ATRIAL NATRIURETIC PEPTIDE IN AGING RATS:

EVIDENCE FOR ALTERED PROCESSING, SECRETION AND RECEPTOR BINDING

by

JONATHAN KAO

B.Sc, University of Toronto, 1985

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 required standard

Urs Steinbrecher

Dr. Jean Shapiro

Dr. Simon Rabkin

Dr. Ralph Keeler

Dr. Steve Pelech THE UNIVERSITY OF BRITISH COLUMBIA

March, 1990

© Jonathan Kao, 1990 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.

^ Medicine Department of

The University of British Columbia Vancouver, Canada

Date April 30, 1990

DE-6 (2/88) ABSTRACT

The recently discovered atrial natriuretic peptide (ANP) has potent , natriuretic and hypotensive effects, and is believed to be involved in the maintenance of in both normal and pathological conditions.

The mammalian aging process is associated with a host of abnormalities that include, among others, a compromised ability to regulate sodium homeostasis.

There are reports that demonstrate a positive correlation between plasma ANP levels and age in man; accordingly, the aim of this study was to examine whether age-related sodium imbalance is associated with disturbances in the homeostasis of ANP. Specifically, the intracellular storage, processing and secretion of ANP from the atrium was studied and associated with circulating

ANP concentrations and ANP receptor binding kinetics.

Studies were conducted with four groups of male Wistar rats designated as 1-,

3-, 10-, and 20-month-old. 24-hour renal clearances were conducted to assess

age-related changes in renal functions. GFR and UNaV increased steadily from

1 to 10 months of age and decreased in the 20-month-old, while fractional

of (FE^) and sodium (FE„a) declined initially (from 1 to 10 months) and then rose in the 20-month-old group.

Circulating ANP levels in the rats was significantly correlated with the increase in age (N = 147, r = 0.59, p < 0.0005). Atria of the animals were isolated and superfused with a modified Langendorff apparatus. The spontaneous release of

ANP increased from 1 to 3 months, and steadily decreased after 3 months. The results indicate that ANP secretion increases with maturation and thereafter declines with advancing age.

- ii - ANP concentrations in the right and left atria were also quantified. The results revealed that atrial ANP content increased from 1 to 3 months and decreased progressively with age. There was a positive correlation between the rate of ANP release and atrial ANP content (N= 42, r=0.50, p<0.01), suggesting that the release of ANP from the right atrium was associated with the atrial content. The concurrence of a reduction in ANP secretion but with elevation in plasma ANP concentration in the aged (20-month-old) rats, suggests that there may be an impairment in renal of ANP.

It was established that the main molecular species present in the atrium was y-ANP and that this was unaffected by age as assessed by reverse-phase high performance liquid chromatography (RP-HPLC) coupled with radioimmunoassay.

The molecular forms of ANP secreted by the atrium consisted of predominantly a-ANP, with a smaller amounts of y-ANP. y-ANP constituted only 1% of the total secreted ANP in the 1-, 3-, or 10-month-old rats, but up to 8% was detected in 20-month-old rats. Although both a-ANP and y-ANP were present in the circulation, the ratio of y-ANP/a-ANP increased significantly with age.

The concentration of y-ANP in the plasma of the 20-month-old rats was two- to three-fold higher than In the two younger groups (1- and 3-month-old). These data imply that the post-transcriptional processing of prohormone y-ANP to active a-ANP is altered with age.

Radio-ligand binding experiments were carried out using glomerular ANP receptors to determine whether the age-related alterations in plasma ANP levels has an effect on the binding of ANP to its target tissues. Both the receptor

density (Bmax) and the equilibrium dissociation constant (kj increased from 1 to

3 months but decreased from 3 to 20 months.

- iii - Collectively, these results suggest that:

1) Aging affects atrial ANP content and consequently influences the release

of ANP from the isolated atria.

2) The processing of prohormone y-ANP to active a-ANP is modified with

age.

3) Plasma levels of ANP increase with age, which may result in down-

regulation of ANP receptor density and increased efficacy in receptor

binding affinity. These may represent the compensatory responses.

- iv - TABLE OF CONTENTS

PAGE

ABSTRACT ii

TABLE OF CONTENTS v

LIST OF FIGURES viii

LIST OF TABLES xi

ACKNOWLEDGEMENTS xii

1.0 INTRODUCTION

1.1 Historical Aspects of ANP 1

1.2 Molecular Structure of ANP 2

1.3 Release of ANP 4

1.4 ANP Receptors 6

1.5 Intracellular Signals Mediating ANP Action 8

1.6 Physiology of ANP 9

1.6.1 Effects on Vascular Smooth Muscle 9

1.6.2 Effects on Renal Hemodynamics and

Sodium Excretion 10

1.6.3 Effects on Pressure and Blood Volume . . 11

1.6.4 Effects on --

Aldosterone System 12

1.7 Pathophysiological Implications 13

1.8 Aging 14

2.0 MATERIALS AND METHODS

2.1 Materials 19

2.1.1 Rats 19

- v - 2.1.2 Chemicals 19

2.2 Methods 19

2.2.1 24-Hour Clearance Studies 19

2.2.2 Measurement of Plasma ANP Levels 20

2.2.3 Spontaneous Release of ANP 21

2.2.4 Determination of Atrial ANP Content 21

2.2.5 HPLC Analysis of ANP Molecular Species 23

2.2.5.1 Plasma Samples 24

2.2.5.2 Perfusates 24

2.2.5.3 Atrial ANP 24

2.2.6 Radioimmunoassay for ANP 24

2.2.6.1 Preparation of Radiolabeled ANP .. . 24

2.2.6.2 Radioimmunoassay 25

2.2.7 Receptor Studies 27

2.2.7.1 Preparation of Glomerular

Microsomes 27

2.2.7.2 Receptor Binding Assay 28

2.3 Data Analysis 30

3.0 RESULTS

3.1 Physical Characteristics of the Rats 31

3.2 Effect of Age on Renal Excretory Function 31

3.3 Radioimmunoassay of ANP 35

3.4 Effect of Age on Plasma ANP Levels 35

3.5 Effect of Age on ANP Secretion Rate 38

3.6 Effect of Age on Atrial ANP Content 43

- vi - 3.7 Effect of Age on Molecular Species

of Circulating ANP 46

3.8 Effect of Age on Molecular Species

of Secreted ANP 51

3.9 Effect of Age on Molecular Species

of Atrial ANP 51

3.10 Effect of Age on Glomerular Receptor

Binding Characteristics 57

3.10.1 Optimal Conditions for

Radiolabelled Receptor Assay 57

3.10.2 Competition and Scatchard Plots 57

4.0 DISCUSSION

4.1 Effect of Age on Renal Function 64

4.2 Effect of Age on Plasma ANP Levels 65

4.3 Effect of Age on ANP Secretion 67

4.4 Effect of Age on Atrial ANP Content 68

4.5 Effect of Age on Molecular Species

of Circulating ANP 70

4.6 Effect of Age On Molecular Species of Secreted ANP .... 73

4.7 Effect of Age on Molecular Species of Atrial ANP 75

4.8 Effect of Age on Glomerular ANP Receptor

Binding Characteristics 75

5.0 SUMMARY 83

6.0 REFERENCES 86

- vii - LIST OF FIGURES

FIGURE PAGE

1 Schematic representation of the processing of y-ANP into N-terminal y-ANP fragment (10K), N-peptide, and a-ANP (3K), the active circulating form 3

2 Schematic overview of ANP synthesis, storage, release, response, and manipulation 5

3 Schematic representation of the modified Langendorff apparatus used for perfusing atria 22

4 Chromatographic separation of the 125I-ANP and 125I

peaks in the labelling of ANP with iodine-125 26

5 Growth curve of the male Wistar rats 32

6 Representative standard curve of a-ANP. B/Bo, Bound

to free ratio 36

7 Recovery of a-ANP added to a pool of rat plasma 37

8 Effect of age on circulating ANP levels 39

9 Relationship between plasma ANP levels and age 40

10 Effect of age on the rate of ANP secretion from isolated right atrium 41 11 Summary of the amount of ANP released from each age

group 42

12 Effect of age on atrial ANP content 44

13 Relationship between amount of ANP released and the atrial ANP content 45 14 Elution positions of synthetic a-ANP and 0-ANP, and purified y-ANP 47 15 Representative RP-HPLC profiles of the plasma IR-ANP. A) 1-month-old B) 3-month-old C) 10-month-old D) 20-month-old 48

- viii - 16 Schematic illustrations of individual plasma concentrations of a-ANP and y-ANP in the total plasma IR-ANP concentration 49

17 Schematic illustrations of average percentages of a-ANP and y-ANP in the total plasma IR-ANP concentration 50

18 Representative RP-HPLC profiles of the perfusate IR-ANP. A) perfusate from 1-month-old rats. B) perfusate from 3-month-old rats. C) perfusate from 10-month-old rats. D) perfusate from 20-month-old rats 52

19 Schematic illustrations of individual perfusate concentrations of a-ANP and y-ANP in the total perfusate IR-ANP concentration 53

20 Schematic illustrations of average percentages of a-ANP and y-ANP in the total perfusate IR-ANP concentration 54

21 Representative RP-HPLC profiles of right auricular extracts of the four age groups of rats. A) extract from a 1-month-old rat. B) extract from a 3-month-old rat. C) extract from a 10-month-old rat. D) extract from a 20-month-old rat 55

22 Representative RP-HPLC profiles of left auricular extracts of the four age groups of rats. A) extract from a 1-month-old rat. B) extract from a 3-month-old rat. C) extract from a 10-month-old rat. D) extract from a 20-month-old rat 56

23 Line graph showing the effect of incubation time on the binding of 125I-ANP to membrane fraction from rat in each age group 58

24 Line graph showing effect of the amount of receptor proteins on the specific binding of 125I-ANP 59

25 Representative competitive inhibition plot of 125I-ANP binding to membrane fractions from rat kidneys in each age group 60

26 Representative Scatchard plot of 125I-ANP binding to membrane fractions from rat kidneys in each age group 62

- ix - 27 Summary of the receptor binding - equilibrium dissociation constant (KJ and maximum binding

capacity (Bm J 63

28 Summary of possible age-related alterations in the homeostatic balance of ANP in aged rats 84

- x - LIST OF TABLES

TABLE PAGE

I Age, body and organ weights in the four age groups of rats 33

II Summary of 24-hour renal clearance data 34

- xi - ACKNOWLEDGEMENT

I would like to first thank Dr. Norman Wong for allowing me to work in his laboratory, and for his continual support as a teacher, inspirer, and friend. I would also like to thank him for his financial sponsorship that enabled me to

complete my graduate studies.

I would like to thank Dr. Eric Wong for his continual friendship and for his

excellent and pertinent advice and technical expertise that assisted me in this

project.

I would like to thank Alice Chan and Edward Mak for their continual friendship

and technical assistance.

I would like to thank David Ridout and Silvia Isakson for their technical

assistance.

- xii - 1.0 INTRODUCTION

1.1 Historical Aspects of ANP

The recent discovery of atrial natriuretic peptide (ANP) is the result of a

lengthy and intensive search for an extrarenal substance or mechanism

that has long been suspected to be involved in the regulation of sodium

and . As early as 1952, Peters (1952) had postulated the

existence of a mechanism that would respond to increase arterial volume

with corresponding increases in the renal excretion of salt. Five years

later, Smith (1957) suggested that the cardiac atria participate in sodium-

volume regulation. Based on this speculation, Gauer, Henry and Seiker

(1961) later demonstrated that a distention of the heart's right atrium

caused both a and natriuresis.

In 1956, Kisch (1956) reported the presence of endocrine-like granules in

tissue of the cardiac atria, which were characterized by Jamieson and

Palade (1964). However, more than ten years passed before it was

observed that granularity of the atrial cells undergoes changes in response

to sodium or volume deprivation and adrenalectomy (Cantin et al., 1975;

De Bold et al., 1978). It was not until 1981 that the significance and

relevance of these atrial granules was demonstrated, de Bold and his

colleagues (1981) found that infusing rat atrial tissue extracts into rats

induced an impressive natriuresis and a reduction in arterial pressure.

They also observed a significant rise in haematocrit. Such effects were

not observed when ventricular extracts were infused into rats.

- 1 - When other investigators (Keeler, 1982; Trippodo et al.,1982; Sonnenberg

and Cupples, 1982) confirmed de Bold's findings, the existence of a potent

natriuretic and vasorelaxant (Currie et al.,1983; Kleinert et al.,1984)

substance in the heart's atrial granules was confirmed. Research based

on these findings became intense, making atrial natriuretic peptide the

most aggressively pursued peptide discovered to date.

1.2 Molecular Structure of ANP

Soon after the confirmation of ANP as a natriuretic peptide, the isolation,

purification, sequencing, and chemical synthesis was accomplished. The

active form of ANP in the circulation is a 28-amlno acid peptide (a-ANP)

in both man and animals (Miyata et al.,1987). This peptide is synthesized

in man as the C-terminus of a 151 polypeptide (in rat 152

amino acid). perpro-ANP, encoded on chromosome 4 (Lewicki et al.,1986).

Post-translational cleavage of the hydrophobic leader sequence generates

a 126 amino acid prohormone, pro-ANP (ANP^^g) or y-ANP, which is

stored in granules within the atrial myocyte (Kangawa et al., 1984b) (Fig.

1). It appears that immediately before, during, or promptly after release,

y-ANP is cleaved by a processing to yield the active C-terminal

peptide a-ANP (ANP^^g). The amino acid sequence of human a-ANP is

same to rat a-ANP, except a single substitution of isoleucine for

methionine at position 12 (Kangawa et al., 1984a). a-ANP contains a 17-

amino acid disulphide ring and a Phe-Arg sequence at the carboxy-

termlnal, which are required for its biological activity (Currie et al., 1984;

Sugiyama et al., 1984). Its half-life in the circulation is short, ranging

from about 30 seconds to about 2.5 minutes (Katsube et al., 1986; Luft

et al., 1986; Yandle et al., 1986). The N-terminal 98 amino acid fragment,

- 2 - peptide

99 \ #126 •s-sH r-hANP

o c— s-sh a-hANP N-Peptide

(10K N-terminal fragment) , (j28 iS Si i i 'S s« 28/ % 1 /9-hANP

Fig. 1 Schematic representation of the processing of y-ANP into N-terminal y-ANP fragment (10K), N-peptide, and a-ANP (3K), the active circulating form. Adapted from Itoh et al., 1988.

- 3 - ANP,.^, of the pro-ANP is co-secreted with a-ANP (Imada et al.,1987).

Although it has a longer half-life (Thibault et al., 1988), it has no clear

biological activity. A unique antiparallel dimer of ANP^.^, comprising 56

amino acids (p-ANP) has also been identified in human plasma (Miyata et

al.,1987). Though it has a slower and longer natriuretic-diuretic action

than a-ANP (Miyata et al.,85; Kangawa et al., 1985), the physiological

significance of this dimer is not clear at this time.

1.3 Release of ANP

The release of ANP into the circulation seems to be stimulated by direct

atrial stretch caused by increases in central blood volume and/or right or

left atrial pressure (de Bold et al.. 1986; Weidman et al., 1989) (Fig. 2).

Increased secretion of ANP is observed in response to intravenous loading

with saline or water (Haller et al., 1987; Yamaji et al., 1986), an acute

increase in sodium intake (Weidman et al., 1988, Hollister et al., 1986),

dynamic exercise (Tanaka et al., 1986) or increased heart rate induced by

pacing (Espiner et al., 1986). In addition, a central shift of volume

produced by the change from the upright to the supine position (Haller

et al., 1987; Hollister et al, 1986) or by water immersion (Epstein et al,

1986, Tajima et al., 1988) also caused an increase in ANP secretion.

There is some evidence that ANP release may also be influenced by

humoral agents. Sonnenberg and Veress (1984) reported that incubation

of isolated rat atria in vitro with arginine (AVP) and

adrenaline but not with the non-pressor analogue of vasopressin (dVAVP),

results in an increased release of ANP. AVP, phenylephrine,

angiotensin II, isoprenaline and carbacholine infusion in vivo produced a

- 4 - volume expansion vasoconstrictors TRAP Fluid Storage Form TUNaV' Y-ANP^ TGFR

Homeostasis T selective Release/Cleavage enzyme ^ 1 Circulating Forms C-Terminus N-Terminus

ANPV98 1/2 a-ANP la30 0 sec. GTP cGMP Renal uncoupled degradation Clearance

Fig. 2 Schematic overview of ANP synthesis, storage, release, response, and manipulation. Abbreviations: ANP, atrial natriuretic peptide; C-terminus, carbosy terminus of the prohormone; N-terminus, amino terminus of prohormone; All, angiotensin II; AVP, arginine vasopressin; PVH, paraventricular nucleus of the ; RAP, right atrial pressure. Adapted from Needleman P, ed., 1988. marked increase in immunoreactive ANP levels (Manning et al., 1985).

Using an isolated heart model, Ruskoaho et al. (1985, 1986) have

demonstrated a role of phosphoinositide system, cytosolic and the

cyclic AMP pathway in the regulation of ANP release. This suggests that

factors which alter the concentration of calcium in heart muscle cells may

influence ANP secretion. In the heart muscle cells, both adrenaline and

acetylcholine alter ANP secretion rate by exerting inotropic and

chronotropic effects on the heart, thereby suggesting the participation of

autonomic nerves in the regulation of ANP release (Toth et al., 1986).

1.4 ANP Receptors

Once released, a-ANP binds and interacts with specific receptors located

in numerous target organ tissues (Fig. 2). ANP receptors have been found

in rat renal glomeruli, medulla and papillary vasa recta (Bianchi et al.,

1985), rat adrenal glomerulosa (Schiffrin et al., 1985), rat mesenteric

artery (Schiffrin et al., 1985), rabbit aorta (Napier et al., 1984), cultured

established epithelial (LLC PK,) cells derived from pig kidney (Napier et al.,

1984), and cultured rat aortic smooth muscle (Hirata et al., 1984). In the

central nervous system, specific high-density binding sites were found in

the subfornical organ, median eminence, area postrema, nucleus tractus

solitarius, and hypothalamic regions, including the anteroventral region

of the third ventricle (Quirion et al., 1984).

