THE ROLE OF ENDOTHELIN IN THE MULTfflORMONAL REGULATION OF FLUID RETENTION IN CONGESTIVE HEART FAILURE by Wendy H. Y. Wong B.Sc, The University of British Columbia, 1990 A THESIS SUBMITTED IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF MASTER OF SCIENCE in THE FACULTY OF GRADUATE STUDIES (Department of Medicine) We accept this thesis as conforming ^to the requiretLjstandard THE UNIVERSITY OF BRITISH COLUMBIA March 1996 © Wendy H. Y. Wong, 1996 In presenting this thesis in partial fulfilment of the requirements for an advanced degree at the University of British Columbia, I agree that the Library shall make it freely available for reference and study. I further agree that permission for extensive copying of this thesis for scholarly purposes may be granted by the head of my department or by his or her representatives. It is understood that copying or publication of this thesis for financial gain shall not be allowed without my written permission. Department of M pci \ f-i n c The University of British Columbia Vancouver, Canada Date April ^ . iqq^ DE-6 (2/88) Abstract Abnormal fluid retention is characteristic of congestive heart failure (CHF). The mechanisms responsible for this phenomenon are unclear. Recently, the role of endothelin (ET) in the pathophysiology of CHF has been given much attention. Studies were conducted to elucidate how ET may contribute to salt and water retention. Cardiomyopathic (CM) hamsters with a moderate degree of heart failure were employed for in vivo and in vitro trials. Clearance methods were used to determine the level of renal function in CM hamsters in comparison with age- and sex-matched control animals. Plasma and urine samples were collected for determination of haematocrit, plasma protein concentration, glomerular filtration rate (GFR), and sodium and water excretion while mean arterial blood pressure (MAP) was monitored intermittently. Radioligand binding studies were carried out to determine ET receptor distribution in the inner medullary collecting duct (IMCD) of CM and control hamsters. The effects of chronic enalapril therapy on renal function and ET receptors were investigated to deduce any interaction between the renin-angiotensin-aldosterone system (RAAS) and the ET hormonal system. Studies were also performed to measure the stimulation response of IMCD cells to ET-1 or to angiotensin II administration. The results demonstrated that renal excretion of sodium and water was diminished in the CHF hamsters in response to atrial natriuretic peptide (ANP) infusion and the fractional excretion of these variables decrementally decreased with increasing severity of heart failure. Binding studies revealed an equal distribution of ET A and ET B receptor subtypes in the IMCD of the hamsters and these receptors, particularly the B variety, were downregulated in the diseased animals. A similar negative correlation was apparent between the receptor density and the degree of cardiac dysfunction — the number of ET receptors declined as heart failure became more pronounced. Following chronic treatment ii with the angiotensin converting enzyme (ACE) inhibitor, enalapril, an improved renal excretory response to ANP was observed with simultaneous restoration of ET receptor density in the IMCD cells. Furthermore, the correlations of the severity of heart failure, fractional excretion of electrolytes, and receptor density, were abolished with enalapril. Cells of the IMCD in CM hamsters exhibited lower basal levels of cGMP accumulation than those of the normal controls. Stimulation of the IMCD cells with ET-1 showed a dose-dependent increase in cGMP production in both CM and healthy animals, but the responses were attenuated in the former. This blunted response may represent a reduction in ET receptor number in the CM hamster kidneys. Incubation of rat IMCD cells with low-concentration angiotensin II displayed a downregulation of ET receptors with an increase in receptor affinity. Co-incubation with the angiotensin receptor antagonist, saralasin, prevented this downregulation and the ET-1 binding affinity remained constant. These results suggest that angiotensin II may participate in the regulation of ET receptor expression. When the data are taken together, there is a strong implication that ET, in combination with angiotensin II, contributes to the regulation of salt and water homeostasis. That renal excretory function is improved with concomitant restoration of ET density after enalapril therapy is convincing evidence that these two hormones interact to promote abnormal fluid retention in the setting of CHF. In conclusion, in the CM hamster model, it is the insufficient ET receptor density, secondary to angiotensin II activation, that results in the unchecked reabsorption of electrolytes by the kidneys in CHF. iii TABLE OF CONTENTS Abstract ii Table of Contents..... iv List of Figures vii List of Tables ix List of Abbreviations x List of Letter Symbols xii Acknowledgements xiii Chapter One Introduction 1.1 Structural and Physiological Changes in CHF 2 1.2 Renal Adaptation and Excessive Salt and Water Retention 5 1.3 Endothelin in CHF 6 1.3.1 The endothelin family 6 1.3.2 Structure of the ET family 7 1.3.3 Biosynthesis and expression of ETs 7 1.3.4 Endothelin receptors 12 1.3.5 Localization of ET receptors 13 1.3.6 Signal transduction from ET receptors... 14 1.4 Potential Roles for ET in the Electrolyte Derangement in CHF 17 1.4.1 Effects of ET on haemodynamics and renal function 17 1.4.2 ETs in the integrated handling of fluid homeostasis in CHF 19 1.5 Angiotensin Converting Enzyme (ACE) Inhibitors 23 1.5.1 Chemical structure and classification 24 1.5.2 Pharmacokinetics 24 1.5.3 ACE inhibitors in CHF 25 1.6 ET Receptor Antagonist 25 1.7 Rationale.... 26 1.8 Objectives and Approach 28 Chapter Two Materials and Methods 2.1 Animals 33 iv 2.1.1 Determination of severity of.CHF 33 2.2 Reagents 33 2.3 In vivo Studies 34 2.3.1 Clearance protocol 34 2.3.2 Chronic enalapril treatment 35 2.3.3 Blood and urine analysis 35 2.3.3.1 Haematocrit and plasma protein concentration 35 2.3.3.2 Ion concentrations 35 2.3.3.3 Glomerular filtration rate 35 2.3.3.4 Urinary cGMP measurement 36 2.4 In vitro Studies....:..'.............! 36 2.4.1 Tissue preparation 36 2.4.1.1 Inner medullary collecting duct (IMCD) cell extraction 36 2.4.1.2 Preparation of IMCD for binding studies 37 2.4.2 Receptor binding studies 37 2.4.2.1 Acid-washed receptor studies 38 2.4.3 Stimulation studies 38 2.4.3.1 ET-induced cGMP accumulation 38 2.4.3.2 Angiotensin II stimulation of IMCD 39 2.5 Statistics : 42 Chapter Three Results 3.1 Categorization of CM hamsters 43 3.2 In vivo Studies : 50 3.2.1 Golden Syrian hamsters as a time control.. 50 3.2.2 Untreated CM hamster studies 50 3.2.2.1 Baseline and ANP-phase haemodynamic characteristics 50 3.2.2.2 Renal effects of ANP administration 50 3.2.2.3 Heart failure severity and urinary excretion rates 53 3.2.3 Enalapril-treated CM hamster studies 53 3.2.3.1 Baseline and ANP-phase haemodynamic characteristics 53 3.2.3.2 Renal effects of ANP administration 64 3.2.3.3 Heart failure severity and urinary excretion rates 71 3.3 In vitro Studies 71 3.3.1 Receptor binding assays 71 v 3.3.1.1 ET receptor distribution in untreated CM hamsters 71 3.3.1.2 ET receptor distribution in enalapril-treated CM hamsters 79 3.3.1.3 Effect of CHF on ET receptor density 79 3.3.1.4 Effect of acid-washing on ET receptor density 79 3.3.2 Stimulation studies 91 3.3.2.1 ET-stimulated cGMP accumulation in IMCD cells 91 3.3.2.2 Effects of angiotensin stimulation on IMCD cells 91 Chapter Four Discussion 4.1 Cardiomyopathy Hamster as the CHF Model of Choice 107 4.2 Physical Characteristics of the Hamsters 109 4.3 The Importance of a Time Control 109 4.4 Haemodynamic and Renal Function in Untreated CM Hamsters... 110 4.5 ET Receptor Distribution in the EVICD of Untreated CM Hamsters 112 4.6 Haemodynamic and Renal Effects of Enalapril in CHF 115 4.7 ET Receptor Distribution in the IMCD of Enalapril-treated CM Hamsters 117 4.8 Angiotensin II Stimulation of IMCD cells 119 Chapter Five Summary 121 Chapter Six Conclusions 123 References 124 Appendix 150 vi LIST OF FIGURES Figure 1.1 Flow chart delineating the adaptive mechanisms which occur during the course of congestive heart failure 3 Figure 1.2 The structure and homology of ET isoforms and the sarafotoxins 8 Figure 1.3 Schematic diagram of post-translational processing of ET peptides. 10 Figure 1.4 Schematic representation of the putative G protein-linked ET receptor subtypes and the multiple transmembrane signal transduction cascades evoked by ET ligand binding 15 Figure 1.5 Neurohumoral systems involved in body fluid homeostasis 27 Figure 1.6 Schematic summary of the hypothetical role of ET in salt and water handling by the kidney in CHF. 29 Figure 1.7 Schematic summary of the approach employed in the present study 32 Figure 2.1 Effects of varying concentrations of angiotensin II on ET receptor density 40 Figure 3.1 Body weights of normal and cardiomyopathic (CHF) hamsters prior to and after enalapril therapy 44 Figure 3.2 Heart weights of normal and cardiomyopathic hamsters prior to and following enalapril therapy 46 Figure 3.3 Heart weight/body weight ratios of normal and cardiomyopathic hamsters prior to and after enalapril therapy 48 Figure 3.4 Fractional excretion of sodium in normal and cardiomyopathic hamsters...: 54 Figure 3.5 Fractional excretion of water in normal and cardiomyopathic hamsters 56 Figure 3.6 Urinary cGMP excretion in normal and cardiomyopathic hamsters 58 Figure 3.7 Correlations between heart weight/body weight ratios and excretory indices in hamsters 60 Figure 3.8 Fractional excretion of sodium in normal and cardiomyopathic hamsters following enalapril therapy 65 Figure 3.9 Fractional excretion of water in normal and cardiomyopathic hamsters chronically treated with enalapril 67 Figure 3.10 Urinary excretion of cGMP in normal and cardiomyopathic hamsters after chronic enalapril therapy : 69 Figure 3.11 Absence of correlative relationships between the heart weight/body weight ratios and excretory indices following enalapril therapy 72 Figure 3.12 Effect of CHF on the density of ET receptor distribution in cardiomyopathic hamster IMCD cells 76 vii Figure 3.13 Effect of enalapril treatment on the density of ET receptor distribution in EMCD cells of cardiomyopathic hamsters 80 Figure 3.14 Relationship between heart weight/body weight ratios and ET receptor density in IMCD cells 83 Figure 3.15 Absence of correlative relationships between heart weight/body weight ratios and ET receptor distribution in IMCD cells following enalapril treatment 87 Figure 3.16 Effect of acid washing on ET receptor density 92 Figure 3.17 ET-stimulated cGMP accumulation in IMCD cells 95 Figure 3.18 Effect of low-concentration angiotensin II on ET receptor density in the IMCD cells of Long-Evans rats 97 Figure 3.19 Prevention by saralasin of angiotensin II-induced ET receptor downregulation 100 Figure 3.20 ET receptor distribution in control IMCD cells and in saralasin-treated IMCD cells of Long -Evans rats 103 viii LIST OF TABLES Table 3.1 Haemodynamic and renal excretory function in golden Syrian hamsters 51 Table 3.2 Haemodynamic and renal excretory function in response to ANP 52 Table 3.3 Haemodynamic and renal excretory function of enalapril-treated hamsters in response to ANP 63 ix LIST OF ABBREVIATIONS A C Adenylate cyclase A C E Angiotensin converting enzyme A N P Atrial natriuretic peptide A V P Arginine vasopressin Bmax Maximum binding capacity B N P Brain natriuretic peptide [Can Intracellular calcium concentration c A M P Adenosine 3',5'-cyclic monophosphate C C D Cortical collecting duct c D N A Complementary deoxyribonucleic acid c G M P Guanosine 3',5'-cyclic monophosphate C H F Congestive heart failure C M Cardiomyopathic C N P C-type natriuretic peptide D A G Diacylglycerol E C E Endothelin converting enzyme E C F V Extracellular fluid volume E D T A Ethylenediaminetetraacetic acid E T Endothelin E T A Endothelin receptor subtype A E T B Endothelin receptor subtype B E T C Endothelin receptor subtype C F C S Fetal calf serum F E N a Fractional excretion of sodium f E H 2 o Fractional excretion of water G Heterotrimeric G protein G F R Glomerular filtration rate G T P Guanosine triphosphate I B M X Isobutylmethylxanthine I M C D Inner medullary collecting duct IP 3 Inositol 1,4,5-trisphosphate K d Dissociation constant kf Ultrafiltration coefficient MAP Mean arterial pressure MAPK Mitogen-activated protein kinase mRNA Messenger ribonucleic acid NO Nitric oxide PA Phosphatidic acid. PBS Phosphate buffered saline PC Pho sphatidylcholine P G E 2 Prostaglandin E 2 PGI 2 Prostaglandin I2 PI Phosphoinositide PKC Protein kinase C P i A Phospholipase Aj PLC Phospholipase C , PLD Phospholipase D PMSF Phenylmethylsulfonyl fluoride RAAS Renin-angiotensin-aldosterone system ROC Receptor operated calcium channel RPF Renal plasma flow rpm Revolutions per minute RVR Renal vascular resistance SEM Standard error of the mean SMC Smooth muscle cell STX Sarafotoxin T X A 2 Thromboxane U C G M P Urinary cGMP uv Urine flow uNav Urinary sodium excretion V D C C Voltage-dependent calcium channel xi LIST OF LETTER SYMBOLS Three-letter and One-letter Symbols for Amino Acids Alanine Ala A Cysteine Cys C Aspartic acid Asp D Glutamic acid Glu E Phenylalanine Phe F Glycine Gly G Histidine His H Isoleucine He I Lysine Lys K Leucine Leu L Methionine Met M Asparagine Asn . N Proline Pro P Glutamine Gin Q Arginine Arg R Serine Ser S Threonine Thr T Valine Val V Tryptophan Trp w Tyrosine Tyr Y ACKNOWLEDGEMENTS I extend my sincerest thanks to my supervisor, Dr. Norman Wong, for providing me with the opportunity to complete this study. His continual guidance and encouragement have been a constant source of support throughout this endeavour. I also wish to express my deepest, gratitude.to Alice Chan and .Dr. Eric Wong for their assistance with the binding experiments, and my appreciation to Edward Mak for performing iodinations, Mohammed Hasham and Leo Smyrnis for their assistance with computer graphics, and to Anita Keister for her administrative efforts. Thanks to all my friends and the many others who helped make this an enjoyable experience. Finally, I am indebted to my family, especially Hubert and Bea, for their love and support. xiii Chapter One Introduction The syndrome of congestive heart failure (CHF) is defined as the inability of the heart to sufficiently pump blood to meet the metabolic requirements of the body. Typically, one side of the heart fails first, followed;by the other. The term "congestive" refers to the backing up of blood in the pulmonary veins: leading to congestion of the lungs and viscera Symptoms and signs of CHF include cardiac hypertrophy, hepatomegaly, oedema, dyspnea, weakness, prolonged circulation time, neck vein distension, and reduced exercise capacity. A wide array of disturbances can lead to CHF. Myocardial damage due to ischaemic heart disease, idiopathic dilated cardiomyopathy, and myocarditis are examples of intrinsic heart disease that result in heart failure. Toxin-induced injury such as that from drug overdose or alcoholism or viral infection can lead to cardiac dysfunction. Other insults to the heart may be of pressure or volume overload in nature. For example, chronic systemic hypertension and mitral valve regurgitation also stress the heart to the degree of failure. Despite continued and recent advances in the understanding and treatment of cardiovascular disease, CHF remains a major health concern. The prevalence of CHF is estimated to be 4 million persons in the United States alone (1), with an incidence of 400 000 persons annually (2). Morbidity is substantial, accounting for 6% of all hospital discharges while the mortality rate is greater than 1 quarter million individuals per year. The prognosis of CHF is grim, with the likelihood of 5-year survival less than 50% (3) from first diagnosis. Many factors have been associated with survival time (4-7). 1 1.1 Structural and Physiological Changes in CHF As heart failure develops, adaptive processes occur within the heart to compensate for reduced pumping efficiency (8,9). Initially, there is a gradual increase in the chamber size of the heart which is mainly due to 'slippage' of the myocardial fibres. As the heart continues to dilate, destruction and scarring of the myocytes curb this slippage; with the consequence that greater forces of contraction, are required to achieve shortening•(Frank-. -Starling Law). Second, cardiac hypertrophy ensues to minimize wall stress. This increase in cell mass maintains adequate cardiac output for some time but at the expense of increased oxygen consumption and depressed contractility. Compensatory mechanisms in CHF are complex (Figure 1.1). Not only is there remodelling of the heart, but neural, hormonal, haemodynamic, and renal adaptations are manifest as well (3, 10-13). Sympathetic activity is increased upon unloading of arterial baroreceptors by diminished blood pressure or stroke volume (14-16). This is accompanied by a rise in the activity of the renin-angiotensin-aldosterone system (RAAS) (17-19) and in the release of arginine vasopressin (AVP) (20-22). These neuroendocrine changes are appropriate to maintain proper organ function through vasoconstriction and ; volume retention. Yet these responses tend to become magnified, resulting in increased afterload and volume load to the already compromised heart. These circumstances create such clinical features as oedema, ascites, and hepatomegaly. Endothelin (ET), a more recently discovered vasoconstrictor, has also been implicated to contribute in the exaggerated responses in CHF since the levels of this peptide are elevated in CHF patients (23-29). Natriuretic and vasodilating factors are concomitantly produced to counter-regulate the vasoconstrictive and fluid-retaining actions of the above systems in CHF. The principal one is the natriuretic peptide family (30) which thus far includes four members. These are Impaired Heart ^ Cardiac Output 1 Ca rdiac Neurohumoral 1 Renal f / ^ C o n t r a c t i l i t y ^ S y m p a t h e t i c Af lenal /fAVP ^ E T ^ R e n i n Nervous Nerve i System Activity Activity Astroke Volume A Arterial Constriction ^ N-' ^After load ^ R B F | G F R AFF ^ Aldosterone A Sodium & Water ^ Re absorption /^Central Filling Pressures >fECFV Oedema J | A N P — Vasodilation Natriuresis &Diuresis Figure 1.1 Flow chart delineating the adaptive mechanisms which occur during the course of congestive heart failure. See text for a detailed discussion. 3 atrial natriuretic peptide (ANP) (31), brain natriuretic peptide (BNP) (32), C-type natriuretic peptide (CNP) (33) and urodilatin (34). That the levels of ANP and BNP are elevated in CHF are well documented (35-41). ANP and BNP share similar structure and biological actions (42). Unlike ANP and BNP, CNP does not exhibit natriuretic properties (43) but may be important in paracrine regulation of central systems related to pressure control and vascular tone. Urodilatin appears to be a more potent natriuretic agent than ANP (44). Prostaglandins E 2 (PGE2) and I2 (PGI2) (45,46) and dopamine (47,48) are thought to play a more minor role in minimizing the degree of vasoconstriction and extracellular fluid volume (ECFV) expansion. Cardiac output and blood pressure are relatively preserved at rest during the early stages of CHF. As the degree of CHF progresses to moderate or severe, cardiac output is reduced and it fails to rise normally upon exercise with the consequence that proportionally less flow is available to the exercising muscle (10). In addition, there is marked vasoconstriction of the cutaneous, renal, and splanchnic beds such that redistribution of the blood flow occurs to provide adequate nourishment to. the brain and coronary vessels: However, compensation can only be sustained for a limited period of. time. The heart tends to decompensate, with either left or right ventricular failure occurring. Left heart failure will result in increased pulmonary venous pressure and thus pulmonary congestion and oedema. Right heart failure leads to elevated venous and capillary hydrostatic pressure and thus peripheral oedema, hepatomegaly, and ascites. Eventually, both sides of the heart may fail. The vasculature in heart failure adapts by increasing tone in the venous capacitance and arteriolar resistance systems (8). This increase involves components of the sympathoadrenal systems, the RAAS, and the impaired function of the endothelium in producing the vasodilator nitric oxide (NO). Hypertrophy of the vascular smooth muscle 4 and increased salt and water content in the vessel wall may also contribute to the reduced compliance of the vessels. 1.2 Renal Adaptation and Excessive Salt and Water Retention The kidney plays a central role in the pathogenesis of CHF. When the heart is impaired, a diminished effective circulating blood volume is perceived by pressure and volume sensors located in the atria, ventricles, great veins and arteries, lungs, and the juxtaglomerular apparatus of the kidney. The abnormally gross retention of salt and water retention may be attributed to the adaptation of the kidney to the weakened cardiac function, leading to the hallmark features of dependent oedema, ascites, hepatomegaly, etc. Both intrarenal and neurohumoral responses occur to enhance sodium and water reabsorption by the kidneys which results in detrimental extracellular fluid volume expansion. One consequence of CHF is that there is a reduction in renal plasma flow (RPF) which parallels the decrease in • cardiac output (49-51). An increase in the glomerular transcapillary hydrostatic pressure is required to maintain an adequate glomerular filtration rate (GFR) and this is effected by a disproportionate increase in efferent arteriolar resistance that is mediated by angiotensin II constriction (52). The resulting increased filtration fraction raises the oncotic pressure in the post-glomerular circulation, thereby abnormally favouring the reabsorption of tubular fluid from the proximal tubule. Increased sympathetic tone has been commonly observed in CHF (53-56). It is beneficial in maintaining blood pressure but excessive venoconstriction may add to the already diminished RPF, further enhancing electrolyte conservation. Nord et al (57) demonstrated that the sympathetic nervous system directly influences tubular reabsorption via stimulation of the Na+-H+ antiport. 5 The RAAS also has a major role in directing the kidney to retain salt and water in CHF. Renin secretion is elevated early in the course of heart failure (58-62) and facilitates the conversion of angiotensinogen to angiotensin I which is followed by angiotensin II formation. Angiotensin II, as discussed above, is a potent vasoconstrictor as well as a potentiator of catecholamine action (49). Studies have shown that angiotensin II directly stimulates sodium reabsorption in the proximal tubule (63,64). Aldosterone levels are increased which promote sodium reabsorption at distal Na+/K+ or Na+/H+ exchange sites of the nephron (64). Arginine vasopressin levels are generally elevated in patients with CHF (20-22,65). This small peptide hormone facilitates water reabsorption in the collecting ducts of the kidney by signalling the insertion of water channel proteins into the apical membrane (66). 