing the efficacies (acutely and chronically) and selectivities of the antagonists l employed in my main studies. One outcome of these preliminary studies was to employ an ETA selective receptor antagonist in addition to PD156707 as an experimental tool. This decision was based on concerns about the effectiveness of PD156707, probably related to it’s very short plasma half-life. In the cases of ABT-627, A-192621 177 and A-182086, my preliminary data indicated these dnigs had adequate effects and receptor selectivity for use in the in vivo studies. ET-1, especially under conditions of increased salt intake, is required for the maintenance of experimental Ang II induced hypertension The data obtained in my thesis project is integrated with previous findings in figure 8.1 to generate a new model representing the interrelationships between salt-intake, Ang II, and ET-1 in the pathogenesis of hypertension. This model focuses on actions of Ang II and ET-1 on the vasculature, and on the kidney. Each peptide has other potential targets that could exert an influence on short- or long-term control of blood pressure. These were not investigated in my studies and therefore are not included in the model. My study, and the model, were predicated on the assumption that minute-to-minute control of MAP is primarily achieved by changing vascular smooth muscle tone, whereas long-term steady- state blood pressure levels are determined by both vascular mechanisms and the natriuretic and diuretic functions of the kidney (Hall, 1986). Accordingly, inferences about ET-1 mediated vascular mechanisms were derived from both in vitro contractile studies, and from acute responses to ET—1 receptor antagonists in vivo. Similarly, inferences about ET-1 mediated changes in sodium and water balance were derived from steady-state changes in blood pressure and sodium excretion during chronic administration of ET-1 receptor antagonists. 178 179 180 The main new findings from my research that are incorporated into the model are: vascular pressor effects mediated by ETA receptor activation are amplified by Ang II, but not high salt intake; renal pressor effects mediated by ETA receptor activation are amplified by the combination of high salt intake and Ang II; vasodilation mediated by ETB receptor stimulation is amplified by high salt intake; Ang II modifies the response to vascular ETB receptor activation in a salt- dependent manner; and renal effects of ETB receptor activation are not significantly influenced by Ang II (Table 8.1). These findings will be discussed in more detail in the following sections. 181 182 Vascular Mechanisms Renal Mechanisms Ang II Salt Ang II Salt ETA pressor response I (Tl) -- (I) II ETB depressor response ii 183 Ang II induced hypertension depends primarily on the actions of ET-1 on ETA receptors, and this effect is greater under conditions of high salt intake I chose to investigate first the effect of ETA receptor activation on Ang II induced hypertension because this receptor subtype was the best characterized at the onset of this thesis project. ETA receptors were known to be expressed highly in the vascular smooth muscle cells and kidney, but not in endothelial cells (lnagami et al., 1993; Haynes and Webb, 1998), suggesting an important role for this receptor in acute and chronic blood pressure control. In vivo work in humans demonstrated that endogenous ET-1 confers basal vasoconstrictor tone via ETA receptor activation (Webb et al., 1998). Most importantly, studies in humans with essential hypertension revealed increased ETA mediated vasoconstriction compared to normotensive controls (Cardillo et al, 1999). In a later study, this increased ETA mediated vasoconstriction was linked to activation of Ang II AT1 receptors (Ghiadoni et al., 2000). Previous work by others showed that specific blockade of ETA receptors significantly attenuated the development of Ang II induced hypertension (D’Uscio et al., 1997; Rajagopalan et al., 1997). However, none of these earlier studies addressed the potential effects of salt intake, or the effects of ETA receptor activation on chronic sodium and water excretion. In this thesis, I present the novel findings that selective ETA receptor blockade lowered MAP in Ang II induced hypertension within one hour of drug administration, whereas the dmg had no effect on blood pressure in rats not 184 receiving Ang II. This effect was not modified in rats on differing salt intakes. l interpreted these results to indicate that chronic infusion of Ang II increased vascular ETA receptor activation, independent of salt intake. In related studies using a two-hour intravenous infusion protocol, pressor responses to Ang II were significantly inhibited by prior blockade of ETA receptors in rats on both high and normal salt intake. There are at least two possible mechanisms to explain these data. Ang II could increase ET-1 tissue concentrations and/or increase vascular reactivity to endogenous ET—1. The former has been demonstrated in earlier studies of Ang II induced hypertension and was not directly evaluated in my work. The latter mechanism was investigated further in a series of in vitro vascular reactivity studies. In rat superior mesenteric arteries, where I showed that ET-1 mediated contraction is due entirely to ETA receptor activation, vascular contractile responses to ET-1 were dramatically affected by chronic Ang II infusion. In arteries from rats on high salt intake and Ang II, vascular contractile responses to ET-1 were significantly reduced compared to high salt controls. Conversely, in arteries from rats on normal salt intake and Ang II, vascular contractile responses to ET-1 were significantly augmented compared to normal salt controls. Salt alone did significantly affect ET-1 induced contractile responses. The precise reasons for this differential response to Ang II are not known. Based on previous work, chronic Ang II infusion has at least three important effects on the arterial vasculature: increased