Two distinct ANP receptor subtypes have been identified and characterized

in various tissues and types. The ANP B-receptor is a 120,000-dalton

subunit protein that recognize only biologically active ANP fragments

(Leitman and Murad, 1986). It has been linked with the generation of

- 6 - guanosine 3', 5'-cyclic monophosphate (cGMP) In response to ANP binding

(Waldman et al., 1984). The intracellular region of the receptor features homology with protein kinase catalytic domains, soluble guanylate cyclase and a brain adenylate cyclase (Krupinski et al., 1989). It is suggested that binding of ANP to the extracellular domain of its receptor initiates a conformational change in the protein kinase-like domain, resulting in derepression of guanylate cyclase activity (Clunkers et al., 1989).

A second receptor, termed the ANP C-receptor, is distinct from the B- receptor in structure, physiological role, and ligand-binding specificity (16,

Maack et al., 1987). The receptor is consisted of a single 60,000-dalton subunit that appears to exist predominantly as a homodimer on the cell surface (Pandey et al., 1987). Its structure is proposed to consist of a large amino-terminal extracellular ligand binding domain adjacent to a single transmembrane anchor and a short hydrophilic cytoplasmic tail

(Fuller et al., 1988). It is believed that the extracelluar domain of this receptor is alone sufficient for specific high affinity ligand binding; no contribution is necessary from other membrane components or from the membrane or cytoplasmic domains (Porter et al., 1989). The C-receptor binds native ANP as well as various truncated and internal ring- contracted analogs of ANP with high affinity (Leitman and Murad, 1986).

This receptor is not involved in the stimulation of particulate guanylate cyclase and does not appear to mediate directly any of the known biological effects of ANP. However, it has been suggested that the receptor is a clearance site that mediates the metabolic clearance and degradation of this (Maack et al., 1987).

- 7 - 1.5 Intracellular Signals Mediating ANP Action

Binding of ANP to its specific cell-surface receptors activate guanylate

cyclase, resulting in the accumulation of guanosine 3', 5'-cyclic

monophosphate (cGMP) levels fWaldman et al., 1984) (Fig. 2). It has been

suggested that this cyclic nucleotide is the intracellular mediator of many,

if not all, of the physiological actions of ANP (Waldman et al., 1984).

Injection of atrial extracts into rats resulted in a rise of plasma and

urinary cGMP levels (Hamet et al., 1984). ANP-induced cGMP

accumulation in the kidney was found to correlate well with the

distribution of particulate guanylate cyclase (Garcia et al., 1985). The

specific action of ANP on particulate guanylate cyclase activity has also

been reported in rat kidney, aorta, lung, liver, intestine, and testes

(Waldman et al., 1984). In cultured vascular smooth muscle cells and

isolated rat renal glomeruli, a strong correlation between ANP binding to

specific receptors and cGMP accumulation has been demonstrated

(Ballermann et al., 1985).

There are, however, major discrepancies between the kinetics of ANP

binding and the dose-response curve of ANP induced cGMP accumulation

fWaldman et al., 1984; Ballermann et al., 1985). Moreover, in vascular

smooth muscle and cultured endothelial cells, some analogs of ANP that

effectively compete for specific binding sites do not antagonize the ANP-

induced increase in cGMP (Leitman and Murad, 1986). These data raise

the possibility that cGMP is not the sole mediator of ANP actions.

Inhibition of adenylate cyclase activity by ANP has also been reported. In

aorta, mesenteric and renal artery, the basal and hormone-stimulated

- 8 - adenylate cyclase activity was inhibited by ANP in a dose-dependent

fashion (Anand-Srivastava et al., 1984). ANP inhibition of adenylate

cyclase activity in anterior and posterior pituitary has also been reported

(Anand-Srivastava et al., 1985). Others, however, did not find effect of

ANP on adenylate cyclase activity in kidney (Waldman et al., 1984) or on

urinary cAMP excretion (Hamet et al., 1984). This suggests that

inhibition of adenylate cyclase may not be a universal phenomenon.

1.6 Physiology of ANP

ANP has powerful natriuretic and vasorelaxant effects that can be divided

into four categories: effects on vascular smooth muscle; alterations of

renal hemodynamics and sodium excretion; changes of blood pressure

and volume; and effects on renin-angiotensin-aldosterone system (Fig. 2).

1.6.1 Effects on Vascular Smooth Muscle

A prominent physiological effect of ANP is the inhibition of smooth muscle

contraction. In a variety of preconstricted tissues as well as in isolated

arteries and arterioles from several vascular beds, application of atrial

extracts caused a reduction in contractile tone (Kleinert et al., 1984). The

aorta, renal vasculature, and facial vein of the rabbit are very sensitive to

ANP's vasorelaxant action (Currie et al., 1983). In human subjects,

infusions of the ANP have been associated with significant and dose-

dependent increases in blood flow to the skin of the forearm (Bussien et

al., 1986). ANP has also been shown to be particularly effective in

antagonizing angiotensin II-induced contractions of the rabbit aorta

(Kleinert et al., 1984). Maack el al. (1984) have reported that the

vasorelaxant effect of ANP can not be blocked even with a high

- 9 - concentrations of angiotensin II (9.7 nM) given to anaesthetized dogs.

1.6.2 Effects on Renal Hemodynamics and Sodium Excretion

ANP has been demonstrated to produce a marked and sustained increase

in glomerular rate (GFR) in isolated perfused rat kidneys

(Camargo et al., 1984), kidneys of intact animals (Maack et al., 1984) and

human subjects (Weidman et al., 1986). The marked increase in filtration

fraction following ANP suggests that it either selectively constricts the

efferent or dilates the afferent arterioles of the , thereby

enhances the filtration rate (Maack, 1985). Alternatively, the hormone

may act on glomerular membrane to directly increase its permeability

(Maack, 1985). Consistent with the latter possibility is the presence of

high-affinity ANP receptors in the rat glomeruli (Ballermann et al., 1985).

An impressive physiological effect of ANP is its ability to enhance renal

sodium and fluid excretion in anaesthetized dogs (Maack et al., 1984).

However, since the natriuretic effect of the hormone can be abolished by

partial occlusion of the renal artery (Sosa et al., 1986), it suggests that

the increased glomerular filtration rate produced by ANP may be a

necessary component of the natriuresis. In addition, the hormone causes

a washout of the medullary osmotic gradient by altering intrarenal

distribution of blood flow (Maack et al., 1985), thus increasing medullary

flow (Fried et al., 1985). As a result of an increased distal delivery of

filtrate (Maack et al., 1984), renal excretion of sodium is enhanced. The

observation of reduced osmolality without a change in free water

clearance during ANP-induced natriuresis supports such a view (Maack

et al., 1984).

- 10 - Although the renal hemodynamic actions of ANP appear to be essential

for its ability to induce natriuresis, other tubular mechanisms cannot be

excluded. Tubular effects of ANP that have been proposed include a

specific inhibition of normal sodium from the medullary

collecting ducts (Sonnenberg and Cupples, 1982). This may involve

altering the passive permeability of the papillary collecting ducts

(Sonnenberg, 1986), or inhibiting apical sodium permeability (Cantiello

and Ausiello, 1986).

1.6.3 Effects on Blood Pressure and Blood Volume

Administration of ANP not only induces an increase in glomerular

filtration rate and sodium excretion, but also consistently produces a

dose-dependent fall in arterial pressure in several animal species (Maack

et al., 1984, Garcia et al., 1985) and In men (Bussien et al., 1986). The

blood pressure declines immediately after the beginning of infusion and

returns to basal levels soon after termination of infusion (Maack et al.,

1984). This may suggest that ANP acts directly upon the systemic

vascular system to decrease blood pressure rather than indirectly through

volume depletion.

The fall in blood pressure has been ascribed to a decrease in cardiac

output (Breuhaus et al., 1985; Maack et al., 1984). This is thought to

bring about by ANP-induced relaxation of venous smooth muscles, leading

to an augmentation of venous capacitance and a reduction of venous

return (Breuhaus et al., 1985). Since ANP infusion also produces a rapid

rise in haematocrit (Maack et al., 1984), the effect of ANP on venous

return may reflect an alteration of permeability. This causes

- 11 - Intravascular fluid to shift into the interstitium. Regulation of

intravascular volumes and pressures through changes of capillary

permeability could be one of the major functions of ANP (Laragh, 1985).

1.6.4 Effects on Renin-Angiotensin-Aldosterone System

ANP has been reported to produce a marked and sustained suppression

of both renal renin secretion and in anaesthetized

and conscious dogs with inferior vena cava ligation (Maack et al., 1984,

Freeman et al., 1985). These actions may be due to ANP-induced

vasodilation of renal vasculature which blocks renin release (Maack et al.,

1984). Increased glomerular filtration that exerts a negative feedback

signal to reduce renin secretion (Maack et al., 1984), or a direct action of

ANP on juxtaglomerular cells to suppress renin secretion (Krutz et al.,

1986) may also be important.

ANP directly and selectively lowers basal secretion of aldosterone by a

direct action on adrenal glomerulosa cells. This has been shown to block

angiotensin's stimulation of aldosterone release both in vitro (Goodfriend

et al., 1984) and in vivo (Maack et al., 1984) studies. Suppression of

aldosterone in vivo is amplified by the concurrent suppression of renin

secretion (Maack et al., 1984). In this manner, ANP has four distinct

presumed actions on the renin system (Laragh, 1985b): it reduces renin

secretion; it blocks aldosterone secretion; it opposes angiotensin IPs

vasoconstrictive effect; and it decreases aldosterone's sodium-retaining

action. Although not every one of these actions is important in acutely

induced natriuresis, all may play some role in the long-term regulation of

sodium balance and blood pressure.

- 12 - 1.7 Pathophysiological Implications of ANP

The available data briefly summarized above strongly implicate that ANP

is involved in the regulation of blood pressure and sodium balance in

healthy men and in patients with diseases characterized by volume

overload. Administration of ANP produces a dose-dependent increase in

diuresis and natriuresis in patients with chronic renal failure or nephrotic

syndrome (Suda et al., 1988, Windus et al., 1989). In congestive heart

failure patients, ANP infusion caused an acute decrease in preload and

peripheral vascular resistance, an increase in cardiac output, a mild

reduction in blood pressure, a lowered plasma aldosterone, and an

enhanced diuresis and natriuresis (Riegger et al., 1986). In patients with

essential hypertension, infusions of ANP produce an exaggerated diuresis

and electrolyte excretion compared to normotensive subjects (Cody et al.,

1987).

Collectively, these data suggest that an excess or absence of the hormone

may be involved in the of several clinical disorders.

Inadequate ANP secretion, for example, might be implicated in low-renin

essential hypertension and in other states that are characterized by

excessive sodium-volume retention (Laragh, 1985). High levels of

circulating ANP have been reported in patients with congestive heart

failure (Ding et al., 1987), cirrhosis (Gerbes et al., 1985), hypertension

(Cantin et al., 1988), renal failure (Ogawa et al., 1987), and atrial

arrhythmias (Yamagi et al., 1985b). Whether this alteration in plasma

ANP levels reflects an involvement of ANP in the pathogenesis of these

disorders or a consequence of such disorders remains unclear.

- 13 - 1.8 Aging

The physiological changes that occur with aging have a significant impact

on the health of elderly individuals. Changes in renal function are

particularly important, because the kidney plays a central role in drug

excretion, fluid regulation and electrolyte balance.

There are a host of renal structural and functional abnormalities

associated with advancing age. The kidney as a whole progressively loses

mass with age. Studies have shown that the normal kidney loses about

20 percent of its mass between the ages of 40 and 80 years (Tauchi et al.,

1971) . This loss is much greater in the cortex than in the medulla, with

vascular changes accompanying the loss of tissue (Takazakura et al.,

1972) . Histologic examinations of the aging kidney frequently show

complete destruction of the preglomerular arterioles in the cortex, and the

formation of shunts between the afferent and efferent arterioles of the

juxtamedullary area (Ljungqvist and Lagergren, 1962). There is also age-

related chronic glomerular lesions observed in rats, hamster, and mice.

With age, there is a thickening of the glomerular basement membrane due

to accumulation of basement membrane-like material on the epithelial

side of the lamina densa (Yagihashi, 1978). There is a thinning of the

lamina rarae interna and externa, and an increasing of mesangial matrix

with age. Finally, there is an increase in the collagen content of whole

glomeruli and basement membrane with age (Kalant et al., 1977). In

addition to these structural abnormalities, there is a functional loss of

glomeruli with preservation of blood flow to the (Luft et al.,

1985). The human kidney contains 800,000 to 1,000,000 glomeruli from

birth to age 40, following which this number may be reduced by one-third

- 14 - to one-half of its original number by the time the subject reaches 70

(Moore, 1943).

These pathological alterations in the glomeruli may contribute, in part, to the observed age-related decline in renal function after the age of 30

(Davies and Shock, 1950). Bricker (1969) was the first investigator to speculate that decreases in specific renal functions with age parallel the decrease in glomerular filtration rate. In 70 healthy male subjects, Davies and Shock (1950) reported that renal blood flow decreases from 600 ml/min at age 40 to 300 ml/min at age 85. As a result, glomerular filtration rate falls from 120 ml/min to 60 - 70 ml/min at age 85.

Creatinine clearance, which closely parallels glomerular filtration rate, also decreases with age. This decrease begins at age 40 and continues in a more or less linear fashion at 0.8 ml/min per year per 1.73 m2 of body surface area (Rowe et al., 1976).

Since virtually all the ingested NaCl are eliminated by the kidneys (Luft and Weinberger, 1982), alterations in renal functions due to aging may have major influences on sodium homeostasis. When subjected to salt deprivation, older individuals are unable to conserve sodium as rapidly and efficiently as younger individuals (Epstein and Hollenberg, 1976).

Although the exact cause for this is not clear, it may be related to alterations in the renin-aldosterone system. In addition, elderly subjects have lower plasma renin activities and urinary aldosterone excretion when placed on an unrestricted diet or immediately following sodium restriction

Weidman et al., 1975). On the other hand, elderly subjects are more likely to develop an exaggerated natriuresis after adminstration of a water

- 15 - or saline load than younger subjects (Lindeman et al., 1970; Schalekamp et al., 1971). Collectively, the data indicates that the ability of the aging kidney to regulate sodium excretion in response to volume contraction and expansion is compromised.

In addition to the inappropriate responses in maintenance of sodium homeostasis, the aging kidney can neither dilute nor concentrate urine as well as a younger kidney (Rowe et al., 1976). One explanation attributes such a change to the age-related decrease in the number of functional thus causing an increase in solute load per . Hence, an obligate diuresis limits the capacity of the kidney to concentrate urine maximally (Rowe et al., 1976b). Another possible explanation for the reduction in concentrating ability relates to changes in distribution of renal blood flow. An increase in the medullary blood flow causes a washout of the normally hypertonic fluid in the renal medulla and thereby reduce the concentrating capacity of the countercurrent system

(Takazakura et al., 1972). The ability to concentrate urine also depends on the response to vasopressin. The aging kidney is less sensitive to the effects of vasopressin (Philips et al., 1984). In vitro experiments have shown that with aging the cyclic AMP generation is reduced in response to vasopressin, and this might also play a role in the age-related decline in urine concentrating ability (Goddard et al., 1984). Further studies have demonstrated that serum vasopressin levels are actually higher in older people than in younger subjects with similar levels of dehydration

(Miller et al., 1953). These findings indicate that although the elderly are able to produce an adequate amount of vasopressin, their kidneys do not respond to vasopressin as well as those of younger people.

- 16 - Recently, there have been several reports that document an elevated ANP

concentration In the plasma of the aged individual, and have established

a significant positive correlation between plasma ANP levels and age

(Haller et al., 1987; Ohashi et al., 1987; Richards et al., 1986; Sagnella

et al., 1986; Larochelle et al., 1987; Ezaki et al., 1988). In addition, it was demonstrated that infusion of 500 ml of 0.15M NaCl produces a more

elaborate increase in ANP concentration in elderly men relative to young

men (Ohashi et al., 1987). Haller et al. (1987) reported that the increase

in circulating ANP levels following saline loading is correlated with age.

Yamasaki et al. (1988) showed that infusion of hypertonic saline (2.5%

NaCl) caused a 119% increase in plasma ANP concentration in elderly men (51 - 61 years) compared to 37% in young men (20 - 22 years).

Moreover, Heim et al. (1989) demonstrated that a small (33 mg) intravenous administration of a-ANP produced an exaggerated hypotensive, diuretic, natriuretic, and calciuretic responses in the elderly men. In another study, Tajima et al. (1988) reported that head-out water immersion induced a pronounced release of ANP in the elderly relative to the young. This immersion-induced increase in plasma ANP levels was related to exaggerated natriuretic and diuretic responses in the elderly

(Epstein et al., 1987). Collectively, these findings suggest that the homeostatic balance of ANP and the sensitivity to ANP in aged individuals may somehow be altered.

Since aging is associated with a compromised ability in maintaining

sodium balance, it is of interest to investigate if this is related to the alteration in ANP homeostasis. Thus, the present experiments were designed to examine some of the putative age-related disturbances

- 17 - in ANP homeostasis. Specifically, the intracellular storage of ANP, the processing of pro-ANP to active a-ANP, the secretion of ANP from the atrium, circulating ANP concentrations, and ANP receptor binding kinetics were studied. Atria from rats of different rats were isolated and superfused in a modified Langendorff apparatus to measure the rate of spontaneous ANP secretion. The atrial tissue and plasma was quantitatively evaluated for ANP content. Reverse-phase high performance liquid chromatography (RP-HPLC) was employed to analyze the various molecular species of ANP in the atrial tissue, perfusing medium and circulation, in order to assess age-related modifications. ANP receptors from glomerular membranes were isolated and radioligand binding studies performed to appraise if receptor affinity and density are affected by age.