1.3 Endothelin in CHF Plasma endothelin levels are raised 2 to 3-fold in CHF (67-73). These levels correlate with the severity of heart failure or pulmonary hypertension (29,74-77) which implicates this peptide in the pathophysiological processes of CHF and perhaps.other cardiovascular diseases (78). Its diverse biologic actions (79-81), including profound vasoconstriction, make ET a prime candidate in contributing to the reduced systemic and renal perfusion characteristic of CHF. 1.3.1 The endothelin family The discovery of a novel endothelium-derived constricting factor was by Hickey and colleagues (82) in 1985. In the subsequent two years, this peptidergic vasoconstrictor was isolated, cloned, and sequenced by Yanagisawa et al (83) and given the name, endothelin, since it was derived from culture supernatant of porcine aortic endothelial cells. Further analysis revealed that three separate genes for ET existed and that each encoded a distinct 6 isopeptide- ET-1, ET-2, and ET-3 (84). Most research has been focussed on ET-1 as the expression of the other isoforms is more limited (85). A fourth peptide, vasoactive intestinal contractor or ET-B, discovered through genomic cloning of mice (86), is considered to be the murine and rat form of human ET-2 since its precursor mRNA is similar to that of prepro-ET-2 and not of prepn> ET-1 or -ET-3 (87). Sarafotoxins, from the venom of the burrowing asp, Attractaspis engaddensis? are four more peptides that share high homology and bioactivity with the ETs (88). Such conservation of the peptides amongst species underscores the physiologic importance of ET. 1.3.2 Structure of the ET family Considerable sequence homology exists between all constituents of the ET family with between 52 % to 81% sequence similarity (89). Each member is composed of a 21-amino acid single chain containing, at identical positions, four cysteine residues that form two disulfide bridges to hold the peptide in a hairpin loop configuration. Polar, charged side chains in the hairpin loop and a hydrophobic COOH-terminus are also characteristic of ET peptides (83,90,91). The flexible hexapeptide C-terminus is greatly conserved and must be intact to confer bioactivity. Figure 1.2 depicts the proposed structure and sequence homology of the ET and sarafotoxin families. 1.3.3 Biosynthesis and expression of ETs Endothelin is synthesized in a manner analogous to other hormonal peptides. From a single open reading frame, ET-1 is translated into a prepropeptide of 203 residues that undergoes sequential proteolytic cleavages to yield the final mature molecule (79,91). Processing begins with cleavage at, by a dibasic pair-specific endopeptidase, residues Lys52-Arg53 and at Arg92-Arg93 which removes the N-terminus containing the secretory signal sequence and the C-terminus to produce the prohormone intermediate, big ET-1, of 38 (human) or 39 (porcine) amino acids. A putative ET-converting enzyme (ECE) then 7 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 ET-1 C S c S S L M D K E C V Y F C H L D I I W ET-2 C S c S S W L D K E C V Y F C H L D I I W ET-3 C 0 c F T Y K D K E C V v 0 C H L D I I W Mouse Rat ET-2 C s c N D W L D K E C V Y F C H L D I I W S6b C s c K D M T D K E C | ~ L | Y F C H | 3 D 0 I W Figure 1.2 The structure and homology of ET isoforms and the sarafotoxins. Upper panel: a free hydrophobic C-terminus and the hairpin loop configuration formed by the two disulfide bonds is common to all members of the ET and sarafotoxin families. Lower panel: the highly conserved sequence of the ET isopeptides and a representative member (S6b) of the sarafotoxins is depicted. Non-conserved residues are enclosed within the boxes. (Adapted from Simonson (ref. 81)). 8 cuts big ET between the unique Trp21-Val22 couplet to give the potent 21-residue peptide (Figure 1.3). The nature of this ECE remains ill-defined although inhibition by phosphorimadon and characterization studies suggest it to be a neutral metalloprotease (92-96). Expression and secretion of ET was originally believed to, be localized exclusively to the vascular endothelium. However, it is now apparent that ET is produced in numerous tissue and cell types, including, but not limited to, endothelium of umbilical vein, glomerulus, corpus cavernosum, and aorta, keratinocytes, macrophages, bone marrow mast cells, astrocytes, mesangial cells, cardiomyocytes, epithelium of the breast, lungs, brain, uterus, stomach, kidney, testis, skeletal muscle, and intestine (reviewed in 80,81,85). Secretion of ET is largely constitutive (83, 97-100) although some evidence indicates there may be a regulated pathway via secretory granules (101). Whether the secretion is constant or regulated may be a function of cell type. Wagner et al (102) reported that ET secretion is polar, given that approximately 80% of ET-1 is secreted basolaterally in cultured endothelial cells.. Many stimuli have been demonstrated to upregulate ET secretion and expression. Growth factors and cytokines such as thrombin (100,103-105), transforming growth factor 13 (100,106-108), interleukin-1 (107,108), tumour necrosis factor alpha (109), and insulin (110,111) have been shown to increase the mRNA levels of ET. Raised message levels and secretion of ET were also observed in response to endotoxin (112,114), phorbol esters (105,114), high glucose (115), fluid mechanical shear stress (116), angiotensin II (117,118), arginine vasopressin (118,119), bradykinin (105), oxidized low-density protein (120), hypoxia (121), cyclosporine (122), and on occasion, ET itself (123). 9 Signal Sequence Mature ET H 2 N - LK -R [R-R 1 20 51 A 54 74 91 A 94 Dibasic Endopeptidase Cleavage C O O H PreproET-1 203 L C O O H ET Converting Enzyme ProET-1 (Big ET-1) H 2 N C O O H 21 Mature ET-1 Figure 1.3 Schematic diagram of post-translational processing of ET peptides. Large preprohormones are cleaved by dibasic pair-specific endopeptidases to give distinct prohormones that are subsequently cleaved by a novel ET converting enzyme at a specific trp-val recognition site to form the mature 21-residue ET isopeptide. (Adapted from Simonson (ref. 81)). 10 There are also inhibitory factors to regulate the production of ET. Indeed, ANP and other nitrovasodilators prevent ET secretion from cultured endothelial cells (122,124). In contrast to these in vitro data, one recent investigation found that under circumstances of vasodilator-induced hypotension, there was enhanced systemic release of ET-1 (125), suggesting that the source was of non-endothelial origin. Sandok et al (126) reported that the rise in circulating levels of ET in a model of acute renal ischaemia occurred only in the-, presence of prostaglandin inhibition or the absence of ANP. Furthermore, nitric oxide (NO) and prostacyclin (PGI2) released after thrombin stimulation provide negative feedback on ET production via cGMP and cAMP formation, respectively (127,128). Kohan and Padilla (129) confirmed that NO and cGMP reduce ET-1 production and noted that interferon gamma is also a potent inhibitor of ET-1 release. The groups of Stewart et al (130) and Cade et al (131) have reported that vascular smooth muscle cells release an inhibitory substance that reduces ET production by endothelial cells. The chemical properties of this substance await determination. The levels of local and circulating ET are adjusted through elimination by at least three mechanisms. First, there is hydrolytic degradation by a neutral metalloendopeptidase, EC 24.11, that is present on the cell surface of various kidney, pulmonary, intestinal, and central nervous tissues (132). This enzyme, which inactivates several regulatory peptides, cleaves ET and hydrolyzes its N-terminus, rendering inoperative products (133). Sarafotoxins are four-fold less susceptible to this endopeptidase than ET which may partially explain why the former are so much more toxic than the latter (134). Lysosomal degradation appears to be another mode in which ET is catabolized as lysosomal vesicles in bovine pulmonary endothelial cells have been shown to be rich in ET that has probably been endocytosed (135). The pulmonary circulation was predominantly responsible for the removal of circulating ET isopeptides after ventricular injection in the rat although 11 there was uptake by the kidneys and liver as well (136,137). Within minutes of injection, more than 60% of the ET-1 was eliminated. Fukuroda et al (138) demonstrated that clearance of exogenous ET by the rat lung and kidney was mediated through E T B receptors (see below). While ET is cleared in the lungs of rats and guinea pigs, it is the kidneys, spleen, and hindlimb that clear ET in the pig (139), suggesting a species-specific pathway for degradation. 1.3.4 Endothelin receptors Prior to molecular evidence of different ET receptor subtypes, pharmacological studies revealed that different agonist affinities and ET isopeptide selectivities for binding sites existed, indicating there was a population of multiple receptor subtypes (140). Differences in physiological responses to the ET isopeptides were also observed. The potency of ET-1 and ET-2 as pressor agents was much greater than that of ET-3 (114) while ET-1 and ET-3 were equipotent in causing the transient vasodilatation (141,142). Two receptor subtypes, termed E T A and ET B , were proposed to distinguish between vasoconstriction and vasodilation mediation, respectively. In 1990, Arai and colleagues (143), and Sakurai and colleagues (144), independently cloned the E T A receptor from bovine lung and E T B receptor from rat lung, respectively. The human A-type receptor was subsequently cloned and characterized (145). Numerous studies have confirmed that ET A is selective for ET-1 over ET-3 and sarafotoxin S6c (STXS6c) while E T B shows similar affinities for all three ET isopeptides (143,144,146). The homology between human and rat E T A and E T B is about 91% and 88%, respectively while that between human ETAand ET B is roughly 55% (140). The development of a wide repertoire of pharmacological tools has enabled further division of the E T B receptor classification. It was shown that this receptor exhibited both vasodilating and vasoconstricting abilities (147) and thus given the designations E T B 1 and 12 ET B 2 , respectively. Others have distinguished between the ET B subtypes by characterizing their respective pharmacological profiles (148,149). However, no definitive biochemical or molecular evidence for the subtypes is available. Binding studies have revealed the preferential binding of ET-3 over ET-1 by an ET receptor in a number of tissues (150-152). This receptor has been classified as the putative ETC receptor and has been cloned from the dermal melanophores of Xenopus laevis (153). A mammalian version of this receptor has not yet been isolated. Sequencing of the cDNA clones has demonstrated that ET receptors are peptides of approximately 426-444 residues containing secretory signal sequences of 20-26 amino acids (143,144,153,154). They belong to the superfamily of G-protein-coupled, seven-domain-membrane-spanning receptors. G-proteins are guanine-nucleotide-binding regulatory proteins. 1.3.5 Localization of ET receptors The distribution of ET receptor subtypes varies between and among species and tissue type (140). For example, in rat heart, mRNAs for both the A-type and the B-type were widespread in myocardial muscle, but only the A variety was present on coronary blood vessels (155). In human kidney, the predominant form is the ET B subtype which is concentrated in the collecting system (156). Similar results have been obtained in the rat and dog in some studies (157,158). However, the receptor profiles in rat kidney cortex and papilla were reversed when compared to that of dog (158,159). Vasculature in the kidney has been found to contain primarily A-type receptors (155,160,161). In general, ET A receptors are expressed more abundantly on vascular smooth muscle cells while ET B ones are expressed primarily on endothelial and other cell types. 13 1.3.6 Signal transduction from ET receptors The effects of ETs are transduced via cell surface receptors coupled to various heterotrimeric G-proteins (81,85). Both E T A and E T B subtypes are able to couple to at least four types of G-proteins: G s , G q , G n , and G; (pertussis toxin-sensitive) (162,163). Binding of ETs to their receptors stimulates the GTP turnover of these proteins which then regulate (activate or inhibit) one or more effectors. Such effectors include phospholipase C (PLC) (164-167), phospholipase A 2 (137), phospholipase D (PLD) (168), and adenylyl cyclase (AC) (163) (Figure 1.4). Activation of PLC results in the hydrolysis of phosphatidylinositol to the second messengers inositol trisphosphate (IP3) and diacylglycerol (DAG) (165,169) although hydrolysis of phosphatidylcholine by PLC or phosphatidic acid degradation by PLD provide another source for the latter (170). n»3 mobilizes Ca 2 + from the sarcoplasmic reticulum or calciosome, thereby raising intracellular concentrations of free calcium [Ca2+];. The increase, along with influx of extracellular Ca 2 + through voltage-dependent and non-voltage-dependent (receptor-operated) ion channels, appears to mediate ET-induced smooth muscle cell (SMC) contractions (85,140). DAG, with cofactors Ca 2 + and phosphatidylserine, activates protein kinase C (PKC) which may sensitize the contractile apparatus to Ca 2 + (171,172). PKC also mediates the ET-1 stimulation of Na +-H + exchange that leads to intracellular alkalinization in vascular SMCs (173). In contrast to the vasoconstricting mechanisms discussed above, a signalling pathway for vasodilation has been observed (174,175). Via E T B 1 , ET stimulates a pertussis toxin-sensitive G protein that results in the release of nitric oxide (NO), which in turn facilitates the relaxation of vascular SMCs through its second messenger, cGMP (176). 14 Figure 1.4 Schematic representation of the putative G protein-linked ET receptor subtypes and the multiple transmembrane signal transduction cascades evoked by ET ligand binding. Isopeptide selectivity of the ET receptor subtypes is shown. Stimulation of the phosphoinositide (PI) pathway leading to protein kinase C (PKC) activation and release of calcium from intracellular stores is well characterized. Extracellular calcium entry through receptor operated (ROC) and voltage dependent (VDCC) calcium channels also increases intracellular calcium following ET receptor activation. Other effector systems affected by ET include phospholipase Aj (PLAj), phospholipase D (PLD), adenylate cyclase (AC), and the electroneutral Na+-H+ antiporter. ET-1, ET-2, ET-3: endothelin-1, endothelin-2, endothelin-3, respectively; ETA, ETB, ETC: endothelin receptor subtypes A, B, C, respectively; G, heterotrimeric G protein; PC, phosphatidylcholine; PA, phosphatidic acid; DAG, diacylglycerol, IP3, inositol 1,4,5-trisphosphate; [Ca2+]„ intracellular calcium concentration; NO, nitric oxide; cGMP, cyclic guanosine monophosphate; cAMP, cyclic adenosine monophosphate; PGE2, PGI2, prostaglandins E 2 and I2; TXAj, thromboxane A^ MAPK, mitogen activated protein kinase; c-fos/c-jun, immediate early genes. 15 16 The signalling pathways mediating long-term effects of ETs, such as mitogenesis, have not been fully elucidated. However, elevated [Ca2+] ;, mitogen-activated protein kinase (MAPK) stimulation, and enhanced PKC and Na +-H + antiporter activity have been suggested to play a role (177). Consistent with this notion is the finding that ETs stimulate the MAPK cascade and 90kD S6 kinase (RSK) (178,179). In addition, increased [Ca2+]; was directly involved in the ET-1-induced rise in the mRNA levels of several fos/jun genes (180). 1.4 Potential Roles for ET in the Electrolyte Derangement in CHF 1.4.1 Effects of ET on haemodynamics and renal function As vasoconstriction is a prominent feature of CHF, it is logical to surmise that this increased tone may be the result of the potent actions of ET, the quantities of which are raised in this pathophysiological state (67-77). Indeed, such observations have been noted in a number of studies (181-184). Systemic infusion of ET-1 has repeatedly caused increases in systemic and renal vascular resistance (RVR) and decreases in RPF and GFR (181,183,185,186). It is controversial whether ET actions are mediated in an autocrine/paracrine or circulating mode. Due to the involvement of several baroreceptor reflexes in the regulation of kidney function, intrarenal infusion of ET-1 has been completed to separate systemic from intrarenal influences of the peptide. Prolonged and dose-dependent decreases in RPF and GFR and increases in RVR were evident with this route of administration as well (187,188). ET-induced vasoconstriction in isolated perfused rat kidney was a direct effect of ET on the renal vasculature and was mediated through both the A and B subtypes of the ET receptor (189). 17 Weak pressor doses of ET-1 did not alter GFR in a study by King et al (181) but higher doses caused extreme contraction of both efferent and afferent arterioles in addition to a depressed glomerular flow rate and ultrafiltration coefficient (Kf), thus reducing GFR (181,190). Utilizing micropuncture studies, these authors found that ET-1 diminishes Kf although they could not deduce whether it was due to a change in glomerular capillary surface area or hydraulic conductivity. In this regard, Simonson and Dunn (191) reported ET-1 contracts glomerular mesangial cells, so that it may reduce filtering surface area. In view of the decline in RPF and in GFR produced by ET-1, it would be expected that the filtered load of sodium would be smaller and thereby reduce excretion, provided tubular transport were unaffected. Furthermore, peritubular Starling forces would favour sodium reabsorption. Both natriuretic (181, 192) and antinatriuretic (186, 193,194) responses have been elicited with systemic infusion of ET-1. The increase in sodium excretion occurred despite reduced RPF and, in another study , GFR (195). Pretreatment with nifedipine, the calcium channel blocker, prevented ET-1 induced renal vasoconstriction and sodium retention (194), implying that ET-1 plays a role in volume equilibrium. Clavell et al (196) found systemic infusion of ET-1 resulted in renal vasoconstriction, diuresis, and antinatriuresis but a natriuretic effect was unmasked with intrarenal BQ123 treatment. Furthermore, the same study found that the A-subtype ET receptor contributed to the vasoconstriction whereas the B-subtype was mainly responsible for the diuresis independent of sodium excretion. Similar results with regard to the roles of the ET receptor subtypes in vasoconstrictor and diuretic responses were reported by Kamphuis et al (197) and Pollock and Opgenorth (198). High-dose intrarenal infusion and isolated perfused kidney studies with lowered GFR also resulted in natriuresis (187,188,199,200). These data suggest that ET induces natriuresis in the kidney, but this may be modulated by other factors that could be intra- or extrarenal in nature. 18 One mechanism by which ET-1 may decrease sodium absorption is through direct tubular action. Zeidel et al (201) demonstrated that ET-1 evoked natriuresis in vivo by inhibiting the Na+/K+-ATPase in epithelial cells of the inner medullary collecting duct (IMCD), likely via a PGE 2 pathway. An earlier report by Jabs et al (202) stated that this prostaglandin diminished ATPase activity in rabbit EVICD. Garvin and Sanders (203) showed that suppression of the Na+/K+-ATPase by ET concomitantly inhibited fluid and bicarbonate transport in the proximal straight tubule. Thus ET affects transport in both the proximal nephron and the distal concentrating segments. Other routes of ET action involve interplay with various neurohumoral systems. 1.4.2 ETs in the integrated handling of fluid homeostasis in CHF As discussed earlier, there is a balance between vasoconstriction and vasodilation, and between fluid retention and fluid excretion to regulate ECFV and blood pressure. Not only do ET levels reach pathophysiological concentrations in CHF, but enhanced release of other hormones and peptides is either stimulated by ET-1 or vice versa in an attempt to maintain functional homeostasis Furthermore, the physiological effects; of ET and the various neurohumoral substances may potentiate or interfere with one another, thereby modulating their respective actions. High plasma levels of ANP are found in CHF. Although the primary stimulus for ANP release is atrial stretch (204), it is also produced (205) and secreted (205-207) by isolated atria and cultured myocytes in response to ET-1. Two recent studies determined that this secretion of ANP was mediated via E T A rather than E T B receptors since BQ123, an A-subtype-selective antagonist, inhibited ET-1-stimulated ANP secretion while ETB-specific agonists did not activate ANP secretion (208,209). By enhancing circulating levels of ANP, ET theoretically would indirectly cause natriuresis since ANP blocks sodium reabsorption at the collecting ducts and increases GFR (210). Despite higher quantities of 19 circulating ANP, a blunted renal response to this peptide is consistently seen in those with CHF (211,212). The cause(s) of this attenuation has not been determined, but one may speculate that it is due to poor renal blood flow which would result in poor delivery of this hormone to the distal sites of the nephron. In addition, intrinsic end-organ resistance or antagonistic fluid-retaining systems may predominate in this setting. Therefore, some investigators argue that the purpose of ET-stimulated ANP secretion is, instead, to oppose the contractile effects of ET (81). Some studies have suggested that ANP modulates the actions of ET (213,214). . Jaiswal (215) found ET inhibits ANP-stimulated cGMP production while Cernacek et al (216) found no interaction between ET and ANP-induced cGMP expression. It is possible that ET and ANP inhibit distal tubular sodium reabsorption via separate mechanisms. ANP also indirectly minimizes sodium reabsorption by antagonizing the release of renin and aldosterone. / In addition, ANP inhibits angiotensin II-stimulated fluid absorption in the proximal nephron through a mechanism involving cGMP-dependent protein kinase (217). Other studies.have focussed on the interactions between ANP and AVP, the levels of which are also abnormally high in CHF (20,22,65). ANP has been shown to suppress AVP release (218) while AVP has the reverse effect on ANP (219). Moreover, AVP attenuated the renal effects of ANP (220) while ANP counteracted the antidiuretic effect of AVP in patients with moderate CHF (221), albeit through unknown mechanisms. There appear to be two components comprising the RAAS. The circulating RAAS seems to be activated in acute situations of homeostasis disturbance and then returns to basal levels of activity after cardiovascular compensation (222). On the other hand, the autocrine/paracrine tissue component appears to mediate longterm effects that influence cardiovascular function and structure. 