ET-1 synthesis, increased ETA receptor number, and decreased nitric oxide activity (Hatakeyama et al., 1994; D’Uscio et al., 1997; 185 Rajagopalan et al., 1997; D’Uscio et al., 1998). Thus, the expected ET-1 contractile response of arteries from Ang II infused rats would be the result of the net effect of these three changes. Increased ET-1 content in arteries would be expected to decrease contractile responses to exogenous ET-1 due to ETA receptor downregulation (D’Uscio et al., 1997). Increased ETA receptor number would be expected to increase ET-1 contractile responses. Diminished NO activity would be expected to increase contractile responses to most agonists, including ET-1. However, a long-term suppression of NO activity would also be expected to increase arterial ET-1 synthesis (Boulanger and Li‘ischer, 1990). High salt intake alone does not affect vascular ET-1 synthesis (Ikeda et al, 1995; D’Uscio et al., 1997; Barton et al., 1998). The influence of high salt intake on ETA receptor number is not known. However, high salt intake has been shown to decrease NO activity in arterial resistance vessels (Boegehold, 1995). l speculate that Ang II infusion during high salt intake shifts the balance of these three vascular effects in favor of increased ET-1 synthesis, and thus diminished vascular reactivity to exogenous ET-1 because of ETA receptor downregulation. I further speculate that, under normal salt conditions, the balance of the effects produced by Ang II favor increased ETA receptor number and diminished NO mediated vasodilation. Thus, vascular responsiveness to ET-1 would be increased. 1 Previous studies showed increased ET-1 synthesis in the kidneys of rats with Ang II induced hypertension (Barton et al., 1997; Barton et al., 1998). In my rats receiving long-term Ang II infusion, chronic treatment with ABT—627 186 completely normalized blood pressure within one day. This effect was well- maintained over the 5 day treatment period in rats on high salt intake, but gradually subsided over 5 days in rats on normal salt intake. In control rats on either salt intake, chronic ETA receptor blockade had no effect on blood pressure or sodium excretion. This suggests that chronic Ang II infusion shifts the pressure-natriuresis relationship to a higher pressure level in part by activation of renal ETA receptors and that this effect is greater on high salt intake. Consistent with my results, in hypertensive Dahl salt-sensitive rats ETA receptor blockade also caused a shift in the pressure-natriuresis relationship to a lower pressure level (Kassab et al., 1998). This effect of chronic renal ETA receptor blockade could be due to either inhibition of ETA receptor mediated renal hemodynamic effects or to unopposed ETB receptor mediated natriuresis and diuresis. The second possibility was addressed in additional studies in my thesis project, and may provide an explanation for the increased chronic antihypertensive effect of the ETA receptor antagonist under high salt conditions. The initial hypotensive effect of ETA receptor blockade in Ang II induced hypertension is not due to a diuretic effect A comparison of the time course and antihypertensive effects of ETA receptor blockade and thiazide diuretic treatment in Ang II induced hypertension was performed to determine if the primary antihypertensive response to ETA receptor blockade was caused by effects on the vasculature or the kidney. My study of the antihypertensive effect of trichlormethiazide in Ang II induced 187 hypertension served as a positive control for the effects of ETA receptor blockade. The effect of a diuretic drug had not been previously tested in Ang II induced hypertension, and thus the time course and blood pressure effects of this treatment were not known. The reduction in blood pressure resulting from ETA receptor blockade occurred rapidly and was associated with a tendency for increased sodium and water balance during the first 24 hours. This pattern of response closely resembles that seen with chronic administration of minoxidil, a direct vasodilator, to rats with Ang II induced hypertension (Melaragno and Fink, 1996b). On the other hand, the peak blood pressure lowering effect of the thiazide diuretic required 72 hours and was preceded by a large loss of sodium and water. Thus, it is clear that ETA receptor antagonism does not lower blood pressure initially in Ang II induced hypertension via a diuretic effect on the kidney. ETB receptor activation opposes Ang II induced hypertension In contrast to ETA receptor mediated events, ETB receptor activation tends to decrease MAP by causing release of endothelial vasodilators and promoting renal loss of sodium and water. Furthermore, recent studies showed that a naturally occurring deletion of the ETB receptor gene (Gariepy et al., 2000) and chronic blockade of ETB receptors cause salt-sensitive hypertension in rats (Pollock, 2000). Previous reports demonstrated that ETB receptor activation opposes increases in MAP in both human essential hypertension (Cardillo et al., 1999) and in DOCA-salt rats (Pollock et al., 2000). 188 E My thesis work was the first investigation of the effects of a selective ETB receptor antagonist in Ang II induced hypertension. My findings, in agreement with those from other models of hypertension, showed that ETB receptor activation opposes the rise in MAP seen in Ang II induced hypertension. Moreover, selective blockade of ETB receptors resulted in a significant increase in MAP within minutes of the initiation of drug therapy (suggesting a vascular mechanism) in all four groups of rats studied, regardless of salt intake or Ang II infusion. It should be noted, however, that the increases in MAP were largest in rats receiving high salt intake, suggesting high salt diet enhances ETB receptor mediated vasodilation. The mechanism for this salt-dependent difference in ETB mediated vasodilation is unknown. None of my in vitro vascular reactivity experiments were designed to investigate ETB mediated