- 18 - 2.0 MATERIALS AND METHODS

2.1 Materials

2.1.1 Rats

Male Wistar rats were purchased from Charles River Breeding

Laboratories, Wilmington, MA, and housed in climate-controlled rooms.

They were fed with standard rat chow (Ralston Purina Co., St. Louis,

MO) and had free access to tap water. They were studied at the desired

ages. The rats were divided into four age groups: group 1 rats, 1-

month-old (aged 33+1 days); group 2 rats, 3-month-old (84 + 3 days)

and is used as the control group; group 3 rats, 10-month-old (289 ± 12

days); and group 4 rats, 20-month-old (596 ± 4 days). In some

instance, rats whose age fall outside of the four specified age groups

were also used for the determination of plasma ANP levels.

2.1.2 Chemicals

Unless otherwise stated, all chemicals were purchased from Sigma

Chemicals, St. Louis, MO.

2.2 Methods

2.2.1 24-Hour Clearance Studies

Rats from each age group were housed in metabolic cages and had free

access to tap water and rat chow. After readjustment to the new

environment in the metabolic cages for 7 days, the first baseline

clearances were determined. Twenty-four hour urine and free-flowing

tail vein blood were collected from each rat. Sodium was analyzed with

a flame photometer (Instrumentation Laboratory 943, Milan, Italy).

- 19 - Plasma and urinary were determined with a VP Abbott

spectrophotometer (Abbott Laboratory, Dallas, Tex). The daily sodium

intake for the rats varied from 7.2 to 14.8 g.

2.2.2 Measurement of Plasma ANP Levels

The rats were anaesthetized with sodium pentobarbital (65 mg/kg)

(M.T.C. Pharmaceutical, Cambridge, Ontario) administered

intraperitoneally. Blood (2-3 ml) was withdrawn from the abdominal

aorta after the rats were anaesthetized for at least 45 minutes. The

samples were transferred to chilled siliconized Vacutainer tubes

containing 50 ul aprotinin (1,000 kallikrein inactivator units/ml) and

EDTA (ethylenediaminetetraacetic acid) (1 mg/ml), then immediately

placed on ice and promptly centrifuged at 4°C at 3,000 rpm.

Plasma samples were first subjected to ANP extraction before analyzed

by radioimmunoassay (RIA) methods . Extraction was carried out using

Sep-Pak C-18 cartridges ( Association Inc. Milford, MA) activated

with 100% methanol (5 ml) and washed with distilled water (12 ml).

Plasma samples were diluted in equal amounts of 0.1 N Hcl, and then

allowed to pass through the cartridge twice by gravity. The cartridge

was then washed with water (12 ml) and the wash was discarded.

Elution of the absorbed ANP was carried using 2 ml of 80% methanol.

The effluent was collected and air dried. The dry sample was

reconstituted in RIA buffer containing 0.1% BSA, 0.01%

triton X-100, and 0.1% NaN3, and analyzed for ANP concentration. The

remaining sample was retained for HPLC analysis.

- 20 - 2.2.3 Spontaneous Release of ANP

After the blood samples were taken, the heart was removed. Both atrial

appendages were quickly excised, and the right atrium was incubated

at 37°C in 10 ml of Tyrode's (in Mm: CaC1.2H20, 1.8; Kcl ,2.7;

MgC1.6H20, 1.1; NaCl, 13.7; Na^PO^ 0.4; D-, 5.6). The left

atrium was save for later determination of ANP content. Both kidneys

were also removed for receptor binding studies. The perfusing medium

was gassed continuously with 95% 02 and 5% C02. Atrial appendages

from the 4 different age groups of rats were perfused simultaneously in

separate chambers in a modified Langendorff apparatus schematically

represented in Fig. 3. The reservoir chamber consisted of a 3-ml

syringe with an inflow tube (PE-100) inserted into the top of the syringe

for the addition of perfusate and held in place with a rubber stopper.

A similar tube was connected to the tip of the syringe to allow outflow

of perfusate. The entire chamber was secured to a stand by a clamp.

The perfusion rate was set at a constant flow rate of 0.5 ml/min by a

peristaltic pump (model PP03, Scientific Instruments, Inc., New York).

The atria were perfused for 40 minutes to allow the hormone secretion

rates to stabilize. After the stabilization period, the perfusate was

collected at 5-rninutes intervals for 100 minutes. The perfusate was

collected by LKB fraction collectors (Bromma, Sweden) and immediately

placed on ice until assay the same day.

2.2.4 Atrial ANP Content Determination

To determine atrial ANP content, both the right and left atria obtained

from the perfusion studies were placed in 2 ml of 0.1 M acetic acid and

kept at -20°C until assay. On the day before assay, the atria were

- 21 - Incubator (37 c 5% C02 Air)

Fig. 3 Schematic representation of the modified Langendorff apparatus used for perfusing atria.

- 22 - homogenized by VisTishear tissue grinder (VisTis Co., Gardiner, N.Y.)

separately for 2 minutes in acetic acid and centrifuged for 30 minutes

at 30,000 rpm. The pellet was discarded and the supernatant was

stored at -20 °C overnight. On the day of assay, the supernatant was

thawed and centrifuged again at 30,000 rpm for 20 minutes to further

disrupt the membranes. The supernatant was then collected for

radioimmunoassay.

2.2.5 HPLC Analysis of ANP Molecular Species

Plasma, perfusate, and tissue samples were subjected to reverse-phase

high-performance liquid chromatography (RP-HPLC) using a Whatman

Partisil ODS-3 Ci8 column (Waters Association Inc. Milford, MA). The

column was first equilibrated with 10% acetonitrile (CH3CN) in 0.1%

trifluoroacetic acid (TFA). The gradient elution was carried out over 55

minutes using a linear gradient (slope 0.4%/min) of 19% - 40% CH3CN

in 0.1% TFA for the first 24 minutes and then at a constant 60%

CH3CN in 0.1% TFA for the remaining period . The flow rate was

maintained at 1 ml/min (Shimadzu model LC-6A pumps, Kyoto, Japan).

The column effluent was monitored at 215 nm (Shimadzu model SPD-

6AV spectrophotometric detector, Kyoto, Japan) and collected at 1-

minute intervals (LKB fraction collector, Bromma, Sweden). The column

was maintained at a constant temperature of 30 °C (Shimadzu model

CTO-6A oven, Kyoto, Japan), and the column pressure ranged from 90

to 120 kgf/cm2. Fifty pi of bovine serum albumin (0.2 g/1) was added

to each collected fraction (50 ,ul) before freezing. After freeze-drying, the

fractions were resuspended in RIA buffer, and the ANP level in each

fraction was measured by radioimmunoassay. Calibration was

-23 - performed against synthetic a-ANP (#9103, Peninsula Laboratories,

Belmont, CA), synthetic |3-ANP (#9105, Peninsula Laboratories, Belmont,

CA) and y-ANP purified from rat atrium using the method of Kangawa

et al. (1985).

2.2.5.1 Plasma Samples

The extracted plasma samples were frozen and lyophilized overnight.

The dry sample was reconstituted in 50 ul of HPLC solvent (60% CH3CN

in 0.1% TEA) and injected into the HPLC system by autoinjector

(Shimadzu model SIL-6A, Shimadzu Corp. Kyoto, Japan).

2.2.5.2 Perfusate

Effluents from an isolated rat atrium collected over the 100-minute

perfusion period were pooled together, and lyophilized. The dry powder

was then reconstituted in 200 ul of HPLC solvent, centrifuged, and 25

ul sample was injected into the HPLC system.

2.2.5.3 Atrial ANP

The homogenized atrial suspension was diluted 20 times in HPLC

solvent, centrifuged, and a 20-ul aliquot was analyzed by the HPLC

system.

2.2.6 Radioimmunoassay for ANP

2.2.6.1 Preparation of Radiolabeled ANP

Radiolabelling of ANP was achieved by the Chloramine-T method

(Greenwood and Hunter, 1963). A small amount of rat a-ANP^ (1-2

ug) was pipetted into a small plastic vial, together with 20 pi of 0.05 M

- 24 - phosphate and 0.15 M NaCl buffer, Ph 7.4. Fifty ul of 0.5 M phosphate

buffer, pH 7.0, was then added to the mixture. One mCi of carrier-free,

high-specific activity Na125I (about 2 - 3 ul) (Amersham, Oakville, Ont)

was added and thoroughly mixed by finger-flicking the tube. Freshly

prepared Chloramine-T (2.5 ug in 5 ul of 0.15 M NaCl, 0.05 M

phosphate buffer, pH 7.4) was added to the mixture to start the

reaction. The reaction was allowed to continue for 10 - 20 seconds

while the vial was generously mixed by finger-flicking the tube. The

reaction was stopped by adding 300 ul of bovine serum albumin (BSA)

(10 g/1) and the contents of the reaction vial was then quickly

transferred to a gel-filtration chromatography column (0.5 x 15 cm)

packed with Sephadex G-10 beads, to separate 125I-ANP from the free

125I. The eluted fractions were collected at 5 minutes intervals. A small

aliquot (2 ul) from each fraction was counted in LKB 1275 minigamma

counter (Wallac, Finland) to determine the positions of the 125I-ANP and

the 125I peaks (Fig. 4). The 125I-ANP fraction was stored in 25 ul aliquots

at -70°C.

2.2.6.2 Radioimmunoassay (RIA) of ANP

Measurement of plasma, perfusate, and tissue ANP levels were

performed using RIA method. ANP antibodies were purchased from

Peninsula Laboratory Co. (Belmont, CA). The antibodies bind to the C-

terminal end of the peptide and has a 100% cross-reactivity with

human a-ANP, rat a-ANP, and atriopeptin III (Peninsula Laboratory

catalog). The radioimmunoassay was performed in RIA phosphate buffer

containing 0.1% BSA, 0.01% triton X-100, and 0.1% NaN3. Synthetic

rat a-ANP at a concentration of 1000, 500, 250, 125, 62.5, 31.25, 15.6,

-25 - Radioactivity (cpm x million) 5 i

[125-ll-ANP

Fraction No.

Fig. 4 Chromatographic separation of the 125I-ANP and 125l peaks in the labelling of ANP with fodine-125.

- 26 - 7.8, 3.9, 1.95 pg/tube was used to construct standard curves. The

incubation mixture consisted of 100 ul of standard or sample and 100

)il of antibody, and was incubated overnight at 4°C. Iodinated ANP (100

jil) (approximately 24,000 cpm) was added, mixed, and incubated

overnight at 4°C. Bound and free ligands were separated by the double-

antibody method utilizing goat anti-rabbit gamma-globulin (100;ul, 1:50)

in the presence of 1% pooled normal rabbit serum. After the addition

of 750 pi of 5% polyethylene glycol 8000 to facilitate precipitation, the

tubes were centrifuged at 3,000 rpm at 4°C for 30 minutes. The

supernatant was aspirated using a vacuum suction and the radioactivity

in the precipitate was measured in LKB 1275 minigamma counter

(Bromma, Sweden). The radioactivity in the sample was converted to

picogram of ANP by fitting the counts in the unknown samples to the

standard curve by using Least Squares Fit computer program.

2.2.7 Receptor Studies

2.2.7.1 Preparation of Glomerular Membranes

Kidneys obtained previously from the perfusion experiments were used

to prepare glomerular membrane. The kidneys were dissected

longitudinally, and the medulla and papilla discarded. The resulting

cortical rims were finely minced with a razor blade and gently pressed

with the blunt end of a syringe inner piston. The tissue was pressed

through a stainless steel sieve (Bellco Biotechnology, Vineland, NJ) with

a pore size of 150;um, and then rinsed from the undersurface through

stacked sieves with pore sizes of 100 jum (to remove large debris) and

75 jum (to retain glomeruli), respectively. Glomeruli retained on the 75-

pm sieve were thoroughly washed with ice-cold Dulbecco's Phosphate-

- 27 - Buffered Saline (PBS) (Gibco Laboratories, Grand Island, NY) and

collected into a 50-ml centrifuge tube. A small suspension was placed

under light microscope to check for purity. Usually the glomerular

preparation was >95% pure. The glomeruli were washed twice by

centrifugation at 1,000 rpm at 4 °C and resuspended in 50 mM Tris HC1

buffer, pH 7.4. The glomeruli were then kept on ice and homogenized

with a Caframo stirrer (Wiatron, Ont) at setting 8 for 90 seconds. The

homogenate was sedimented at 1,000 rpm for 10 minutes to remove

unbroken cells, nuclei, and debris. The resulting supernatant was

centrifuged at 15,000 rpm for 30 minutes. The pellet was suspended

in Tris buffer with 1% Tyrode salt and vortexed to disperse the pellet.

The glomerular membrane obtained from the kidneys of one rat were

suspended in 2.0 ml of Tris-Tyrode buffer. Two 50 vol aliquots were

taken for protein determination by the Lowry method (Lowry et al.,

1951). The glomerular membrane suspension was kept frozen in 1 ml

aliquots at -70 °C.

2.2.7.2 Receptor Binding Assay

a) Dissociation Curves

The glomerular membrane fractions were thawed and diluted in

assay buffer to yield a final concentration of 0.5 mg/ml

membrane protein. The assay buffer contains 50 mM Tris HC1

(pH 7.4), 1% Tyrode salt, 1 pM aprotinin, 0.4%

phenylmethylsulfonyl fluoride (PMSF), and 0.2% bovine serum

albumin (BSA). To determine the effect of incubation time,

glomerular membranes were incubated with 120 fmol 125I-ANP

(70,000 cpm) for varying periods up to 120 minutes. Dissociation

- 28 - rates were determined by adding 100 nM unlabelled ANP at binding equilibrium and determining the remaining radioactivity

as a function of time up to 120 minutes.

To determine the optimal amount of receptor protein on the specific binding of 125I-ANP, increasing amount of glomerular membrane fraction (0.01 to 1.0 mg/tube) were incubated with

120 fmol (70,000 cpm) for 10 minutes at 20°C in the presence or absence of 100 nM of unlabelled ANP.

Competitive Binding Study

Synthetic unlabelled rat a-ANP was prepared by diluting in assay buffer to concentrations of 100,000, 4,000, 2,000. 1,000, 750,

500, 250, 62.5, 15.6, and 3.9 pg/20 ;ul to determine glomerular receptor density and equilibrium dissociation constant using competitive binding techniques, . The experiment was started by adding 20 pi of unlabelled ANP to each assay tube to saturate the binding sites on the membrane. Each tube was extensively vortexed and incubated in water bath at 20 °C. After 5 minutes,

100 ^ll of radiolabelled ANP (60,000 - 80,000 cpm) was added to each tube and vortexed. The incubation was continued in the water bath at 20 °C for another 10 minutes to allow competition

of labelled and unlabelled ANP for the receptor binding sites.

Glass microfiber filters (Boehringer Mannheim, Indianapolis, IN) were placed into 1% polyethylenimine and soaked for 2 to 4 hours at room temperature. At the end of the incubation period, the pre-soaked filters were placed on a filtration apparatus, and

- 29 - excess polyethylenlmine was removed by briefly applying suction.

Twelve ml of ice-cold wash-buffer (PBS containing 0.2% BSA)

were added to the first tube, vortexed, and poured onto the first

filter, followed by a 12-ml wash of the first tube. Suction was

then applied to separate bound radioactivity from free, and the

filter was washed with an additional 12 ml of wash buffer. In the

same fashion, one duplicate set of standards was separated first,

followed by all samples, and then by the second set of standard

tubes, also in duplicate. Filter-associated radioactivity was

counted in a LKB 1275 minigamma counter (Wallac, Finland)

with 75% counting efficiency. Averages of duplicate

determinations of bound 125I-ANP were used for data analysis.

2.3 Data Analysis

Results are expressed as mean + standard error of the mean (SEM) for

separate experiments. Glomerular ANP receptor density (B^) and

equilibrium dissociation constant (KJ were determined from the binding

data using a computer-based LIGAND program (Munson and Rodbard,

1980). Linear regression analysis was used to determine correlations

between variables. Statistical analysis was performed using unpaired

Student's t test or analysis of variance when appropriate, to evaluate

the significance of the difference between means. P value of less than

0.05 was considered to be significant.

- 30 - 3.0 RESULTS

3.1 Physical Characteristics of the Rats

The rate of growth of the Wistar rats was carefully observed over the

duration of the present study. This is illustrated in Fig. 5. The rats

exhibited two growth phases: an initial rapid-growth phase, as the rat

rapidly gained weight from birth to approximately 100 days of age; and

a plateau phase, in which a slower growth rate was observed.

Physical characteristics of the four age groups of rats: whole body

weights, whole heart, right atrium, left atrium, and kidneys, are

summarized in Table 1.

3.2 Effect of Age on Renal Excretory Function

Table 2 summarizes glomerular nitration rate (GFR), 24-h urinary

sodium excretion (UNaV), fractional sodium excretion (FEpjJ, plasma

sodium (PNa), and fractional excretion of water (FE^o) from the 24-hour

clearance study. GFR steadily increased from 1 to 10 months (both p

< 0.005 vs 3-month-old), and declined with advancing age to 20 months

(p < 0.05 vs 3-month-old, p < 0.0005 vs 10-month-old).

The changes in UNaV seemed to parallel the changes in GFR UNaV

increased from 1 to 10 months (both p < 0.05 vs 3-month-old), and

decreased at 20 months (p < 0.05 vs 3-month-old). The difference

between the 10- and 20-month-old groups was statistically significant

(p < 0.05).