20 Activation of the RAAS, in patients with CHF is most likely in response to perceived low renal plasma flow and pressure which may be due, in part, to the exquisite sensitivity of the kidney (223) to the potent vasoconstricting properties of ET-1. Yet the effects of ET-1 on the RAAS have varied, depending on its source. Infusion of ET-1 has been shown to elevate circulating aldosterone (186,193) which may be through direct stimulation of the adrenal glomerulosa.cells (224). However,, plasma renin activity is raised after systemic infusions of ET-1 and would therefore activate the angiotensinogen-angiotensin-aldosterone cascade. Whatever the stimulus for aldosterone secretion, enhanced sodium reabsorption by the distal tubules and collecting ducts should ensue. Moreover, angiotensin II formation is also increased, and this peptide directly stimulates sodium reabsorption from the proximal tubule. In contrast, local intrarenal ET appears to inhibit the RAAS. ET-1 production in the glomeruli and IMCD has been documented (225). A number of studies has established that ET-1 inhibits renin secretion in vitro by a calcium-dependent pathway (226-229). One group recently found that ETs selectively inhibit cAMP-stimulated renin secretion in murine juxtaglomerular cells through a calcium-sensitive process mediated by E T B receptor activation (230). Presumably, aldosterone secretion and angiotensin II formation would decrease following depressed renin activity, leading to increased sodium output. Thus Simonson (81) has suggested ET is natriuretic when locally released but antinatriuretic when circulating levels of ET are elevated, as in CHF. This rise in circulating ET may reflect spillover from autocrine/paracrine secretion. While ET may activate the RAAS, one component of the RAAS, angiotensin II, also exerts effects on ET. Angiotensin II has been shown to increase ET-1 synthesis in endothelial cells (118) as well as upregulate ET receptor and mRNA levels for the A subtype in vascular smooth muscle cells (231). These data further support the notion of a 21 link between the RAAS and ET. In contrast, Wilkins et al (232) found that the pressor and renal actions of ET in vivo apparently were not mediated by the RAAS since results with captopril treatment were similar to those without it. Water metabolism is most notably regulated through the actions of AVP, but current evidence indicates ET also participates in fluid homeostasis. The finding that volume depletion in rats and increased osmolality reduced ET-1 production in rat IMCD cells in a dose-dependent fashion supports the idea that ET may provide a communication link between volume status and sodium and water transport (233). That immunoreactive ET peptides were present in secretory vesicles in the rat posterior pituitary and that the vesicles were depleted during water deprivation also implies an active role for ET in body fluid balance (234). Although AVP and ET stimulate the production of one another (186,235), they are also mutually (antagonistic in the renal collecting system. AVP downregulated ET-1 binding to E T B receptors in the rat cortical collecting duct via a protein kinase A-dependent mechanism (236) while ET-1 inhibited the antidiuretic effect of AVP (185). More specifically, endothelin inhibited the A VP-induced accumulation of cAMP in the cortical collecting duct;, outer and inner medullary collecting ducts, and the papillary tubules of the microdissected rat nephron via a pathway involving PKC (237,238). Subsequent studies demonstrated that endothelin inhibits AVP-stimulated water permeability in rat IMCD by blocking cAMP generation (239,240). Edwards and colleagues (241) have since revealed that the inhibition by ET of AVP-induced increases in water permeability and cAMP accumulation is achieved through activation of the E T B receptor rather than the A subtype. A current publication by Ozaki et al (242) demonstrates the coupling of E T B receptors in porcine kidney tubular epithelial LLC-PK t cells to two distinct signal transduction cascades. The increase in cGMP in response to ET-1 was pertussis toxin-insensitive while the decrease of AVP-induced cAMP was 22. pertussis toxin-sensitive, suggesting that the latter response was mediated by a pertussis toxin-sensitive G ; protein. As cGMP has been shown to inhibit amiloride-sensitive sodium transport in LLC-PKj cells (243), these authors have suggested that ET-stimulated cGMP increases may also inhibit sodium channels in epithelial cells to evoke natriuresis. Thus ET may act as a natriuretic factor by inhibiting A VP-induced cAMP and by stimulating. cGMP production. Collectively, the above data indicate that many complex interactions may be occurring simultaneously during the course of heart failure with the increased activity of several neurohormonal systems. With the observation that salt and water retention is characteristic of CHF, it is possible that the effects of one or more fluid-retaining systems are overriding those of the excretion-promoting systems. This may be achieved through increased sensitivity of the vasculature and kidneys to the vasoconstricting factors or decreased responsiveness to the vasodilating substances, but the explicit mechanism(s) of how fluid retention persists remain a mystery. To elucidate the retentive mechanisms, the relative importance of each hormonal system in contributing to this pathophysiology must be determined. Utilization'of. inhibitors of the various systems, such as an angiotensin converting enzyme (ACE) inhibitor, may help to delineate the impact of each hormone on ECF volume expansion in CHF. 1.5 Angiotensin Converting Enzyme (ACE) Inhibitors Angiotensin converting enzyme (ACE) inhibitor is a peptidyl dipeptidase whose primary action is to prevent the conversion of angiotensin I to angiotensin II, both in the circulation and in the autocrine/paracrine tissue system. The discovery of the nonapeptide inhibitor, teprotide, from the snake venom of Bothrops jararaca (244) provided the first observation of inhibition of ACE. Since then, synthetic ACE inhibitors have been produced that are employed in animal and clinical studies. 23 1.5.1 Chemical structure and classification Angiotensin converting enzyme inhibitors interact with a zinc ion that is present in the active site of ACE (245). To ensure efficient catalytic conversion, ACE inhibitors are designed with specific chemical groups that will bind effectively to the zinc ion. Thus, ACE inhibitors are classified, on the:basis, of their zinc ligand. Three classes of ACE inhibitors have been produced with the following moieties: sulfhydryl-containing, carboxyl-containing, and phosphoryl-containing. Captopril, enalapril, and fosinopril are examples of ACE inhibitors in each class, respectively. The extent of inhibitory action, or its potency, is determined by the strength of binding to the zinc ion and the presence of additional binding sites. Carboxyl groups bind less tightly to the zinc ion than sulfhydryl groups. However, binding by enalapril is strengthened by the addition of two more binding sites, thereby increasing duration of action. Several ACE inhibitors are administered as prodrugs to enhance absorption of the drug. Prodrugs are esterified forms of the molecules which are activated by, hydrolysis to the diacids in the liver, following intestinal absorption. Enalapril is one compound that is orally delivered as a prodrug. 1.5.1 Pharmacokinetics Following oral ingestion, ACE inhibitors may be 25% to 80% bioavailable. The presence of food decreases absorption with some compounds. Generally, ACE inhibitors are bound to plasma proteins and the amount of binding varies between 10% to 95%. The majority of ACE inhibitors are eliminated predominantly via the kidneys whether they are metabolized or not. Enalapril is approximately 60% to 70% bioavailable and food does not affect its absorption (244). Maximum levels of enalapril are attained within one-half to one and one-half hours after ingestion while those of the active enalaprilat form are achieved within 2 to 4 hours after ingestion. Both forms circulate virtually protein-free 24 and peak activity occurs 4 to 6 hours after administration. The effective half-life of enalaprilat is about 11 hours. Numerous studies have shown that ACE inhibitors exhibit a steep dose-response curve, in contrast to previously-held notions (246). Only low dosages are required to block angiotensin conversion^ and the effect of increasing the dose is to augment the duration but not the intensity of inhibition (245). High dosages have been used to maximize efficacy and duration (246). Nussberger et al (247,248) observed a dissociation between the quantities of plasma angiotensin II and the antipressor effect with some ACE inhibitors. This dissociation could be related to other consequences of ACE inhibitor action such as attenuation of sympathetic activity , increased vasodilating hormones and parasympathetic activity, and inhibition of the tissue component of angiotensin (245,249). 1.5.3 ACE inhibitors in CHF Several intensive studies have been conducted that indicate ACE inhibitors benefit patients with varying degrees of heart failure (250). The CONSENSUS (251) demonstrated that mortality from CHF decreased in patients with severe heart failure while the SOLVD trial (252) gave similar results in patients with mild-to-moderate symptomatic heart failure. Furthermore, the SOLVD prevention study (253) showed that ACE inhibitors could circumvent any progression to heart failure in asymptomatic patients with left ventricular dysfunction. 1.6 ET Receptor Antagonist BQ123, an ETA-selective antagonist, was designed based on the structure of a cyclic pentapeptide, found in the fermentation products of Streptomyces misakiensis, which behaved as an ET-receptor antagonist. Structure-function studies led to the development 25 of BQ123 cyclo[D-Asp-Pro-D-Val-Leu-D-Trp] which is recognized by the first extracellular loop of the ET A receptor (85). BQ123 binds to ETAwith an IC5 0 of 22 nM. 1.7 Rationale In light of the fact that ET infusion has been repeatedly demonstrated to affect haemodynamic properties as well as renal function, the elevated levels of this peptide in CHF make it a prime candidate as a major contributor to the pathophysiological process. Furthermore, interplay between ET and other neurohormonal systems, including the RAAS, natriuretic peptides, and AVP, has been documented in numerous studies. Each component is likely able to modulate the effects of one or more of the other systems in an antagonistic or synergistic manner, depending on the extent of activation and on signals that are received by various sensors in the body. In the environment of heart failure, ET, the RAAS, and AVP (and also the sympathetic nervous system) act jointly to increase vascular tone while ANP opposes it. The RAAS and AVP also act to conserve fluid by the kidney while ANP tries to promote its loss. Normally, ET is believed to be natriuretic and diuretic, but in the course of CHF, when the circulating levels are anomalously high, it appears to work in reverse. In addition, most of the hormones are able to regulate the expression of one another, adding another dimension of interaction. Therefore, which physiological responses will prevail in CHF is determined by the balance of forces created by the different regulatory elements (Figure 1.5). When the available data is taken together, a logical approach to take would be to determine how the neuroendocrine systems interact with one another in CHF. By administering an ACE inhibitor, one can dissect out the importance of the RAAS to some extent. The effect of angiotensin II blockade on ET can be examined through characterizing the ET receptor distribution in the IMCD of CM hamsters. Because angiotensin II potently stimulates ET- production and release, the circulating and tissue 26 vaso Fluid Excretion Fluid Retention Figure 1.5 Neurohumoral systems involved in body fluid homeostasis. An imbalance between the vasodilating/natriuretic and vasoconstricting/anti-natriuretic forces, favouring fluid retention as observed in CHF, is shown above. 27 levels of E T wil l rise. I f E T and its receptors have a typical hormone-receptor relationship, the high circulating levels of E T would be expected to downregulate the E T receptors. Indeed, certain populations of E T receptors have been found to be downregulated in heart failure (73,254). We hypothesize that the increased levels of E T in C H F downregulate the E T B receptors, which have been shown to antagonize the actions of A V P , thereby removing the tonic inhibition, of A V P action. Downregulation of E T receptors would also decrease sodium excretion which is probably mediated by thesereceptors as well. B y decreasing the E T receptor density in the I M C D , A V P will stimulate c A M P accumulation which wil l in turn direct insertion of water-channel proteins into the apical membrane for increased water reabsorption. Fewer E T receptors would result in a diminished capacity to mediate salt output. Such consequences could lead to the hallmark fluid retention of C H F . This hypothesis was tested. Figure 1.6 summarizes the current hypothesis. 1.8 Objectives and Approach The overall objective of this thesis is to gain further insight.into the pathophysiology of salt and water retention Specifically, one objective was to determine whether E T participates in this fluid retention, and i f so, by what mechanism(s). Second, the study also aimed at answering whether angiotensin II plays a role in mediating the actions of E T . The approach of the study is outlined below. Both in vivo and in vitro experiments were conducted to correlate physiological responses with physical differences in the kidneys of normal and C M hamsters. Standard clearance measurements were obtained from C M and normal control hamsters for basal values and to demonstrate a blunted response to A N P in C M hamsters as compared to normal. The I M C D were then carefully isolated from the kidneys of both groups to characterize E T 28 Figure 1.6 Schematic summary of the hypothetical role of ET in salt and water handling by the kidney in CHF. The rise in ET levels, secondary to angiotensin II and vasopressin stimulation, results in downregulation of ET receptors in the kidneys of CHF individuals. Reduction in ET receptor density leads to enhanced electrolyte reabsorption. See text for discussion. 29 30 receptor subtype distribution and density through radioligand binding methods. Binding experiments were performed using I 2 5I-labelled ET-1 and the specific receptor antagonist, BQ123. The clearance and radioligand binding studies were then repeated on additional C M and normal animals that had been treated with the A C E inhibitor, enalapril, for one week prior to the experiments. Chronic enalapril therapy was administered to determine whether A C E inhibition would restore the natriuretic response by cardiomyopathic ( C M ) hamsters to exogenous A N P . Urine samples were collected for basal values pre- and post-enalapril, and to ascertain the response to A N P before and after enalapril treatment via measurement of its second messenger, c G M P (Figure 1.7). Stimulation studies were completed on the I M C D of C M and normal hamsters to assess whether there was a difference in the response of the respective cells to ET-1 administration. Rat I M C D were also stimulated with low-concentrations of angiotensin II to clarify whether this peptide has regulatory functions for E T receptor expression. It is expected that this research will aid in a better understanding of the physiological and molecular mechanisms underlying the predominant expanded extracellular fluid volume in C H F . A greater understanding of these mechanisms would be important in designing therapeutic regimes. 31 A. Cardiomyopathic Hamsters Untreated Enalapril-treated Clearance Methods Urine Plasma Collection Collection Glomerular Filteration Rate Fractional Excretion of N a + Fractional Excretion of H 2 0 cGMP B. Cardiomyopathic Hamsters Untreated Enalapril-treated Extract IMCD I Co"agenase Digested Tissue 1 2 5 I - E T - 1 Radioligand Binding Studies i ET Receptor Subtype Distribution Figure 1.7 Schematic summary of the approach employed in the present study. (A) Flow chart of the in vivo scheme. (B) Flow chart of the in vitro scheme. 32 Chapter Two Materials and Methods 2.1 Animals Male UM-X7.1 strain cardiomyopathic (CM) Syrian hamsters and age/genetically-matched inbred albino normal controls were purchased at 290 days of age (Canadian Hybrid Farms, King's County, Nova Scotia). All animals, weighing between 110 and 160 grams, were fed regular laboratory chow (Ralston Purina Co., St. Louis, MO) and tap water ad libitum. They were housed in standard plastic cages in a climate-controlled room (22°C) and allowed to adapt to the new environment before use for studies. 2.1.1 Determination of severity of CHF Careful assessment of the degree of heart failure was performed on each animal based on a previously established grading scale (255,256). Grade 0 designates no lesions. Grade 1 is considered to be mild heart failure with minimal subcutaneous oedema, no measurable exudates in the body cavities, and liver enlargement. Moderate heart failure may be Grade 2 when there is diffuse subcutaneous oedema and mild pleural effusions and ascites , or Grade 3 when ascites and pleural effusions are at measurable volumes of 2.5 ml or less. Grade 4 represents severe heart failure and symptoms include profound generalized oedema over the entire body and face in addition to conspicuous ascites and pleural effusions that measure more than 2.5 ml. 2.2 Reagents Inactin was obtained from Byk Gulden Konstanz (Germany). Enalapril was generously donated by Merck Frosst Canada Inc. Endothelin (ET)-l and the ETA-specific antagonist, BQ123, were purchased from Peninsula Laboratories Inc., Belmont, California. The following compounds were obtained from Sigma Chemical, St. Louis, MO.: atrial natriuretic peptide (ANP), saralasin (Sarlle), an angiotensin II receptor antagonist, 33 polyethylene glycol, and tris-HCl buffer. [3H]-inulin-methoxy was from NEN Dupont (Boston, MA) and Iodine-125 (125I) was from Dupont Canada Inc. The other chemicals and reagents were obtained from Fisher. 2.3 In vivo Studies 2.3.1 Clearance protocol Each hamster, whether untreated or enalapril-treated, was anaesthetized with Inactin (25 mg/kg) by intraperitoneal injection and carefully weighed. Surgical anaesthesia typically ensued within 10-20 minutes. The animal was then placed on a temperature-regulated table and body temperature was monitored with a rectal probe. A tracheostomy using polyethylene tubing (PE 160) was performed to maintain a clear airway. The external jugular vein and carotid artery were cahnulated with polyethylene catheters (PE 50) for infusion and blood sampling, respectively. The carotid cannula was connected to a Gilson recorder (Middleton, WI) connected to a pressure transducer to record blood pressure. Through a suprapubic incision, a polyethylene catheter was inserted into the bladder for urine collection. An intravenous infusion (compact infusion pump, Harvard Apparatus Co., Inc., Millis MA) of 0.9% saline containing [3H]inulin was administered at a rate of 0.0201ml/min for the duration of the experiment. Following a 60-minute equilibration period, two 30-minute baseline clearance collections were made. In the next phase, a bolus dose (1 ug/kg) of ANP was given followed by its addition to the saline infusion at a concentration of 1 ug/kg/min for 90 minutes. Two 30-minute clearance periods were obtained 30 minutes after the initiation of ANP infusion. Blood samples were drawn into heparinized tubes for haematocrit and plasma protein concentration determination at the midpoint of each clearance phase. Urine was collected into preweighed tubes for gravimetric volume calculation. At the end of each experiment, animals were sacrificed by 34 removing their hearts for weight measurement. Kidneys were harvested for receptor studies. Time control experiments were conducted using analogous clearance methods. Cannulated male golden Syrian hamsters were continuously infused with [3H]inulin-saline. Blood pressure was monitored as above. Following 45 minutes of equilibration, three 30-minute clearance collections at 60-minute intervals were performed. Blood samples were obtained during each collection phase for calculation of glomerular filtration rate (GFR). 2.3.2 Chronic enalapril treatment Enalapril (2.5 mg/kg) was administered subcutaneously once per day, for six days, prior to study. On the seventh day of treatment, clearance studies were performed as described above. 2.3.3 Blood and urine analysis 2.3.3.1 Haematocrit and plasma protein concentration Ffaematocrit was determined using a micro haematocrit reader (Phillips-Drucker, Astoria Oregon, USA) while plasma protein was ascertained with a hand refractometer (Erma, Tokyo). 2.3.3.2 Ion concentrations Plasma and urinary sodium and potassium were measured with the IL943 flame photometer (Instrumentation Laboratory, Lexington Mass.). 2.3.3.3 Glomerular filtration rate 10 ul of plasma or urine were aliquoted to plastic scintillation vials and 5 ml of Scinti Verse (Fisher Scientific, USA) were added for quenching. Radioactivity was measured by a liquid scintillation counter (Beckman Scientific, Irvine CA). 35 2.3.3.4 Urinary cGMP measurement cGMP was measured with a commercially available radioimmunoassay kit (NEN, Du Pont) after acetylation. 2.4 In vitro Studies 2.4.1. Tissue preparation 2.4.1.1 Inner medullary collecting duct (IMGD) cell extraction Kidneys were excised and thoroughly rinsed in ice-cold PBS buffer after removal of perirenal fat and capsules. After bisection with a sterile scalpel, the renal medulla and papilla were carefully dissected out and minced in 1 ml of collagenase (1.5 mg/ml) (United States Biochemical, Ohio) dissolved in 37°C 1640 medium. The minced tissues were transferred to a tube containing 5 ml of 1640 medium and collagenase and then incubated at 37°C for 30 minutes. To prevent clumping, the digested tissues were pipetted in and out with a pasteur pipet. Digestion was stopped with the addition of an equal volume of 37°C 1640 medium with. 10% fetal calf serum (FCS) and the mixture was centrifuged at 1000 rpm for 3 minutes. Once the supernatant was discarded, the.digested tissues were rinsed with 5 ml of the same medium and centrifuged at 1000 rpm for a further 3 minutes. The supernatant was aspirated and 10 ml of 37°C 1640 medium with 10% FCS was added and mixed. The mixture was further incubated for one hour and then centrifuged again at 1000 rpm for 3 minutes. The supernatant was discarded and for a) stimulation studies, the pellet was resuspended with 37°C 1640 medium (about 1 ml/100 ug protein) containing isobutylmethylxanthine (50 ul/ml) and for b) receptor binding studies, the pellet was resuspended with 2 ml of 1% Tris-Tyrode buffer and frozen at -80°C until use for studies. A sample of the IMCD cell preparation was sent to the Division of Anatomic Pathology at the University of British Columbia for histological identification. Histological examination of tissue sections confirmed that the cells were low columnar with distinct cell borders and 36 formed tubular structures, and that brush borders were absent. Sections stained with mouse monoclonal antibodies specific for epithelial membrane antigen (Dako, Santa Barbara, CA), low molecular weight cytokeratin (Enzo, New York), and high molecular weight cytokeratin (Enzo, New York) were positive for the first two but not the latter. These results were consistent with the cells being of collecting duct origin (257) 2.4.1.2 Preparation of IMCD for binding studies The IMCD cells obtained above were homogenized with a Caframo Stirrer (Wiatron, Ontario) for 2 minutes at 4°C. Low speed (1000 rpm) centrifugation for 5 minutes at 4°C sedimented the cellular debris, unbroken cells, and nuclei which were discarded. The remaining supernatant was kept in Tris-Tyrode buffer (1%) and subjected to high speed centrifugation of 15 000 rpm for 20 minutes at 4°C. The resulting supernatant was discarded and the pellet resuspended with 8.5 ml Tris-Tyrode (1%). This suspension was sonified at low speed for 5 seconds while on ice. A 20 ul aliquot of this suspension was taken for protein assay by the Lowry method. 2.4.2 Receptor binding studies Homogenized LMCD cells were used for binding studies to determine the ET receptor profile in normal and cardiomyopathic hamsters. 