vascular effects. Future studies could be conducted to investigate the effects of S6c, the ETB selective receptor agonist , on resistance arteries from rats maintained on varying salt intakes. Possible explanations for an apparent increase in ETB mediated vasodilation in rats on high salt intake include: increased ETB receptor number or affinity for ET-1, increased vascular cyclooxygenase activity, and increased vascular reactivity to either NO, PGI2, or other endothelial-derived vasodilators. An important new finding of my thesis research was that Ang II appears to attenuate ETB receptor mediated vasodilation in rats on high salt intake, yet augment this effect in rats on normal salt intake. The former effect plays a major role in promoting hypertension in rats receiving chronic Ang II infusion and high 189 salt intake. Conversely, the ability of Ang II to increase ETB mediated vasodilation probably plays a role in preventing hypertension in rats on normal salt intake. The mechanism for this salt-dependent difference in the effects of Ang II on ETB receptor mediated vasodilation is unknown. This could be addressed in future studies by investigating the vascular contractile responses to S6c of resistance arteries from rats receiving both chronic Ang II infusions and varying salt intakes. Possible explanations for the effects of Ang II on ETB receptor mediated vasodilation in high salt rats are that: combined effects of high salt and Ang II to decrease NO activity in resistance arteries could impair ETB mediated vasodilation, which is mediated in part by NO; or, combined effects of high salt and Ang II could increase ET-1 synthesis, which would lead to downregulation of ETB receptors. A possible explanation for the effects of Ang II on ETB receptor mediated vasodilation in normal salt rats is increased ETB receptor number or affinity. Ang II was shown to increase ETB receptor expression in cardiac myocytes In vitro (Kanno et al., 1993). Combined blockade of both ETA and ETB receptors with A-182086 produced a decrease in MAP that was presumably due to the net effect of inhibiting both ETA mediated vasoconstriction and ETB mediated vasodilation. Since the overall result of combined ET-1 receptor blockade was a slight drop in blood pressure only in rats receiving chronic Ang II infusion, it can be assumed that the ETA mediated vasoconstriction predominates over ETB mediated vasodilation in Ang II induced hypertension. This is supported by my findings that both the in vivo pressor and the in vitro vascular contractile effects are 190 exclusively due to ETA receptor activation. The acute depressor response to combined ET-1 receptor blockade in rats receiving Ang II was presumably larger in high salt rats than in normal salt rats because ETA mediated vasoconstriction is similar regardless of salt intake, whereas ETB mediated vasodilation is significantly greater in rats on normal salt intake. Published studies show chronic Ang II infusion increases renal ET-1 content (Barton et al., 1998), while the effect of salt intake on renal ET-1 formation is controversial (Firth et al., 1995; Melo et al., 1998; Morita et al., 1999). As discussed above, it is likely that renal actions mediated by ETA receptors contribute to the sustained increase in MAP observed in rats with Ang II induced hypertension. ETB receptor activation has been reported to cause natriuresis and diuresis (Kohan et al., 1993; Clavell et al., 1995). My data suggest that ETB receptor mediated natriuretic and diuretic effects are not influenced by chronic Ang II infusion. This is somewhat surprising in view of evidence that Ang II infusion increases renal ET-1 synthesis (Barton et al., 1998). Other work indicates that high salt intake increases ETB receptor mediated natriuretic and diuretic effects (Pollock, 2000; Gariepy et al, 2000), and my data tend to support this idea. The mechanism of the effect is not known, but could involve increased renal NO activity secondary to high salt intake (Schultz and Tolins, 1993). Natriuretic responses elicited by ETB receptor stimulation are mediated in part by NO (Hoffman et al., 2000). As noted earlier in this discussion, increased ETB receptor mediated natriuretic and diuretic effects could 191 account for the relatively better antihypertensive efficacy of selective ETA receptor antagonism in high versus normal salt rats. Conclusions Ang II induced hypertension is a well-established salt-sensitive model of hypertension. ETA selective receptor antagonists have been shown to lower blood pressure in salt-sensitive models of hypertension (Schiffrin, 1999) and to prevent the development of Ang II induced hypertension (D’Uscio et al., 1997; Rajagopalan et al., 1997). The data presented in this thesis show that selective ETA receptor blockade alone normalizes MAP in Ang II induced hypertension, especially under conditions of high salt intake, presenting a strong case that ET-1 acting at ETA receptors is an important mechanism of the salt-sensitivity of Ang II induced hypertension. Conversely, decreased ETB receptor function has been shown to increase MAP in other models of hypertension (Pollock et al., 1999; Matsumura et al., 2000; Cardillo et al., 2000). My thesis work was the first to show that this is also true in Ang II induced hypertension. Furthermore, the depressor actions of ETB receptor stimulation in the vasculature and the kidney are increased by high salt intake. Thus, combined blockade of both ETA and ETB receptors is less effective at lowering blood pressure than selective ETA receptor antagonism under conditions of high salt intake. An implication of this conclusion is that selective ETA receptor blockade should be more effective than combined ET-1 receptor blockade at lowering blood pressure in salt-sensitive subgroups of human 192 hypertensive patients, such as African Americans and the elderly. Similarly, combined ET-1 receptor antagonists should be most effective in subgroups that are salt-resistant or on a low-salt diet. 193 References Abbott Laboratories, personal communication, 1998. Ando K, Sato Y, Fujita T. Salt sensitivity in hypertensive rats with angiotensin II administration. Am J Physiol. 