-31 - Body weight (g) 800

600

m a W 0 OB

400

200

m

0 0 100 200 300 400 500 600 700 Age (day)

Fig. 5 Growth curve of male Wistar rats. Rats were purchased at 1 or 3 months old and allow to grow to the desire age. Each value represents the mean of 2 to 5 rats. TABLE I

AGE. BODY AND ORGAN WEIGHTS IN THE FOUR AGE GROUPS OF RATS

AGE GROUP (month)

1-Month 3-Month 10-Month 20-Month

No. of Rats 36 48 29 21

Age (days) - Range 30-36 70-95 200 - 350 580 - 620 - mean ± SEM 33 ± 1 84 ±3 289 ± 12 596 ±4

Weight (g) - Range 126 - 165 280 - 360 550-710 520 - 655 - mean ± SEM 150 ±3 334 ±7 600 ± 15 576 ± 19

Heart Weight (g) 0.65 ± 0.05 0.94 ± 0.02 1.66 ± 0.04 1.55 ± 0.04

Right Atrium 23.2 ± 1.1 33.7 ± 0.8 54.7 ± 2.3 59.4 ± 2.2 Weight (mg)

Left Atrium 17.4 ±0.9 22.7 ± 1.0 42.1 ± 2.7 41.0 ±3.9 Weight (mg)

2 Kidneys 1.53 ±0.05 2.17 ±0.05 3.80 ±0.14 3.40 ± 0.21 Weight (g)

Values are expressed as mean ± SEM

-33 - TABLE II SUMMARY OF 24-HOUR RENAL CLEARANCE DATA

AGE GROUP (month)

1-Month 3-Month 10-Month 20-Month

No. of Rats 15 22 14 10

a b PNa (mEq) 143 + 0.3 142 ±0.3 144 ±0.2" 145±0.3 t

GFR (ml/min) 1.90 ± 0.07 b 3.21 ± 0.07 4.50 ± 0.10 b 2.43 ± 0.06 a f

b a a UNaV (mEq/24h) 1.79 ± 0.04 2.32 ± 0.06 2.72 ± 0.07 2.01 ± 0.06

b b b FENa (%) 0.66 ± 0.07 0.51 ± 0.03 0.42 ± 0.03 0.57 ± 0.05

FE^ (%) 0.71 ± 0.05 b 0.50 ± 0.04 0.44 ± 0.05 b 0.55 ± 0.04 b

Values are expressed as mean + SEM

Abbreviations:

PNa - plasma sodium GFR - glomerular filtration rate

UNaV - 24-h urinary sodium excretion

FENa - fractional sodium excretion

FEH;>ri - fractional excretion of water

a - p < 0.05 as compared to 3-month-old b - p < 0.005 as compared to 3-month-old t - p < 0.05 as compared to 10-month-old

- 34 - Similarly, FE^ and FEj^q values were altered due to the changes in

GFR. Both FElia and FE^q was increased in a stepwise fashion from 1

to 10 months of age (both p < 0.005 vs 3-month-old), and decreased

with advancing age to 20 months (p < 0.005 vs 3-month-old).

Plasma sodium concentration (PNa) decreased from 1 to 3 months (p <

0.05), and increased at 10 months and 20 months of age (both p <

0.001). Between 10- and 20-month-old, PNa was statistical different (p

< 0.05).

3.3 Radioimmunoassay

As shown in Fig. 6, binding of 125I-a-ANP to the anti-ANP serum was

inhibited by unlabelled a-ANP.

The 50% binding intercept of the standard curve was 45 ± 12 pg/tube

for 22 determinations. The minimum detectable quantity was 5

pg/tube. The inter- and intra-assay coefficients of variation of IR-ANP

measurements were less than 12%. The recoveries of various quantities

(1.25 to 900 pg) of a-ANP added to a pool of rat plasma or buffer are

shown in Fig. 7. The regression line calculated from the amount of

a-ANP added vs. the amount recovered was almost superimposable on

the theoretical line (100% recovery).

3.4 Effect of Age on Plasma ANP Levels

Sodium pentobarbital was chosen in the present study to anaesthetize

the animals because it is an anaesthetic agent widely used and has the

least effect on the circulating ANP levels (Horky et al., 1985). Only a

-35 - B/Bo {%)

Fig. 6 Representative standard binding curve of a-ANP. B/Bo, Bound to free ratio.

- 36 - ANP found (pg/tube) 1200 i

0 200 400 600 800 1000 ANP added (pg/tube)

100% recovery Found

Fig. 7 Recovery of a-ANP added to a pool of rat plasma. The dotted line represent 100% recovery. The results shown are the mean+SEM of triplicate determinations.

- 37 - small amount of blood (2-3 ml) was taken from each animal to

minimize any effect of volume depletion on the release of ANP. During

the anaesthetization and blood withdrawal process, extreme care was

exercised to avoid any stimulation to the rats.

The mean plasma irnrnunoreactive ANP (IR-ANP) concentration in

1-month-old rats was 75 + 4 pg/ml (N = 31, range: 46 to 124 pg/ml),

and this was increased to a mean of 91 ±4 pg/ml (N = 48, range: 51

to 132 pg/ml) in 3-month-old rats (p < 0.005). The 10-month-old rats

had a mean level of 115 ± 8 pg/ml (N = 27, range: 52 to 210 pg/ml, p

< 0.005 vs 3-month-old). The highest level of ANP was found in the 20-

month-old, at 160 ± 17 pg/ml (N = 18, range: 91 to 328 pg/ml) and it

was significantly higher than the other three younger age groups (p <

0.001) (Fig 8). Together with additional data obtained from rats with

various ages, there was a significant positive correlation between plasma

ANP levels and age (N = 147, r = 0.59, p < 0.0005) (Fig 9).

3.5 Effect of Age on ANP Secretion Rate

The spontaneous release of ANP from the isolated right atrium in each

age group is illustrated in Fig. 10. The release of ANP increased from

5.74 + 0.42 pg/min/mg to 10.0 ± 0.65 pg/min/mg as the rat matured

from 1 to 3 months (p < 0.005). However, as the rat aged, the release

of ANP was significantly decreased to 7.30 ± 0.43 pg/min/mg in the 10-

month-old (p < 0.005 vs 3-month-old) and 4.70 ± 0.25 pg/min/mg in

the 20-month-old (p < 0.005 vs 3-month-old). Fig 11 illustrates the

mean secretion rate in the different groups of rats. There was no

apparent correlations between the rate of ANP release and the plasma

- 38 - Plasma IR-ANP (pg/ml) 200 i

1-month 3-month 10-month 20-month Age

Fig. 8 Effect of age on circulating ANP levels. The results shown are mean+SEM. N = 31, 48, 27, and 18, for 1-, 3-. 10-, and 20-month-old rats, respectively. * p < 0.005 as compared to 3-month-old

- 39 - Plasma IR-ANP level(pg/ml)

N - 147 r * 0.59 p < 0.0005 Y ' 0.15 X * 78.2

a »

8 \f

0 100 200 300 400 500 600 700 Age (day)

Fig. 9 Relationship between plasma ANP levels and age. (N • 147, r = 0.59, p < 0.0005). Rate of IR-ANP secretion (pg/min/mg)

14 -

12 -

2 -

0 H 1 1 1 1 1 r— 1 1 1 r~ 0 10 20 30 40 50 60 70 80 90 100 Time (minute)

A"- 1-month 3-month -Q-10-month -i^-20-month

Fig. 10 Effect of age on the rate of ANP secretion from isolated right atrium. The results shown are mean + SEM. N - 15, 24,16, and 12, for 1-, 3-, 10-, and 20-month-old rats, respectively. Rate of IR-ANP secretion (pg/min/mg)

1-month 3-month 10-month 20-month Age

Fig. 11 Summary of ANP released from each age group. The results shown are mean+SEM. N = 15, 24, 16, and 12, for 1-, 3-. 10-, and 20-month-old rats, respectively. * p < 0.005 as compared to 3-month-old.

- 42 - ANP concentration (N = 44, r = 0.057, p > 0.5), suggesting that high

plasma ANP concentrations found in the older rats were not due to

inappropriate secretion.

3.6 Effect of Age on Atrial ANP Content

The concentration of ANP in right and left atrium of the four age groups

of rats is illustrated in Fig. 12. The average right and left atrium of the

1-month-old rats contained 230 ±14 and 182 ± 6 ng ANP/mg tissue,

respectively; this was increased to 360 ±15 and 315 ± 9 ng ANP/mg

tissue, respectively, in the 3-month-old rats (p < 0.005 in both atria).

On the other hand, ANP content in right and left atrium were slightly

lower but did not reach statistical significance in the 10-month-old (329

±17 and 286 ±12 ng/mg tissue, respectively, p > 0.05 in both atria vs

3-month-old). In 20-month-old, the ANP content were further reduced

to 175 ± 9 and 140 ± 8 ng/mg tissue, respectively, (p < 0.005 in both

atria vs 3-month-old). ANP content in both atria was lower in the 20-

month-old group than the 10-month-old group (p < 0.005). In all age

groups, the right atrial ANP concentration was significantly greater than

the left (p < 0.05).

Interestingly, there was a positive correlation between the rate of ANP

release and the right atrial ANP content (N = 42, r = 0.50, p < 0.01)

(Fig. 13). This suggests that the release of ANP from the right atrium

may depend on the atrial ANP content. On the other hand, there was

no correlation between plasma ANP levels and atria ANP concentrations

(N = 41, r = 0.034, p > 0.5), again confirming the previous results that

elevated plasma ANP levels found in the older rats may not be due to

- 43 - Atrial IR-ANP concentration (ng/mg) 400 i

Fig. 12 Effect of age on atrial ANP content. The results shown are mean+SEM. N = 15, 24,16, and 12, for 1-, 3-, 10-, and 20-month-old rats, respectively. * p < 0.005 as compared to 3-month-old; " p < 0.005 as compared to 10- month-old.

- 44 - Fig. 13 Relationship between amount of ANP released and the atrial ANP content. (N = 42, r = 0.50, p < 0.01). altered secretion.

3.7 Effect of Age on Molecular Species of Circulating ANP i To characterize the molecular species of plasma ANP in the different age

groups of rats, plasma extracts were analyzed by RP-HPLC coupled with

RIA. The elution positions of standard a-, B-, and purified y-ANP are

depicted in (Fig. 14). Representative RP-HPLC profiles of plasma

extracts from each age group of rats are illustrated in Fig 15. The

plasma IR-ANP in all age groups consisted of two major components

with different molecular weights. The first peak eluted at the position

of synthetic a-ANP, and the second emerged with an apparent molecular

weight (13 K daltons) of y-ANP.

In 5 plasma extracts from the 1- and 3-month-old rats, about

70 - 100% of the IR-ANP in the plasma had a molecular size similar to

that of a-ANP (Fig 16). A smaller but distinctive peak was eluted at an

apparent molecular weight of y-ANP in all but two of the samples, and

represented between 8% to 30% of the total IR-ANP. With increasing

age, the proportion of the two molecular species of ANP was changed.

Fig. 17 summarizes the mean percentage of a- and y-ANP, The result

represents the average percentage from 5 plasma extracts analyzed

separately. The mean percentage of y-ANP in the total IR-ANP was 13

± 4%, 16 ± 7%, 26 ± 7%, and 39 ± 4% for 1-, 3-, 10- and 20-month-old,

respectively. Thus, the y-ANP concentration and the percentage of y-

ANP in IR-ANP were markedly Increased in 20-month-old rats (p < 0.01

vs 3-month-old).

- 46 - IR-ANP (fmol/fraction) CH3CN gradient {%) 300 | 1 70

Alpha-ANP 60 250

200

Gamma-ANP 150

100

6 11 16 21 26 31 36 41 Fraction No.

IR-ANP CH3CN gradient

Fig. 14 Elution positions of synthetic a-ANP, synthetic p-ANP, and purified y-ANP on

a C18 Partisil column eluted with a linear gradient of 0.1% TFA-acetonitrile. The a-IR-ANP level in each fraction was assayed by the RIA for a-ANP.

- 47 - A) 1-month-old B) 3-month-old

IR-ANP (fmol/fraction) CH3CN gradient (%) IR-ANP (fmol/fraction) CH3CN gradient (%) 50 i 1 100 35 100

Alpha-ANP Alpha-ANP 30 80 80

25

60 60 20 -

15 40 - 40

Gamma-ANP Gamma-ANP

Q j I 1 I I I I | I I I 1 | I I I I | I I I I | IVI I | I I I I | I I I I | I I I lQ 1 6 11 16 21 26 31 36 41 1 6 11 16 21 26 31 36 41 Fraction No. Fraction No.

• IR-ANP CH3CN gradient IR-ANP CH3CN gradient

c) 10-month-old D) 20-month-old

IR-ANP (fmol/fraction) CH3CN gradient (%) IR-ANP (fmol/fraction) CH3CN gradient (%) 30 100 25 100 Alpha-ANP Alpha-ANP 25 80 80

20 Gamma-ANP 60

15 -

Gamma-ANP 40

Q I I I I | I I I I | I I I I I I | I I I I | I I I I | I I I I | I I I I | I I I Q 0 | I I I I | •• I I | I I I I | I • I • | n IVJCPI I I | I t I I | I I i I | I I I I Q |Tl I 1 6 11 16 21 26 31 36 41 1 6 11 16 21 26 31 36 41 Fraction No. Fraction No.

• IR-ANP • CH3CN gradient • IR-ANP CH3CN gradient

Fig. 15 Representative RP-HPLC profiles of the plasma IR-ANP. A) 1-month-old B) 3-month-old C) 10-month-old D) 20-month-old. The analysis was carried

out in C18 Partisil column eluted with a linear gradient of 0.1 % TFA-acetonitrile. The cc-IR-ANP level in each fraction was assayed by the RIA for a-ANP.The elution positions of synthetic a-ANP and purified y-ANP are indicated.

- 48 - Fig. 16 Schematic illustrations of individual plasma concentrations of a-ANP and y-ANP in the total plasma IR-ANP concentration. * p < 0.01 as compared to 3-month-old. % in plasma IR-ANP

Fig. 17 Schematic illustrations of average percentages of a-ANP and y-ANP in the total plasma IR-ANP concentration from 5 RP-HPLC runs in each group. * p < 0.01 vs 3-month-old

- 50 - 3.8 Effect of Age on Molecular Species of Secreted ANP

In all four age groups, virtually all of the immunoreactlve ANP secreted

by the isolated right atrium had a molecular size similar to that of

a-ANP (Fig. 18). A small percentage of perfusate (1% - 8%) was

chromatographically identical to y-ANP. Fig 19 illustrates the relative

percentage of a- and y-ANP from each RP-HPLC and RIA analysis. The

mean of 3 analyses in each age group is summarized in Fig 20. The y-

ANP represented approximately 1% of the total perfusate ANP in the 1-,

3- and 10-month-old groups, but close to 8% of total ANP in the 20-

month-old was y-ANP (p < 0.05 vs 3-month-old). It indicates that the

cosecretional processing mechanism of y-ANP into a-ANP is influenced

by age.

3.9 Effect of Age on Molecular Species of Atrial ANP

Representative RP-HPLC profiles of atrial extracts from the right or left

atrium in each age group are shown in Figs. 21 and 22, respectively.

Evidently, the only component of IR-ANP was y-ANP like material in all

4 age groups, and neither a- nor p-ANP were present in either atrial

tissues. There was no significant differences in elution pattern between

the right and left atrium. No major difference was seen between the

four groups of animals.

Previous reports demonstrated that y-ANP may undergo partial

degradation during the extraction procedure to form a-ANP or (3-ANP

(Sugawara et al., 1988). Synthetic a-ANP may also dimerize to form

p-ANP during its exposure to the conditions used for the extraction

(Kangawa et al., 1985). In the present study, the atrial form of ANP was

- 51 - A) 1-month-old B) 3-month-old

IR-ANP (Imol/fraction) CH3CN gradient {%) IR-ANP (tmol/fraction) CH3GN gradient (*) 100 1400 100 Alpha-ANP 1000 1200 - 80 800 1000

60 800 600

600 40 400

400 -

200 - 20 Gamma-ANP 200 -

" 111111 11 16 21 26 31 36 41 1 6 11 16 21 26 31 36 . 41 Fraction No. Fraction No.

IR-ANP CH3CN gradient -e- IR-ANP CH3CN gradient

C) 10-month-old D) 20-month-old

IR-ANP (fmol/fraction) CH3CN gradient («) IR-ANP (fmol/fraction) CH3CN gradient («) 100 3501 1100 1600 Alpha-ANP Alpha-ANP

300 80 80 1200 250

60 60 900 200

150 - 40 40 600

100 Gamma-ANP

300 Gamma-ANP 20 20 50

Otf 11 16 21 26 31 36 41 1 6 11 16 21 26 31 36 41 Fraction No. Fraction No.

• IR-ANP - CH3CN gradient •IR-ANP - CH3CN gradient

Fig. 18 Representative RP-HPLC profiles of the perfusate IR-ANP. A) perfusate from 1-month-old rats. B) perfusate from 3-month-old rats. C) perfusate from 10- month-old rats. D) perfusate from 20-month-old rats. The elution positions of synthetic a-ANP and purified y-ANP are indicated. - 52 - R-ANP Concentration (pmol/mg tissue) 10

8

6 - 7\

4 -

CO

0 \/wmA \A \A V 1-month 3-month 10-month 20-month Age

CZH Alpha-ANP Hi Gamma-ANP

Fig. 19 Schematic illustrations of individual perfusate concentrations of a-ANP and yANP in the total perfusate IR-ANP concentration. Alpha-ANP {%) Gamma-ANP {%) 10

100 -

8

80-

6 60

- 4 40 -

-2 20

0 0 1-month 3-month 10-month 20-month Age

Alpha-ANP H Gamma-ANP

Fig. 20 Schematic illustrations of average percentages of a-ANP and "y-ANP in the total perfusate IR-ANP concentration from 3 RP-HPLC runs in each group. * p < 0.05 as compared to 3-month-old.

- 54 - A) 1-month-oId B) 3-month-old

IR-ANP (Imol/lraction) CH3CN gradient («) IR-ANP <(mol/1raction) CH3CN gradient («) 300 ICO 350

250 300 "

200 250

200 150

150 100 lOO

1 6 11 16 21 26 31 36 41 1 6 11 16 21 26 31 36 41 Fraction No. Fraction No.

• IR-ANP - CH3CN gradient •IR-ANP • CH3CN gradient

C) 10-month-old D) 20-month-old

IR-ANP (fmol/lractloo) CH3CN gradient (%) IR-ANP (fmol/fraction) CH3CN gradient (*) 300 100 3001 1100

Gamma-ANP

250 260 o 80

200 200 60

150 150

40 100 100

20

U111 imprwTmjffflTrTTrTrfnnpnm 111 t*wprmfni 11111q 1 6 11 16 21 26 31 36 41 1 6 11 16 21 26 31 36 41 Fraction No. Fraction No.