125I-ET-1 was prepared by the chloramine T method (258) and served as the hot ligand while ET-1 served as the cold ligand. BQ123 blocked the E T A receptor. The binding buffer used consisted of 1% Tris-Tyrode with 1 mM PMSF (pH 7.4). A standard curve for ET-1 was constructed with concentrations ranging from 0 to 100 ng/20 pi buffer. The experiments were carried out in duplicate in plastic culture tubes with a final volume of 240 pi (20 pi BQ123 (lOOng), 100 pi homogenate, 100 pi (120 000 cpm) hot label, 20 pi cold ET-1) incubated at room temperature for 30 minutes. BQ123 was allowed to completely bind all E T A subtype receptors in the homogenate before cold and hot ET-1 were added to the mixture. At the 37 end of the reaction, bound and unbound labelled peptides were separated by double antibody precipitation; 50 ul of 10% normal rabbit serum, 100 ul of goat anti-rabbit IgG, and 1 ml of 5% polyethylene glycol 8000 were added sequentially. The mixture was left at room temperature for 15 minutes before centrifugation at 3000 rpm for 30 minutes at 4°C. The supernatant was aspirated by vacuum suction and the pellet radioactivity was counted by an LKB minigamma counter (Wallec, Finland). These counts were entered into the computer Ligand Program (305) to calculate the dissociation constant (Kd) and maximum binding capacity (Bmax) of ET receptors for ET-1. 2.4.2.1 Acid-washed receptor studies To show that the ET receptors were not previously occupied by elevated levels of endogenous ET-1, the IMCD samples were first washed with acid to ensure any occupied cell-surface receptors were free. IMCD cells were isolated as described above and homogenized for 2 minutes on ice. The homogenate was then centrifuged at 1000 rpm for 10 minutes at 4°C. The supernatant was retained and centrifuged at 15 000 rpm for 16 minutes at 4°C. Then the resulting pellet was resuspended with 2 ml of sodium acetate buffer, pH 5.0, and allowed to stand for 10 minutes at room temperature. This suspension was again centrifuged as in the preceding step. A gentle rinse with 2 ml of 1% Tris-Tyrode was performed to remove any residual acid without disturbing the pellet. Finally, the pellet was resuspended with an appropriate amount of 1% Tris-Tyrode solution. The cells were now ready for binding studies as described above. 2.4.3 Stimulation Studies 2.4.3.1 ET-induced cGMP Accumulation Fresh, digested IMCD cells were pre-conditioned in IMDM culture medium with 10% FCS at 37°C for 1 hour prior to stimulation with ET-1. They were then spun down at 1000 rpm for 3 minutes at 4°C. The supernatant was discarded and the cells resuspended 38 with an appropriate volume of IMDM medium. A 100 pi aliquot of the cell suspension was transferred to each of sixteen labelled tubes in a 37°C water bath. Various concentrations (0 M, 10"9M, 10"8M, 10-7M) of ET-1 in stimulating medium (IMDM + 1 mM EBMX, 5 mM Na acetate, 3 mM lactic acid) were added to the labelled tubes and the mixture was incubated for 5 minutes. The tubes were then placed into an ice bath and the reaction stopped by the addition of 10 pi of 100 mM EDTA solution. Samples were frozen at -80°C for 15 minutes and thawed to rupture cell membranes, allowing the release of soluble cGMP. After a vigorous mixing, the freezing, thawing, and mixing procedure was repeated. Addition of 300 pi of Tris-EDTA (50 mM Tris, 4 mM EDTA) buffer brought the final volume to 0.5 ml and the sample was centrifuged at 3000 rpm for 30 minutes followed by acetylation. The supernatant was retained for cGMP measurement by radioimmunoassay and the pellet for protein content by the Lowry method. 2.4.3.2 Angiotensin II Stimulation of IMCD Cells IMCD cells of Long-Evans rats were extracted as above. The digested cells were grown to confluence in 5 ml of 37°C 1640 medium with 10% FCS in 6-well culture dishes. Incubation of IMCD cells with different concentrations (10-10M, IO"9 M, 10"8 M, IO"7 M, IO"6 M) of angiotensin II indicated it had little effect on ET receptor number in the IMCD cells. However, IMCD cells that were incubated with 10"11 M angiotensin II showed a distinct change in ET receptor density (Figure 2.1). This concentration of angiotensin II was chosen for further studies as it represents a more physiological concentration. Angiotensin II (10-11 M) was added to the cell cultures for 18 hours and grown at 37°C in 1640 medium plus 10% FCS. Cells were then harvested and centrifuged at low speed for 3 minutes. The supernatant was discarded and the pellet resuspended with 2 ml of the same medium. The mixture was centrifuged again and the supernatant aspirated. Two ml of Tris-Tyrode was then added and the suspension was homogenized for 2 minutes at 39 Figure 2.1 Effects of varying concentrations of angiotensin II on ET receptor density. Ratios of ET receptor density in IMCD cells treated with different concentrations of angiotensin II compared to control (no angiotensin II) are illustrated. At higher concentrations, little effect was seen on receptor number. Experiments were performed in duplicate or triplicate. 40 F r a c t i o n a r C h a n g e in ET Recepto r Dens i ty control 10-11 10-10 10-9 10-8 10-7 10-6 Angiotens in II (M) -41 4°C. The supernatant was collected and centrifuged at high speed (15 000 rpm) for 20 minutes at 4°C. The resulting supernatant was discarded and the pellet resuspended with an appropriate volume of 1% Tris-Tyrode. Receptor studies were performed as described above. Another set of experiments was conducted as outlined above except with one additional modification. The same experiment was repeated but in the presence of the angiotensin receptor antagonist, saralasin. 2.5 Statistics All data were expressed as the mean ± standard error of the mean (SEM). Paired student's t-test was used to determine differences between control versus experimental phases within the same animals. Unpaired student's t-test was used to compare the means between two populations. Linear regression analysis ( method of least squares) was used to estimate correlation coefficients. Differences were considered significant at the level of p<0.05. 42 Chapter Three Results, 3.1 Categorization of CM Hamsters Based on the grading system described previously (255,256), all CM hamsters included in this study presented with a moderate degree of heart failure (Grades. 2 and 3) with the exception of one manifesting severe heart failure. The former had subcutaneous oedema accompanied by pleural effusions, minor body cavity exudates, and liver congestion while the latter exhibited more gross oedema and organ pathology. Dilatation of the centrolobular veins and central cyanosis with bluish tongue were evident. Despite fluid retention, the CM animals were smaller in body size than the normal ones, with a mean body weight of 133 ± 4 g compared to 150 ± 1 g (p<0.0001) (Figure 3.1). In contrast, the total heart weight of the CM group was 735 ± 42 mg compared to 577 ± 6 mg in the normal controls (p<0.001) (Figure 3.2). There was overt hypertrophy of the atria and ventricles of the CM group. A comparison of the total heart/body weight ratios between the two groups showed a significant difference (CHF: 5.5 ± 0.3 mg/g, control: 3.8 ± 0.1 mg/g, p<0.000.1) (Figure 3.3). No difference was observed between the body weights of untreated (150 ± 1 g) and enalapril-treated (153 ± 2 g) control hamsters or between that of the untreated (133 ± 4 g) and enalapril-treated (132 ± 2 g) CM animals (Figure 3.1). Thus, enalapril had no effect on body mass. On the other hand, the total heart weights of the enalapril-treated CM hamsters (604 ± 1 3 mg) were significantly lower (p<0.01) than that of their untreated counterparts (735 ± 42 mg), and both these values were higher than those of either of the controls (Figure 3:2). Furthermore, there was a parallel decrease (p<0.01) in the total heart/body weight ratio of the enalapril-treated CM hamsters (4.5 ± 0.1 mg/g) compared to that of those without treatment (5.5 ± 0.3 mg/g) (Figure 3.3). 43 Figure 3.1 Body weights of normal and cardiomyopathic (CHF) hamsters prior to and after enalapril therapy (2.5 ug/kg/day s.c. for one week). Values are the mean ± SEM of 6 (untreated CHF) or 8 animals. *p<0.0001 vs. normal. 44 45 Figure 3.2 Heart weights of normal and cardiomyopathic hamsters prior to and following enalapril therapy (2.5 pg/kg/day s.c. for one week). Values are the mean ± SEM of 6 (untreated CHF) or 8 animals (untreated or treated normal, treated CHF). *p<0.05 vs. normal. **p<0.001 vs. normal. +p<0.01 vs. untreated CHF. 46 Total Heart Weight (mg) 1000 r 8 0 0 6 0 0 4 0 0 2 0 0 0 Norma l No rma l C H F + I C H F Unt rea ted Ena lap r i l - t r ea ted 47 Figure 3.3 Heart weight/body weight ratios of normal and cardiomyopathic hamsters prior to and after enalapril therapy (2.5 ug/kg/day s.c. for one week). Values are the mean ± SEM of 6 (untreated CHF) or 8 animals (untreated or treated normal, treated CHF). *p<0.0001 vs. normal. +p<0.01 vs. untreated CHF. 48 49 3.2. In vivo Studies 3.2.1 Golden Syrian hamsters as a time control Table 3.1 contains haemodynamic and renal excretion indices as a function of time. Values represent three collection phases as described in the methods section. There was a slight tendency for mean arterial pressure (MAP) to decline (from 126 ± 6 to 120 ± 6 to 111 ± 4 mmHg, p<0.05) from phase I to phase III while glomerular filtration rate (GFR) remained quite stable ( from 0.53 ± 0.07 to 0.53 ± 0.09 to 0.52 ± 0.07 rnl/min). With the exception of an increase in the urine flow rate during the last phase (23.2 ± 3 . 4 ul/min, p<0.05), there was no significant difference in the patterns of all other excretory parameters. 3.2.2 Untreated CM hamster studies 3.2.2.1 Baseline and ANP-phase haemodynamic characteristics The haemodynamic characteristics of CM and normal hamsters are summarized in Table 3.2. The baseline MAP of CF£F hamsters (92 ± 3 mm Hg) was significantly lower (p<0.01) than those of controls (119 ± 8 mm Hg) as were the haematocrit (47 ± 1 % versus 52 ± 1 %, p<0.01) and plasma protein concentration (4.3 ± 0.2 g/dl versus 4.9 ± 0.1 g/dl, p<0.04). The GFR, however, did not differ significantly between the two groups (CHF: 0.55 ± 0.12 ml/min; control: 0.40 ± 0.07 ml/min). Administration of exogenous ANP resulted in a 25% drop in MAP of both groups (CHF, 70 ± 4 mm Hg; control, 88 ± 6 mm Hg), but no significant change in GFR (CHF: 0.52 ± 0.08 ml/min; control: 0.55 ± 0.10 ml/min). 3.2.2.2 Renal effects of ANP administration Baseline urine flow (4.2 ± 1.0 versus 6.0 ± 1.6 ul/min) as well as absolute and fractional excretion rates of sodium (1.42 ± 0.41 versus 1.88 ± 0.50 %; 2.3 ± 0.5 versus 1.9 ± 0.3 %) and water (1.1 ± 0.2 versus 0.8 ± 0.1 %) did not differ significantly between the 50 Table 3.1 Haemodynamic and renal excretory function in golden Syrian hamsters (n=ll) Phase I Phase II Phase III MAP, mm Hg 126 ± 6 120 ± 6 111 ± 4 * GFR, ml/min 0.53 ±0.07 0.53 ±0.09 0.52 ±0.07 P N a mmol/1 142.2 ± 1.3 140.5 ± 1.8 139.8 ± 1.6 UV, ul/min 12.7 ±2.5 16.3 ±3.9 23.2 ±3 .4* U N a V, mmol/min 4.43 ± 0.68 4.36 ±0.42 4.81 ±0.54 F E N a , % 6.6 ± 1.2 6.4 ±0.5 6.9 ±0 .7 F E H20> % 2.7 ±0.5 3.5 ± 1.0 3.9 ±0.6 Values are means ± SEM. MAP, mean arterial pressure; GFR, glomerular filtration rate; P N a , plasma sodium concentration; UV, urine flow; U N a V, urinary sodium excretion; F E N a , fractional excretion of sodium; F E H 2 G , fractional excretion of water. *p<0.05 vs. phase I. 51 Table 3.2 Haemodynamic and renal excretory function in response to ANP Control Group (n = 8) CHF Group (n= 6) Baseline ANP Baseline ANP MAP, mmHg 119 ± 8 88 ± 6 * 9 2 ± 3 f 70 ± 4*t Haematocrit, % 52 ± 1 ND 4 7 ± If ND Plasma protein, g/dl 4.9 ±0.1 ND 4.3 ± 0 . 2 | ND GFR, ml/min 0.40 ±0.07 0.55 ±0.10 0.55 ±0.12 0.52 ±0.08 GFRa, ml/min/kg 2.7 ±0.5 3.3 ± 0.6 3.9 ±0.8 3.5 ±0 .8 UV, ul/min 4.2 ± 1.0 21.4 ±3.9* 6.0 ± 1.6 8.9±2.5f U N a V, mmol/min 1.42 ±0.41 4.36 ±0.74* 1.88 ±0.50 2.08±0.64f F E ^ , % 2.3 ±0.5 5.9 ±0.8* 1.9 ±0.3 2 .7±0.5f F E H 2 0 ' 0 / 0 1.1 ±0 .2 3.2 ±0 .4* 0.8 ±0.1 1 .7±0 .3+ f UCGMP> pmol/ml 7.6 ± 1.3 33.4 ±4 .3* 11.7 ± 1.9 23.8 ± 3.5Jt Values are means ± SEM. ANP, atrial natriuretic peptide; MAP, mean arterial pressure; GFR, glomerular filtration rate; GFRa, glomerular filtration rate normalized to body weight; UV, urine flow; U N a V, urinary sodium excretion; F E N a , fractional excretion of sodium; F E H 2 G , fractional excretion of water; U ^ p , urinary cyclic guanosine monophosphate; ND, not determined. *p<0.002 vs. baseline. +p<0.02 vs. baseline. fp<0.01 vs. control. Jp<0.03 vs. baseline. 52 normal and CHF hamsters (Table 3.2 and figures 3.4 and 3.5). Urinary cGMP levels (7.6 ± 1 . 3 versus 11.7 ± 1.9 pmol/ml) were also not significantly different (Figure 3.6). However, there was a marked difference in the response to ANP infusion by the two groups. In the control group, there were significant rises in all of the parameters (urine flow: 21.4 ±3 .9 ul/min, p<0.02, F E N a : 5.9 ± 0.8%, p<0.02, F E H 2 0 : 3.2 ± 0.4%, p<0.02, cGMP. 33.4 ± 4.3 pmol/ml, pO.OOl) but only marginal increases in the CM animals (urine flow: 8.9 ± 2.5 ul/min, F E N a : 2.7 ± 0.5%, F E H 2 Q : 1.7 ± 0.3%, cGMP: 23.8 ± 3.5 pmol/ml, p<0.03) (Figures 3.4, 3.5, 3.6 and Table 3.2). The natriuretic response to ANP by the normal animals occurred despite a decrease in MAP and no increase in GFR. 3.2.2.3 Heart failure severity and urinary excretion rates Retention of salt and water as well as a diminished response to ANP were evident in the CM hamsters. There was a significant negative correlation between the degree of heart failure and ANP-induced fractional sodium excretion (Figure 3.7A) and of ANP-induced cGMP excretion (Figure 3.7B). A more severe degree of heart failure was expressed as a larger heart weight/body weight ratio. A negative correlation was also observed between the stage of heart failure and ANP-evoked fractional water excretion although this did not achieve statistical significance. 3.2.3 Enalapril-treated CM hamster studies 3.2.3.1 B aseline and ANP-phase haemodynamic characteristics The baseline MAP and haematocrit of CHF animals improved to 114 ± 5 mmHg and 52 ± 1 % (Table 3.3) after chronic enalapril treatment although these values were slightly lower than that of the control group (123 ± 8 mmHg; 54 ± 1 %). The plasma protein concentration (4.8 ± 0.2 g/dl) of CHF hamsters was no longer diluted when compared to 53 Figure 3.4 Fractional excretion of sodium in normal and cardiomyopathic hamsters. Baseline values and responses to acute ANP infusion (1 pg/kg/min) are shown. Data are expressed as the mean ± SEM of 6 (CHF) or 8 (control) animals. *p<0.002 vs. respective baseline. **p<0.01 vs. respective normal phase. 54 55 Figure 3.5 Fractional excretion of water in normal and cardiomyopathic hamsters. Baseline values and responses to acute ANP infusion (1 ug/kg/min) are shown. Data are expressed as the mean ± SEM of 6 (CHF) or 8 (control) animals. *p<0.002 vs. respective baseline. +p<0.02 vs. respective baseline. **p<0.01 vs. respective normal phase. 56 57 Figure 3.6 Urinary cGMP excretion in normal and cardiomyopathic hamsters. Baseline values and the responses to acute ANP infusion (1 ug/kg/min) are shown. Data are expressed as the mean ± SEM of 6 (CHF) or 8 (control) animals. *p<0.001 vs. respective baseline. **p<0.03 vs. respective baseline. 58 59 Figure 3.7 Relationships between heart weight/body weight ratios and excretory indices in hamsters. (A) Negative correlation between heart/body weight ratios and the fractional excretion of sodium, (B) Negative correlation between heart/body weight ratios and the urinary excretion of cGMP. Open symbols = control animals; closed symbols = CM animals. (n=14). 60 61 B. 62 Table 3.3 Haemodynamic and renal excretory function of enalapril-treated hamsters in response to ANP . Control Group (n -8) CHF Group (n= 8) Baseline ANP Baseline ANP MAP, mm Hg 123 ± 8 88 ± 5 * 114 ± 5 81 ± 4 * Haematocrit, % 54 ± 1 58± 1* 52 ± 1 53 ± It Plasma protein, g/dl . 5.1 ±0.1 4.8 ±0.3 4.8 ±0.3 4 .5±0 .1* GFR, ml/min 0.52 ± 0.08 0.61 ±0.04 0.65 ± 0.04 0.59 ±0.03 GFRa, ml/min/kg 3.4 ±0.6 ' 4.0 ± 0.3 4.8 ±0.2$ 4.3 ±0.3 UV, pl/min 8.5 ± 1.5 21.4 ± 1.1* 2 1 . 6 ± 2 . 3 | 23.3 ±2 .2 U N a V, mmol/min 2.53 ±0.58 5.30 ±0.39* 6.06±0.55f 6.07 ±0.78 F E N a , % 3.4 ±0.5 6.7 ±0 .5* 5 .5±0.5f 7.5 ±0.6* F E H20> % 1.7±0.2 3.8 ±0 .3* 2 .7±0.3f 3 .9±0 .1* U c G M p , pmol/ml 6.2 ±0 .6 33.8 ±2 .3* 10.9± l . l f 39.1 ± 1.6* Values are mean ± SEM. Abbreviations are as in Table 3.2. *p<0.01 vs. baseline. fp<0.01 vs. control. Jp<0.02 vs. control. 63 the control value (5.1 ± 0.1 g/dl). No difference was displayed in the GFR of the CHF and controls (0.65 ± 0.04 ml/min vs. 0.52 ± 0.08 ml/min, respectively). A larger than 25% decrease in MAP occurred in both groups following ANP infusion (control: 88 ± 5.4 ; CHF: 81 ± 4 mmHg) while GFR remained unaffected. The haematocrit was raised to a greater extent in the control than the CHF group whereas the plasma protein concentration fell slightly in both cases after exogenous ANP (Table 3.3). 3.2.3.2 Renal effects of ANP administration In comparison with the control group (8,5 ± 1 . 5 ul/min), enalapril therapy resulted in a higher basal urine flow in the CHF group (21.6 ± 2.3 ul/min, p<0.01) (Table 3.3). This was also accompanied by-higher fractional and absolute rates of sodium excretion (5.5 ± 0.5 versus 3.4 ± 0.5 %; 6.06 ± 0.55 versus 2.53 ± 0.58 mmol/min, p<0.01) as well as of fractional water excretion (2.7 ± 0.3 versus 1.7 ± 0.2 %, p<0.01) (Figures 3.8 and 3.9). Baseline cGMP levels of the CHF animals (10.9 ± 1.1 pmol/ml) were significantly higher (p<0.01) than those of normal ones (6.2 ± 0 . 6 pmol/ml) (Figure 3.10). The effects of ANF infusion were no longer blunted in the CHF group with enalapril. The ANP-induced levels of the above variables, with respect to the CHF group (urine flow: 23.3 ± 2.2 Ul/min, F E N a : 7.5 ± 0 . 6 %, F E H 2 Q : 3.9 ± 0.1%, cGMP: 39.1 ± 1.6 pmol/ml), were at least comparable, if not marginally greater, than those of the normal controls (21.4 ± 1.1 ul/min, 6.7 ± 0.5%, 3.8 ±0.3%, cGMP: 33.8 ± 2.3 pmol/ml) (Table 3.3 and figures 3.8, 3.9,3.10). 64 Figure 3.8 Fractional excretion of sodium in normal and cardiomyopathic hamsters following enalapril therapy (2.5 pg/kg/day s.c. for one week). Baseline values and responses to acute ANP infusion (1 pg/kg/min) are shown. Data are expressed as the mean ± SEM of 8 animals. *p<0.01 vs. respective baseline. **p<0.01 vs. normal baseline. 65 66 Figure 3.9 Fractional excretion of water in normal and cardiomyopathic hamsters chronically treated with enalapril (2.5 ug/kg/day s.c. for one week). Baseline values and responses to acute ANP infusion (1 ug/kg/min) are shown. Data are expressed as the mean ± SEM of 8 animals. *p<0.01 vs. respective baseline. **p<0.01 vs. normal baseline. 67 6 8 Figure 3.10 Urinary excretion of cGMP in normal and cardiomyopathic hamsters after chronic enalapril therapy (2.5 ug/kg/day s.c. for one week). Baseline values and responses to acute ANP infusion (1 ug/kg/min) are shown. Data are expressed as the mean ± SEM of 8 animals. *p<0.01 vs. respective baseline. **p<0.01 vs. normal baseline. 69 70 3.2.3.3 Heart failure severity and urinary excretion rates Animals treated with the ACE inhibitor, enalapril, did not exhibit the same relationship between heart failure severity and ANP-induced fractional sodium and water excretion (Figures 3.11A and 3.1 IB) as those untreated. The negative correlation between the degree of heart failure and ANP-induced cGMP excretion was also abolished upon enalapril therapy. (Figure 3.11C). 3.3 In vitro Studies 3.3.1 Receptor binding assays 3.3.1.1 ET receptor distribution in untreated CM hamsters Total ET receptor number (Bmax) and E T B receptor number were much lower (p<0.005, p<0.003, respectively) in CM hamsters (532 ± 77; 214 ± 26 fmol/mg protein, respectively) when compared to normal controls (959 ± 154; 483 ± 88 fmol/mg protein, respectively) (Figure 3.12A). Blocking of ET A receptors with receptor-specific BQ123 revealed a 50 % distribution of E T A (476 ± 81 fmol/mg protein) and of E T B (483 ± 88 fmol/mg protein) receptors in the IMCD of normal hamsters (Figure 3.12B). However, the percentage of A and B receptors in the. CHF animals was 60% and 40%, respectively, with 318 ± 57 fmol/mg protein being the former and only 214 ± 26 fmol/mg protein comprising the latter. The number of A receptors did not differ significantly (p=0.07) between the two groups but the number of the B subtype did (p<0.003). Thus the downregulation of ET receptors was attributed more to the downregulation of E T B receptors. Although the maximum binding capacity, Bmax, was depressed in the IMCD cells of CHF hamsters, the affinity of the receptors did not differ between the two groups. The dissociation constant, Kd, of control animals was 0.496 ± 0.087 nM while that of CHF animals was 0.675 ± 0.147 nM, p = 0.16. The affinity of the E T B receptors was similar between the two groups: normal Kd = 0.203 nM, CHF Kd = 0.302 nM, p - 0.15. These values fall within the range of those reported by others. Please see appendix. 71 Figure 3.11 Absence of correlative relationships between heart weight/body weight ratios and excretory indices following enalapril therapy (2.5 pg/kg/day s.c. for one week). Negative correlations were abolished between heart/body weight ratios and (A) fractional sodium excretion, (B) fractional water excretion, and (C) urinary cGMP excretion. Open symbols = control animals; closed symbols = CM animals. (n=16). 72 F E Na (%) 10 r • o 0 -A. 0 0 0 0 0 • r= -5.8E-02 p= ns J I I I J L_ I _l L 3 3.2 3.4 3.6 3.8 4 4.2 4.4 4.6 4.8 5 5.2 5.4 Hear t W t / Body Wt (mg/g) 73 B. F E H 2 0 (%) 6 5 4 3 2 0 -0 0 • — A. • • — 4 — - i 2 • _ o o 0 - r= 9 . 4 E - 0 2 i i i i p=ns I I . i i i i i i 3 3.2 3.4 3.6 3.8 4 4.2 4.4 4.6 4.8 5 5.2 5.4 Heart W t / B o d y Wt (mg/g) 74 c. c G M P (pmol/ml) 40 r 35 -• • 20 h 0 • 15 -r= 0.21 p= ns -j Q I I I I I I I I I I I I I 3 3.2 3.4 3.6 3.8 4 4.2 4.4.4.6 4.8 5 5.2 5.4 Heart W t / B o d y Wt (mg/g) 75 Figure 3.12 Effect of CFIF on the density of ET receptor distribution in cardiomyopathic hamster IMCD cells. (A) Comparison of total ET receptor and E T B receptor density and (B) comparison of E T A and E T B receptor density in normal and cardiomyopathic hamsters. Values are the mean ± SEM of 8 animals. Experiments were performed in duplicate as described in Materials and Methods. *p<0.005 vs. normal. **p<0.003 vs. normal. 76 A. fmol/mg protein 1200 r 1000 800 600 400 200 0 Normal Normal CHF E T III E T B 77 78 3.3.1.2 ET receptor distribution in enalapril-treated CM hamsters After enalapril therapy, the total number of ET receptors in CHF animals was 1016 ± 140 fmol/mg protein which was similar to the 1040 ± 2 1 6 fmol/mg protein in normal controls (Figure 3.13A). Addition of BQ123 showed that there was no difference in the density of E T B receptors between normal and CM animals (559 ± 72 fmol/mg and 582 ± 110 fmol/mg protein, respectively) (Figure 3.13A), and the percentage of each receptor subtype was roughly 50% (Figure 3.13B). This indicates there was no longer downregulation of ET receptors in the CHF group. Again, the affinity of the receptors was not different between groups. The Kd for the normal controls was 0.983 ± 0.228 nM and 1.162 ± 0.189 nM for the CHF animals, p = 0.28. Similarly, the Kd for E T B receptors was not significantly higher in the CHF group (0.712 ± 0.169 nM vs. 0.666 ± 0.201 nM, p = 0.43). 3.3.1.3 Effect of CHF on ET receptor density The number of ET receptors declined as the severity of CHF became more pronounced. The inverse relationship was highly significant in reference to the total ET receptor density (Figure 3.14A) and a parallel interdependence, was observed with regard to the ET A or E T B receptor density (Figures 3.14B and 3.14C). These negative correlations were abolished (Figures 3.15A and 3.15C) or rendered non-significant (Figure 3.15B) in animals treated with enalapril, suggesting enalapril prevents downregulation of ET receptors. 