1990; 259: R1012-R1016. Ando K, Sato Y, lto Y, Ogata E, Fujita T. Effect of salt loading on aldosterone response to long-term infusion of angiotensin II in rats. J Cardiovasc Pharmacol. 1991; 17: 386-389. Bader M, Paul M, Femandez-Alfonso M, Kaling M, Ganten D. Renin-angiotensin system. A: Molecular biology of the renin-angiotensin system. In: Swales JD, ed. Textbook of Hypertension. Oxford: Blackwell Scientific Publications; 1994: 214-232. Balakrishnan SM, Wang HD, Gopalakrishnan V, Wilson TW, McNeill JR. Effect of endothelin antagonist on hemodynamic responses to angiotensin II. Hypertension. 1996; 28: 806-809. Banks RO, Pollack DM, Novak J. The renal and systemic hemodynamic actions of endothelin. In: Highsmith, RF, ed. Endothelin: Molecular Biology, Physiology, and Pathology. New Jersey: 1998; 167-188. Barton M, Shaw S, D’Uscio LV, Moreau P, LUSChBI' TF. Angiotensin II increases vascular and renal endothelin-1 and functional endothelin converting enzyme activity in vivo: role of ETA receptors for endothelin regulation. Biochem Biophys Res Comm. 1997; 238: 861 — 865. Barton, M., 8. Shaw, L.V. D’Uscio, P. Moreau, and TF. Luscher. Differential modulation of the renal and myocardial endothelin system by angiotensin II in vivo. Effects of chronic selective ETA receptor blockade. J Cardiovasc Pharmacol. 1998; 31: $265-$268. Baylis C. Acute interactions between endothelin and nitric oxide in the control of renal haemodynamics. Clin Exp Pharmacol Physiol. 1999; 26: 252-257. Berthold H, Munter K, Just A, Kirchheim HR, Ehmke H. Stimulation of the renin- angiotensin system by endothelin subtype A receptor blockade in conscious dogs. Hypertension. 1999; 33(6): 1420-1424. Bigaud M, Pelton JT. Discrimination between ETA and ETB receptor-mediated effects of endothelin-1 and [Ala1'3'11'151endothelin-1 by 80123 in the anesthetized rat. BrJ Pharmacol. 1992; 107: 912-918. 194 Boegehold MA. F low-dependent arteriolar dilation in normotensive rats fed low- or high-salt diets. Am J Physiol. 1995; 269: H1407-H1414. Boulanger CM, Lt'Jscher TF. Release of endothelin from the porcine aorta: inhibition by endothelium-derived nitric oxide. J Clin Invest. 1990; 85: 587-590. Brody MJ, Vamer KJ, Vasquez EC, Lewis SJ. Central nervous system and the pathogenesis of hypertension. Hypertension. 1987; 18(5): lll7-lll12. Brooks DP, Jorkasky DK, Freed Ml, Ohlstein EH. Pathophysiological role of endothelin and potential therapeutic targets for receptor antagonists. In: Highsmith, RF, ed. Endothelin: Molecular Biology, Physiology, and Pathology. New Jersey: 1998; 223-268. Brown AJ, CasaIa-Stenzel J, Gofford S, Lever AF, Morton JJ. Comparison of fast and slow pressor effects of angiotensin II in the conscious rat. Am J Physiol. 1981; 241(10): H381-H388. Brown JH, Taylor P. Muscarinic receptor agonists and antagonists. In: Hardman JG, Limbird LE, eds. Goodman & Gilman’s The Pharmacological Basis of Therapeutics, Ninth Edition. New York: 1996; 141-160. Brunner HR. Endothelin inhibition as a biologic target for treating hypertension. Am J Hypertens. 1998; 11: 1038-1098. Burt VL, Whelton P, Roccella EJ, Brown C, Cutler JA, Higgins M, Horan MJ, Labarthe D. Prevalence of hypertension in the US adult population: Results from the Third National Health and Nutrition Examination Survey, 1988-1991. Hypertension. 1995; 25(3): 305-313. Campese VM. Salt sensitivity in hypertension: Renal and cardiovascular implications. Hypertension. 1994; 23: 531-550. Cardillo, 0., CM. Kilcoyne, M. Waclawiw, R.O. Cannon III, and J.A. Panza. Role of endothelin in the increased vascular tone of patients with essential hypertension. Hypertension. 1999; 33: 753-758. Champion HC, Estrada LS, Estrada LN, Filep JG, Kadowitz PJ. Analysis of effects of bosentan (Ro 47-0203), a non-peptide endothelin ETA/ETB receptor antagonist, in the hindlimb vascular bed of the cat. Can J Physiol Pharmacol . 1998; 76: 141-147. Chapleau, MW. Arterial baroreflexes. In: Taubert, KA, executive ed. Izzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 83-86. 195 Clavell, A., A Stingo, K Margulies, R. Brandt, and J. Burnett. Role of endothelin receptor subtypes in the in vivo regulation of renal function. Am J Physiol. 1995; 268: F455-F460. Clozel M, Breu V, Gray GA, Kalina B, Loffler B-M, Burri K, Cassal J-M, Hirth G, Muller M, Neidhardt W, Ramuz H. Pharmacological characterization of bosentan, a new potent orally active nonpeptide endothelin receptor antagonist. J Pharmacol Exp Ther. 1994; 270 (1): 226-235. Clozel M, Gray GA, Breu V, Loffler B-M, Osterealder R. The endothelin ETB receptor mediates both vasodilation and vasoconstriction in vivo. Biochem Biophys Res Comm. 1992; 186: 867-873. Coleman TG, Hall JE. Systemic and regional blood flow regulation. In: Taubert, KA, executive ed. lzzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 79-82. Cowley AW, Jr., Roman RJ. The role of the kidney in hypertension. JAMA. 1996; 275(20): 1581 -1 589. Cristol J-P, Warner TD, Thiemerrnann C, Vane JR. Mediation via different receptors of the vasoconstriction effects of endothelins and sarafotcxins in the systemic circulation and renal vasculature of the anesthetized rat. Br J Pharmacol. 1993; 108: 776-779. Csiky, B., and G. Simon. Synergistic vascular effects of dietary sodium supplementation and angiotensin II administration. Am J Physiol. 1997; 273: H1275-H1282. Cushman, WC. Hypertension in the elderly. Curr Opin Cardiol. 1994; 9: 561- 567. D’uscio LV, Moreau P, Shaw S, Takase H, Barton M, Lt‘ischer TF. Effects of chronic ETA-receptor blockade in angiotensin Il-induced hypertension. Hypertension. 1997; 29(part 2): 435-441. D’Uscio LV, Shaw 8, Barton M, Luscher TF. Losartan but not verapamil inhibits angiotensin ll-induced tissue endothelin-1 increase: Role of blood pressure and endothelial function. Hypertension. 