• IR-ANP CH3CN gradient • IR-ANP - OH3CN gradient

Fig. 21 Representative RP-HPLC profiles of right auricular extracts of the four age groups of rats. A) extract from a 1-month-old rat. B) extract from a 3-month- old rat. C) extract from a 10-month-old rat. D) extract from a 20-month-old rat. The elution position of purified y-ANP is indicated.

- 55 - A) 1-month-old B) 3-month-old

IR-ANP ((mol/fraction) CH3CN gradient (*) IR-ANP (fmoi/fraction) OH3CN gradient (*) 350 4001 1100

300

3O0 - 250

200

200 - 150 -

1O0 100 -

1 6 11 16 21 26 31 36 41 1 6 11 16 21 26 31 36 41 Fraction No. Fraction No.

•IR-ANP • CH3CN gradient -e- IR-ANP CH3CN gradient

C) 10-month-old D) 20-month-old

„ IR-ANP (f mol/fraction) OH3CN gradient (%) IR-ANP (fmol/fraction) CH3CN gradient {%) 3501 l_i 2 100 100

250 - 300 -

250 - 200

200 - 150 -

150 -

100 100 -

6 11 16 21 26 31 36 41 1 6 11 16 21 26 31 36 41 Fraction No. Fraction No.

• IR-ANP ' CH3CN gradient -e- IR-ANP CH3CN gradient

Fig. 22 Representative RP-HPLC profiles ot left auricular extracts of the four age groups of rats. A) extract from a 1-month-old rat. B) extract from a 3-month- old rat. C) extract from a 10-month-old rat. D) extract from a 20-month-old rat. The elution position of purified y-ANP is indicated.

-56- found to be predominant y-ANP with little or no a-ANP present. No

p-ANP was detected in plasma, perfusate or atrial extract samples. This

suggests the extraction condition used in this study do not alter the

natural molecular form of ANP.

3.10 Effect of Age on Glomerular Receptor Binding Characteristics

3.10.1 Optimal Conditions for Radiolabelled Receptor Assay

Specific binding of 125I-ANP to the glomerular membrane fraction was a

time-dependent process in all four groups of the rats (Fig 23). When

determined at 20 °C in the presence of 0.12 nM labelled ligand

(70,000 cpm), the specific binding increased rapidly and reached a peak

stable equilibrium value within 5 minutes. This was stable for at least

another 15 minutes before a gradual decline in binding was observed.

After 120 minutes of incubation, the specific binding had decreased to

60% of the maximum binding.

At 20°C, the specific binding of 125I-ANP to the glomerular membrane

fraction was proportional to the membrane protein concentration from

0.05 to 0.20 mg/tube (Fig. 24).

3.10.2 Competition and Scatchard Plots

A representative plot of the competitive inhibition binding study using

glomerular membranes from 1-, 3-, 10-, and 20-month-old rats is

depicted in Fig. 25. Nonspecific binding was determined by the addition

of 100 nM of unlabelled ANP. In this series of competitive binding

inhibition experiments, nonspecific binding was linear and accounted

for 2 - 3% of total radioactivity. Binding was not further reduced by

- 57 - 125-1 ANP bound (fmol/mg protein) 500

400 -

300

200 CO

100

ir 0 20 40 60 80 100 120 Incubation Time (min)

1-month -^3-month -B- 10-month 20-month

Fig. 23 Line graph showing the effect of incubation time oh the binding of 12SI-ANP to the rat renal membrane fractions from the four age groups of rats. Membrane suspension (0.1 mg/tube) was incubated with 70,000 cpm (0.12 nmol) 125I-ANP at 20°C. Values are mean of triplicate determinations. Specific Binding of 1251-ANP (cpmxIOOO) 16 i :

0 0.2 0.4 0.6 0.8 1 Glomerular membrane (mg protein/tube)

Rg. 24 Line graph showing effect of the amount of receptor proteins on the specific binding of ^l-ANP. Increasing amount of the renal membrane fraction (0.01 to 1.0 mg/tube) were incubated with 70,000 com (120 fmol) for 10 minutes at 20°C in the presence or absence of 100 nM of unlabelled ANP. Values are means of triplicate determinations.

- 59 - Bound/Total 0.09

0.08 -

0.07 -

0.06

0.05 -

0.04 -

0.03

0.02 •10 -9.5 -9 -8.5 -8 -7.5 -7 -6.5 Log Total ANP (M)

^- 1-month -0- 3-month

Q- 10-month -^f- 20-month

Fig. 25 Representative competitive inhibition plot of 12SI-ANP binding to membrane fractions from rat kidneys in each age group. Membranes (0.1 mg/tube) were incubated with increasing amount of unlabelled ANP (0 to 100,000 pg/tube). Each value represents the mean of duplicate determinations.

- 60 - higher (up to 100 uM) concentrations of unlabelled ANP. In the absence of unlabelled ANP, 6 - 9% of total added radioactivity was specifically bound to the glomerular membrane. Specific binding of 125I-ANP was saturable with increasing concentrations of 125I-ANP. Glomerular membranes isolated from all four groups of rats exhibit a similar pattern. Scatchard analysis of the above data revealed that all 4 groups of rats has a single high class of binding sites with an apparent equilibrium dissociation constant (KJ (Fig. 26).

The result from all competitive binding inhibition experiments is summarized in Fig. 27. Glomerular ANP receptor density increased as the rats matured from 1 to 3 months of age and consistently decreased with advancing age. On the other hand, receptor binding affinity declined (higher k,,) from 1 to 3 months of age and enhanced at 20 months of age.

- 61 - Bound/Free 0.1 i

0 2 4 6 8 10 12 14 16 Bound (pmol/L)

•A- 1-month 3-month •S- 10-month 20-month Fig. 26 Representative Scatchard plot of ,25I-ANP binding to membrane fractions from rat kidneys in each age group. Membranes (0.1 mg/tube) were incubated with increasing amounts of unlabelled ANP (0 to 100,000 pg/tube). Each value represents the means of duplicate

determinations. In this example, k,, and Bmjix are 0.14 nM and 162 fmol/mg for 1-month-old (triangle), 0.30 nM and 485 fmol/mg for 3-month-old (diamond), 0.29 nM and 353 fmol/mg for 10-month-old (square), and 0.078 nM and 221 fmol/mg for 20-month-old (star), respectively.

-62- Bmax (fmol/mg) Kd (nM) 500 f—

Fig. 27 Summary of the receptor binding results - equilibrium dissociation constants (K„) and maximum binding capacities (B^). The results shown are means+SEM of 11 experiments. * p < 0.001 as compared to 3-month-old. ** p < 0.05 as compared to 3-month-old.

- 63 - 4.0 DISCUSSION

Using senescent (the period around or beyond the median life span)

laboratory animals to assess the potential age-related changes in ANP

physiology may clearify any understanding of this hormone in both

normal and pathological states. The results obtained in this aging

model may also provide a natural extension to the current

understanding in young adult rats. The life span of laboratory rats is

relatively short, approximately 2-3 years, allowing gerontological

studies of this species to be conducted within a reasonable time-frame.

The male rat reaches puberty at around 50 ± 10 days, and the body

weight of the young adult is around 300 - 400 grams (Baker et al.,

1979). It is generally accepted that senescence is about 18 months or

older (Dax, 1987). The selection of age groups in the present study (1-,

3-, 10-, and 20-month-old) is based on this information.

4.1 Effect of Age on Renal Functions

Results from the 24-hour clearance studies clearly indicate the kidney's

ability to excrete salt and water is affected by age. Maturation of rats,

from 1 to 3 months, was associated with an increase in GFR and UNaV,

and a decrease in PNa, FE,^, and FE^Q (Table 2). GFR was at the

highest value when the rat reached about 10 months of age; at which

time both UNaV and PNa were increased while FE,^ and FE^Q were

further reduced. With the aging of rats to 20 months of age, GFR and

UNaV were significantly reduced, resulting in elevation of FE^, FE^o and

PNa. Values obtained in these clearance study are comparable to other

published results (Peiico et al., 1989; Soejima et al., 1988; Hirata et al.,

1985).

- 64 - These observations are consistent with the fact that newborn and young

animal of many species is characterized by an attenuated natriuretic

response to sodium loading (Kleinman, 1982). Saline expansion of the

newborn animal is often associated with the development of edema,

weight gain, and increase in the serum concentration of sodium

(McCance and Widdowson, 1957). These are thought to be caused by

inappropriate reabsorption of sodium in the distal nephron of the kidney

(Kleinman and Bank, 1983). On the other hand, a host of abnormalities

in renal structure and function accompany advancing age (Papper,

1973). Senescence, for example, results in renal atrophy with a

reduction in renal cortical mass (Darmady et al., 1973) and a reduction

in renal blood flow and filtration rates (Davies and Shock, 1950).

Furthermore, a reduction in the tubular maximum capacity (Tm) for

glucose (Miller et al., 1952), and an impairment in the kidney's capacity

to concentrate urine (Miller et al., 1952) and handle an acid load (Adler

et al., 1968) have been associated with aging. These alterations in

kidney function have been implicated to alter the kidney's ability in

maintaining sodium homeostasis.

4.2 Effect of Age on Plasma ANP Levels

Plasma levels of ANP measured by radioimmunoassay vary widely from

approximately 60 to 1650 pg/ml in rats and from 25 to 100 pg/ml in

humans (Lang et al., 1985; Gutkowska et al., 1984; de Bold, 1985).

The discrepancies may be accounted for by differences in anaesthetic

agents used (Horky et al., 1985), procedures for collecting and storing

samples, varying recoveries of extraction methods (Rankin et al., 1987),

or specificity of the antibody employed. The plasma ANP concentrations

- 65 - measured in this study ranged from 50 to 350 pg/ml, which are comparable to most of the published reports.

Although a few studies noted that circulating ANP levels in healthy men do not vary with age (Weil et al., 1986; Rascher et al., 1987). the majority of reports described a tendency for ANP levels to increase with age. Ohashi et al. (1987) demonstrated an eight-fold increase in plasma

ANP levels in elderly men (64 - 91 years old) compared to young men

(24 -28 years old) (120 ± 22 vs 25 ± 5 pg/ml). Haller et al. (1987) observed a 7 times higher ANP concentration in older subjects than in younger subjects (167 + 31 vs 24 + 3 pg/ml). Furthermore, plasma levels of ANP were positively and significantly correlated with age in healthy volunteers (r = 0.71, p < 0.001 by Sagnella et al., 1986; r =

0.35, p < 0.02 by Larochelle et al., 1987; r = 0.82, p < 0.001 by Haller et al., 1987; r = 0.61, p < 0.001 by Richards et al, 1986; and r = 0.46, p < 0.01 by McKnight et al., 1989).

However, whether this relationship also exists in rats has not been previously investigated. The present study demonstrated that plasma

ANP levels in rats is correlated with age. The aged rats (20-month-old) have significantly higher levels of plasma ANP than the other three younger groups, 1-, 3-, or 10-month-old (160 ± 17 vs 75 ± 4, 91 ± 4, and 115 ± 8 pg/ml, respectively). The mean plasma ANP levels in the

20-month-old was 76% greater than in the 3-month-old rats. In addition, a close relationship between plasma ANP levels and age was found (Fig. 9), indicating to the importance of age as a determinant of plasma ANP levels.

- 66 - The reasons for the elevation in plasma ANP concentration in the aged

are not clear. It may be speculated that the elevation is due to a

secondary effect to hyperaldosteronism (Wong et al., 1989; Lendenson

et al., 1987), chronic heart failure (Ezaki et al., 1988), chronic renal

failure (Suda et al., 1988), or hypertension (Sagnella et al., 1986; Cantin

et al., 1988), which are conditions often associated with old age (Rowe

and Minaker, 1985). Alternatively, aging can affect the sensitivity of

physiological regulatory centres thereby causing a raise in basal levels

of circulating (Helderman et al., 1978; Kirkland et al., 1984;

Rondeau et al., 1982). It is also possible that the set-point of ANP basal

levels in the plasma is raised in the aged subjects. Finally, the raised

plasma ANP level may reflect alterations in metabolic clearance of the

circulating peptides. This aspect will be discussed further in the

following section.

4.3 Effect of Age on ANP Secretion

The elevated plasma ANP levels detected in the aged rats may be a

result of increased release of ANP from the atria or reduced clearance

of ANP from the circulation. To determine which mechanism is

responsible, isolated atria were perfused in a modified Langendorff

apparatus to measure the in vitro secretion of ANP. The results

suggested that ANP secretion increased with maturation (from 1 to 3

months old) and progressively decreased with age from 3 to 20 months

(Fig. 11). ANP secretion was reduced by 27% in the 10-month-old and

53% in the 20-month-old when compared to the 3-month-old. These

data clearly indicate that the spontaneous release of ANP increases with

maturation and decreases with aging.

- 67 - Although the spontaneous ANP secretion in the aged rats were

decreased, their plasma ANP levels remained elevated; this suggests that

the metabolic clearance of ANP was altered. Since the kidney has been

demonstrated to be an important site for the catabolism of atrial

peptides (Berg et al., 1988; Tang et al., 1984; Sonnenberg et al., 1988)

and that aging has been shown to be accompanied by a variety of

abnormalities in the structure and function of the kidney (Papper,

1973), it is conceivable that the renal degradation of ANP is

compromised by age. Studies have indirectly demonstrated that when

the renal clearance function is impaired, ANP concentration in the

plasma quickly rise. For example, in nephrectomized rats (Petersen et

al., 1988; Smith et al., 1986) and chronic renal failure patients (Shenker

et al., 1987), the plasma ANP concentration was found to be

significantly raised.

4.4 Effect of Age on Atrial ANP Content

The result of the current study clearly indicated there was age-related

changes in atrial ANP content. Atrial ANP contents in both right and

left atrium of the rats were elevated from 1 to 3 months of age, and

declined slightly but not reaching statistical significance at 10 months.

At 20 months, a dramatic reduction was seen; the atrial ANP

concentration was only 49% of the 3-month-old (Fig. 12). This data is

consistent with previous reports that described a lower atrial ANP

content in the newborn (19 - 21 days old) compared to adults (8-16

weeks old) (Pollock and Banks, 1985; Imada et al., 1985). However, the

changes between adult and older animals have not been previously

examined. This study also revealed that within the same age group the

- 68 - concentration of ANP was greater in the right atrium than in the left atrium (Fig. 12), agreeable with many published reports (Imada et al.,

1985; Cantin and Genest, 1985; Katsube et al., 1985; Tsunoda et al.,

1986). On the other hand, only a few reports shown that ANP concentration between the right and left atrium is similar (Dolan et al.,

1989; Wei et al., 1987).

Since tissue ANP concentration is determined by synthesis, storage and secretion, the age-associated decrease in ANP concentration in the atrium per se does not imply a decrease in synthesis. A depletion of atrial tissue due to excessive secretion could have been the cause.

However, results obtained from the present study indicate that diminish synthesis is the cause. The aged atrium (20-month-old) not only contained the least amount of ANP in the atrial tissue, but also had the lowest secretion rate compared to the other three groups (Figs. 11, 12).

A significant correlation was also found between the rate of ANP release and the atrial ANP concentration regardless of age group (Fig. 13), suggesting that the aging atrium secretes the same proportion of ANP with respect to its ANP content as do atrium from the other three younger groups. Therefore, it is reasonable to conclude that the reduction in ANP secretion by the aging heart is a consequence of decreased synthesis.

In an earlier study. Pollock and Banks (1985) reported a significantly lower level of ANP content in atrial extract prepared from newborn rats

(19 days of age) than from adult animals (56 days of age). When comparing the natriuretic activities of the extracts, they found that the

- 69 - newborn atrial extracts have lower natriuretic activity, and speculated

that the compromised ability of newborn animals to excrete a sodium

load is related to a deficiency of ANP in the atrium. The data obtained

in the present study is consistent with their results. Comparing to the

adults (3-month-old), the young rats (1-month-old) were found to

contain a lower concentration of ANP in the atrium concurrently with

a low sodium excretion (low UNaV) by the kidney (Table 2). In another

study, Inscho et al. (1987) noted a significantly lower hypotensive and

natriuretic activities of atrial extracts prepared from 290-day-old than

from 15-, 39-, or 56-day-old animals. They also observed that the atrial

extracts from the old rats (290 days old) was found to contain more ANP

per tissue DNA level but had reduced biological activities. This

indicates that the composition of individual extracts may be changed

with age. Accordingly, this may provide yet another reason as to why

the ability of the aging kidney to excrete sodium is curtailed.

4.5 Effect of Age on Molecular Species of Circulating ANP

Chromatographic analyses of healthy human plasma samples

consistently showed single ANP peaks that correspond to the active

a-ANP molecules (Yamaji et al., 1985; Sugawara et al., 1985; Theiss et

al., 1987). No other forms of ANP were detected. In normal rat plasma,

one major peak that corresponds to a-ANP was detected, along with

smaller quantity of the large molecular-weight y-ANP (Kangawa et al.,

1984; Misono et al., 1984). In pathological conditions such as chronic

renal failure or congestive heart failure, y-ANP was found to increase

significantly in proportion to the total ANP concentration (Ogawa et al.,

1987; Ding et al., 1987).

- 70 - The result from the present study clearly indicates that both a- and y- molecular forms of ANP are present in rat plasma extracts from all four age groups (Figs. 15, 16). In a total of 20 analyses, no {3-ANP was detected (Fig. 15). This is in accordance with other's findings (Kangawa et al., 1984; Misono et al., 1984). However, y-ANP increases significantly in proportion to the total ANP concentration with age (Fig.

17). Close examination of Fig. 16 revealed that the increase in total plasma ANP concentration with age consisted of a smaller increase in the a component and a larger increase in the y component. This may have pertinent significance; as y-ANP is not biologically active, it may be one of the reasons why age-related reduction in renal sodium excretion occurs in the presence of elevated plasma ANP levels.