3.3.1.4 Effect of acid-washing on receptor density Downregulation of ET receptors in CHF tissue was still observed after an acid wash to remove ligands from previously-occupied receptors. The numbers of total ET (725 ± 67 fmol/mg protein) and E T B receptors (419 ± 100 fmol/mg protein) were significantly lower than those on normal cells (1029 ± 91 fmol/mg protein and 686 ± 70 fmol/mg protein, 79 Figure 3.13 Effect of enalapril treatment (2.5 mg/kg/day s.c. for one week) on the density of ET receptor distribution in IMCD cells of cardiomyopathic hamsters. (A) Comparison of total ET receptor and E T B receptor density and (B) comparison of E T A and E T B receptor density in normal and cardiomyopathic hamsters. Values are the mean ± SEM of 8 animals. Experiments were performed in duplicate. 80 A. fmol/mg protein 1 4 0 0 r 1200 1000 800 600 400 200 0 .Ji Normal Normal CHF C H F E T E T B 81 Figure 3.14 Relationship.between heart weight/body weight ratios and ET receptor density in IMCD cells. Negative correlations are apparent between the heart/body weight ratios and (A) total ET receptor number, (B) E T A receptor number and, (C) E T B receptor number. Open symbols = control animals; closed symbols = CM animals. (n=16). 83 84 85 86 Figure 3.15 Absence of correlative relationships between heart weight/body weight ratios and ET receptor distribution in IMCD cells following enalapril treatment (2.5 mg/kg/day s.c. for one week). Negative correlations between heart/body weight ratios and (A) total ET receptor density, (B) E T A receptor density, and (C) E T B receptor density were abolished after chronic enalapril therapy. Open symbols = control animals; closed symbols = CM animals. (n=16). 87 fmol /mg protein 2000 r o -0. 1500 ' 1000 500 -0 0 0 0 0 . o 0 • 5.2E-03 p= ns J i i i i i i 3 3.2 3.4 3.6 3.8 4 4.2 4.4 4.6 4.8 5 5,2 5.4 Heart W t / Body Wt (mg/g) 88 B. 1200 fmol /mg protein . ' 1000 800 600 -400 200 0 0 0 0 0 r= -0.10 p= ns 0 0 0. _l J_ J L J I I I I 3 3 . 2 3 . 4 3 . 6 3 . 8 . 4 4 .24 .44 .64 .8 5 5 .25 .45 .65 .8 6 Heart W t / Body Wt (mg/g) 89 c . fmol/mg: p ro te in ' 1 4 0 0 r 1200 1000 800 600 h 400 200 0 0 0 r= 0.12- p= ns. J L J L 3 3.23:43.63.8 .4,4.24,44.64.8 5 6 .25 ,45 .65 .8 6 Hear t W t / Body Wt (mg/g) 90 p<0.01 and p<0.05, respectively) (Figure 3.16A). The number of E T A receptors was comparable between the two groups (306 ± 62 fmol/mg protein vs. 343 ± 42 fmol/mg protein) (Figure 3.16B). 3.3.2 Stimulation studies 3.3.2.1 ET-stimulated cGMP accumulation in IMCD cells Basal levels of cGMP production were 6.34 ± 0.37 fmol/pg/5 min in cells derived from CHF IMCD. This was significantly less than the 7.60 ± 0.35 fmol/pg/5 min produced in IMCD cells of normal controls. Cyclic GMP accumulation was dose-dependent in the cells of both groups of animals. At each dose of ET given, radioimmunoassay detected a lower level of cGMP in the CHF cells when compared to the normal control cells. Figure 3.17 illustrates the dose-dependent effect of ET stimulation. 3.3.2.2 Effects of angiotensin stimulation on IMCD cells Figure 3.18A shows that angiotensin II at a concentration of 1011 M mediated downregulation of ET receptors in IMCD tissue (p<0.02 versus control) and that E T B receptors were substantially fewer by 29% (p<0.003 versus control). Nearly 80% of the ET receptors in these Long-Evans rat IMCD cells were of the B variety. Endothelin A receptors, of which there were about 20%, were downregulated by 39% (Figure 3.18B). Furthermore, the affinity of the ET receptors increased (Kd decreased to 1.16 ± 0.38 nM from 1.98 ± 0.43 nM, (p<0.001)) after angiotensin II treatment. Similarly, the E T B receptor affinity increased (Kd decreased from 1.43 ±0 .31 nM to 0.932 ± 0.346 nM, (p<0.02)). Cells co-incubated with angiotensin II and saralasin did not exhibit this downregulation (Figures 3.19A and 3.19B) nor the change in affinity. In fact, a comparison of receptor number in control and saralasin/angiotensin II - stimulated cells showed no difference between the two (Figures 3.20A and 3.20B) and the dissociation constants were quite comparable (1.98 ± 0.43 nM vs. 1.64 ± 0.18 nM (p=0.25) and 91 Figure 3.16 Effect of acid washing on ET receptor density. IMCD cells were washed with sodium acetate (pH 5.0) for 10 minutes and then rinsed with Tris-Tyrode before completion of receptor binding studies as delineated in Materials and Methods. Removal from previously occupied ET receptors of endogenous ET shows a true downregulation of total ET receptor and ET B receptor numbers in cardiomyopathic hamsters (A). Downregulation of ET A receptors was minimal in cardiomyopathic hamsters (B) Values are expressed as the mean ± SEM of 5 animals. Experiments were performed in duplicate. *p<0.01 vs. normal. **p<0.05 vs. normal. 92 93 94 Figure 3.17 ET-stimulated cGMP accumulation in IMCD cells. Cells from either normal or cardiomyopathic hamsters were incubated with different concentrations of ET-1 for 5 minutes and cGMP was measured by radioimmunoassay as described in Materials and Methods. Values are the mean ± SEM of 5 animals with experiments performed in quadruplicate. *p<0.01 vs. normal at respective ET-1 concentrations. 95 c G M P (fmol/ug/15 min) 12 r 0 1 0 - 9 1 0 - 8 Endothel in (M) 1 0 - 7 - e - Normal — 0 - CHF 96 Figure 3.18 Effect of low-concentration angiotensin II on ET receptor density in the IMCD cells of Long-Evans rats. (A) Downregulation of total ET and E T B receptors and (B) total and E T A receptors following incubation of IMCD cells with IO - 1 1 M angiotensin II. Ang II, angiotensin II. Values are the mean ± SEM of 5 experiments. *p<0.02 vs. control. **p<0.003 vs. control. 97 A. 3000 fmol/mg protein 2500 2000 1500 1000. 500 0 Control Control Ang Ang II ET E T B 98 B. 3000 fmol/mg protein 2500 2000 1500 1000 500 0 Control Control Ang II Ang ET I I ETA 99 Figure 3.19 Prevention by saralasin of angiotensin II-induced ET receptor downregulation. IMCD cells from Long-Evans rats co-incubated with saralasin (10-9 M) and angiotensin II (IO1 1 M) show no downregulation of (A) total ET and E T B receptors or of (B) total ET and E T A receptors. Ang II, angiotensin II; Ang + Sarlle, angiotensin II + saralasin. *p<0.02, **p<0.05 vs. angiotensin II alone. 100 A. 2500 fmol/mg protein 2000 1500 1000 500 0 Ang II ~ i — — i — — r Ang II . Ang + S a r l l e A n g + Sar i ie E T E T B 101 B. 2500 fmol/mg protein 2000 1500 1000 500 0 Ang Ang II Ang + Sar l le Ang + Sari Ie ET ETA 102 Figure 3.20 ET receptor distribution in control IMCD cells and in saralasin-treated IMCD cells of Long-Evans rats. (A) Total ET receptor and E T B receptor densities and (B) total ET and E T A receptor densities are similar in untreated cells and in cells co-incubated with saralasin (10-9M) and angiotensin II (1011 M). Ang II + Sarlle, angiotensin II + saralasin. Values are the mean ± SEM of 6 experiments. 103 A, 3000 fmol/mg protein 2500 2000 1500 1000 500 0 Control Control Ang + Sar l le Ang + Sar l le E T E T B 1 0 4 B. 3000 fmol/mg protein 2500 h 2000 h 1500 1000 -500 -0 Control Control Ang + Sa r l l eAng + Sar i Ie ET ETA 105 1.43 ± 0.31 nM vs. 1:48 ± 0.37 nM (p=0.46), control ET and ETBvs. saralasin/angiotensin II ET and ET B , respectively). 106 Chapter Four Discussion Through the use of ACE inhibitors, the cycle of CHF can be interrupted via inhibition of angiotensin II-mediated vasoconstriction. In addition, this blockade would impede the sodium and water retention normally induced by angiotensin II and aldosterone. It has been demonstrated that ACE inhibitors can prolong the lives of patients with CHF (250), although the exact mechanism has not been determined. This study was designed to elucidate how enalapril could improve renal function in CHF hamsters by combining in vivo and in vitro methods. By drawing comparisons between the in vivo and in vitro characteristics of untreated with enalapril-treated CM hamsters, the mechanism of ACE inhibition could be derived. The study was based on the premise that the increased levels of angiotensin II would stimulate ET production which, in turn, would downregulate ET receptors and thereby diminish the natriuretic effects of ET. This is the first line of evidence showing that ET and its receptors play a major role in the neurohormonally-integrated excessive fluid retention in CHF. 4.1 Cardiomyopathic Hamster as the CHF Model of Choice Cardiomyopathic hamsters were initially introduced over thirty years ago by Homburger et al (259). Establishment of various strains (eg. Bio 14.6, Bio FIB, Bio TO-2, UM-X7-1) of CM hamsters was derived through the cross-breeding of diseased animals with unrelated hamsters followed by the inbreeding of affected animals in the F2 generation. Transmission of the hereditary heart and skeletal muscle disorder is through an autosomal recessive gene and animals homozygous for this allele show complete phenotypic penetrance. Focal myocytolytic necrosis develops within 30 days post-partum and maximal degeneration and necrosis is seen 30 to 60 days thereafter (259-261). Scarring, hypertrophy, and dilatation of the heart occur, with heart failure ensuing by about 200 days after birth. The estimated lifespan of CM hamsters is 320 to 360 days although many 107 die at a younger age (250 days). Classic CHF is the cause of death in most animals, with multiple organ congestion generally manifest. In choosing a model to study various aspects of CHF, the obvious choice would be human subjects. However, heterogenicity of the disorder (with respect to ethnic diversity and to cause and development in-patients) and ethical considerations place restrictions on their use. Consequently, researchers have turned to animal models of CHF, of which there are several. Examples include dogs with tricuspid insufficiency (212), inferior vena caval constriction (182), or rapid ventricular pacing (67) and rats with high output CHF by aorto-caval shunting (263) or ventricular infarct by coronary artery ligation (264). However, these models are also heterogeneous and the development of CHF varies in relation to the insult to the heart. Furthermore, these models necessitate the use of complicated and time-consuming procedures to prepare the animals for experimentation, and thereby may produce artificial disturbances in the animals which could lead to unreliable results. The use of CM hamsters can circumvent these impediments to some extent as the disorder is fairly homogeneous with a predictable clinicopathological course in these animals. Another advantage is the natural occurrence of the disorder rather than by surgical induction: Cardiomyopathic hamsters of the UM-X7.1 line were employed in this study. Hamsters of a specific age (290 days) were used to minimize any differences in developmental stages of CHF. The hamsters use in this study had a moderate level of heart failure. The degree of heart failure was assessed based on the grading scale established previously as discussed in the Materials and Methods section. This grading system can assure homogenicity of the hamsters by excluding those animals identified as having asymptomatic CHF. In addition, in view of previous studies applying this grading system, comparison of current and previous findings is facilitated. 108 4.2 Physical Characteristics of the Hamsters The total body weights of the CM hamsters were significantly lower than those of the age-and sex-matched control animals. One explanation for this lower weight may be due to poorer feeding and decreased nutrient delivery to body tissues secondary to inadequate circulatory function. Total heart weights between the CM and normal hamsters also differed significantly but in this instance, it was the former that had higher heart weights. That the increase in heart weight was in accordance with body size can be ruled out since the diseased animals displayed higher total heart/body weight ratios. This increased heart weight is consistent with previous reports (265-267). Higher heart weights probably reflect congestion of the heart as well as the increase in cell mass (hypertrophy), features characteristic of CHF. That animals treated with enalapril presented with lower heart weights and lower heart/body weight ratios indicates that ACE inhibition can slow or reverse the process of compensatory cardiac hypertrophy, although not completely. This growth suppression probably stems from the reduced proliferative effects of diminished angiotensin II levels. 4.3 The Importance of a Time Control It is necessary to determine whether the physiological functions of animals undergoing anaesthesia and surgical manipulations remain stable with time. The use of this type of control ensures that any differences in physiological parameters observed during the course of an experiment can be attributed to the treatments applied rather than to changes that occur as a result of systemic perturbations brought on by prolonged anaesthesia. In this study, haemodynamic and renal indices of function were relatively unaltered over a 4-hour period. Although there was a mildly significant decrease in MAP, the GFR was maintained by the kidneys. The increase in urine flow rate and the high percentage of sodium excretion could be ascribed to activation of compensatory mechanisms in response 109 to volume expansion induced by a high rate of saline infusion. Nevertheless, the data indicate the preparations were stable. 4.4 Haemodynamic and Renal Function in Untreated CM Hamsters Mean arterial pressures (MAP) were markedly lower in the CM hamsters in comparison with the normal control group. A lower MAP was not unexpected as the. effective circulating volume is usually diminished in CHF and this is in agreement with observations made by others (67,182,263,268,269). Glomerular filtration was relatively preserved in the heart failure animals and thus was similar to that in the control group. In heart failure dogs and rats, the GFR was reduced when compared to normal animals (67,182,268,269) which could be due to differences in species, stage of CHF, or methodology. Both haematocrit and plasma protein concentration in the diseased animals were also decreased relative to that in the controls, reflecting extracellular fluid volume expansion. Infusion of ANP had similar effects on MAP and GFR in CHF and control hamsters; there was a dramatic drop in MAP with no significant change in GFR. The steady GFR indicated the preparation was stable. Similarly, Isnard and colleagues (270) saw MAP fall in both control and CHF rabbits after ANP dosing while Koepke et al (212) also found no differences in the effects of ANP infusion on haemodynamic variables between control and heart failure dogs. The vasodilating effects of ANP were clearly evident in these studies. Similar to the results of studies in other animals (212,268,269), baseline measurements of urine flow and sodium and water excretion were diminished in CHF hamsters. The lower excretion rates may be explained by the lower MAP which may have resulted in poorer renal blood flow and perfusion pressure. Urinary cGMP levels at baseline were also comparable between normal and CM animals, with a nonsignificant greater value in the former. In agreement with the finding by Isnard et al (270), this higher cGMP production 110 by the sick animals could reflect stimulation of ANP receptors by higher endogenous levels of ANP. According to Wong et al (271), urinary cGMP is a biological marker of renal activity of ANP. These values imply that renal function in the CHF hamsters was relatively preserved, although there was a tendency toward mild electrolyte retention. Attenuated responses to exogenous ANP in animals with CHF have been repeatedly reported (212,268,269) and this study further confirmed this observation. The measurements of all the above parameters following ANP administration were significantly lower in CHF versus normal animals. The reasons for the blunted response are unclear. Several explanations have been proposed. It is possible that the attenuated renal effects may be due to the overactivation of the RAAS (270) or other neuroendocrine systems since the antagonism between ANP and angiotensin II or AVP has been documented (217,220). Another hypothesis involves increased ANP degradation or incomplete processing, resulting in loss of biological activity; Isnard et al (270) showed that the ANP structure was intact in CHF rabbits. Others have claimed that renal resistance to ANP is due to downregulation of ANP receptors after chronic plasma ANP elevation since decreased receptor density has been observed in a number of studies (264,272,273). On the other hand, there have also been reports of no ANP receptor downregulation occurrence in CHF and that renal resistance is not due to second messenger defects (270, 274). A limitation imposed by most of the previously reported studies is the lack of investigation into the receptor subtype(s) responsible for regulation. The discrepancies between the published reports may be partially accounted for by the possibility of species or severity-dependent differences. The negative correlation between the degree of heart failure and response to ANP found in this study supports the concept that severity of CHF is an important determinant in responsiveness to ANP activity. Since the available data cannot be reconciled between the various studies, it is a likely possibility that other factors 111 are contributing to the observed results, depending on the model chosen and the degree of CHF. Perhaps other neuroendocrine systems, activated according to the stage of heart failure, modulate the effects of ANP. Endothelin is a definite contender in this regard as its plasma levels correlate with the extent of CHF (74) and is known to affect ANP release (205-209). 4.5 ET Receptor Distribution in the IMCD of Untreated CM Hamsters Consistent with previous reports (73,254), downregulation of ET receptors in CHF was observed in this study; the IMCD of the CHF hamsters exhibited a 45% decline in receptor number in comparison to the normal controls. Blockade of E T A receptors with the antagonist BQ123 indicated that there was roughly equal distribution of the A and B receptor subtypes in the normal controls. The pattern of distribution in the CHF animals favoured the expression of the A subtype by about 10%. The downregulation in the earlier studies was selective; decreased density of receptors was seen in mesenteric arteries but not in myocardium of CHF rats (254) while downregulation occurred in the ventricles and kidneys of CHF rabbits. However, neither the receptor subtype distribution in these studies nor the nephron segments in the latter were specified and thus offer no point of reference for comparison. Alternatively, collation of the present data with characterization studies in canines (158) and rats (159,275) shows a deviation of ET receptor distribution in the medulla of CM hamsters from these species. Canine and rat species show a preponderance of the E T B subtype in medullary preparations. Another study (276) examining the ET binding sites in CM hamsters found no difference between these and matched controls in the amount of ET-1 binding in a number of tissues. However, the experiments do not allow for direct comparison as they were conducted in another strain and at a much younger (90 days) age. Neither the total receptor affinity nor the E T B receptor affinity in the CHF animals differed significantly from that in the controls 112 in our study. In contrast, the binding affinity of ET-1 receptors was increased after downregulation in a rabbit model of heart failure as compared to the control (73). Although the reason for the change in binding affinity is uncertain, it may represent a mechanism in which the decrease in receptor number is compensated by an enhanced binding affinity. The revelation that ET receptors are downregulated in the present study lends support to the hypothesis that ET levels are raised in CHF and thus effect downregulation and/or desensitization of its receptors. Albeit that levels of ET were not measured in this investigation, it has been widely documented that ET concentrations are raised in the plasma of CHF patients (67-77) as well as in the plasma and hearts of CM hamsters (277). Moreover, the capability of most renal cell types to produce ETs (278-281) could increase local concentrations in the kidney that serve to downregulate the ET receptors in an autocrine or paracrine mode. Reduced clearance or metabolism of ET may also contribute to the elevated levels of the peptide and thereby effect downregulation. Prior receptor occupancy by endogenous ET could be excluded in this study as a stringent acid wash of the IMCD cells also showed receptor downregulation. The mechanism(s) by which ET receptor downregulation occurs have not been extensively examined. Receptor-mediated endocytosis is likely to be at least one pathway for decreasing receptor density. In cultured smooth muscle cells incubated with ET, the observed downregulation was postulated to occur as a result of internalization of the ET receptor with its tightly bound ligand (282). However, regulation of receptor number may be at the level of transcription or translation. Indeed, downregulation of the E T B receptor mRNA in cultured osteoblasts, after treatment with sufficient ET, was reported by Sakurai et al (283). The reduced quantities of E T B mRNA could be ascribed to decreased mRNA stability rather than to inhibition of transcription since the RNA polymerase inhibitor, 113 actinomycin D, did not affect mRNA levels. Activation of PKC appears to be involved in the downregulation of ET receptors by increasing the susceptibility of the mRNA to degradation. This may be accomplished by preventing the synthesis of an ET receptor mRNA stabilizing factor (283,284). Further studies addressing this aspect of downregulation are required Stimulation experiments conducted in this study showed that ET-1-stimulated cGMP production by IMCD cells was lower in CF£F animals when compared with that in normal controls, both basally and in response to several concentrations of ET-1. That this cGMP production evoked by ET-1, which occurred in a dose-dependent manner, was diminished in the CM animals suggests that the E T B receptor was decreased in number since cGMP production is known to be increased by activation of this receptor subtype (176,285). If this is the case, these results provide further confirmation that downregulation of the E T B receptors occurs in the setting of CHF. However, it cannot be excluded that the hyposensitivity is at the level of the second messenger rather than at the receptor. Since ET is known to inhibit .AVP-stimulated water permeability, and cAMP accumulation (237-241) via the E T B receptor, downregulation of this receptor subtype in particular will abate the normally diuretic effect of ET with the consequence of increased water retention. Reduced density of the A-receptor subtype in IMCD cells may be synergistic in permitting exaggerated water reabsorption. A preliminary report demonstrated that BQ123 abolished the inhibitory effect of IO"10 M ET on AVP-stimulated water permeability in rat IMCD (286), suggesting that E T A normally assumes a role in water homeostasis as well. Furthermore, sodium excretion appears to be mediated by the A-subtype (78). The inhibition by ET of the Na+/K+-ATPase in the rat proximal nephron and collecting duct (201-203) may well be mediated through the A receptor although the finding that this inhibition may occur via a PGE 2 suggests otherwise (202). A decrease in 114 the density of this receptor subtype would thus leave the pump activity unchecked and allow unimpeded sodium reabsorption. On the other hand, the recent report by Clavell and colleagues (196) suggests that natriuresis is not regulated by the A-receptor subtype based on the finding that ET-1 infusion alone elicited no change in sodium excretion but blockade of this receptor resulted in a natriuretic action at the level of the proximal tubule. These authors proposed that ET regulation of sodium.excretion.was independent of both the A and B receptor and that there may be an unidentified subtype responsible. Other investigators have conducted experiments that indicate the B-receptor subtype is responsible for ET-induced natriuresis as well as diuresis. Renal artery infusion of STXS6c, an E T B agonist, resulted in a significant rise in fractional excretion of sodium in the dog and simultaneous infusion of BQ123 with ET-1 produced a similar result, possibly due to unmasking of E T B effects (158). Pollock and Opgenorth (198) found that BQ123 had no effect on the diuretic and natriuretic response to big ET-1, supporting possible ETB-mediated inhibition of tubular reabsorption. If the B-subtype regulates natriuresis, then downregulation of this receptor subtype would also lead to decreased sodium output, as seen in CHF. Additional studies are required to resolve what role, if any, the A receptor plays in the renal handling of sodium balance. 