1995; 31: 1305-1310. Delemarre FM, de Jong D, Didden MA, de Jong PA. Effect of angiotensin infusion on plasma endothelin in pregnancy. Eur J Obstet Gynecol Reprod Biol. 1998; 77: 33-35. 196 Department of Health and Human Services. Healthy People 2000: National Health Promotion and Disease Prevention Objectives for the Nation. Washington DC: Public Health Service; 1991. Deshmukh R, Smith A, Lilly LS. Hypertension. In: Lilly LS, ed. Pathophysiology of Heart Disease; Second edition. Baltimore: Williams and Wilkins; 1998: 267- 288. Dohi Y, Hahn AWA, Boulanger CM, Buhler FR, Li'ischer TF. Endothelin stimulated by angiotensin II augments contractility of spontaneously hypertensive rat resistance arteries. Hypertension. 1992; 19: 131-137. Dowell FJ, Henrion D, Benessiano J, Poitevin P, Levy B. Chronic infusion of low- dose angiotensin II potentiates the adrenergic response in vivo. J Hypertens. 1996; 14: 177-182. Dustan HP, Valdes G, Bravo E, Tarazi RC. Excessive sodium retention as a characteristic of salt-sensitive hypertension. Am J Med Sci. 1986; 292: 67-74. Edwards RM, Stack EJ, Pullen M, Nambi P. Endothelin inhibits vasopressin action in rat inner medullary collecting duct via the ETB receptor. J Pharmacol Exp Ther. 1993; 267: 1028-1033. Ely D, Caplea A, Dunphy G, Daneshvar H, Turner M, Milsted A, Takiyyudin M. Spontaneously hypertensive rat Y chromosome increases indexes of sympathetic nervous system activity. Hypertension. 1997; 29(2): 613-618. Emori T, Hirata Y, Ohta K, Kanno K, Eguchi S, Imai T, Shichiri M, Marumo F. Cellular mechanism of endothelin-1 release by angiotensin and vasopressin. Hypertension. 1991; 18: 165-170. Ergul S, Parish CD, Puett D, Ergul A. Racial differences in plasma endothelin-1 concentrations in individuals with essential hypertension. Hypertension. 1996; 28: 652-655. Farquharson CA, Struthers AD. Spironolactone increases nitric oxide bioactivity, improves endothelial vasodilator dysfunction, and suppresses vascular angiotensin II angiotensin II conversion in patients with chronic heart failure. Circulation. 2000; 101(6): 594-597. Ferri C, Bellini C, Desideri G, Giuliani E, De Siati L, Cicogna S, Santucci A. Clustering of endothelial markers of vascular damage in human salt-sensitive hypertension: Influence of dietary sodium load and depletion. Hypertension. 1998; 32: 862-868. 197 Ferri C, Desideri G, Baldoncini R, Bellini C, Valenti M, Santucci A, De Mattia G. Angiotensin II increases the release of endothelin-1 from human cultured endothelial cells but does not regulate its circulating levels. Clin Sci (Colch). 1999; 96: 261-270. Firth, J., K Schricker, p. Ratcliffe, and A. Kurtz. Expression of endothelins 1 and 3 in the rat kidney. Am J Physiol. 1995; 269: F522-F528. F lack JM, Yunis C. Ethnicity and socioeconomic status in hypertension. In: Taubert, KA, executive ed. lzzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 153-155. Force, T. Mechanisms of endothelin-induced mitogenesis in vascular smooth muscle. In: Highsmith, RF, ed. Endothelin: Molecular Biology, Physiology, and Pathology. New Jersey: 1998; 121-166. Frishman WH. ls-Adrenergic Blockers. In: Taubert, KA, executive ed. lzzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 297-300. Fukuroda T, Ozaki S, Ihara M, lshikawa K, Yano M, Nishikibe N. Synergistic inhibition by BQ-123 and BQ-788 of endothelin-1-induced contractions of the rabbit pulmonary artery. BrJ Pharmacol. 1994; 113: 336-338. Gariepy, C.E., T. Ohuchi, S.C. \Mlliams, J.A. Richardson, and M. Yanagisawa. Salt-sensitive hypertension in endothelin-B receptor-deficient rats. J Clin Invest. 2000; 105(7): 925-933. Gavras H. Angiotensin-Converting Enzyme Inhibitors. In: Taubert, KA, executive ed. lzzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 309-310. Geisterfer AA, Peach MJ, Owens GK. Angiotensin II induces hypertrophy, not hyperplasia, of cultured rat aortic smooth muscle cells. Circ Res. 1985; 62: 749- 756. Graham RM. a-Adrenergic blockers. In: Taubert, KA, executive ed. lzzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 300-303. Gray GA. Generation of endothelin. In: Gray GA, Webb DJ, eds. Molecular Biology and Pharmacology of the Endothelins. Austin: R.G. Landes; 1995: 13- 32. 198 Griendling KK Alexander RW. Renin-Angiotensin System. C: Cellular mechanisms of angiotensin II action. In: Swales JD, ed. Textbook of Hypertension. Oxford: Blackwell Scientific Publications; 1994: 244-253. Grossman E, Hoffman A, Keiser HR. Sodium intake modulates renal vascular reactivity to endothelin-1 in Dahl rats. Clin Exp Pharmacol Physiol. 1990; 17: 121-128. Gurbanov K, Rubinstein I, Hoffamn A, Abassi A, Better O, Winaver J. Differential regulation of renal regional blood flow by endothelin-1. Am J Physiol. 1996; 271: F1166-72. Guyton AC, Langston JB, Navar G. Theory for renal autoregulation by feedback at the juxtaglomerular apparatus. Circ Res. 1964; 14/15(supp 1): l187—l197. Guyton AC. Abnormal renal function and autoregulation in essential hypertension. Hypertension. 1991; 18(5): llI49-III53. Hall JE, Guyton AC, Smith MJ, Coleman TG. Blood pressure and renal function during chronic changes in sodium intake: Role of angiotensin. Am J Physiol. 1980; 239: F271-F280. Hall JE. Control of Na+ excretion by angiotensin II: Intrarenal mechanism and blood pressure regulation. Am J Physiol. 1986; 250: R960-R972. Hatakeyama H, Miyamori l, Yamagishi S, Takeda Y, Takeda R, Yamamoto H. Angiotensin II upregulates the expression of type A endothelin receptor in human vascular smooth muscle cells. Biochem Mol Biol Int 1994; 34: 127-134. Haynes WG, Webb DJ. Endothelin as a regulator of cardiovascular function in health and disease. J Hypertens. 1998; 16: 1081 -1098. Henrion D, Laher I, Laporte R, Bevan JA. Angiotensin II amplifies arterial contractile response to norepinephrine without increasing Ca++ influx: role of protein kinase C. J Pharmacol Exp Ther. 1992a; 26: 835-840. Henrion D, Laher I, Laporte R, Bevan JA. Further evidence from an elastic artery that angiotensin II amplifies noradrenaline-induced contraction through activation of protein kinase C. Eur J Pharmacol. 