In contrast to most studies where y-ANP was found to account for only a small percentage of total ANP in the plasma, the present study showed y-ANP represented as much as 39% of the total ANP (Fig. 17).

This discrepancy may be explained by the fact that the rats used in most studies were relatively young, ranging from 150 to 350 g in body weight. According to the growth curve of the rats used in the present experiment, this can be translated to an age of 1 to 3 months (Fig. 5).

Hence, it is only logical that results obtained from these young rats are compared to 1- and 3-month-old groups in the present study. In these two groups, the average percentage of y-ANP in the plasma were about

14% (Fig. 17), agreeing well with the published results. Larger quantities of y-ANP were found only in the older groups (10- and 20- month-old), which had not been previously examined.

- 71 - In a human study done by Ohashi and his colleagues (1987), it was demonstrated that the increase in plasma ANP in the elderly was due to an increase in a-ANP and not the y-ANP molecule. Species differences could have accounted for the discrepancy. As mentioned before, y-ANP is usually not found in normal human plasma (Yamaji et al., 1985; Sugawara et al., 1985; Theiss et al., 1987), while it is known to exist in normal rat plasma (Kangawa et al., 1984b; Misono et al.,

1984).

Increase in circulating y-ANP may be due to the fact that a-ANP and

y-ANP have different half-lifes (T,/2) in the circulation. T1/2 of a-ANP was calculated to be around 30 seconds (Katsube et al., 1986; Luft el al., 1986), while y-ANP, in contrast, is thought to have a much longer

T1/2. Because a complete molecule of y-ANP has not been synthesized

in vitro, the precise T1/2 of y-ANP is difficult to determine. However, T1/2 of other large fragments of ANP molecules derived from the y-ANP, such

as ANPi3.104 and N-terminal peptide (ANPj.gg), have been evaluated.

ANPig.,^ lacks most of the biologically active C-terminal portion of the

molecule (ANP99.126), but retains the majority of the remaining N-terminal

region; its T,/2 was estimated to be 9 times longer than that of a-ANP

(Katsube et al., 1986). N-terminal peptide (ANP^gg) has the intact N

terminal of the y-ANP molecule but lacks the active C terminal; its T1/2 was 150 seconds, which was 5 times longer than that of a-ANP

(Thibault et al., 1988). Since y-ANP (ANPi.126) has a molecular size

larger than both ANP13.104 or ANP,.^, it is appropriate to assume that its

T1/2 is at least the same, if not greater, than that of ANP13.,04 or ANP,.^.

- 72 - Thus, a diminished degradation due to its longer T1/2 and/or increased

y-ANP secretion, y-ANP's concentration in the plasma may quickly rise.

It is possible that the significant increase in the y-ANP concentration in

the present study is due to reduced renal clearance function as a result

of age-related abnormalities in the structure and function of the kidney.

Since ANP has been shown to be eliminated predominantly by the

kidney (Tang et al., 1984; Berg et al., 1988; Sonnenberg et al., 1988),

a decrease in the renal clearance would easily lead to an accumulation

of y-ANP.

Recently, a bound form of ANP have been detected both in men (Miyata

et al., 1987; Richards et al., 1987) and in rats (Kato et al., 1988).

Another explanation as to why plasma y-ANP may appear to accumulate

in the 10- and 20-month-old rats In the present study is that a-ANP

may exist as a bound form in the circulation. This bound form of ANP

may resemble a large y-ANP molecule in the HPLC analysis, thus

leading to the illusion of excess y-ANP concentration in the plasma.

Also, it is conceivable that age may alter the ratio of free to bound form

of plasma ANP in some unknown way, as it is reported in some

pathological conditions (Kato et al., 1988). Other studies are required

to elucidate the cause and ascribe a significance for the apparent

accumulation of y-ANP in the circulation.

4.6 Effect of Age on Molecular Species of Secreted ANP

Reverse phase-HPLC analyses of the perfusate coupled with

radioimmunoassay demonstrated that the predominant form of ANP

released was chromatographically identical to a-ANP in all four age

- 73 - groups {Fig. 18). However, a minor peak that is chromatographically identical to y-ANP also appeared in many perfusate analyses. The relative proportion of y-ANP to a-ANP was increased with age (Figs. 19,

20). In the 1-, 3-, and 10-month-old groups, y-ANP accounted for approximately 1% of the total ANP; in the 20-month-old group, close to

8% was y-ANP. This suggests a possible age-related defect in the processing of y-ANP to the active a-ANP form prior to secretion.

This finding is consistent with previous observations that the predominant molecular form of ANP found in the perfusion medium collected from isolated adult (Saito et al., 1986; Lang et al., 1985; Currie et al., 1984) or neonatal (Shields and Glembotski, 1987) rat hearts is a low molecular-weight material. The precursor protein represented only minimal amount, if any, of total ANP concentration (Lang et al., 1985;

Currie et al., 1984; Ruskoaho 86, Sugawara 1985 & 1986). Similar results were obtained in studies on human, where the main molecular form of ANP in the plasma taken at the coronary sinus during cardiac catheterization was a-ANP (Sugawara et al., 1985; Yandle et al., 1986b).

Together with our results, these observations support the theory that proteolytic conversion of the prohormone to circulating ANP occurs mainly within cardiac myocytes, and that the low molecular-weight a-ANP found in plasma is released directly from the heart.

There are evidences to support the speculation that the prohormone y-

ANP is released into the circulation due to increased biosynthesis and/or defective conversion or storage. In renal juxtaglomerular apparatus, both active renin and inactive pro-renin have been shown to

- 74 - be released (Hsueh et al., 1983), and their ratio in plasma is altered in

certain physiological and pathological conditions (Delevia et al., 1976;

Day and Luetscher, 1974). Similarly, both the precursor and the active

circulating forms of were detected in the plasma (Carroll et al.,

1988; Cohen et al., 1988; Robbins et al., 1984). Therefore, the

increasing concentration of y-ANP in the perfusate reflects a possible

abnormality in the processing of the precursor hormone to the active

a-ANP in the aging heart.

4.7 Effect of Age on Molecular Species of Atrial ANP

Accumulated evidences have established that in both man and animal

the predominant form of ANP in the heart is the high molecular-weight

y-ANP (Nakao et al., 1984, 1987; Thibault et al., 1987; Kangawa et al.,

1984b). In the present study, y-ANP is the predominant and the only

form of ANP present in all four groups of atrial tissues (Figs. 23 and

24). Apparently, age has no effect on the molecular species of ANP in

the atrial tissue. This is an intriguing observation; since data from the

present isolated atrial perfusion study indicated that perfusing medium

contains predominantly a-ANP, it is apparent that the post-translational

cleavage to form a-ANP occur within cardiocytes and does not require

protease from the circulation. With aging, the atrium secretes an

increasing proportion of y-ANP, suggesting that this processing

mechanism is somehow altered by age. This may imply the capacity of

the putative processing is exceeded in the aged subjects.

4.8 Effect of Age on Glomerular ANP Receptor Binding Characteristics

The actions of ANP, like those of other peptide hormones, are believed

- 75 - to be mediated through binding to specific receptors on cell plasma membranes. It is commonly accepted that circulating peptide hormones may directly regulate the numbers of receptors at target cells, and changes in the ambient concentrations of many peptide hormones are commonly associated with reciprocal changes in the density of their specific receptors in target tissue (Lefkowitz and Michel, 1985).

To explain the diminished GFR and reduced sodium excretion in the presence of chronic elevation in circulating ANP levels, the possibility of an abnormality at postglomerular level was investigated. Glomerular membranes were chosen because many investigators (Ballermann et al.,

1985; Ogura et al., 1989; Garcia et al., 1989) have previously reported that high affinity binding sites of ANP existed in the rat kidney, especially in the by radiolabeled receptor assay (Ogura et al., 1985) and autoradiography (Yamamoto et al., 1987). Furthermore, since this tissue contains high density of specific ANP receptors and it is easily accessible, the renal glomerular ANP receptor is ideal for studying alterations in receptor density and affinity that may be accompanied by age-related changes in circulating ANP levels.

The specific binding of glomerular membrane to 125I-ANP was linear between 0.05 and 0.2 mg of tissue (Fig. 24), subsequently, a concentration of 0.1 mg of glomerular membrane per assay tube was used throughout the study. In addition, the ANP binding capacity was greatly influenced by incubation time (Fig. 23), as Napier et al. (1984) and Ogura et al. (1989) have previously reported. Since the maximum binding was achieved in 5 minutes and remained stable for 15 minutes.

- 76 - the ANP radiolabelled receptor assay was conducted for 10 minutes at

20°C.

When data from competitive binding inhibition studies (Fig. 25) were

subjected to computerized Scatchard analysis, it was apparent that ANP

interacted with one population of sites of a single affinity in glomeruli

isolated from all four age groups of rats (Fig. 26). The receptor binding

to ANP was saturable (Fig. 25) and with high affinity. Nonspecific

binding was low, and contributed to less than 3% of the total count.

The glomerular receptor density (Bmax) in the four age groups of rats was

calculated to be ranged from 180 to 480 fmol/mg tissue protein (Fig.

27), values that are almost identical with those reported by Ballermann

et al. (1985) (116 to 426 fmol/mg protein) and Gauquelin et al. (1988)

(103 to 500 fmol/mg protein). The B,^ in 1- and 3-month-old rats

were calculated to be 182 and 446 fmol/mg protein, respectively, in

excellent agreement with values reported by Garcia and his colleagues

(1989) of 170 fmol/mg protein for 4-week-old and 503 fmol/mg protein

for 12-week-old Wistar rats. Similarly, equilibrium dissociation constant

(Kj) of the ANP binding sites in glomerular membrane ranged from 0.06

to 0.35 nM (Fig. 27), agree well with values reported by Ballermann et

al. (1985) (0.02 - 0.55 nM), and many other groups (Gauquelin et al.,

1988; Garcia et al., 1988; Ogura et al., 1989).

Glomerular ANP receptor density was greater in the 3-month-old than

in the 1-month-old (Fig. 27), which is probably due to the adaptive

changes of the rat as a result of maturation. In agreement with this

- 77 - observation is the recent study that documented a progressive increase

in the density of the glomerular ANP binding sites with age, from 4- to

16-week-old rats (Garcia et al., 1989). The glomerular receptor affinity,

on the other hand, is lower (higher kj in the 3-month-old than in the

1-month-old (Fig. 27). These observations suggest that even though the

density of ANP receptors was lower in the 1-month-old, it is

compensated by a more effective ANP binding (higher affinity, lower KJ.

Consistent with this finding is the observation that extracts prepared from atria of adult rats (>2 months of age) contained significantly greater natriuretic activity than extracts from pups (19 days of age)

(Pollock and Banks, 1985). This up-regulation of receptor density may be an indication of the still developing and "tuning-up" of many physiological processes in the young rats.

With advancing age (from 3 to 20 months), the receptor density steadily

declined. The oldest group of rats, 20-month-old, contained the least

amount of receptor compared to the other 3 age groups (Fig. 27). This lower density of glomerular ANP receptors in the 20-month-old rats was accompanied by a higher binding affinity (lower kj. In the 10-month- old glomerulus, although the binding affinity was similar to the 3- month-old, the receptor density was significantly lower, thus reflecting regulation of receptors as a result of aging. With further reduction in the receptor density in the 20-month-old (to about 55% of the 3-month- old), the receptor affinity was dramatically increased, which indicates the body's attempt to compensate for the down-regulation in receptor density.

- 78 - Down regulation of ANP receptors have been previous reported in various experimental, physiological and clinical conditions in which plasma ANP levels are elevated (Morton et al., 1987; Takayanagi et al.,

1986; Roubert et al., 1987; Lynch et al. 1986; Ballermann et al. 1985;

Hirata et al. 1985). For instance, Lynch et al. (1986) demonstrated that water deprivation, which depresses plasma ANP levels, augments binding receptor number, but salt loading, which increases plasma ANP level, decreases receptor density in the kidney of the rats. In spontaneous hypertensive rats (SHR), a marked (60%) down-regulation of ANP receptors in aortic smooth muscles and adrenal capsules was observed to correspond to an elevation in plasma ANP level (Takayanagi et al., 1986). In cultured rat aortic smooth muscle cells pretreated with

ANP, a significantly lower binding capacity was observed when subsequently exposed to 125I-ANP (Roubert et al., 1987).

This apparent decrease in efficiency or capacity of physiological processes is a common feature of aging. The changes in the number of receptors with age may be due to modulations in the rate of receptor synthesis and degradation, or alterations in receptor regulation

(Scarpace and Abrass, 1988). Similarly, the down-regulation of the glomerular receptor in the aged rats may due to age-associated alterations in the structure, composition or function of the membrane

(Naeim and Walford, 1985; Heron et al., 1980). On the other hand, the responsiveness of a variety of tissues to stimulation by hormones, neurotransmitters, antigens, and mitogens may decrease with age

(Walford et al., 1981), and this may explains the blunted responses of the aged rats to a chronically elevated circulating ANP. In adrenergic

- 79 - receptors (Scarpace and Abrass, 1988; Forn et al., 1970), corticosteroids and androgen receptors (Roth, 1976; Robinette and Mawhinney, 1977), there are many reports that document a reduction in biological responsiveness concurrent with a decline in receptor binding.

Taken together, these observations support the notion that the number of ANP receptors changes reciprocally in response to changes in plasma

ANP, and may provide an insight into the mechanism whereby binding sites of ANP in the kidney are reduced in old rats. There are several mechanisms that have been postulated to explain this receptor down- regulation phenomenon, but available evidence favours two hypotheses.

On hypothesis postulated that the total number of receptors in the cell is normal, but hormone-mediated internalization has removed a sufficient fraction of receptor such that fewer receptors are on the surface of the cell available for binding (Krupp and Lane, 1981). The other suggested that the total number of receptors in the cell (as well as the number of receptors on the cell surface) is reduced as a result of increased degradation of the internalized receptors (Jacobs and

Cuatrecasas, 1983). Which mechanism is responsible for down- regulation in the glomerular receptor of 10- and 20-month-old rats in the present study cannot be deduced from the data, and the exact mechanism remains to be elucidated.

Recently, two subtypes of ANP receptors have been found: a B- or R,- receptor subtype, in which the receptor is coupled to guanylate cyclase, and is related to physiological effects in certain tissues (Takayanagi et al., 1987; Leitman and Murad, 1986); and a C- or Ra-receptor subtype,

- 80 - which recognizes biological active ANP fragments as well as inactive

truncated forms such as ANP102.121 (so called C-ANP) (Maack et al., 1987;

Leitman and Murad, 1986). Although the latter receptor type represented more than 90% of the total number of ANP binding sites, the receptor does not generate biological responses and seems to serve merely as a "clearance receptor" that helps to regulate ANP concentration in the circulation (Maack et al., 1987). Thus, modifications in the proportion of these receptors in target tissue could lead to profound physiological and pathophysiological effects.

The present data of ANP receptor down-regulation caused by age-related elevation in plasma ANP levels do not allow one to differentiate between the two populations of receptors, since a-ANP binds to both subtypes of ANP receptors. However, Maack et al. (1987) have postulated that

ANP receptor regulation largely represents the regulation of the ANP clearance receptor (C-receptor). This speculation is further supported by results obtained from studies done by Hirata et al. (1987), Kihara et al., (1985) and Ben-Ishay et al. (1973). This being the case, then, a down-regulation of C-receptor density may, in part, explain the experimental data that elderly subjects are more likely to develop exaggerated natriuresis and diuresis compared to younger subjects following a water or saline load (Lindeman et al., 1970; Schalekamp et al., 1971), infusion of a small dose of a-ANP (Heim et al., 1989), or subjected to head-out water immersion (Tajima et al., 1988; Epstein et al., 1987). Then, a reduction in the number of C-receptors would allow the increase in endogenous ANP to interact with B-receptors, thereby producing exaggerated responses.

- 81 - Therefore, If down-regulation of the ANP receptor observed in the present study represents a decrease in the number of clearance receptors, then this may partially explain the age-related accumulation of ANP in the plasma of the 20-month-old rats because less ANP has been removed from the circulation. The fact that chronically elevated

circulating ANP levels in the 20-month-old rats correspond with

diminished glomerular filtration rate and reduced natriuresis in the kidney suggests the existence of age-related alterations in receptors.

Decreases in receptor regulation, either in synthesis or degradation,

changes in structure composition and function of the membrane, or

decreases in responsiveness to stimulation by hormones may play an

important part in the hormone-receptor interaction and in the generation of functional responses.

- 82 - 5.0 SUMMARY

In summary, data obtained from this study suggests that age may affect

the processing, secretion, circulating levels, and receptor binding of ANP

on 4 levels (Fig. 28).

The first level of modification may occur at the site of intracellular

storage. A depletion of ANP concentration in the storage granules

within the aging atria may occur as a result of reduced synthesis. The

major molecular form of ANP in the atrium was y-ANP and remained

unchanged with age.

The second level of alteration may reside in the secretion of ANP. A

relationship was established between the rate of ANP release and the

atrial tissue level, suggesting that reduced ANP secretion is a result of

lower atrial content. Although the molecular forms of ANP in the atrium

were not altered by age, ANP released in the perfusate showed an

increasing amount of y-ANP with the progression of age. This was

taken to indicate an impaired post-translational processing of y-ANP to

active a-ANP, which seems to take place prior to secretion.

A third site of modulation may take place at the circulating levels of

ANP. The plasma ANP levels correlated positively with age. An

increased plasma ANP concentration is not a consequence of

inappropriate secretion, but instead may be caused by an age-related

reduction in the renal clearance of ANP. The elevation in total ANP

concentration may, in part, be due to the rise in y-ANP concentration.

Since y-ANP appears to have a greater half-life in the circulation, this

- 83 - I ANP Content

i ANP Release Secretion t y-ANP/a-ANP Ratio

t Total ANP Level T y-ANP/a-ANP Ratio

ANP Accumulation

Receptor i ANP Degradation

IB IK max Compensation

l Clearance C-ANP B-ANP Receptor Receptor No Change

Fig. 28 Summary of possible age-related alterations in the homeostatic balance of ANP in aged rats

- 84 - may partially contribute to the accumulation of plasma ANP.