4.6 Haemodynamic and Renal Effects of Enalapril in CHF Contrary to other published studies (287-289), ACE inhibitor treatment resulted in an increase in MAP of the CHF animals in the present study. This is an unexpected finding given the known effects of angiotensin II blockade. One explanation for this increase may be due to an improved cardiac output by ACE inhibition without a significant change in vascular resistance, thereby raising MAP. Enalapril did not change the GFR of the hamsters. Infusion of ANP resulted in a large drop in MAP while it did not affect GFR. 115 Thus, the effect of ANP on these haemodynamic functions remained the same as in the untreated hamsters. In agreement with the study of Winaver et al (268), enalapril restored the natriuretic and diuretic responses to ANP in the sick hamsters as evidenced by the increased urine flow, sodium and water excretion, and.urinary cGMP levels. The increase in each of the parameters was similar to or greater than that in the normal controls after ANP infusion. Moreover, the baseline measurements of these variables were significantly improved after enalapril treatment. The raised baseline values are likely attributed to the activity of already elevated levels of endogenous ANP in the CHF animals, and infusion of ANP further enhanced fluid excretion. In addition, the baseline plasma protein concentration and haematocrit were restored to nondiluted values comparable to those of the normal animals. The haematocrit increased in response to the diuretic effects of exogenous ANP. Chronic enalapril therapy abrogated the relationship between the severity of heart failure and ANP-induced responses. Absent were the negative correlations between the degree of cardiac dysfunction, and ANP-induced cGMP, and fractional sodium and water excretion. The improvement of haemodynamic and renal excretory functions following ACE inhibition was likely due, in part, to the mitigated effects of angiotensin II and aldosterone. By curtailing angiotensin II formation, the intense systemic and renal vasoconstriction can be relaxed, allowing better perfusion to the kidneys. Furthermore, the direct reabsorptive actions of angiotensin II are diminished as well as its indirect actions via angiotensin-stimulated aldosterone. Indeed, ACE inhibition with lisinopril improved haemodynamic indices in patients with left ventricular dysfunction such that RBF increased while GFR decreased, thereby lowering filtration fraction and thus fluid reabsorption (290). The same 116 study reported lisinopril lowered plasma concentrations of angiotensin II, aldosterone, and ANP. In another study by Tepel and colleagues (291), captopril and enalapril inhibited the cytosolic free calcium increase induced by angiotensin II, platelet derived growth factor, ET, and AVP in mesahgial cells. These results suggest that ACE inhibition may confer renal protective effects through modulation of calcium-dependent contractile responses to angiotensin II, ET, or other pressor agents in the kidneys. 4.7 ET Receptor Distribution in the IMCD of Enalapril-treated CM Hamsters Comparison of ET receptor density in heart failure animals and in healthy controls showed no difference between the two groups after chronic enalapril administration. Addition of the ETA-specific antagonist, BQ123, revealed that the density of the E T B subtype was also similar between the two groups, and there was equal distribution of the two receptor subtypes in the IMCD. Thus, downregulation of ET receptors did not occur and this was probably the result of ACE inhibition. To date, these are the only data showing the distinct downregulation of ET receptors in the EMCD of a CHF model and the restoration to normal density following ACE inhibition therapy. In addition to the beneficial effects of ACE inhibition on angiotensin II actions, the fact that ET receptor density was no longer compromised in CHF animals after enalapril suggests that ACE inhibition may have lowered the levels of ET indirectly by blockade of angiotensin II-stimulated ET production. In light of a number of reports demonstrating that angiotensin II stimulates synthesis and release of ET-1 (118,292), as well as upregulation of ET-1 mRNA (293), it is plausible to surmise that angiotensin II blockade would lower ET-1 levels. Supporting this hypothesis are recent reports investigating the possible interaction of angiotensin II with ET and vice versa (294). Captopril was shown to suppress ET-1 release in cultured human endothelial cells perhaps through a bradykinin pathway (295,296). Furthermore, in patients with essential hypertension, captopril 117 diminished circulating concentrations of ET-1 (297). With the advent of lowered ET concentrations upon enalapril administration, the ET receptors are no longer subjected to the desensitizing effects of pathophysiological elevations of ET. Thus, the observed ET receptor density in the IMCD of the diseased hamsters is similar to that in the IMCD of the healthy controls. That improved renal function is coincident with restoration of ET receptor number in the CM hamsters following ACE inhibitor therapy has at least two implications. First, this finding suggests that, as discussed above, angiotensin II does exert influence on ET levels and consequently, ET receptor density. Second, re-establishment of sufficient ET receptor density enables the kidney to promote appropriate sodium and water excretion in response to both endogenous and exogenous ANP. Normalization of ET receptor number, especially of the B variety, can allow regulated reabsorption of water by the IMCD in response to AVP stimulation. The E T B receptor accomplishes dose-dependent inhibition of AVP-induced cAMP accumulation or increased water permeability through the activation of PKC (237,238). However, the inhibition of AVP-stimulated increase in water permeability appears to be mediated.through at least one other pathway. The latest publication by Garcia et al (298) shows that. NO decreases this water permeability by lowering cAMP levels subsequent to the activation of a cGMP-dependent protein kinase (PKG) by augmented cGMP formation. NO release after E T B activation by ETs has been documented (176,285,299). Whether the A or B subtype, both, or neither is involved in sodium balance is controversial. The data accumulated to date favour the association of the B subtype. To lend credence to this theory is the finding that NO appears to contribute to the regulation of renal excretory function (300). Inhibition of NO production decreased absolute and fractional excretion of sodium after renal nerve stimulation in anaesthetized dogs, 118 suggesting it has a modulatory role in electrolyte homeostasis. New reports assert that nitric oxide blocks basal and AVP-induced sodium reabsorption in cortical collecting tubules (301,302). Taken together, the data show enalapril therapy restored the E T B receptor density, and activation of these receptors prompted .diuresis through a PKC-dependent and/or PKG-dependent mechanism and natriuresis through a nitric oxide pathway. The total receptor and E T B affinities"for ET-1 did not differ significantly between the CM and control hamsters after enalapril administration. However, these affinities were decreased relative to those in the untreated hamsters. The reasons for this discrepancy are open to interpretation. One explanation is that the increased affinity of the receptors in the untreated hamsters was a compensatory response to the diminished number of ET receptors and that the affinity decreased once the ET receptor population returned to the usual density. Yet this does not account for the same pattern seen in the healthy group. It is conceivable that enalapril may have had a direct influence on the binding characteristics of ET receptors, and that the differences in the affinities observed were not a function of counterbalancing mechanisms. Whether ACE inhibitors exert direct effects on ET receptor binding affinities must be examined. In addition, there is the remote possibility that the inconsistencies may be attributable to variations of binding efficiency amongst the hamsters. 4.8 Angiotensin II Stimulation of IMCD Cells To conclusively determine whether a link exists between angiotensin II and ET, this assay was carried out. Incubation of IMCD cells with angiotensin II showed a significant downregulation of ET receptors when compared to controls without angiotensin II. Eighty percent of the ET receptors were of the B variety while the A subtype made up the "119 remainder. There was substantial reduction of both the A and B subtypes after angiotensin II treatment, further corroborating that angiotensin II does cause downregulation of ET receptors. This finding is in agreement with that of Roubert and associates (303) whose work revealed a downregulation of ET-1 binding sites by angiotensin II in the VSMC of rat thoracic aorta. Nevertheless, recent publications have disclosed the upregulating effects of angiotensin II on the E T B receptor subtype mRNA in rat cardiomyocytes (304) and on the E T A receptor subtype in human VSMC cultivated from the pulmonary artery (231). Furthermore, the latter study reported no change in receptor affinity for ET-1 after incubation with angiotensin II while the current study found an increase in receptor affinity. The enhanced receptor affinity observed in the present study likely represents a compensatory response to counterbalance the decline in receptor number. The reasons for these opposing results are not clear. Angiotensin II may differentially regulate ET receptor populations depending on the species or cell type involved. In addition, the increased expression of the type A ET receptor in the above study may be related to the concentrations of angiotensin II utilized. Upregulation of the E T A receptors was most prominent at higher concentrations (> 10-10 M) of angiotensin II but the present study showed downregulation at a concentration of 10"11 M. Thus the dosage of angiotensin II may be a determining factor. Co-incubation of rat IMCD cells with angiotensin II and the angiotensin non-specific receptor antagonist, saralasin (Sarlle), prevented downregulation of ET receptors in the current study. Furthermore, the previous increase in receptor affinity of the saralasin-minus IMCD cells was not observed in IMCD cells incubated with saralasin. These results definitively demonstrate that angiotensin II is an important participant in the regulation of ET receptor expression. 120 Chapter Five Summary Clearance experiments performed in this study have demonstrated that CM hamsters with moderate heart failure exhibit a blunted response to exogenous ANP infusion in comparison to that of age- and sex-matched controls. The urinary excretion of sodium, water, and cGMP were all diminished after ANP dosing in the CHF animals. A significant negative correlation was observed between these measured parameters and the severity of heart failure. Chronic treatment with the ACE inhibitor, enalapril, heightened hydromineral output as well as cGMP production by the CHF hamsters after ANP stimulation. No correlation was present between these variables and the degree of heart failure following enalapril therapy. Therefore, ACE inhibition was beneficial in relieving the symptoms due to volume overload. In vitro studies revealed that ET receptor distribution of the A and B subtypes was nearly equal at about 50% each in the IMCD cells of the normal hamster controls. The ET receptors of CM hamsters were downregulated relative to the binding site density seen in normal control animals and the level of downregulation had an inverse relationship with the degree of heart failure. This decrease in receptor density no longer occurred in the CHF hamsters after they were treated with enalapril for one week. Enalapril was also instrumental in abolishing the negative correlation between ET receptor density and cardiac dysfunction. The dissociation constants for ET-1 binding did not differ significantly between the diseased and healthy animals prior to enalapril treatment and this was also the case in enalapril-treated hamsters. However, the dissociation constants were higher (decreased affinity) in the enalapril-treated animals when compared to those values of the non-treated. 121 Stimulation with ET-1 of a primary culture of IMCD cells in CHF and normal hamsters showed an attenuated response in the former as determined by the quantification of the second messenger, cGMP. The response to ET-1 stimulation was dose-dependent. Moreover, basal accumulation of cGMP was significantly lower in the CHF animals, suggesting that ET receptor density may have been decreased. Finally, stimulation studies established there is indeed interaction between the RAAS and ET system. Angiotensin II, at low concentration, was shown to cause downregulation of ET receptors of both the A and B subtype in the IMCD cells of rats. The ET receptor distribution in rat IMCD was composed of approximately 80% B subtype and 20% A subtype. Binding affinity for ET-1 increased following incubation of the IMCD cells with angiotensin II. However, the simultaneous addition of angiotensin II and the angiotensin receptor antagonist, saralasin, precluded any decrease in binding sites for ET-1 in the IMCD cells. Not only was the maximum binding capacity similar between control and cells with receptor inhibition by saralasin, but the dissociation constants remained at comparable values. These experiments illustrate that angiotensin II is involved in the regulation of ET receptor expression. 122 Chapter Six Conclusions The results obtained in this study have elucidated that ET, in conjunction with other hormones like angiotensin II, plays a role in the pathophysiologic salt and water retention characteristic of CHF. The data present compelling evidence for participation by ETs in the regulation of hydromineral balancein that downregulation of ET receptors is strongly. associated with abnormal sodium and water reabsorption in the setting of CHF: That enalapril concomitantly restores appropriate renal function and ET receptor density provides a solid argument that angiotensin II and ET synergistically promote increased fluid reabsorption to the detriment of the body. 123 REFERENCES 1. Ho KKL, Pinsky JL, Kannel WB, Levy D. The epidemiology of heart failure: The Framingham experience. J Am Coll Cardiol 1993; 22:6A-13A. 2. Smith WMcF. Epidemiology of congestive heart failure. Am J Cardiol 1985; 55:3A-5A. 3. Shmuely H, Sclarovsky S, Rosenfeld JB. Management of congestive heart failure: neuroendocrine approach. Isr JMedSci 1993; 29:6-10. 4. Valdemarsson S, Bergdahl A, Edvinsson L. Relationships between plasma levels of catecholamines and neuropeptides and the survival time in patients with congestive heart failure. Journal of Internal Medicine 1994; 23 5:595-601. 5. Massie BM, Conway M. Survival of patients with congestive heart failure: past, present and future prospects. Circulation 1987; 75(Suppl IV):IIV-1 l-IV-18. 6. Haber HL, Leavy JA, Kesseler PD, Kukin ML, Gottlieb SS, Packer M. The erythrocyte sedimentation rate in congestive heart failure. N Engl J Med 1991; 324:353-358. 7. Omland T, Terje R, Aakvaag A, Aarsland T, Dickstein K. Plasma endothelin determination as a prognostic indicator of 1-year mortality after acute myocardial infarction. Circulation 1994; 89:1573-1579. 8. Just H, Drexler H, Hasenfuss G. Pathophysiology and treatment of congestive heart failure. Cardiology 1994; 84(Suppl 2):99-107. 9. Francis GS, McDonald KM, Cohn JN. Neurohumoral activation in preclinical heart failure. Circulation 1993; 87(Suppl IV):IV90-IV-96. 10. Burkart F, Kiowski W. Circulatory abnormalities and compensatory mechanisms in heart failure. Amer JMed 1991; 90(Suppl 5B):19S-22S. 11. Riley DJ, Weir M, Bakris GL.1 Renal adaptation to the failing heart. Postgraduate Med 1994; 95(8): 141-150. 12. Awazu M, Ichikawa I. Alterations in renal function in experimental congestive heart failure. Sent Nephrol 1994; 14(5):401-411. 13. Forfar JC. Neuroendocrine activation in congestive heart failure. Am J Cardiol 1991; 67:3C-5C. 124 14. Di Bona GF. The functions of the renal nerves. Rev Physiol Biochem Pharmacol 1982; 94:76-88. 15. Leimbach WN, Wallin G, Victor RG, Aylward PE, Sundlof G, Mark AL. Direct evidence from intraneural recordings for increased central sympathetic outflow in patients with heart failure. Circulation 1986; 73:913-919. 16. Hasking GJ, Esler MD, Jennings GL, Burton D, Korner PI. Norepinephrine spillover to plasma in patients with congestive heart failure: Evidence of increased overall and cardiorenal sympathetic activity. Circulation 1986; 73:615-621. 17. Curtiss C, Cohn IN, Vrobel T, Franciosa JA. Role of the renin-angiotensin system in the systemic vasoconstriction of chronic congestive heart failure. Circulation 1978; 58:763-769. 18. Dzau VJ, Colucci WS, Hooenberg NK, Williams GH. Relation of renin-angiotensin-aldosterone system to clinical state in congestive heart failure. Circulation 1981; 63:645-651. 19. Francis GS, Goldsmith SR, Levine B, Olivari MT, Cohn IN. The neurohumoral axis in congestive heart failure. Ann Intern Med 1984; 101:370-377. 20. Goldsmith SR, Francis GS, Cowley AW Jr, Levine TB, Cohn JN. Increased plasma arginine vasopressin levels in patients with congestive heart failure. J Am Coll Cardiol 1983; 1(6): 1385-1390. 21. Uretsky BF, Verbalis JG, Generalovich T, Valdes A, Reddy PS. Plasma vasopressin response to osmotic and hemodynamic stimuli in heart failure. Am J Physiol 1985; 248:H396-H402. 22. Sved AF, Ottenweller JE, Tapp WN, Thompson ME, Natelson BH. Elevated plasma vasopressin in cardiomyopathic hamsters. Life Sci 1985; 37:2313-2317. 23. McMurray JJ, Ray SG, Abdullah I, Dargie HJ, Morton JJ. Plasma endothelin in chronic heart failure. Circulation 1992; 85:1374-1379. 24. Stevenson LW, Fonarow GC. Endothelin and the vascular choir in heart failure. J Am Coll Cardiol 1992; 20(4):854-857. 25. Cody RJ. The potential role of endothelin as a vasoconstrictor substance in congestive heart failure. Euro Heart J1992; 13:1573-1578. 26. Zimmerman RS. A potential role for endothelin in congestive heart failure. Mayo Clin Proc1992; 67:801-803. 125 27. Simonson MS, Dunn MJ. Endothelins: A family of regulatory peptides. Hypertension 1991; 17(6):856-862. 28. Hutchison FN. Endothelin: A new role for an old friend? J Lab Clin Med 1993; 122:126-127. 29. Wei C-M, Lerman A, Rodeheffer RJ, McGregor CGA, Brandt RR, Wright S, Heublein DM, Kao PC, Edwards WD, Burnett JC Jr. Endothelin in human congestive heart failure. Circulation 1994; 89:1580-1586. 30. Wei C-M, Heublein DM, Perrella MA, Lerman A, Rodeheffer RJ, McGregor CGA, Edwards WD, Schaff HV, Burnett JC Jr. Natriuretic peptide system in human heart failure. Circulation 1993; 88:1004-1009. 31. De Bold AJ, Borrenstein HB, Veress AT, Sonnenberg H. A rapid and potent natriuretic response to intravenous injection of atrial myocardial extract in rats. Life Sci 1981; 28:89-94. 32. Sudoh T, Kangawa K, Minaminp N, Matsuo H. A new natriuretic peptide in porcine brain. Nature 1988; 332:78-81. 33. Sudoh T, Minamino N, Kangawa K, Matsuo H. C-type natriuretic peptide (CNP): a new member of natriuretic peptide family identified in porcine brain. Biochem Biophys Res Commun 1990; 168:863-870. 34. Valentin J-P, Humphreys MH. Urodilatin: A paracrine renal natriuretic peptide. Semin Nephrol 1991; 4:597-601. 35. Franch HA, Callahan LT, Blaine EH: Plasma and atrial content of atrial natriuretic factor in cardiomyopathic hamsters. Life Sci 1986; 39:1151-1159. 36. Ding J, Thibault G, Gutkowska J, Garcia R, Karabatsos T, Jasmin G, Genest J, Cantin M. Cardiac and plasma atrial natriuretic factor in experimental congestive heart failure. Endocrinology 1987'; 121:248-257. 37. Burnett JC, Kao PC, Hu DC, Heser EW, Hueblein D, Granger JP, Opgenorth TJ, Reeder GS. Atrial natriuretic peptide elevation in congestive heart failure in the human. Science 1986; 231:1145-1147. 38. Brandt RR, Wright RS, Redfield MM, Burnett JC Jr. Atrial natriuretic peptide in heart failure. J Am Coll Cardiol 1993; 22(Suppl A):86A-92A. 39. Cantin M, Thibault G, Ding J, Gutkowska J, Garcia R, Jasmin G, Hamet P, Genest J. ANF in experimental congestive heart failure. Am J Pathol 1988; 130:552-568. 126 40. Moe GW, Grima EA, Wong NLM, Howard RJ, Armstrong PW. Dual natriuretic peptide system in experimental heart failure. 1 J Am Coll Cardiol 1993; 22(3):891-898. 41. Naruse M, Takeyama Y, Tanabe A, Hiroshige J, Naruse K, Yoshimoto T, Tanaka M, Katagiri T, Demura H. Atrial and brain natriuretic peptides in cardiovascular disease. Hypertension 1994; 23(Suppl I):I-231-1-234. 42. Yoshimura M, Yasue H, Morita E, Sakaino N, Jougasaki M, Kurose M, Mukoyana M, Saitou Y, Nakao K, Imura H. Hemodynamic, renal, and hormonal responses to brain natriuretic peptide infusion in patients with congestive heart failure. Circulation 1991; 84:1581-1588. 43. Hunt PJ, Richards AM, Espiner EA, Nicholls MG, Yandle TG. Bioactivity and metabolism of C-type natriuretic peptide (CNP) in normal man. J Clin Endocrinol Metab 1994; 78:1428-1435. 44. Abassi ZA, Powell Jr, Golomb E, Reiser HR. Renal and systemic effects of urodilatin in rats with high-output heart failure. Am J Physiol 1992; 262:F615-F621. 45. Oliver JA, Scicca RR, Pinto J, Cannon PJ. Participation of the prostaglandins in the control of RPF during acute reduction of cardiac output in the dog. J Clin Invest 1981; 67:229-237. 46. Dzau VJ, Packer M, Lilly LS, Swartz SL, Hollenberg NK, Williams GH. Prostaglandins in severe congestive heart failure: relation to activation of the renin-angiotensin system and hyponatremia. NEngl JMed 1984; 310:347-352., 47. Francis GS, Goldsmith SR, Pierpont G, Cohn J. Free and conjugated plasma catecholamines in patients with congestive heart failure. J Lab Clin Med 1984; 102:393-398. 48. Viquerat CE, Daly P, Swedberg , Evers C, Curran D, Parmley WW, Chatterjee K. Endogenous catecholamine levels in chronic heart failure: Relation to the severity of hemodynamic abnormalities. Am JMed 1985; 78:455-460. 49. Suki WN. Renal hemodynamic consequences of angiotensin-converting enzyme inhibition in congestive heart failure. Arch Intern Med 1989; 149:669-673. 50. Mokotoff R, Ross G, Leiter L. Renal plasma and sodium reabsorption and excretion in congestive heart failure. J Clin Invest 1948; 27:1-9. 51. Merrill AJ. Edema and decreased ,renal blood flow in patients with chronic congestive heart failure: evidence of "forward failure" as the primary cause of edema. J Clin Invest 1946; 25:389-400. 127 52. Ichikawa I, Pfeffer JM, Pfeffer MA, Hostetter TH, Brenner BM. Role of angiotensin II in the altered renal function of congestive heart failure. Cir Res 1984; 55:669-675. 