1992b; 224: 13-20. Herizi A, Jover B, Bouriquet N, Mimran A. Prevention of the cardiovascular and renal effects of angiotensin II by endothelin blockade. Hypertension. 1998; 31(part 1): 10-14. 199 Hickey KA, Rubanyi GM, Paul RJ, Highsmith RF. Characterization of a coronary vasoconstrictor produced by cultured vascular endothelial cells. Am J Physiol. 1985; 248: C550-C556. Hoffman A, Abassi ZA, Brodsky S, Ramadan R, Winaver J. Mechanisms of big endothelin-1 diuresis and natriuresis: role of ET(B) receptors. Hypertension. 2000; 35(3): 732-739. Hoffman 88, Lefkowitz RJ. Catecholamines, sympathomimetic drugs, and adrenergic receptor antagonists. In: Hardman JG, Limbird LE, eds. Goodman & Gilman’s The Pharmacological Basis of Therapeutics, Ninth Edition. New York: 1996; 249-264. Howard PG, Plumpton C, Davenport AP. Anatomical localization and pharmacological activity of mature endothelins and their precursors in human vascular tissue. J Hypertens. 1992; 10: 1379-1386. Ikeda M, Kohno M, Takeda T. Endothelin production in cultured mesangial cells of spontaneously hypertensive rats. Hypertension. 1995; 25: 1196-1201. Imai T, Hirata Y, Emori T, Yanagisawa M, Masaki T, Marumo F. Induction of endothelin-1 gene by angiotensin and vasopressin in endothelial cells. Hypertension. 1992; 19: 753-757. lnagami, T. Endothelin. In: Taubert, KA, executive ed. lzzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 33-34. lnoue A, Yanagisawa M, Kimura S, Kasuya Y, Miyauchi T, Goto K, Masaki T. The human endothelin family: Three structurally and pharrnacologically distinct isopeptides predicted by three separate genes. Proceedings of the National Academy of Science, USA. 1989; 86: 2863-2867. INTERSALT Cooperative Research Group. INTERSALT: an international study of electrolyte excretion and blood pressure: Results for 24 hour urinary sodium and potassium excretion. Br Med J. 1988; 297: 319-328. Izzo, JL, Jr. The sympathetic nervous system in human hypertension. In: Taubert, KA, executive ed. Izzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 109-112. Jackson EK, Garrison JC. Renin and angiotensin. In: Hardman JG, Limbird LE, eds. Goodman 8. Gilman’s The Pharmacological Basis of Therapeutics, Ninth Edition. New York: 1996; 733-758. 200 F‘— Johnson RJ, Fink GD, Galligan JJ. Mechanisms of endothelin-induced venoconstriction in isolated guinea pig mesentery. J Pharmacol Exp Ther. 1999; 289: 762-767. Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. The sixth report of the joint national committee on prevention, detection, evaluation, and treatment of high blood pressure (JNC V). National Institutes of Health: National Heart, Lung, and Blood Institute: National High Blood Pressure Education Program. NIH Publication No. 98-4080. 1997. Kanagy, N.L., C.M. Pawloski, and GD. Fink. Role of aldosterone in angiotensin II induced hypertension in rats. Am J Physiol. 1990; 259: R102-R109. Kannel WB, Vifilson PWF. Cardiovascular risk factors and hypertension. In: Taubert, KA, executive ed. Izzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 199-202. Kanno K, Hirata Y, Tsujino M, Imai T, Shichiri M, Ito H, Marumo F. Up-regulation of ETB receptor subtype mRNA by angiotensin II in rat cardiomyocytes. Biochem Biophys Res Commun. 1993; 194(3): 1282-1287. Kaplan NM. Salt and blood pressure. In: Taubert, KA, executive ed. lzzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 167-169. Kaplan, NM. Primary hypertension: pathogenesis. In: Clinical Hypertension, 7‘h edition. Williams and Wilkins Publications; 1998b; 41-99. Kaplan, NM. Renal parenchymal hypertension. In: Clinical Hypertension, 7th edition. Williams and Wilkins Publications; 1998a; 281-299. Kassab, 8., MT. Miller, J. Novak, J. Reckelhoff, B. Clower, and JP. Granger. Endothelin-A receptor antagonism attenuates the hypertension and renal injury in Dahl salt-sensitive rats. Hypertension. 1996; 31: 397-402, 1998. Kawaguchi H, Sawa H, Yasuda H. Effect of endothelin on angiotensin converting enzyme activity in cultured pulmonary artery endothelial cells. J Hypertens. 1991; 9: 171-174. Kenyon CJ, Morton JJ. Experimental models of hypertension. 8: Experimental steroid-induced hypertension. In: Swales JD, ed. Textbook of Hypertension. Oxford: Blackwell Scientific Publications; 1994: 494-500. 201 Kiss P, Horvath l, Szokodi I, Toth P, Kekesi V, Juhasz-Nagy A, Toth M. Endothelin does not interact with angiotensin II in the coronary vascular bed of anesthetized dogs. J Cardiovasc Pharmacol 1998; 31: S103—S-105. Klein H, Abassi Z, Keiser HR. Effects of angiotensin II and phenylephrine on urinary endothelin in normal female volunteers. Metabolism. 1995; 44: 115- 118. Kohan, D.E., E. Padilla, and AK Hughes. Endothelin 8 receptor mediates ET- 1 effects on cAMP and PGE2 accumulation in rat IMCD. Am J Physiol. 1993; 265: F670-F676. Kohno M, Yokokawa K, Horio T, Yasunari K, Murakawa K, Takeda T. Atrial and brain natriuretic peptides inhibit the endothelin-1 secretory response to angiotensin II in porcine aorta. Circ Res 1992; 70: 241-247. Koletsky S, Rivera-Velez JM, Pritchard WH. Production of hypertension and vascular disease by angiotensin. Arch Pathol. 1966; 82: 99-106. Kotchen TA. To salt, or not to salt? Am J Physiol. 1999; H1807-H1810. Krieger JE, Roman RJ, Cowley AW. Hemodynamics and blood volume in angiotensin II salt-dependent hypertension in dogs. Am J Physiol. 1989; 257: H1402-H1412. LaCroix AZ. Gender effects and hypertension in women. In: Taubert, KA, executive ed. Izzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 150-153. Lariviere R, Lebel M, Kingma l, Grose JH, Boucher D. Effects of losartan and captopril on endothelin-1 production in blood vessels and glomeruli of rats with reduced renal mass. Am J Hypertens. 1998; 11: 989-997. Li 0, Dale WE, Hasser EM, Blaine EH. Acute and chronic angiotensin hypertension: Neural and nonneural components, time course, and dose dependency. Am J Physiol. 