A fourth site of age-associated alteration may occur at the level of receptor binding. An elevation of plasma ANP in the aging rats caused a down-regulation of the glomerular receptor density, and the observed increase in binding affinity of the receptors may represents its own compensatory response. If the reduction of ANP receptors represent a down-regulation of the ANP clearance receptor (C-receptor), this may account for the accumulation of ANP in the circulation. Down- regulation of C-receptor density may also explain the observations that intravenous ANP infusion, head-out water immersion, water or saline loading in the elderly produce an exaggerated diuretic and natriuretic responses compared to the young. Most of the available receptors are guanylate cyclase-coupled B-receptors in the elderly. Alternatively, exaggerated responses by the aged kidney may be due to increased affinity of the glomerular receptors.

- 85 - REFERENCES

Adler S, Llndeman RD, and Yienst MJ. (1968) Effect of acute acid loading on urinary acid excretion by the ageing human kidney. Journal of Laboratory Clinical Medicine 72: 278-289.

Akimoto K, Miyata A, Kangawa K. Koga Y, Hayakwa K. and Matsuo H. (1988) Molecular forms of atrial natriuretic peptide in the atrium of patients with cardiovascular disease. Journal of Clinical Endocrinology and Metabolism 67: 93-97.

Anand-Srivastava MB, Franks DJ, Cantin M, and Genest J. (1984) Atrial natriuretic factor inhibits adenylate cyclase activity. Biochemical Biophysical Research Communications 121: 855-862.

Anand-Srivastava MB, Cantin M, and Genest J. (1985) Inhibitory effect of atrial natriuretic factor on adenylate cyclase activity in adrenocortical membranes. Life Sciences 36: 1873- 1879.

Arendt RM, Gerbes AL, Stangl E, Glatthor C, Schimak M, Ritter D, Riedel A, Zahringer J, Kemkes B, and Erdmann E. (1987) Baseline and stimulated ANF plasma levels: is an impaired stimulus-response coupling diagnostically meaningful? KUnishe Wochenschrift 65(Suppl VIII): 122-126.

Baker HJ, Lindsey JR. and Weisbroth SH, Eds (1979) The laboratory rat Academic Press, Appendix 1.

Ballermann BJ, Hoover RL, Karnovsky MJ, and Brenner BM. (1985) Physiologic regulation of atrial natriuretic peptide receptors in rat renal glomeruli. Journal of Clinical Investigation 76: 2049-2056.

Ballermann BJ, Bloch KD, Seidman JG. and Brenner BM. (1986) Atrial natriuretic peptide transcription, secretion, and glomerular receptor activity during mlneralocortlcoid escape in the rat. Journal of Clinical Investigation 78: 840-843.

Berg JA, Hayashi M, Fujli Y, and Katz Al. (1988) Renal metabolism of atrial natriuretic peptide in the rat. American Journal of Physiology 255: F466-F473.

Bianchi C, Gutkowska J, Garcia R, Thibault G, Genest J, and Cantin M. (1985) Radioautographic localization of 125I-atrial natriuretic factor (ANF) in rat tissues. Histochemistry 82: 441-446.

Breuhaus BA, Saneii HH, Brandt MA, and Chimoskey JE. Atriopeptln II lowers cardiac output in conscious sheep. American Journal of Physiology 249: R776-R780.

Briker NS. (1969) On the meaning of the intact nephron hypothesis. American Journal of Medicine 46: 1-11.

Cantiello HF and Ausiello DA. (1986) Atrial natriuretic factor and cGMP inhibit amiloride- sensitive Na+ transport in the cultured renal epithelial cell line. LLC-PT. Biochemical Biophysical Research Communications 134: 852-860.

- 86 - Cantin M, Benchimol S, Castonguay Y, Berlinquet JC, and Huet M. (1975) Ultrastructural cytochemistry of atrial muscle cells. V. Characterization of specific granules in human left atrium. Journal of Ultrastructure Research 52: 179-192.

Cantin M. and Genest J. (1985) The heart and atrial natriuretic factor. Endocrinology Review 6: 107-127.

Cantin M, Garcia R, Thibault G, Kuchel O, Gutkowska J, Larochelle P, Hamet P, Schiffrin EL, and Genest J. (1988) Atrial natriuretic factor in experimental and human hypertension. European Heart Journal 9(suppl H): 21-27.

Carroll RJ, Hammer RE, Chan SJ, Swift HH, Rubenstein AH, and Steiner DF. (1988) A mutant human proinsulin is secreted from islets of Langerhans in increased amounts via an unregulated pathway. Proceedings of the National Academy of Sciences of the USA 85: 8943-8947.

Chinkers M and Garbers DL. (1989) The protein kinase domain of the ANP receptor is required for signaling. Science 245: 1392-1394.

Cody RJ, Atlas SA, Laragh JH. (1987) Physiological and pharmacological studies of atrial natriuretic factor, a human natriuretic and vasoactive peptide. Journal of Clinical Pharmacology 27(Suppl 12): 927-936.

Cohen ML and Berkowitz BA. (1976) Vascular contraction: Effect of age and extracellular calcium. Blood Vessels 13: 139-154.

Cohen RM, and Campus F. (1988) Update on insulinomas or the case of the missing (pro)insulinoma. Care 11: 506-508.

Currie MG, Geller DM, Cole BR (1983) Bioactive cardiac substances: Potent vasorelaxant activity in mammalian atria. Science 221: 71-73.

Currie MG, Sukin D, Geller DM, Cole BR, and Needleman P. (1984) Atriopeptin release from the isolated perfused rabbit heart. Biochemical Biophysical Research Communications 124: 711-717.

Currie MG, Geller DM, Cole BR, Siegel NR. Fok KF, Adams SP, Eubanks SR, Galluppi GR, and Needleman P. (1984) Purification and sequence analysis of bioactive atrial peptides (atriopeptins). Science 223:67-69.

Darmady EM, Offer J, and Woodhouse MA. (1973) The parameters of the ageing kidney. Journal of Pathology 109: 195-207.

Davies DF, and Shock NW. (1950) Age changes in glomerular filtration rate, effective renal plasma flow, and tubular excretory capacity in adult males. Journal of Clinical Investigation 29: 496-507.

Dax EM. (1987) Age-related changes in membrane receptor interactions. Endocrinology and Metabolism Clinics 16: 947-964.

Day RP, and Luetscher JA. (1974) Big renin; a possible prohormone in kidney and plasma of a patient with Wilm's tumour. Journal of Clinical Endocrinology and Metabolism 38: 923- 926.

- 87 - de Bold AJ, Raymond JJ, and Bencosme SA. (1978) Atrial specific granules of the rat heart: Light microscopic staining and histochemlcal reactions. Journal of Histochemistry and Cytochemistry 26: 1094-1101. de Bold AJ, Boresnstein HB, Veress AT, and Sonnenberg H. (1981) A rapid and potent natriuretic response to intravenous injection of atrial myocardial extract in rats. Life Sciences 28: 89-94. de Bold AJ. (1985) Atrial natriuretic factor: A hormone produced by the heart. Science 230: 767-770. de Bold AJ, de Bold ML, and Sarda IR (1986) Functional-morphological studies on in vitro cardionatrin release. Journal of Hypertension 4(Suppl 2): S3-S7.

Delevia A, Christlieb AR Melby JC, Graham CA, Day RP, Luetscher JA, and Zager PG. (1976) Big renin and biosynthetic defect of aldosterone production in diabetes mellitus. New England Journal of Medicine 295: 639-642.

Ding J, Thibault G, Gutkowska J, Garcia R Karabatsos T, Jasmin G, Genest J, and Cantin M. (1987) Cardiac and plasma atrial natriuretic factor in experimental congestive heart failure. Endocrinology 121: 248-257.

Dolan LM, Young CA, Khoury JC, and Dobrozsi DJ. (1989) Atrial natriuretic factor during the perinatal period: Equal depletion in both atria. Paediatric Research 25: 339-341.

Epstein M, and Hollenberg NK. (1976) Age as a determinant of renal sodium conservation in normal man. Journal of Laboratory and Clinical Medicine 87: 411-417.

Epstein M, Loutzenhiser RD, Friedland E, Aceto RM, Camargo MJF, and Atlas SA. (1986) Journal of Hypertension 4(Suppl 2): S93-S99.

Epstein M, Loutzenhiser R Friedland E, Aceto RM Camargo MJF, and Atlas SA. (1987) Relationship of increased plasma atrial natriuretic factor and renal sodium handling during immersion-induced central hypervolemia in normal humans. Journal of Clinical Investigation 79: 738-745.

Espiner EA, Nicholls MG, Yandel TG, Crozier IG, Cuneo RC, McCormick D, and Ikram H. (1986) Studies on the secretion, metabolism and action of atrial natriuretic peptide in man. Journal of Hypertension 4(Suppl 2): S85-S91.

Ezaki H, Matsushita S, Shiraki M, Kuramoto K, and Yamaji I. (1988) Clinical evaluation of the plasma levels of immunoreactive atrial natriuretic peptide in elderly patients with heart diseases. Journal of American Geriatric Society 36: 537-541.

Fliers E, and Swaab DF. (1983) Activation of vasopressingergic and oxytocinergic neurons during aging in the Wistar rat. Peptides 4: 165-171.

Forn J, Schonhofer PS, Skidmore IF, and Krishna G. (1970) Effect of aging on the adenyl cyclase and phosphodiesterase activity of isolated fat cells of rat. Biochimica Et Biophysica Acta 208: 304-309.

- 88 - Fried TA, McCog RN, Osgood RW, Reineck HS, and Stein JH. (1985) The effects of atrial natriuretic factor on glomerular dynamics. Clinical Research 33: 587A.

Fuller F, Porter JG, Arfsten AE, Miller J, Schilling JW, Scarborough RM, Lewicki JA, and Schenk DB. (1988) Atrial natriuretic peptide clearance receptor. Complete sequence and functional expression of cDNA clones. Journal of Biological Chemistry 263(19): 9395-9401.

Garcia R, Thibault G, Gutkowska J, Hamet P, Cantin M, and Genest J. (1985) Effect of chronic infusion of synthetic atrial natriuretic factor (ANP 8-33) in conscious two-kidney one- clip hypertensive rats. Proceedings of the Society For Experimental Biology and Medicine 178: 155-159.

Garcia R, Gauquelin G, Cantin M, and Schiffrin EL. (1988) Renal glomerular atrial natriuretic factor receptors in one-kidney, one clip rats. Hypertension 11: 185-190.

Garcia R, Gauquelin G, Thibault G, Cantin M, and Schiffrin EL. (1989) Glomerular atrial natriuretic factor receptors in spontaneously hypertensive rats. Hypertension 13: 567-574.

Gauer OH, Henry JP, and Seiker HO. (1961) Cardiac receptors and fluid volume control. Progress in Cardiovascular Diseases 4: 1-26.

Gerbes AL, Reiner MD, Arendt M. (1985) Plasma atrial natriuretic factor in patients with cirrhosis. New England Journal of Medicine 313: 1609-1610.

Goddard C, Davidson YS, Moser BB. Davies I. and Faragher EB. (1984) Effect of ageing on cyclic AMP output by renal medullary cells in response to arginine vasopressin in vitro in C57BL/Icrfa mice. Journal of Endocrinology 103: 133-139.

Greenwood FC, and Hunter WM. (1963) The preparation of I131-labelled human growth hormone of high specific radioactivity. Biochemistry Journal 89: 114-123.

Guillemette G, Garcia R, Carrier F, Cantin M, Gutkowska J, Thibault G, and Schiffrin EL. (1988) Glomerular ANP receptor regulation during changes in sodium and water metabolism. Journal of Physiology 254: F51-F55.

Gutkowska J, Horky K, Thibault G, Januszewicz M, Cantin M, and Genest J. (1984) Atrial natriuretic factor is a circulating hormone. Biochemical Biophysical Research Communications 125: 315-323.

Hartter E, Kurz R, Woloxzczuk W, and Petzl DH. (1987) Circadian variation and age dependence of human atrial natriuretic peptide levels in hospitalized patients. Hormone & Metabolic Research 19: 490-492.

Haller BGD, Zust H. Shaw S. Gnadinger MP, Uehlinger DE, and Weidmann P. (1987) Effects of posture and ageing on circulating atrial natriuretic peptide levels in man. Journal of Hypertension 5: 551-556.

Heim JM, Gottmann K, Weil J, Strom TM, and Gerzer R. (1989) Effects of a bolus dose of atrial natriuretic factor in young and elderly volunteers. European Journal of Clinical Investigation 19: 265-271.

- 89 - Heldermari JH, Vestal RE, Rowe JW. (1978) The response of arginlne vasopressin to intravenous ethanol and hypertonic saline in man: the impact of aging. Journal of Gerontology 33: 39-43.

Heron DS, Shinitzky M, Hershkowitz M, and Samuel D. (1980) Lipid fluidity markedly modulates the binding of serotonin to mouse brain membrane. Proceedings of the National Academy of Sciences of the USA 77: 7463-7467.

Hirata Y, Tomita M, Yoshimi H, and Ikeda M. (1984) Specific receptors for atrial natriuretic factor (ANF) in cultured vascular smooth muscle cells of rat aorta. Biochemical Biophysical Research Communications 125: 562-568.

Hirata Y, Takata S, Tomita M, and Takaichi S. (1985) Binding, internalization, and degradation of atrial natriuretic peptide in cultured vascular smooth muscle cells of rat. Biochemical Biophysical Research tommunications 132: 976-984.

Hollister AS, Tanaka I, Imada T, Onrot J, Biaggioni I, Robertson D, and Inagami T. (1986) Sodium loading and posture modulate human atrial natriuretic factor plasma levels. Journal of Hypertension 4(Suppl 2): S106-S111.

Horky K, Gutkowska J, Garcia R, Thibault G, Genest J, and Cantin M. (1985) Effect of different anaesthetics on immunoreactive atrial natriuretic factor concentrations in rat plasma. Biochemical Biophysical Research Communications 129: 651-657.

Hsueh WA, Carlson EJ, and Dzau VJ. (1983) Characterization of inactive renin from human kidney and plasma. Evidence of a renal source of circulating inactive renin. Journal of Clinical Investigation 71: 506-517.

Imada T, Takayanagi R, and Inagami T. (1985) Changes in the content of atrial natriuretic factor with the progression of hypertension in spontaneously hypertensive rats. Biochemical Biophysical Research Communications 133: 759-765.

Imada T, Takayanage R, and Inagami T. (1987) Identification of a peptidase which processes atrial natriuretic factor precursor to its active form with 28 amino acid residues in particulate fractions of rat atrial homogenate. Biochemical Biophysical Research Communications 143: 587-592.

Inscho EW, Wilfinger WW, and Banks RO. (1987) Age-related differences in the natriuretic and hypotensive properties of rat atrial extracts. Endocrinology 121: 1662-1670.

Jacobs S and Cuatrecasas P. (1983) Insulin receptors. Annuals of Receptor Pharmacology & Toxicology 23: 461-479.

Jamieson JD and Palade GE. (1964) Specific granules in atrial muscle. Journal of Cellular Biology 23: 151-172.

Kangawa K, Fujuda A, Kubota I, Hayashi Y, and Matsuo H. (1984a) Purification and complete amino acid sequence of alpha-human atrial natriuretic polypeptide (alpha-hANP). Biochemical Biophysical Research Communications 121: 585-591.

- 90 - Kangawa K, Tawaragi Y, Oikawa S, MIzuno A, Sakuragawa Y, Nakazato H, Fukuda A, Minamlno N, and Matsuo H. (1984b) Identification of rat gamma-atrial natriuretic polypeptide and characterization of the cDNA encoding its precursor. Nature 312: 152-155.

Kangawa K, Fukuda A, and Matsuo H. (1985) Structural identification of beta- and gamma- human atrial natriuretic peptide. Nature 313: 397-400.

Kato J, Kida O, Kita T, Nakamura S, Sasaki A, Kodama K, and Tanaka K. (1988) Free and bound forms of atrial natriuretic peptide (ANP) in rat plasma: Preferential increase of free ANP in spontaneously hypertensive rats (SHR) and stroke-prone SHR (SHRSP). Biochemical Biophysical Research Communications 153: 1084-1089.

Katsube N, Schwartz D, Needleman P. (1986) Atriopeptin turnover: Quantitative relationship between in vitro changes in plasma levels and atrial content. Journal of Pharmacology Experimental Therapeutics 239: 474-479.

Keeler R (1982) Atrial natriuretic factor has a direct, -independent action on kidneys. Canadian Journal of Physiology and Pharmacology 61: 996-1002.

Kirkland J, Lye M. Goddard C. (1984) Plasma arginine vasopressin in dehydrated elderly patients. Clinical Endocrinology 20: 451-456.

Kisch B. (1956) Electron microscopy of the atrium of the heart. Experimental Medicine and Surgery 23: 99-112.

Kleinert HD, Maack T, Atlas SA, Januszewicz A, Sealey JE, and Laragh HH. (1984) Atrial natriuretic factor inhibits angiotensin-norepinephrine-, and -induced vascular contractility. Hypertension 6(Suppl I): 1143-1147.

Kleinman LI. (1982) Developmental renal physiology. Federation Proceedings 25:104-110.

Kleinman LI, and Banks RO. (1983) Segmental nephron sodium and potassium reabsorption in newborn and adult dogs during saline expansion. Proceedings of the Society For Experimental Biology and Medicine. 173: 231-237.

Krupinski J, Coussen F, Bakalyar HA, Tang WJ, Feinstein PG, Orth K, Slaughter C, Reed RR and Gilman AG. (1989) Adenylate cyclase amino acid sequence: possible channel- or transporter-like structure. Science 244: 1558-1564.

Krupp M and Lane MD. (1981) On the mechanism of ligand-induced down-regulation of insulin receptor level in the liver cell. Journal of Biological Chemistry 256: 1689-1694.

Lang RE, Tholken H, Ganten D, Luft FC, Ruskoaho H, and Unger T. (1985) Atrial natriuretic factor - a circulating hormone stimulated by volume loading. Nature 314: 264-266.