53. Moe GW, Legault L, Skorecki KL. Control of extracellular fluid volume and pathophysiology of edema formation. In: Brenner BM, Rector FC Jr., eds. The kidney. 4th ed. Philadelphia: Saunders, 1991; 364-383. 54. Thomas JA, Marks BH. Plasma norepinephrine in congestive heart failure. Am J Cardiol 1978; 41:233-243. 55. Levine TB, Francis GS, Goldsmith SR, Simon AB, Cohn JN. Activity of the sympathetic nervous system and renin-angiotensin system assessed by plasma hormone levels and their relation to hemodynamic abnormalities in congestive heart failure. Am J Cardiol 1982; 49:1659-1665. 56. Francis GS, Goldsmith SR, Cohn JN. Relationship of exercise capacity to resting left ventricular performance and basal plasma norepinephrine levels in patients with congestive heart failure. Am Heart J1982; 104:725-731. 57. Nord EP, Howard MJ, Hafezi A, Moradeshagi P, Vaystub S, Insel RA. Alphaj adrenergic agonists stimulate Na+-H^ antiport activity in the rabbit renal proximal tubule. J Clin Invest 1987; 80:1755-1762. 58. Packer M. Neurohormonal interactions and adaptations in congestive heart failure. Circulation 1988; 77:721-730. 59. Watkins L Jr, Burton JA, Haber E, Cant JR, Smith FW, Barger AC. The renin-angiotensin-aldosterone system in congestive heart failure in conscious dogs. J Clin Invest 1976; 57:1606-1617. 60. Riegger GA, Liebau G, Bauer E, Kochsiek K. Vasopressin and renin in high output heart failure of rats: Hemodynamic effects of elevated plasma hormone levels. J Cardiovasc Pharmacol 1985; 7:1-5. 61. Dzau VJ, Colucci WS, Hollenberg NK, Williams GH. The relation of the renin-angiotensin-aldosterone system to clinical state in congestive heart failure. Circulation 1981; 63:645-651. 62. Merrill AJ, Brannon ES. Concentration of renin in renal venous blood in patients with congestive heart failure. Am JMed 1946; 1:468-472. 63. Schuster VL, Kokko JP, Jacobson HR. Angiotensin II directly stimulates sodium transport in rabbit proximal convoluted tubules. J Clin Invest 1984; 73:507-515. 128 64. Hall JE. Control of sodium excretion by angiotensin II: intrarenal mechanisms and blood pressure regulation. Am J Physiol 1986; 250:R960-R972. 65. Riegger GA, Liebau G, Kochsiek K. Antidiuretic hormone in congestive heart failure. Am J Med \9%2; 72(l):49-52. 66. Nielsen S, Agre P. The aquaporin family of water channels in kidney. Kidney Int 1995; 48:1057-1068. 67. Margulies KB, Hildebrand FL Jr, Lerman A, Perrella MA, Burnett JC Jr. Increased endothelin in experimental heart failure. Circulation 1990; 82:2226-2230: 68. Stewart DJ, Cernacek P, Costello KB, Rouleau JL. Elevated endothelin-1 in heart failure and loss of normal response to postural change. Circulation 1992; 85:510-517. 69. Lerman AL, Kubo SH, Tschumperlin LK, Burnett JC Jr. Plasma endothelin concentrations in humans with end-stage heart failure and after heart transplantation. J Am Coll Cardiol'1992; 20:849-853. 70. McMurray JJ, Ray SG, Abdullah I, Dargie HJ, Morton JJ. Plasma endothelin in chronic heart failure. Circulation 1992; 85:1374-1379. 71. Nakamura M, Arakawa N, Yoshida H, Funakoshi T, Chiba M, Yasumi A, Makita S, Aoki H, Hiramori K. Increased plasma endothelin concentrations in patients with acute heart failure after myocardial infarction. Jpn Circ J1993; 57:371-378. 72. Cacoub P, Dorent R, Nataf P, Carayon A, Maistre G, Piette JC, Godeau P, Cabrol C, Gandjbakhch I. Plasma endothelin and pulmonary pressures in patients with congestive heart failure. Am Heart J1993; 126:1484-1488. 73. Loffler B-M, Roux S, Kalina B, Clozel M, Clozel J-P. Influence of congestive heart failure on endothelin levels and receptors in rabbits. J Mol Cell Cardiol 1993; 25:407-416. 74. Rodeheffer RJ, Lerman A, Heublein DM, Burnett JC Jr. Increased plasma concentrations of endothelin in congestive heart failure in humans. Mayo Clin Proc 1992; 67:719-724. 75. Pacher R, Bergler-Klein J, Globits S, Teufelsbauer H, Schuller M, Krauter A, Ogris E, Rodler S, Wutte M, Hartter E. Plasma big endothelin-1 concentrations in congestive heart failure patients with or without systemic hypertension. Am J Cardiol 1993; 71:1293-1299. 129 76. Cody RJ, Haas GJ, Binkley PF, Capers Q, Kelley R. Plasma endothelin correlates with the extent of pulmonary hypertension in patients with chronic congestive heart failure. Circulation 1992; 85:504-509. 77. Tomoda H. Plasma endothelin-1 in acute myocardial infarction with heart failure. Am Heart J1993, 125:667-672. 78. Tamirisa P, Frishman WH, Kumar A. Endothelin and endothelin antagonism: roles in cardiovascular health and disease. Am Heart J\995; 130:601-610. 79. Yanagisawa M, Masaki T. Molecular biology and biochemistry of the endothelins. Trends Pharmacol Sci 1989; 10:374-378. 80. Simonson MS, Dunn MJ. The. molecular mechanisms of cardiovascular and renal regulation by endothelin peptides. J Lab Clin Med 1992; 119(6):622-639. 81. Simonson MS. Endothelins: Multifunctional renal peptides. Physiol Revs 1993; 73(2): 375-411. 82. Hickey KA, Rubanyi GM, Paul RJ, Highsmith RF. Characterization of a coronary vasoconstrictor produced by cultured endothelial cells. Am J Physiol 1985; 248:C550-C556. 83. Yanagisawa M, Kurihara H, Kimura S, Tomobe Y, Kobayashi M, Mitsui Y, Yazaki Y, Goto K, Masaki T. A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature (Lond.) 1988; 332:411-415. 84. Inoue A, Yanagisawa M, Kimura S, Kasuya Y, Miyauchi T, Goto K, Masaki T. The human endothelin family: three structurally and pharmacologically distinct isopeptides predicted by three separate genes. Proc Natl Acad Sci USA 1989; 86:2863-2867. 85. Rubanyi GM, Polokoff MA. Endothelins: molecular biology, biochemistry, pharmacology, physiology, and pathophysiology. Pharmacol Rev 1994; 46(3):325-415. 86. Saida K, Mitsui Y, Ishida N. A novel peptide, vasoactive intestinal contractor, of a new (endothelin) peptide family. Molecular cloning, expression, and biological activity. J Biol Chem 1989; 264:14613-14616. 87. Bloch KD, Hong CC, Eddy RL, Shows TB, Quartermous T. cDNA cloning and chromosomal assignment of the endothelin 2 gene: vasoactive intestinal contractor peptide is rat endothelin 2. Genomics 1991; 10:236-242. 88. Kloog Y, Sokolovsky M. Similarities in mode and sites of action of sarafotoxins and endothelins. Trends Pharmacol Sci 1989; 10:212-214. 130 89. Takasaki C, Yanagisawa M, Kimura S, Goto K, Masaki T. Similarity of endothelin to snake venom toxin. Nature 1988; 335:303. 90. Itoh Y, Yanagisawa M, Ohkubo S, Kimura C, Kosaka T, Inoue A, Ishida N, Mitsui Y, Onda H Fujino M, Masaki T. Cloning and sequence analysis of cDNA encoding the precursor of a human endothelium-derived vasoconstrictor peptide, endothelin: identity of human and porcine endothelin. FEBS Lett 1988; 231:440-444. 91. Yanagisawa M, Akihiro I, Ishikawa T, Kasya Y, Kimura S, Kumagaye S, Nakajima K, Watanabe T, Sakakibara K, Goto K, Masaki T. Primary structure, synthesis, and biological activity of rat endothelin, an endothelium-derived vasoconstrictor. Proc Natl Acad Sci USA 1988; 85:6964-6967. 92. Sawamura T, Kasuya Y, Matsushita Y, Suzuki N, Shinmi O, Kishi N, Sugita Y, Yanagisawa M, Goto K, Masaki T, Kimura S. Phosphoramidon inhibits the intracellular conversion of big endothelin-1 to endothelin-1 in cultured endothelial cells. Biochem BiophysRes Commun 1991; 174:779-784. 93. Sawamura T, Kimura S, Shinmi O, Sugita Y, Kobayashi M, Mitsui Y, Yanagisawa M, Goto K, Masaki T. Characterization of endothelin converting enzyme activities in soluble fraction of bovine cultured endothelial cells. Biochem Biophys Res Commun 1990; 169:1138-1144. 94. Matsumura Y, Ikegawa R, Tsukahara Y, Takaoka M, Morimoto S. Conversion of big endothelin-1 to endothelin-1 by two types of metalloproteinases derived from porcine aortic endothelial cells. FEBS Lett 1990; 272:166-170. 95. Ohnaka K, Takayanagi R, Yamauchi T, Okazaki H, Ohashi M, Umeda F, Nawata H. Identification and characterization of endothelin converting enzyme activity in cultured bovine endothelial cells. Biochem Biophys Res Commun 1990; 168:1128-1136. 96. Xu D, Emoto N, Giaid A. ECE-1: a membrane-bound metalloprotease that catalyzes the proteolytic activation of big endothelin-1. Cell 1994; 78:473-485. 97. Sakamoto H, Sasaki S, Hirata Y, Imai T, Ando K, Ida T, Sakurai T, Yanagisawa M, Masaki T, Marumo F. Production of endothelin-1 by rat cultured mesangial cells. Biochem Biophys Res Commun 1990; 169:462-468. 98. Sakamoto H, Sasaki S, Nakamura Y, Fushimi K, Marumo F. Regulation of endothelin-1 production in cultured rat mesangial cells. Kidney Int 1992; 41:350-355. 99. Resink TJ, Hahn AW, Scott-Burden T, Powell J, Weber E, Buhler FR. Inducible endothelin mRNA expression and peptide secretion in cultured human vascular smooth muscle cells. Biochem Biophys Res Commun 1990; 168:1303-1310. 131 100. Zoja C, Orisio S, Perico N, Benigni A, Morigi M, Benatti L, Rambaldi A, Remuzzi G. Constitutive expression of endothelin gene in cultured human mesangial cells and its modulation by transforming growth factor B, thrombin, and a thromboxane A 2 analogue. Lab Invest 1991; 64:16-25. 101. Yoshizawa T, Shinmi O, Giaid A, Yanagisawa M, Gibson SJ, Kimura S, Uchiyama Y, Polak JM, Masaki T, Kanazawa I. Endothelin: a novel peptide in the posterior pituitary system. Science Wash DC 1990; 247:462-464. 102. Wagner OF, Christ G, Wojta J, Vierhapper H, Parzer S, Nowotny PJ, Schneider B, Waldhausl W, Binder BR. Polar secretion of endothelin-1 by cultured endothelial cells. J Biol Chem 1992, 267 16066-16068. 103. Emori T; Hirata Y, Imai T, Ohta K, Kanno K, Eguchi S, Marumo F. Cellular mechanism of thrombin on endothelin 1 biosynthesis and release in bovine endothelial cells. Biochem Pharmacol 1992; 44:2409-2411. 104. Schini VB, Hendrickson H, Heublein DM, Burnett JC, Vanhoutte PM. Thrombin enhances the release of endothelin from cultured porcine aortic endothelial cells. Eur J Pharmacol 1989; 165:2-3. 105. Marsden PA, Dorfman DM, Collins T, Brenner BM, Orkin S, Ballermann BJ. Regulated expression of endothelin 1 in glomerular capillary endothelial cells. Am J Physiol 1991; 261 :F 117-F125. 106. Kurihara H, Yoshizumi M, Sugiyama T, Takaku F, Yanagisawa M, Masaki T, Hamaoki M, Kato H, Yazaki Y. Transforming growth factor beta stimulates the expression of endothelin mRNA by vascular endothelial cells. Biochem Biophys Res Commun 1989;. 159:1435-1440. 107. Ohta K, Hirata Y, Imai T, Kanno K, Emori T, Shichiri M, Marumo F. Cytokine-induced release of endothelin-1 from porcine renal epithelial cell line. Biochem Biophys Res Commun 1990; 169:578-584. 108. Maemura K, Kurihara H, Morita T, Hashi Y, Yazaki Y. Production of endothelin 1 in vascular endothelial cells is regulated by factors associated with vascular injury. Gerontology 1992; 38(suppl l):29-35 109. Lamas S, Michel T, Collins T, Brenner BM, Marsden PA. Effects of interferon gamma on nitric oxide synthase activity and endothelin 1 production by vascular endothelial cells. J Clin Invest 1992; 90:879-887. 110. Hu RM, Levin ER, Pedram A, Frank HJ. Insulin stimulates production and secretion of endothelin from bovine endothelial cells. Diabetes 1993; 42:351-358. 132 111. Oliver FJ, De La Rubia G, Feener EP, Lee ME, Loeken M, Shiba T, Quertermous T, King GL. Stimulation of endothelin gene expression by insulin in endothelial cells. J Biol Chem 1991; 266:23251-23256. 112. Sugiura M, Inagami T, Kon V. Endotoxin stimulates endothelin-release in vivo and in vitro as determined by radioimmunoassay. Biochem Biophys Res Commun 1989; 161:1220-1227. 113. Nambi P, Pullen M, Slivjak MJ, Ohlstein EG, Storer B, Smith EF III. Endotoxin-mediated changes in plasma endothelin concentrations; renal endothelin receptor and renal function. Pharmacology 1994; 48:147-156. 114. Inoue A, Yanagisawa M, Takuwa Y, Mitsui Y, Kobayashi M, Masaki T. The human preproendothelin 1 gene. Complete nucleotide sequence and regulation of expression. J Biol Chem 1989; 264:14954-14959. 115. Yamauchi T, Ohnaka K, Takayanagi R, Umeda F, Nawata H. Enhanced secretion of endothelin-1 by elevated glucose levels from cultured bovine aortic endothelial cells. FEBS Lett 1990; 267:16-18. 116. Yoshizumi M, Kurihara H, Sugiyama T, Takaku F, Yanagisawa M, Masaki T, Yazaki Y. Hemodynamic shear stress stimulates endothelin production by cultured endothelial cells. Biochem Biophys Res Commun 1989; 161:859-864. 117. Dohi Y, Hahn AW, Boulanger CM, Buhler FR, Luscher TF. Endothelin stimulated by angiotensin II augments contractility of spontaneously hypertensive rat resistance arteries. Hypertension,1992; 19:131-137. 118. ImaiT, 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. 119. Bakris GL, Fairbanks R, Traish M, Akerstrom V, Kern S. Arginine vasopressin stimulates human mesangial cell production of endothelin. J Clin Invest 1991; 87:1158-1164. 120. Boulanger CM, Tanner FC, Bea ML, Hahn AW, Werner A, Luscher TF. Oxidized low density lipoproteins induce mRNA expression and release of endothelin from human and porcine endothelium. Circ Res 1992; 70:1191-1197. 121. Kourembanas S, Marsden PA, McQuillan LP, Faller DV. Hypoxia induces endothelin gene expression and secretion in cultured human endothelium. J Clin Invest 1991; 88:1054-1057. 133 122. Kohno M, Yokokawa K, Mandal AK, Horio T, Yasunari K, Takeda T. Cardiac natriuretic peptides inhibit cyclosporine-induced production of endothelin in cultured rat mesangial cells. Metabolism 1995; 44(3):404-409. 123. Hahn AW, Resink TJ, Scott-Burden T, Powell J, Dohi Y, Buhler FR. Stimulation of endothelin mRNA and secretion in rat vascular smooth muscle cells: a novel autocrine function. Cell Regul 1990; 1:649-659. 124. Saijonmaa O, Ristimaki A, Fyhrquist F. Atrial natriuretic peptide, nitroglycerine, and nitroprusside reduce : basal and stimulated endothelin production from cultured endothelial cells. Biochem Biophys Res Commun 1990; 173:514-520. 125. Cernacek P, Stewart DJ, Levy M. Plasma endothelin-1 response to acute hypotension induced by vasodilating agents. Can J Physiol Pharmacol 1994; 72:985-991. 126. Sandok EK, Lerman A, Stingo AJ, Perrella MA, Gloviczki P, Burnett JC Jr. Endothelin in a model of acute ischemic renal dysfunction: modulating action of atrial natiruretic factor. J Am Soc Nephrol 1992; 3:196-202. 127. Boulanger C, Luscher TF. Release of endothelin from the porcine aorta. Inhibition by endothelium-derived nitric oxide. J Clin Invest 1990; 85:587-590. 128. Yokokawa K, Kohno M, Yasunari K, Murakawa K, Takeda T. Endothelin-3 regulates endothelin-1 production in cultured human endothelial cells. Hypertension 1991; 18:304-315. 129. Kohan DE, Padilla E. Endothelin-1 production by rat inner medullary collecting duct: effect of nitric oxide,. cGMP, and immune cytokines. Am J Physiol 1994; 266:F291-F297. 130. Stewart DJ, Langleben D, Cernacek P, Cianflone K. Endothelin release is inhibited by coculture of endothelial cells with cells of vascular media. Am J Physiol 1990; 259:H1928-H1932. 131. Cade C, Ilozue V, Rubanyi GM, Parker BL. Smooth muscle cells release a diffusible factor responsible for decreasing big endothelin and endothelin produced by cultured endothelial cells [abstract]. J Vase Med Biol 1990, 2:174. 132. Erdos EG, Skidgel RA. Neutral endopeptidase 24.11 (enkephalinase) and related regulators of peptide hormones. FASEBJ19&9; 163:145-151. 133. Vijayaraghavan J, Scicli AG, Carretero O, Slaughter C, Moomaw C, Hersh LB. The hydrolysis of endothelins by neutral endopeptidase 24.11 (enkephalinase). J Biol Chem 1990; 265:14150-14155. 134 134. Sokolovsky M, Galron R, Kloog Y, Bdolah A, Indig FE, Blumberg S, Fleminger G. Endothelins are more sensitive than sarafotoxins to neutral endopeptidase: possible physiological significance. Proc Natl Acad Sci USA 1990; 87(12):4702-4706. 135. Nakamura S, Naruse M, Naruse K, Demura F£, Uemura H. Immunocytochemical localization of endothelin cultured bovine endothelial cells. Histochemistry 1990; 94:475-477. 136. Anggard E, Galton S, Rae G, Thomas R, McLoughlin L, de Nucci G, Vane JR. The fate of radioiodinated endothelin-1 and endothelin-3 in the rat. J Cardiovasc Pharmacol 1989; 13(suppl 5):S46-S49. 137. de Nucci G, Thomas R, D'Orleans-Juste P, Antunes E, Walder C, Warner TD, Vane JR. Pressor effects of circulating endothelin are limited by its removal in the pulmonary circulation and by the release of prostacyclin and endothelium-derived relaxing factor. Proc Natl Acad Sci USA 1988; 85:9797-9800. 138. Fukuroda T, Fujikawa T, Ozaki S, Ishikawa K, Yano M, Nishikibe M. Clearance of circulating endothelin-1 by E T B receptors in rats. Biochem Biophys Res Commun 1994; 199(3): 1461-1465. 139. Pernow J, Hemsen A, Lundberg JM. Tissue specific distribution, clearance, and vascular effects of endothelin in the pig. Biochem Biophys Res Commun 1989; 161:647-653. 140. Pollock DM, Keith TL, Highsmith RF. Endothelin receptors and calcium signalling. FASEBJ\995; 9:1196-1204. 141. Warner TD, Mitchell JA, Denucci G, Vane JR: Endothelin-1 and endothelin-3 release EDRF from isolated perfused arterial vessels of the rat and rabbit. J Cardiovasc Pharmacol 1989; 13:S85-S88. 142. Gardiner SM, Compton AM, Bennett T. Effects of indomethacin on the regional haemodynamic responses to low doses of endothelins and sarafotoxin. Br J Pharmacol 1990; 100:158-162. 143. Arai H, Hori S, Aramori I, Ohkubo Ff, Nakanishi S. Cloning and expression of a cDNA encoding an endothelin receptor. Nature 1990; 348:730-732. 144. Sakurai T, Yanagisawa M, Takuwa Y, Miyazaki F£, 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. 145. Adachi M, Yang Y-Y, Furuichi Y, Miyamoto C. Cloning and characterization of cDNA encoding human A-type endothelin receptor. Biochem Biophys Res Commun 1991; 180(3):1265-1272. 135 146. Masaki T, Vane JR, Vanhoutte PM. International Union of Pharmacology nomenclature of endothelin receptors. Pharmacol Rev 1994; 46:137-142. 147. Clozel M, Gray GA, Breu V, Loftier B-M, Osterwalder R. The endothelin E T B receptor mediates both vasodilation and vasoconstriction in vivo. Biochem Biophys Res Commun 1992; 186:867-873. 148. Sokolovsky M, Ambar I, Galron R. A novel subtype of endothelin receptors. J Biol Chem 1992; 267:20551-20554. 149. Warner TD, Allcock GH, Corder R, Vane JR. Use of the endothelin antagonists BQ-123 and PD 142893 to reveal three endothelin receptors mediating smooth muscle contraction and the release of EDRF. Br J Pharmacol 1993; 110:777-782. 150. Jeng AY, Savage P, Soriano A, Balwierczak JL. Different affinities and selectivities of endothelin-1 and endothelin-3 binding to various rat tissues. Biochem Int 1990; 22:669-676. 151. Martin ER, Brenner BM, Ballermann BJ. Heterogeneity of cell surface endothelin receptors. J Biol Chem 1990; 265:14044-14049. 152. Douglas SA, Beck GR Jr, Elliott JD, Ohlstein EH. Pharmacological evidence for the presence of three distinct functional endothelin receptor subtypes in the rabbit lateral saphenous vein. Br J Pharmacol 1995; 114:1529-1540. 153. Karne S, Jayawickreme CK, Lerner MR. Cloning and characterization of endothelin-3 specific receptor (ETC receptor) from Xenopus laevi dermal melanophores. J Biol Chem 1993; 268:19126-19133; 154. Elshourbagy NA, Korman DR, Wu HL, Sylvester DR, Lee JA, Nuthalaganti P, Bergsma DJ, Kumar CS, Nambi P. Molecular characterization and regulation of the human endothelin receptors. J Biol Chem 1993; 268:3873-3879. 155. Hori S, Komatsu Y, Shigemoto, Mizuno N, Nakanishi S. Distinct tissue distribution and cellular localization of two messenger ribonucleic acids encoding different subtypes of rat endothelin receptors. Endocrinology 1992; 130(4):1885-1895. 156. Karet F, Kuc R, Davenport AP. Novel ligands BQ123 and BQ3020 characterize endothelin receptor subtypes E T A and E T B in human kidney. Kidney Int 1993; 44:36-42. 157. Takemoto F, Uchida S, Ogata E, Kurokawa K. Endothelin-1 and endothelin-3 binding to rat nephrons. Am J Physiol 1993; 33:F827-F832. 136 158. Brooks DP, DePalma PD, Pullen M, Nambi P. Characterization of canine renal endothelin receptor subtypes and their function. J Pharmacol Exp Ther 1994; 268(3): 1091-1097. 159. Gellai M, DeWolf R, Pullen M, Nambi P. Distribution and functional role of ET receptor subtypes in normotensive and hypertensive rats. Kidney Int 1994; 46:1287-1294. 160. Maguire JJ, Kuc RE, O'Reilly G, Davenport AP. Vasoconstrictor endothelin receptors characterized in human renal artery and vein in vitro. Br J Pharmacol 1994; 113:49-54. 161. Chow LH, Subramanian S, Nuovo GJ, Miller F, Nord EP. Endothelin receptor mRNA expression in renal medulla identified by in situ RT-PCR. Am J Physiol 1995; 38:F449-F457. 162. Bitar KN, Stein S, Omann GM. Specific G proteins mediate endothelin induced contraction. Life Sci 1992; 50:2119-2124. 163. Takigawa M, Sakurai T, Kasuya Y, Abe Y, Masaki T, Goto K. Molecular identification of guanine-nucleotide-binding regulatory proteins which couple to endothelin receptors. Eur J Biochem 1995; 228:102-108. 164. Aramori I, Nakanishi S. Coupling of two endothelin receptor subtypes to differing signal transduction in transfected Chinese hamster ovary cells. J Biol Chem 1992; 267:12468-12474. ' 165. Marsden PA, Danthuluri NR, Brenner BM, Ballermann BJ, Brock TA. Endothelin action on vascular, smooth muscle involves inositol trisphosphate and calcium mobilization. Biochem Biophys Res Commun 1989; 158:86-93. 166. Resink TJ, Scott-Burden T, Buhler FR. Endothelin stimulates phospholipase C in cultured vascular smooth muscle cells. Biochem Biophys Res Commun 1988; 157:1360-1368. 167. Reynolds EE, Mok LL, Kurokawa S. Phorbol ester dissociates endothelin-stimulated phosphoinositide hydrolysis and arachidonic acid release in vascular smooth muscle cells. Biochem Biophys Res Commun 1989; 160:868-873. 168. Billah MM, Anthes JC, Mullmann TJ. Receptor-coupled phospholipase D: regulation and functional significance. Biochem Soc Trans 1991; 19:324-329. 169. Lee TS, Chao T, Hu KQ, King GL. Endothelin stimulates a sustained 1,2 diacylglycerol increase and protein kinase C activation in bovine aortic smooth muscle cells. Biochem Biophys Res Commun 1989; 162:381 -3 86. 137 170. Resink TJ, Scott-Burden T, Buhler FR. Activation of multiple signal transduction pathways by endothelin in cultured human vascular smooth muscle cells. Eur J Biochem 1990; 189:415-421. 171. Hon M, Sato K, Miyamoto S, Ozaki H, Karaki H. Different pathways of calcium sensitization activated by receptor agonists and phorbol esters in vascular smooth muscle. Br J Pharmacol 1993; 110:1527-1531. 172. Nishimura J, Moreland S, Ahn HY, Kawase T, Moreland RS, Van Breemen C. Endothelin increases myofilament Ca 2 + sensitivity in alpha toxin permeabilized rabbit mesenteric artery. Ore Res 1992; 71:951-959. 173. Koh E, Morimoto S, Kim S, Nabata T, Miyashita Y, Ogihara T. Endothelin stimulates Na +/H + exchange'in vascular smooth muscle cells. Biochem Int 1990; 20:375-380. 174. Emori T, Hirata Y, Marumo F. Specific receptors for endothelin-3 in cultured endothelial cells and its cellular mechanism of action. FEBS Lett 1990; 263:261-264. 175. Hirata Y, Hayakawa H, Suzuki E, Kimura K, Kikuchi K, Nagano T, Hirobe M, Omata M. Direct measurements of endothelium-derived nitric oxide release by stimulation of endothelin receptors in rat kidney and its alteration in salt-induced hypertension. Circulation 1995; 91:1229-1235. 176. Edwards RM, Pullen M, Nambi P. Activation of endothelin E T B receptors increases glomerular cGMP via an L-arginine-dependent pathway. Am J Physiol 1992; 263(32):F1020-F1025. 177. Simonson MS, Dunn MJ. Cellular signalling by peptides of the endothelin gene family. FASEB J1990; 4:2989-3000. 178. Wang Y, Rose PM, Webb ML, Dunn MJ. Endothelins stimulate mitogen-activated protein kinase cascade through either E T A or ET B . Am J Physiol 1994; 267(36):C1130-C1135. 179. Sadoshima J, Qiu Z, Morgan JP, Izumo S. Angiotensin II and other hypertrophic stimuli mediated by G protein-coupled receptors activate tyrosine kinase, mitogen-activated protein kinase, and 90-kD S6 kinase in cardiac myocytes. Circ Res 1995; 76:1-15. 