1996; 271: R200-R207. Lombardi D, Gordon KL, Polinksy P, Suga S, Schwartz SM, Johnson RJ. Salt- sensitive hypertension develops after short-term exposure to angiotensin II. Hypertension. 1999; 33: 1013-1019. Luft FC, Wilcox CS, Unger T, Kuhn R, Demmert G, Rohmeiss P, Ganten D, Sterzel RB. Angiotensin-induced ypertensionin the rat: Sympathetic nerve activity and prostaglandins. Hypertension. 1989; 14: 396-403. 202 Lund-Johansen P. Central haemodynamics in essential hypertension at rest and during exercise. J Hypertens. 1989; 7(supp 6): $52-$55. Lund-Johansen P. Haemodynamics of essential hypertension. In: Swales JD, ed. Textbook of Hypertension. Oxford: Blackwell Scientific Publications; 1994: 61-76. Maguire JJ, Kuc RE, Davenport AP. Affinity and Selectivity of PD156707, A novel nonpeptide endothelin antagonist, for human ETA and ETB receptors. J Pharmacol Exp Ther. 1997; 280(2): 1102-1108. Manhem PJ, Clark SA, Brown WB, Murray GD, Robertson JI. Effect of chlorothiazide on serial measurements of exchangeable sodium and blood pressure in spontaneously hypertensive rats. Clin Sci. 1985; 69 (5): 511-515. Matsumura, Y., T. Kuro, F. Konishi, M. Takaoka, C.E. Gariepy, and M. Yanagisawa. Enhanced blood pressure sensitivity to DOCA-salt treatment in endothelin ET(B) receptor-deficient rats. BrJ Pharmacol. 2000; 129(6): 1060- 1062. Melaragno MG, Fink GD. Enhanced slow pressor effect of angiotensin II in two- kidney, one-clip rats. Hypertension. 1996a; 25; 288-293. Melaragno MG, Fink GD. Inhibition of the slow pressor effect of angiotensin II contributes to the antihypertensive effect of angiotensin-converting enzyme inhibitors in renovascular hypertension. J Pharrnalcol Exp Ther. 1996b; 278: 297-303. Melo, L.G., A.T. Beress, C.K. Chong, S.C. Pang, T.G. Flynn, and H. Sonnenberg. Salt-sensitive hypertension in ANP knockout mice: potential role of abnormal plasma renin activity. Am J Physiol. 1998; 274: R255-R261. Miyamori l, Takeda Y, Yoneda T, Takeda R. Endothelin release from mesenteric arteries of spontaneously hypertensive rats. J Cardiovasc Pharmacol. 1991; 17 (suppl 7): $408-$410. Miyauchi T, Masaki T. Pathophysiology of endothelin in the cardiovascular system. Ann Rev Physiol. 1999; 61: 391 -41 5. Moreau P, d’Uscio LV, Shaw S, Takase H, Barton M, LIJischer TF. Angiotensin II increases tissue endothelin and induces vascular hypertrophy: Reversal by ETA receptor antagonist. Circulation. 1997; 96: 1593-1597. 203 Morita, H., H. Kurihara, Y. Kurihara, T. Kuwaki, T. Shindo, Y. Oh-hashi, M. Kumada, and Y. Yazaki. Response of blood pressure and catecholamine metabolism to high salt loading in endothelin-1 knockout mice. Hypertens Res. 1999; 22: 11-16. Mortensen LH, Fink GD. Captopril prevents chronic hypertension produced by infusion of endothelin-1 in rats. Hypertension. 1992; 19: 676-680. Mortensen LH, Fink GD. Salt-dependency of endothelin-induced, chronic hypertension in conscious rats. Hypertension. 1992; 19: 549-554. Mortensen LH, Pawloski CM, Kanagy NL, Fink GD. Chronic hypertension produced by infusion of endothelin in rats. Hypertension. 1990; 15: 729-733. Muirhead EE, Leach BE, Davis JO, Armstrong FB, Pitcock JA, Brosius WL. Pathophysiology of angiotensin-salt hypertension. J Lab Clin Med. 1975; 85(5): 734-745. Murphy TJ, Alexander RW, Griendling KK, Runge MS, Bernstein KE. Isolation of a cDNA encoding the vascular type I angiotensin II receptor. Nature. 1991; 351: 233-236. National Heart, Lung, and Blood Institute. Fact Book Fiscal Year 1996. Bethesda, MD: US. Department of Health and Human Services, National Institutes of Health; 1997. Nguyen PV, Parent A, Deng LY, Fluckiger JP, Thibault G, Schiffrin EL. Endothelin vascular receptors and responses in decxycorticostercne acetate- salt hypertensive rats. Hypertension. 1992; 19: Il-98-ll-104. Oates, JA. Antihypertensive agents and the drug therapy of hypertension. In: Hardman JG, Limbird LE, eds. Goodman & Gilman’s The Pharmacological Basis of Therapeutics, Ninth Edition. New York: 1996; 781-808. Ohuchi T, Kuwaki T, Ling GY, Dewit D, Ju KH, Onodera M, Cao WH, Yanagisawa M, Kumada M. Elevation of blood pressure by genetic and pharmacological disruption of the ETB receptor in mice. Am J Physiol. 1999; 276: R1071-R1077. Opgenorth TJ, Adler AL, Calzadilla SV, Chiou WJ, Dayton BD, Dixon DB, Gehrke LJ, Hernandez L, Magnuson SR, Marsh KC, Novodas El, Von Geldem TW, Wessale JL, Vifinn M, Wu-Wong JR. Pharmacological characterization of A- 127722: An orally active and highly potent ETA-selective receptor antagonist. J Pharmacol Exp Ther. 1996; 276: 473-481. 204 Perry HM, Jr. Central and peripheral sympatholytics. In: Taubert, KA, executive ed. lzzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 306-308. Pitt CM, Nally JE. Angiotensin ll-mediated potentiation of endothelin-1-evoked bronchial contractions: a role for Ieukotrienes? Pulm Pharmacol Ther. 1999; 12:7-12. Plato CF, Garvin JL. Nitric oxide, endothelin and nephron transport: Potential interactions. Clin Exp Pharmacol Physiol. 1999; 26: 262-268. Plumpton C, Champeney R, Ashby MJ, Kuc RE, Davenport AP. Characterization of endothelin isoforms in human heart: Endothelin-2 demonstrated. J Cardiovasc Pharmacol. 1993; 22(supp 8): $26-$28. Pollock, D.M. High salt intake increases the hypertension produced by chronic ETB receptor blockade in Sprague-Dawley rats. FASEB J. 14(4): A132 (abstract), 2000. Pollock, D.M., G.H. Allcock, A. Krishnan, B.D. Dayton, and J6. Pollock. Upregulation of endothelin B receptors in kidneys of DOCA-salt hypertensive rats. Am J Physiol Renal Physiol. 1999; 278(2): F279-286. Pollock, DM. Endothelin receptor subtypes and tissue distribution. In: Highsmith, RF, ed. Endothelin. New Jersey: Humana Press Inc; 1998; 1-29. Potter GS, Johnson RJ, Fink GD. Role of endothelin in hypertension of experimental chronic renal failure. Hypertension. 1997; 30: 1578-1584. Puschett JB. Diuretics. In: Taubert, KA, executive ed. lzzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 294-296. Qui Hy, Henrion D, Levy Bl. Endogenous angiotensin II enhances phenylephrine-induced tone in hypertensive rats. Hypertension. 1994; 24: 317- 321. Rajagopalan S, Laursen JB, Borthayre A, Kurz S, Keiser J, Haleen S, Giaid A, Harrison 06. Role for endothelin-1 in angiotensin lI-mediated hypertension. Hypertension. 1997; 30(part1): 29-34. Ramsay LE, Yeo WW, Chadwick IG, Jackson PR. Diuretics. In: Textbook of Hypertension, ed. JD Swales. Oxford, Blackwell Scientific Publishing. 1994; 1046-1058. 205 Robertson JIS, Nicholls MG, eds. The renin-angiotensin system: Volume one: biochemistry and physiology. Gower Medical Publishing; 1993. Rubanyi GM, Polokoff MA. Endothelins: Molecular biology, biochemistry, pharmacology, physiology, and pathophysiology. Pharmacol Rev. 1994; 46 (3): 325-415. Ruschitzka F, Corti R, Noll G, Luscher TF. A rationale for treatment of endothelial dysfunction in hypertension. J Hypertens. 1999; 17 (suppl 1): S25- S35. Ruschitzka F, Noll G, Shaw S, Luscher TF. Endothelin and endothelin receptor antagonists in cardiovascular disease. J Hypertens. 1998; 16(suppl 8): S13- $23. Safar ME, London GM. Arterial and venous compliance in sustained essential hypertension. Hypertension. 1987; 10: 133-139. Sakurai T, Yangisawa M, Takuwa Y, Miyazaki H, Kimura S, Goto K, Masaki T. Cloning of a cDNA encoding a non-isopeptide selective subtype of the endothelin receptor. Nature. 1990; 348: 732-735. Sasaki K, Yamano Y, Bardhan S, Iwai N, Murray JJ, Hasegawa M. Cloning and expression of a complementary DNA encoding a bovine adrenal angiotensin II type-1 receptor. Nature. 1991; 351: 230-232. Sato Y, Ogata E, Fujita T. Role of chloride in angiotensin lI-induced salt- sensitive hypertension. Hypertension. 1991; 18: 622-629. Schiffrin E. Endothelin and endothelin antagonists in hypertension. J Hypertens. 1998; 16: 1891-1895. Schiffrin EL. Role of endothelin-1 in hypertension. Hypertension 1999; 34: 876- 881. Schultz PJ, Tolins JP. Adaptation to increased dietary salt intake in the rat. Role of endogenous nitric oxide. J Clin Invest. 1993; 91: 642-650. Semchenko A, Grauer K, Curry CL, Gums J. American Academy of Family Physicians. Management of uncontrolled hypertension. American Family Physician Monograph No. 2. 1998. Sharifi AM, Schiffrin EL. Apoptosis in aorta of decxycorticostercne acetate salt Hypertensive rats - Effect of endothelin receptor antagonism. J Hypertens. 1997; 15: 1441-1448. 206 Simon G, llIyyes, Csiky 8. Structural vascular changes in hypertension: Role of angiotensin II, dietary sodium supplementation, blood pressure, and time. Hypertension. 1998; 32: 654-660. Sirvio ML, Metsarinne K, Saijonmaa S, Fyrquist F. Tissue distribution and half- life of 125l-endothelin in the rat: Importance of pulmonary clearance. Biochem Biophys Res Comm. 1990; 167: 1191-1195. Sventek P, Turgeon A, Garcia R, Schiffrin EL. Vascular and cardiac overexpression of endothelin-1 gene in one-kidney one-clip goldblatt hypertensive rats but only in the late phase of two-kidney one-clip goldblatt hypertension. J Hypertens. 1996; 14: 57-64. Taddei S, Virdis A, Ghiadoni L, Sudano I, Notari M, Salvetti A. Vasoconstriction to endogenous ET-1 is increased in the peripheral circulation of patients with essential hypertension. Circulation. 1999; 100: 1680-1683. Takanohashi A, Tojo A, Kobayashi N, Yagi S, Matsuoka H. Effect of trichlormethiazide and captopril on nitric oxide synthase activity in the kidney of decxycorticostercne acetate-salt hypertensive rats. Jpn Heart J. 1996; 37(2): 251 -259. Takayanagi R, Ohnaka K, Liu W, Ito T, Nawata H. Molecular biology of endothelin-converting enzyme (ECE). In: Highsmith, RF, ed. Endothelin. New Jersey: Humana Press Inc; 1998; 75-92. Taubes G. The (Political) Science of Salt. Science. 1998; 281: 898-907. The osteopathic medicine’s caring cooperative. Living with high blood pressure: It’s in your control. American Osteopathic Association and SmithKIine Beecham Pharmaceuticals. 1996: 1-6. Tiret L, Poirier O, Hallet V, McDonagh TA, Morrison C, McMurray JJV, Dargie HJ, Arveiler D, Ruidavets J-B, Luc G, Evans A, Cambien F. The Lys198Asn polymorphism in the endothelin-1 gene is associated with blood pressure in overweight people. Hypertension. 1999; 33: 1169-1174. Tsuda T, Griendling KK, Ollerenshaw JD, Lassegue 8, Alexander RW. Angiotensin II and endothelin induced protein phosphorylation in cultured vascular smooth muscle cells. J Vasc Res. 1993; 30: 241 -249. Warner TD, Elliott JD, Ohlstein EH. California dreamin’ ‘bout endothelin: Emerging new therapeutics. Trends Pharmacol Sci. 1996; 17: 177-181. Webb DJ, Monge JC, Rabelink TJ, Yanagisawa M. Endothelin: New discoveries and rapid progress in the clinic. Trends Pharmacol Sci. 1998; 19(1): 5-8. 207 Webb DJ, Strachan FE. Clinical experience with endothelin antagonists. Am J Hypertens. 1998; 1 1: 71 S-7QS. Weinberger MH. Salt sensitivity. In: Taubert, KA, executive ed. lzzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 89-90. Weir MR. Calcium entry blockers. In: Taubert, KA, executive ed. lzzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 311-314. Whelton PK, He J, Klag, MJ. Blood pressure in westemized populations. In: Swales JD, ed. Textbook of Hypertension. Oxford: Blackwell Scientific Publications; 1994: 11-21. Williams, GH. Hypertensive vascular disease. In: Isselbacher KJ, Braunwald E, Wilson JD, Martin JB, Fauci AS, Kasper DL, eds. Harrison’s Principles of lntemal Medicine; Thirteenth edition. New York: McGraw-Hill, Inc; 1994; 1116- 1131. Wyss, JM. Sympathetic nervous system abnormalities in hypertension. In: Taubert, KA, executive ed. lzzo JD Jr., Black HR. ed. Hypertension Primer. American Heart Association Council on High Blood Pressure Research; 1993: 106-108. Yanagisawa M, Kurihawa H, Kimura S, Tomobe Y, Kobayashi M, Mitsui Y, Yazaki Y, Goto K, masaki T. A novel potent vasoconstrictor produced by vascular endothelial cells. Nature. 1988; 332: 411-415. Yoshida K, Yasujima M, Kohzuki M, Kanazawa M, Yoshinaga K, Abe K. Endothelin-1 augments pressor response to angiotensin II infusion in rats. Hypertension. 1992; 20: 292-297. 208