180. Pribnow D, Muldoon LL, Fajardo M, Theodor L, Chen LY, Magun BE. Endothelin induces transcription of fos/jun family genes: a prominent role for calcium ion. Mol Endocrinol 1992; 6:1003-1012. 138 181. King AJ, Brenner BM, Anderson S. Endothelin: a potent renal and systemic vasoconstrictor peptide. Am J Physiol 1989; 256:F1051-F1058. 182. Underwood RD, Aarhus LL, Heublein DM, Burnett JC Jr. Endothelin in thoracic inferior vena caval constriction model of heart failure. Am J Physiol 1992; 263:H951-H955. 183. Sorensen SS, Madsen JK, Pedersen EB. Systemic and renal effect of intravenous infusion of endothelin-1 in healthy human volunteers. Am J Physiol 1994; 266:F411-F418. 184. Kiowski W, Sutsch G, Hunziker P, Muller P, Kim J, Oechslin E, Schmitt R, Jones R, Bertel O. Evidence for endothelin-1-mediated vasoconstriction in severe chronic heart failure. Lancet 1995; 346:732-736. 185. Goetz KL, Wang BC, Leadley RJ, Zhu JL, Madwed J, Bie P. Endothelin and sarafotoxin produce dissimilar effects on renal blood flow, but both block the antidiuretic effects of vasopressin. Proc Soc Exp Biol Med 1989; 191:425-427. 186. Miller WL, Redfield MM, Burnett JC. Integrated cardiac, renal, and endocrine actions of endothelin. J Clin Invest 1989; 83:317-320. 187. Stacy DL, Scott JW, Granger JP. Control of renal function during intrarenal infusion of endothelin. Am J Physiol 1990; 258:F1232-F1236. 188. Katoh T, Chang H, Uchida S, Okuda T, Kurokawa K. Direct effects of endothelin in the rat kidney. Am J Physiol 1990; 258:F297-F402. 189. Warner, TDVBattisthuB, Allcock:GH, Vane JR. Endothelin ETAand E T B receptors mediate vasoconstriction and prostanoid release in the isolated kidney of the rat. Eur J Pharmacol 1993; 250:447-453. 190. Kon V, Yoshioka T, Fogo A, Ichikawa I. Glomerular actions of endothelin in vivo. J Clin Invest 1989; 83:1762-1767. ' 191. Simonson MS, Dunn MJ. Endothelin-1 stimulates contraction of rat glomerular mesangial cells and potentiates 13-aderenergic-mediated cyclic adensoine monophosphate accumulation. J Clin Invest 1990; 85:790-797. 192. Garcia R, Lachance D, Thibault G. Positive inotropic action, natriuresis and atrial natriuretic factor release induced by endothelin in the conscious rat. J Hypertens 1990; 8:725-731. 193. Goetz KL, Wang BC, Madwed JB, Zhu JL, Leadley RJ. Cardiovascular, renal, and endocrine responses to intravenous endothelin in conscious dogs. Am J Physiol 1988; 255:R1064-R1068. 139 194. Rabelink TJ, Kaasjager KAH, Boer P, Stroes EG, Braam B, Koomans HA. Effects of endothelin-1 on renal function in humans: implications for physiology and pathophysiology. Kidney Intl 994; 46:3 76-3 81. 195. Perico N, Cornejo RP, BenigniA, Malanchini B, Ladny JR, Remuzzi G. Endothelin induces diuresis and natriuresis in the rat by acting on proximal tubular cells through a mechanism mediated by lipoxygenase products. J Am Soc Nephrol 1991; 2:57-69. 196. Clavell AL, Stingo AJ, Margulies KB, Brandt RR, Burnett JC Jr. Role of endothelin receptor subtypes in the in vivo regulation of renal function. Am J Physiol 1995; 268:F455-F460. 197. Kamphuis C, Yates NA, McDougall JG. Differential blockade of the renal vasoconstrictor and diuretic responses to endothelin-1 by endothelin antagonist. Clin Exper Pharmacol Physiol 1994; 21:329-333. 198. Pollock DM, Opgenorth TJ. E T A receptor-mediated responses to endothelin-1 and big endothelin-1 in the rat kidney. Br J Pharmacol 1994; 111:729-732. 199. Ferrario RG, Foulkes R, Salvati P, Patrono C. Hemodynamic and tubular effects of endothelin and thromboxane in the isolated perfused rat kidney. Eur J Pharmacol 1989; 171:127-134. 200. Nitta K, Naruse M, Sanaka T, Tsuchiya K, Naruse K, Zeng Z, Demura H, Sugino N. Natriuretic and • diuretic effects of endothelin in isolated perfused rat kidney. Endocrinol Japan 1990; 36:887-890. 201. .Zeidel ML, Brady HR, Kone BC, Gullans SR, Brenner BM. Endothelin, a peptide inhibitor of Na+-K+-ATPase in intact renal tubular epithelial cells. Am J Physiol 1989; 257:C1.101-C1107.. 202. Jabs K, Silva P, Zeidel ML. Prostaglandin E 2 inhibits Na/K-ATPase in rabbit inner medullary collecting duct cells (Abstract). Kidney Int 1988; 33:268A. 203. Garvin J, Sanders K. Endothelin inhibits fluid and bicarbonate transport in part by reducing Na+/K+ATPase activity in the rat proximal straight tubule. J Am Soc Nephrol 1991; 2:976-982. I 204. Dietz JR. Release of natriuretic factor from rat heart-lung preparation by atrial distension. Am J Physiol 1984; 247-R1093-R1096. 205. Gardner DG, Newman ED, Nakamura KK, Nguyen KPT. Endothelin increases the synthesis and secretion of atrial natriuretic peptide in neonatal rat cardiocytes. Am J Physiol 1991; 26LE177-E182. 140 206. Hu JR, Berninger UG, Lang RE. Endothelin stimulates atrial natriuretic peptide (ANP) release from rat atria. Eur J Pharmacol 1988; 158:177-178. 207. Bruneau BG, de Bold AJ. Selective changes in natriuretic peptide and early response gene expression in isolated rat atria following stimulation by stretch or endothelin-1. Cardiovascular Res 1994; 28:1519-1525. 208. Thibault G, Doubell AF, Garcia R, Lariviere R, Schiffrin EL. Endothelin-stimulated secretion of natriuretic peptides by rat atrial myocytes is mediated by endothelin A receptors. Circ Res 1994; 74:460-470. 209. Leite MF, Page E, Ambler SK. Regulation of ANP secretion by endothelin-1 in cultured atrial myocytes: desensitization and receptor subtype. Am J Physiol 1994; 267:H2193-112203 210. Brenner BM, Ballerman BJ, Gunning ME, Zeidel ML. Diverse biological action of atrial natriuretic peptide. Physiol Rev 1990; 70:665-699. 211. Cody RJ, Atlas SA, Laragh JH, Kubo SH, Covit AB, Ryman KS, Shakuovich A, Pondolfino K, Clark M, Carmargo MJF, Scarborough RM, Lewicki JA. Atrial natriuretic factor in normal subjects and heart failure patients. J Clin Invest 1986; 78:1362-1374. 212. Koepke JP, DiBona GF. Blunted natriuresis to atrial natriuretic peptide in chronic sodium-containing disorders. Am J Physiol 1987; 252:F865-F871. 213. Perico N, Dadan J, Gabanelli M, Remuzzi.G. Cyclo-oxygenase,products and atrial natriuretic peptide modulate: renal response to endothelin. J Pharmacol Exp Ther 1990;. 252:1213-1220.: 214. Neuser D, Knorr A, Stasch J-P, Kazda S. Mitogenic activity of endothelin-1 and-3 on vascular smooth muscle cells is inhibited by atrial natriuretic peptides. Artery 1990; 17:311-324. 215. Jaiswal RK. Endothelin inhibits the atrial natriuretic factor stimulated cGMP production by activating the protein kinase C in rat aortic smooth muscle cells. Biochem Biophys Res Commun 1992; 182:395-402. 216. Cernacek P, Legault L, Stewart DJ, Levy M. Specific endothelin binding sites in renal medullary collecting duct cells: lack of interaction with ANP binding and cGMP signalling. Can J Physiol Pharmacol 1992; 70:1167-1174. 217. Garcia NH, Garvin JL. ANF and angiotensin II interact via kinases in the proximal straight tubule. Am J Physiol 1995; 268:F730-F735. 141 218. Clark BA, Elahi P, Fish L, McAloon-Dyke M, Davis K, Minaker KL, Epstein FH. Atrial natriuretic peptide suppresses osmostimulated vasopressin release in young and elderly humans. Am J Physiol 1991; 261:E252-E256. 219. Sonnenberg Ff, Veress AT. Cellular mechanism of release of atrial natriuretic factor. Biochem Biophys Res Commun 1984; 124:443-449. 220. Patel KP. Interaction among, atrial natriuretic factor (ANF), vasopressin, and renal nerves in terms of renal responsiveness in rats. Life Sci 1991; 48:261-267. 221. Nakamura M, Yoshida H, Funakoshi T, Arakawa N, Hiramori K. Renal, haemodynamic and hormonal interactions between atrial natriuretic factor and arginine vasopressin in patients with congestive heart failure. Clin Sci 1992; 82:363-368. 222. Dzau VJ. Tissue renin-angiotensin system in myocardial hypertrophy and failure. Arch Intern Med 1993; 153:937-942. 223. Badr KK, Murray JJ, Breyer MD, Takahashi K, Inagami T, Harris RC. Mesangial cell, glomerular and renal vascular responses to endothelin in the rat kidney. Elucidation of signal transduction pathways. J Clin Invest 1989; 83:336-342. 224. Cozza EN, Gomez-Sanchez CE, Foecking MF, Chiou S. Endothelin binding to cultured calf adrenal zona glomerulosa cells and stimulation of aldosterone secretion. J Clin Invest 1989; 84:1032-1035. 225. Ujiie K, Terada Y, Nonoguchi H, Shinohara M, Tomita K,. Marumo F. Messenger RNA expression and synthesis of endothelin-1 along, rat nephron segments. J Clin Invest: 1992; 90:1043-1048. 226. Takagi M, Matsuoka H, Atarashi K, Yagi S. Endothelin: a new inhibitor of renin release. Biochem Biophys Res Commun 1988; 157:1164-1168. 227. Takagi M, Matusoka H, Atarashi K, Yagi S. Inhibitory effect of endothelin on renin release in vitro. Am J Physiol 1989; 257:E833-E838. 228. Matusumara Y, Nakase K, Ikegawa R, Hayashi K, Ohyama T, Morimoto S. The endothelium-derived vasoconstrictor peptide endothelin inhibits renin release in vitro. Life Sci 1989; 44:149-157. 229. Tejedor A, Moe O, Alpern RJ, Campbell WB, Henrich WL. Endothelin inhibits in vitro renin release (Abstract). Clin Res 1989; 37:585A. 230. Ritthaler T, Scholz H, Ackermann M, Riegger G, Kurtz A, Kramer BK. Effects of endothelins on renin secretion from isolated mouse renal juxtaglomerular cells. Am J Physiol 1995; 268:F39-F45. 142 231. Hatakeyama H, .Miyamori I, Yamagishi S, Takeda Y, Takeda R, Yamamoto H. Angiotensin II up-regulates the expression of type A endothelin receptor in human vascular smooth muscle cells. Bioc Mol Biol Int 1994; 34(1): 127-134. 232. Wilkins FC Jr, Kassab S, Kato T, Mizelle HL, Opgenorth TJ, Granger JP. Chronic endothelin-induced pressor and renal actions in conscious dogs do not require altered ANG II formation. Am J Physiol 1995; 268:R395-R402. 233. Kohan DE, Padilla E. Osmolar regulation of endothelin-1 production by rat inner medullary collecting duct. J din-Invest 1993; 91:1235-1240. . 234. Yoshizawa T, Shinmi O, Giaid A, Yanagisawa M, Gibson SJ, Kimura S, Uchiyama Y, Polak JM, Masaki T, Kanazawa I. Endothelin: a novel peptide in the posterior pituitary system. Science Wash. DC 1990; 247:462-464. 235. Bakris GL, Fairbanks R, Traish AM. Arginine vasopressin stimulates human mesangial cell production of endothelin. JClinlnvest 1991; 87:1158-1164. 236. Takemoto F, Uchida S, Katagiri H, Oka Y, Nakao A, Kurokawa K. Desensitization of endothelin-1 binding by vasopressin via a cAMP-mediated pathway in rat CCD. Am J Physiol 1995; 268:F385-F390. 237. Tomita K, Nonoguchi H, Marumo F. Effects of endothelin on peptide-dependent cyclic adenosine monophosphate accumulation along the nephron segments of the rat. J Clin Invest 1990; 85:2014-2018. 238. Woodcock EA, Land SL. Functional endothelin E T B receptors on renal papillary tubules. Eur J Pharmacol 1993; 247:93-95 239. Oishi R^ Nonoguchi-H,;. Tomita K, Marumo F. Endothelin-1 inhibits AVP-stimulated osmotic water permeability in rat inner medullary collecting duct. Am J Physiol 1991; 26LF951-F956. 240. Nadler SP, Zimpelman JA, Hebert RL. Endothelin inhibits vasopressin-stimulated water permeability in rat terminal inner medullary collecting duct. J Clin Invest 1992; 90:1458-1466. j . 241. Edwards RM, Stack EJ, Pullen M, Nambi P. Endothelin inhibits vasopressin action in rat inner medullary collecting duct via the E T B receptor. J Pharmacol Exp Ther 1993; 267(3): 1028-1033. 242. Ozaki S, Ihara M, Saeki T, Fukami T, Ishikawa K, Yano M. Endothelin E T B receptors couple to two distinct signalling pathways in porcine kidney epithelial LLC-PK! cells. J Pharmacol Exp Ther 1994; 270(3): 1035-1040. 143 243. Cantiello HF, Ausiello DA. Atrial natriuretic factor and cGMP inhibit amiloride-sensitive Na+ transport in the cultured renal epithelial cell line, LLC-PKj. Biochem Biophys Res Commun 1986; 134:852-860. 244. Dzau VJ, Pratt RE. Renin-angiotensin system, Chapter 73 in The Heart and Cardiovascular System, 2nd edit., edited by HA Fozzard et al. Raven Press, Ltd., New York, 1992. 245. Burnier M, Biollaz J. Pharmacokinetic optimisation of angiotensin converting enzyme (ACE) inhibitor therapy. Clin Pharmacokinet 1992; 22(5):375-384. 246. Reid JL. Inhibitors of the renin-angiotensin system. Drug Res 1993; 43 (I):263-264. 247. Nussberger J, BrunnerDB, Waeber B,,Brunner HR. True versus immunoreactive angiotensin II in human plasma. Hypertension 1985; 70(suppl):ll-17. 248. Nussberger J, Juillerat L, Perret F, Waeber B, Bellet M, et al. Need for plasma angiotensin measurements to investigate converting enzyme inhibition in humans. Am Heart J19S9, 117:717-722. 249. Burnier M, Waeber B, Nussberger J, Brunner HR. Pharmacokinetics of angiotensin converting enzyme inhibitors. Br J Clin Pharmacol 1989; 28:133S-140S. 250. Teerlink JR. The evolving role of angiotensin-converting enzyme inhibition in heart failure: expanding the protective envelope. J Cardiovasc Pharmacol 1994; 24(suppl):S32-S37. 251. The CONSENSUS Trial Study Group. Effects of enalapril on mortality in severe congestive heart failure. . Results of the Cooperative North Scandinavian Enalapril Survival Study (CONSENSUS). N Engl JMed 1987; 316:1429-1435. 252. The SOLVD Investigators. Effect of enalapril on survival in patients with reduced left ventricular ejection fractions and congestive heart failure. N Engl J Med 1991; 325:293-402. ' 253. The SOLVD Investigators. Effect of enalapril on mortality and the development of heart failure in asymptomatic patients with reduced left ventricular ejection fraction. NEngl JMed 1992; 327:685-691. 254. Fu L-X, Sun X-Y, Hedner T, Feng Q-P, Liang Q-M, Hoebeke J, Hjalmarson A. Decreased density of mesenteric arteries but not of myocardial endothelin receptors and function in rats with chronic ischemic heart failure. J Cardiovasc Pharmacol 1993; 22:177-182. 144 255. Bajusz E, Homburger F, Baker JR, Bogdonoff P. Dissociation of factors influencing myocardial degeneration and generalized cardiocirculatory failure. Ann NY Acad Sci USA 1969; 156:396-420. 256. Cantin M, Thibault G, Ding J, Gutkowska J, Garcia R, Jasmin G, Hamet P, Genest J. ANF in experimental congestive heart failure. Am J Pathol 1988; 130:552-568. 257. Cordon-Cardo C, Finstad CL, Bander NH, Melamed MR. Immunoanatomic distribution of cytostructural and tissue-associated antigens in the human urinary tract. Am J Pathol 1987; 126:269-284.. 258. Hunter WM, Greenwood FC. The preparation of 131I-labeled human growth hormone of high specific radioactivity. Nature 1962; 194:495-496. 259. Homburger F, Baker JR, Nixon CW,Wilgram G. New hereditary disease of Syrian hamsters. Primary, generalised polymyopathy and cardiac necrosis. Arch Int Med 1962; 110:660-662. 260. Bajusz E, Jasmin G. Hereditary disease model of cardiomyopathy; studies on a new line of Syrian hamsters. Fed Proc 1972; 31:621. 261. Jasmin G, Proseher L. Hereditary polymyopathy and cardiomyopathy in the Syrian hamster. I. Progression of heart and skeletal muscle lesions in the UM-X7.1 line. Muscle Nerve 1982; 5:20-25. 262. Margulies KB, Heublein DM, Perrella MA, Burnett JC. ANF mediated renal cGMP generation in congestive heart failure. Am J Physiol 1991; 260:F562-F568. 263. Qing G, Garcia R. Characterisation.of plasma and tissue atrial natriuretic factor during development of moderate high output heart failure in the rat. Cardiovasc Res 1993; 27:464-470. 264. Tsunoda K, Mendelsohn FA, Sexton PM, Chai SY, Hodsman GO, Johnston CI. Decreased atrial natriuretic peptide binding in renal medulla in rats with chronic heart failure. CirRes 1988; 62:155-161. 265. Bajusz E. Hereditary cardiomyopathy: a new disease model. Am Heart J 1969; 77:686-696. 266. Wong NLM, Wong EFC, Hu DCK. Atrial natriuretic factor release in cardiomyopathic hamsters. Cardiology 1992; 80:12-17. 267. Trippodo NC, Fox M, Natarajan V, Balkrushna CP, Dorso CR, Asaad MM. Combined inhibition of neutral endopeptidase and angiotensin converting enzyme in cardiomyopathic hamsters with compensated heart failure. J Pharmacol Exp Ther 1993; 267(1): 108-116. 145 268. Winaver J, Hoffman A, Burnett JC Jr, Haramati A. Hormonal determinants of sodium excretion in rats with experimental high-output heart failure. Am J Physiol 1988; 254:R776-R784. 269. Abassi ZA, Kelly G, Golomb E, Klein H, Keiser HR. Losartan improved the natriuretic response to ANF in rats with high-output failure. J Pharmacol Exp Ther 1994; 268(l):224-230. 270. Isnard R, Carayon A, Eurin J, Maistre G, Bouanani N, Barthelemy C, Crozatier B, Komajda M, Legrand J-C. Glomerular atrial natriuretic factor receptors in experimental congestive heart failure. Am J Physiol 1993; 265:H923-H928. 271. Wong KR, Xie MH, Shi LB, Liu FY, Huang CL, Gardner CL, Cogan MG. Urinary cGMP as biological marker of the renal activity of atrial natriuretic factor. Am J Physiol 1988; 255:F1220-F1224. 272. Hirata Y, Tomita M, Takada S, Yoshimi H. Vascular receptor binding activities and cyclic GMP responses by synthetic human and rat atrial natriuretic peptides (ANP) and receptor down-regulation by ANP. Biochem Biophys Res Commun 1985; 128:538-546. 273. Bianchi C, Thibault G, Wrobel-Konrad E, De Lean A, Genest J, Cantin M. Atrial natriuretic factor binding sites in experimental congestive heart failure. Am J Physiol 1989; 257:F515-F523. 274. Luk JKH, Wong EFC, Wong NLM. Second messenger changes of atrial natriuretic factor and brain natriuretic peptide in kidneys of cardiomyopathic hamsters. Cardiology 1994; 85:82-87. 275. Chow LH, Subramanian S, Nuovo GJ, Miller F, Nord EP. Endothelin receptor mRNA expression in renal medulla identified by in situ RT-PCR. Am J Physiol 1995; 269:F449-F457. 276. Bolger GT, Berry R, Liard F, Garneau M, Jaramillo J. Cardiac responses and binding sites for endothelin in normal and cardiomyopathic hamsters. J Pharmacol Exp Ther 1992; 260(3): 1314-1322. 277. Inada T, Tanaka M, Hasegawa K, Ohtani S, Doyama K, Fujiwara T. Increased levels of endothelin-1 in plasma and heart tissue of cardiomyopathic Syrian hamsters [Abstract]. Circulation 1994; 90:1-260. 278. Marsen TA, Schramek H, Dunn MJ. Renal actions of endothelin: linking cellular signaling pathways to kidney disease. Kidney Int 1994; 45:336-344. 279. Clavell AL, Burnett JC Jr. Physiologic and pathophysiologic roles of endothelin in the kidney. Curr Opin Nephrol Hyperten 1994; 3:66-72. 146 280. Nassar GM, Badr KF. Endothelin in kidney disease. Curr Opin Nephrol Hyperten 1994; 3:86-91. 281. Karet FE, Davenport AP. Localization of endothelin peptides in human kidney. Kidney Int 1996; 49:382-387. 282. Hirata Y, Yoshimi H, Takaichi S, Yanagisawa M, Masaki T. Binding and receptor down-regulation of a novel vasoconstrictor endothelin in cultured rat vascular smooth muscle cells. FEBS Lett 1988; 239:13-17. 283. Sakurai T, Morimoto H, Kasuya Y, Takuwa Y, Nakauchi H, Masaki T, Goto K. Level of E T B receptor mRNA is down-regulated by endothelins through decreasing the intracellular stability of mRNA molecules. Biochem Biophys Res Commun 1992; 186:342-347. -284. Masaki T. Overview: Reduced sensitivity of vascular response to endothelin. Circulation 1993; 87(suppl V):V33-V35. 285. Owada A, Tomita K, Terada Y, Sakamoto H, Nonoguchi H, Marumo F. Endothelin (ET)-3 stimulates cyclic guanosine 3',5'-monophosphate production via E T B receptor by producing nitric oxide in isolated rat glomerulus, and in cultured rat mesangial cells. J Clin Invest 1994; 93:556-563. 286. Nadler SP, Zimpelmann JA.. Diuresis modulates endothelin action in rat inner medullary collecting duct [Abstract]. J Am Soc Nephrol 1992; 3:796. 287. Abassi Z, Haramati A,. Hoffman. A, Burnett JC Jr, Winaver J. Effect of converting-enzyme inhibition on renal response to ANF in rats with experimental heart failure. Am J Physiol 1990; 259:R84-R89. 288. Wambach G, Schittenhelm U, Stimpel M, Bonner G, Kaufmann W. Natriuretic action of ANP is blunted by ACE inhibition in humans. J Cardiovasc Pharmacol 1989; 13(5):748-753. 289. Boldt J, Menges T, Wollbruck M, Harter K, Hempelmann G. Continuous I.V. administration of the ahgiotensin-converting enzyme inhibitor enalaprit in the critically ill: effects on regulators of circulatory homeostasis. J Cardiovasc Pharmacol 1995; 25(3):416-423. 290. Cleland JGF, Shah D, Krikler S, Dritsas A, Nihoyannopoulos P, Frost G, Oakley CM. Effects of lisinopril on cardiorespiratory, neuroendocrine, and renal function in patients with asymptomatic left ventricular dysfunction. Br Heart J 1993; 69:512-515. 147 291. Tepel M, Heidenreich S, Zhu Z, Walter M, Nofer J-R, Zidek W. Captopril inhibits the agonist-induced increase of cytosolic free Ca 2 + in glomerular mesangial cells. Kidney Int \ 994; 46:696-702. 292. Kohno M, Horio T, Ikeda M, Yokokawa K, Fukui T, Yasunari K, Kurihara N, Takeda T. Angiotensin II stimulates endothelin-1 secretion in cultured rat mesangial cells. Kidney Int 1992; 42:860-866. 293. Chua BHL, Chua CC, Diglio CA, Siu BB. Regulation of endothelin-1 mRNA by angiotensin II in rat heart endothelial cells. Biochim Biophys Acta 1993; 1178:201-206. 294. Egido J. Vasoactive hormones and renal sclerosis. Kidney Int 1996; 49:578-597. 295. Yoshida H, Nakamura M. Inhibition by angiotensin converting enzyme inhibitors of endothelin secretion from cultured human endothelial cells. Life Sci 1992; 50:195-200. 296. Momose N, Fukuo K, Morimoto S, Ogihara T. Captopril inhibits endothelin-1 secretion from endothelial cells through bradykinin. Hypertension 1993; 21:921-924. 297. Uemasu J, Munemura C, Fujihara M, Kawasaki H. Inhibition of plasma endothelin-1 concentration by captopril in patients with essential hypertension. Clin Nephrol 1994; 41:150-152. 298. Garcia NH, Stoos BA, Carretero A, Garvin JL. Mechanism of the nitric oxide-induced blockade of collecting duct water permeability. Hypertension 1996; 27(part 2):679-686. 299. D'Orleans-Juste P, Claing A, Telemaque S, Maurice M-C, Yano M, Gratton J-P. Block of endothelin-1-induced release of thromboxane Aj from the guinea pig lung and nitric oxide from the rabbit kidney by a selective E T B receptor antagonist BQ-788 Br J Pharmacol 1994; 113:1257-1262. 300. Egi Y, Matsumura Y, Miura A, Murata S, Morimoto S. Interaction between nitric oxide and angiotensin II on antidiuresis and norepinephrine overflow induced by stimulation of renal nerves in anesthetized dogs. J Cardiovasc Pharmacol 1995; 25:187-193. 301. Stoos BA, Garcia NH, Garvin JL. Nitric oxide inhibits sodium reabsorption in the isolated perfused cortical collecting duct. J Am Soc Nephrol 1995; 6:89-94. 302. Garcia NH, Pomposiello SI, Garvin JL. Nitric oxide inhibits ADH-stimulated osmotic water permeability in cortical collecting ducts. Am J Physiol 1996; 270:F206-F210. . . 148 303. Roubert P, Gillard V, Plas P, Guillon JM, Chabrier PE, Braquet P. Angiotensin II and phorbol-esters potently down-regulate endothelin (ET-1) binding sites in vascular smooth muscle cells. Biochem Biophys Res Commun 1989; 164:809-815. 304. Kanno K, Hirata Y, Tsujino M, Imai T, Schichiri MK, Ito H, Marumo F. Up-regulation of ETB receptor subtype mRNA by angiotensin II in rat cardiomyocytes. Biochem Biophys Res Commun 1993; 194:1282-1287. 305. Munson PJ, Rodbard D. LIGAND: A versatile computerized approach for characterization of ligand-binding systems. Anal Biochem 1980; 107:220-239. 149 APPENDIX o o c CJ CJ cj 2 P o m 6 C o 10 s c •a cj o o Pi c cs C e o •a o e S3 fi o T3 e co a o T 3 CJ CN IO O -H CN o -H CN r-© i n o CN CO © +1 00 m CN CN e '55 o t« OH OC e i s o .6 -H • s -H i s oo 2 o , e r -S s S I o eg CN CO , "o •)r! '2 ^ o . S w H o -a •a 5 e a. ^ T o .5 w CJ-g . I- > — I 3 oo ca ^ W fl fi 2 o oo 2 13 2 C •a 2 s cj s o a a u Q a u 150 c o oo «s o u > c I T3 2 u c o O 3 •a 3 •a 152 CJ o C cj CJ C\ CO cj O oj •a CO •a eg CJ c n co e —i W a. cj u OJ Ct. ON o 00 T3 Ui o d -H m o o o © -H NO o o o © o o o d -H 00 CN NO o o d -H ON o o d 4) ON ON o CO ,1 Q. 00 6 S3 o 2 a--H I CN C o CN S3 B NO NO ON o o CN CO — • «J O o ' >o -H O NO O £ 2 c o CJ -H ON CN O NO -H r~ NO St "o •a o 5