Laragh JH. (1985) The endocrine control of blood volume, blood pressure and sodium balance: Atrial hormone and renin system interactions. Journal of Hypertension 4(Suppl 2): S143-S148.

Laragh JH. (1985b) Atrial natriuretic hormone, the renin-aldosterone axis and blood pressure-electrolyte homeostasis. New England Journal of Medicine 313: 1330-1340.

- 91 - Larochelle P. Cusson JR, Gutkowska J, Schiffrin EL, Hamet P. Kuchel O. Genest J, and Cantin M. (1987) Plasma atrial natriuretic factor concentrations in essential and renovascular hypertension. British Medical Journal 294: 1249-1252.

Lefkowitz RJ and Michel T. (1983) Plasma membrane receptors. Journal of Clinical Investigation 72: 1185-1189.

Leitman DC and Murad. (1986) Identification of multiple binding sites of atrial natriuretic factor by affinity cross-linking in cultured endothelial cells. Journal of Biological Chemistry 25: 11650-11655.

Lendenson PW, Langevin H, and Michener M. (1987) Plasma atriopeptin concentrations in hyperthyroidism. Euthyroidism and hypothyroidism: studies in man and rat. Journal of Clinical Endocrinology & Metabolism 65: 1172-1176.

Lewicki JA, Greenberg B, Yamanaka M et al. (1986) Cloning, sequence analysis and processing of the rat and human atrial natriuretic factor precursors. Federal Proceedings 45: 2086-2090.

Lindeman RD, Adler S, Yiengst MH, and Shock NW. (1970) Natriuresis and carbohydrate- induced antinatriuresis after overnight fast and hydration. Nephron 7: 289-300.

Ljungqvist A, and Lagergren C. (1962) Normal intrarenal arterial pattern in the aging kidney: A microangiographical and histological study. Journal of Anatomy 96: 285-300.

Luft FC, Grim CE, Fineberg NS, and Weinberger MH. (1979) Effects of volume expansion and contraction in normotensive white, blacks and subjects of different ages. Circulation 59: 643-649.

Luft FC, and Weinberger MH. (1982) Sodium intake and essential hypertension. Hypertension 4(Supple III): III-14-III-19.

Luft FC, Weinberger MH, Grim CE. (1985) Sodium sensitivity and resistance in normotensive humans. American Journal of Medicine 72: 726-733.

Lynch DR, Braas KM, and Snyder SH. (1986) Atrial natriuretic factor receptors in rat kidney, adrenal gland, and brain: Autoradiographic localization and fluid balance dependent changes. Proceedings of the National Academy of Sciences of the USA 83: 3357-3361.

Maack T, Marion DN and Camargo MJF. (1984) Effects of auriculin (atrial natriuretic factor) on blood pressure, renal function and the renin-aldosterone system in dogs. American Journal of Medicine 77: 1069-1075.

Maack T. (1985) Atrial natriuretic factor Structure and functional properties. Kidney International 27: 607-615.

Maack T, Suzuki M, Almeida FA, Nussenzveig D, Scarborough RM, McEnroe GA, and Lewicki JA. (1987) Physiological role of silent receptors of atrial natriuretic factor. Science 238: 675-678.

Manning PT, Schwartz D, Katsube NC, Holmberg SW, and Needleman P. (1986) Vasopressin-stimulated release of atriopeptin; Endocrine antagonists in fluid homeostasis. Science 229: 395-397.

- 92 - Martin ER, Lewicki JA, Scarborough RM, and Ballermann BJ. (1989) Expression and regulation of ANP receptor subtypes in rat renal glomeruli and papillae. American Journal of Physiology 257: F649-F657.

McCance RA, and Widdowson EM. (1957) Hypertonic expansion of the . Acta Paediatric Scandinavian 46: 337-353.

McKnight JA, Roberts G, Sheridan B, and Atkinson AB. (1989) Relationship between basal and sodium-stimulated plasma atrial natriuretic factor, age, sex and blood pressure in normal man. Journal of Human Hypertension 3: 157-163.

Miller JH, McDonald RK, and SHock NW. (1951) The renal extraction of p-aminohippurate in the aged individual. Journal of Gerontology 6: 213-216.

Miller JH, McDonald RK, and SHock NW. (1952) Age changes in the maximal rate of renal tubular reabsorption of glucose. Journal of Gerontology 7: 196-200.

Misono KS, Fukumi H, Grammer RT, and Inagami T. (1984) Rat atrial natriuretic factor: complete amino acid sequence and disulphide linkage essential for biological activity. Biochemical Biophysical Research Communications 119: 524-529.

Miyata A, Kangawa K, Toshimori T, Hatoh T, and Matsuo H. (1985) Molecular forms of atrial natriuretic peptides in mammalian tissues and plasma. Biochemical Biophysical Research Communications 129: 248-253.

Miyata A, Kangawa K, Toshimori T. (1987) Molecular forms of atrial natriuretic polypeptides circulating in human plasma. Biochemical Biophysical Research Communications 142: 461- 467.

Moore RA. (1943) Total number of glomeruli in the normal kidney. Anatomical Record 48: 153-168.

Morii N, Nakao K, Klhara M, Sugawara A, Sakamoto M, Yamori Y, and Imura H. (1986) Decreased content in left atrium and increased plasma concentration of atrial natriuretic polypeptide in spontaneously hypertensive rats (SHR) and SHR-prone. Biochemical Biophysical Research Communications 135: 74-81.

Morton JJ, Lyall F, and Wallace ECH. (1987) Rat atrial natriuretic peptide vascular receptor: effect of alterations in sodium balance and of DOC hypertension. Journal of Hypertension 5: 474-479.

Munson PJ, and Rodbard D. (1980) LIGAND: a versatile computerized approach for characterization of ligand binding systems. Analytical Biochemistry 107: 220-239.

Murthy KK, Thibault G, Schiffrin EL, Garcia R, Chartier L, Gutkowska J, Genest J, and Cantin M. (1986) Disappearance of atrial natriuretic factor from circulation in the rat. Peptides 7: 241-246.

Naeim F, and Walford RL. (1985) Aging and complexes: The lipid bilayer, integral proteins, and cytoskeleton. In: Handbook of the biology of aging. CE Finch, and El Schneider, Eds, Van Nostrand Reinhold , pp. 272-289.

- 93 - Nakao K. Sugawara A, Morii N, Sakamoto M, Suda M, Soneda J, Ban T, Kihara M, Yamori Y, Shimokura M, Klso Y, and Imura H. (1984) Radioimmunoassay for alpha-human and rat atrial natriuretic polypeptide. Biochemical Biophysical Research Communications 124: 815- 821.

Nakao K, Sugawara A. Shiono S, Saito Y, Morii N, Yamada T, Itoh H, Musoyama M, Arai H, Sakamoto M, and Imura H. (1987) Secretory from of atrial natriuretic polypeptide as cardiac hormone in humans and rats. (Canadian Journal of Physiology Pharmacology 65: 1756-1761.

Napier MA, Vandlen RL, Albers-Schonberg G, Nutt RF, Brady S, Lyle T, Winquist R, Faison EP, Heinel LA, and Blaine EH. (1984) Specific membrane receptors for atrial natriuretic factor in renal and vascular tissues. Proceedings of the National Academy of Sciences of the USA 81: 5946-5950.

Needleman P, Ed. (1988) Biological and molecular aspects of atrial factors. Alan Liss Inc., New York, pp. xiv.

Ogawa K, Smith Al, Hodsman GP, Jackson B, Woodcock EA, and Johnston CI. (1987) Plasma atrial natriuretic peptide: Concentrations and circulating forms in normal man and patients with chronic renal failure. Clinical & Experimental Pharmacology & Physiology 14: 95-102.

Ogura T, Mitsui T, Ogawa N, and Ota Z. (1985) Specific receptor binding of atrial natriuretic peptide to rat renal cortex. Research Communications in Chemical Pathology and Pharmacology 49: 353-360.

Ogura T, Yamamoto I. and Ogawa N. (1989) Developmental change of kidney receptor for atrial natriuretic factor in spontaneously hypertensive rat. Hypertension 13: 449-455.

Ohashi M, Fuji! N, Nawata H, Kato K, Ibayashi H. Kangawa K, and Matsuo H. (1987) High plasma concentrations of human atrial natriuretic polypeptide in aged men. Journal of Clinical Endctcrtnology & Metabolism 64: 81-85.

Pandey KN, Inagami T, and Misono KS. (1987) Three distinct forms of atrial natriuretic factor receptors: kidney tubular epithelim cells and vascular smooth muscle cells contain different types of receptors. Biochemical & Biophysical Research Communications 147(3): 1146-1152.

Papper S. (1973) The effect of age in reducing renal function. Geriatrics 28: 83-87.

Perico N, Delaini F, Lupini C, Benigni A, Galbusera M, Boccardo P, and Remuzzi G. (1989) Blunted excretory response to atrial natriuretic peptide in experimental nephrosis. Kidney International 36: 57-64.

Peters JP. (1952) The problems of cardiac edema. American Journal of Medicine 12: 66-76.

Petersen JS, Bech OM, Steiness E, Kirstein D, Korsgard N, Bandrup U, and Christensen S. (1988) Atrial content and plasma levels of atrial natriuretic peptides in rats with chronic renal failure. Scandinavian Journal of Clinical Laboratory Investigation 48: 431-439.

- 94 - Philips PA, Rolls BJ, Ledingham JG. (1984) Reduced after water deprivation in healthy elderly men. New England Journal of Medicine 311: 753-759.

Pollock DM and Banks RO. (1985) Lower atrial natriuretic activity in the newborn versus adult rat. Mineral Electrolyte Metabolism 11: 41-44.

Porter JG, Scarborough RM, Wang Y, Schenk D, McEnroe GA, Kang LL, and Lewickl JA. (1989) Recombinant expression of a secreted form of the atrial natriuretic peptide clearance receptor. Journal of Biological Chemistry 264(24): 14179-14184.

Quirion R, De Lean A, Gutkowska J, Cantin M and Genest J. (1984) Receptors/acceptors for atrial natriuretic factor (ANF) in brain and related structures. Peptides 5:1167-1174.

Rankin AJ, Ledsome JR, Keeler R, and Wilson N. (1987) Extracted and nonextracted atrial natriuretic peptide in rabbits during tachycardia. American Journal of Physiology 253: R696-R700.

Rascher W, Bald M, Kreis J, Tulassay T, Heinrich U, and Sch:arer K. (1987) Atrial natriuretic peptide in infants and children. Hormone Research 28: 58-63.

Richards AM, Tonolo G, Tillman D, Connell JM, Hepburn D, and Robertson JLS. (1986) Plasma atrial natriuretic peptide in stable and accelerated essential hypertension. Journal of Hypertension 4: 790-791.

Richards AM, Tonolo G, Mclntyre GD. (1987) Radio-immunoassay for plasma alpha human atrial natriuretic peptide: a comparison of direct and pre-extracted methods. Journal of Hypertension 5: 227-236.

Riegger AJG, Kromer EP. and Kochsiek K. (1986) Atrial natriuretic peptide in patients with severe heart failure. Klinishe Wochenschrift 64(Suppl VI): 89-92.

Robbins DC, Tager HS, and Rubenstein AH. (1984) Biologic and clinical importance of pro insulin. New England Journal of Medicine 310: 1165-1171.

Robinette CL, and Mawhinney MG. (1977) binding of dihydrotestosterone in young and senile rats. Federal Proceedings 36: 344-348.

Rondeau E, de Lima J, Caillens H. (1982) High plasma antidiuretic hormone in patients with cardiac failure: Influence of age. Mineral Electrolyte Metabolism 8: 267-274.

Roth GS. (1976) Reduced binding site concentration in cortical neuronal perikarya from senescent rats. Brain Research 107: 345-354. Rondeau E, de Lima J, Caillens H. (1982) High plasma antidiuretic hormone in patients with cardiac failure: Influence of age. Mineral Electrolyte Metabolism 8: 267-274.

Roubert P, Lonchampt MO, Chabrier PE, Plas P, Goulin J, and Braquet P. (1987) Down- regulation of atrial natriuretic factor receptors and correlation with cGMP stimulation in rat cultured vascular smooth muscle cells. Biochemical Biophysical Research Communications 148: 61-67.

- 95 - Rowe JW, Andres R, Tobln JD, Norris AH, and Shock NW. (1976) The effect of age on creatinine clearance in men: A cross sectional and longitudinal study. Journal of Gerontology 31: 155-163.

Rowe JW, Shock NW, and DeFronzo RA. (1976b) The influence of age on the renal response to water deprivation in man. Nephron 17: 270-278. Rowe JW, and Minaker KL. (1988) Geriatric Medicine. In: Handbook of the biology of aging. CE Finch, and El Schneider, Eds, Van Nostrand Reinhold, pp. 932-959.

Ruskoaho H, Toth M, and Lang RE. (1985) Atrial natriuretic peptide secretion: Synergistic effect of phorbol ester and A23187. Biochemical Biophysical Research Communications 133: 581-588.

Ruskoaho H, Toth M, Ganten D, Unger T, and Lang RE. (1986) The phorbol ester induced atrial natriuretic peptide secretion is stimulated by forskolin and Bay K8644 and inhibited by 8-bromo-cGMP. Biochemical Biophysical Research Communications 139: 266-274.

Sagnella GA, Markandu ND, Shore AC, and MacGregor GA. (1986) Raised atrial natriuretic peptides in essential hypertension. Lancet 2: 179-181.

Salto Y, Nakao K. Morii N, Sugawara A. Shiono S, Yamada T, Itoh H, Sakamoto M, Kurahashi K, Fujiwara M, and Imura H. (1986) Bay K 8644, a voltage-sensitive calcium channel agonist, facilitates secretion of atrial natriuretic polypeptide from isolated perfused rat hearts. Biochemical Biophysical Research Comirumicatiorts 138: 1170-1176.

Scarpace PJ and Abrass IB. (1988) Alpha- and Beta-Adrenergic Receptor Function in the Brain During Senescence. Neurobiology of Aging 9: 53-58.

Schalekamp MADM, KraussXH, Schalekamp-Kuyken MPA, Kolsters G, and Birkenhager WH. (1971) Studies on the mechanism of hypernatriuresis in essential hypertension in relation to measurements of plasma renin concentration, body and renal function. Clinical Science 41:219-231.

Schiffrin EL, Chartier L, Thibault G, St.-Louis J, Cantin M, and Genest J. (1985) Vascular and adrenal receptors for atrial natriuretic factor in the rat. Circulation Research 56: 801- 807.

Schiffrin EL, St-Louis J. Garcia R, Thibault G. Cantin M, and Genest J. (1986) Vascular and adrenal binding sites for atrial natriuretic factor: Effects of sodium and hypertension. Hypertension 8(Suppl I): 1141-1145.

Schiffrin EL. (1988) Decreased density of binding sites for atrial natriuretic peptide on platelets of patients with severe congestive heart failure. Clinical Science 74: 213-218.

Shenker Y, Port FK, Swartz RD, Gross MD, and Grekin RJ. (1987) Atrial natriuretic hormone secretion in patients with renal failure. Life Sciences 41: 1635-1644.

Shields P, and Glembotski CC. (1987) Characterization of the molecular forms of ANP released by perfused neonatal rat heart. Biochemical Biophysical Research Communications 146: 547-553.

Smith HW. (1957) Salt and water volume receptors. American Journal of Medicine 23:623- 652.

- 96 - Smith S, Anderson S, Ballermann BJ, and Brenner BM. (1986) Role of atrial natriuretic peptide in adaptation of sodium excretion with reduced renal mass. Journal of Clinical Investigation 77: 1395-1398.

Soejima H, Grekin RJ, Birggs JP, and Schnermann J. (1988) Renal response of anaesthetized rats to low-dose infusion of atrial natriuretic peptide. American Journal of Physiology 255: R449-R455.

Sonnenberg H, and Cupples WA. (1982) Intrarenal localization of the natriuretic effect of cardiac atrial extract. Canadian Journal of Physiology Pharmacology 60: 1149-1152.

Sonnenberg H and Veress AT. (1984) Cellular mechanism of release of atrial natriuretic factor. Biochemical Biophysical Research Communications 124: 443-449.

Sonnenberg H. (1986) Mechanisms of release and renal tubular action of atrial natriuretic factor. Federal Proceedings 45: 2106-2110.

Sonnenberg JL, Sakane Y, Jerg AY. (1988) Identification of protease 3.4.24.11 as the major atrial natriuretic factor degrading enzyme in the rat kidney. Peptides 9: 173-180.

Sosa RE, Volpe M, Marion DN. (1986) Relationship between renal hemodynamic and natriuretic effects of atrial natriuretic factor. American Journal of Physiology 19: F520-F524.

Suda S, Weidmann P, Saxenhofer H, Cottier C, Shaw SG, and Ferrier C. (1988) Atrial natriuretic factor in mild to moderate chronic renal failure. Hypertension 11: 483-90.

Sugawara A, Nakao K, Morii N, Sakamoto M, Suda M, Shimokura M, Kiso Y, Kihara M, Yamori Y, Nishimuro K, Soneda J, Ban T, and Imura H. (1985) Alpha-human atrial natriuretic polypeptide is released from the heart and circulates in the body. Biochemical Biophysical Research Communications 129: 439-446.

Sugawara A, Nakao K, Morii N, Yamada T, Itoh H, Shiono SA, Saito Y, Mukoyama M, Aral H, Nishimura K, Obata K, Yasue H, Ban T, and Imura H. (1988) Augmented synthesis of beta-human atrial natriuretic polypeptide in human failing hearts. Biochemical Biophysical Research Communications 150: 60-67.

Sugiyama MH, Fukume H, Gammer RT, Misono KS, Yabe Y, Morisawa Y, and Inagami T. (1984) Synthesis of atrial natriuretic peptides and studies on structural factors in tissue specificity. Biochemical Biophysical Research Communications 123: 338-344.

Tajama F, Sagawa S, Iwamoto J, Miki K, Claybaugh JR, and Shiraki K. (1988) Renal and endocrine responses in the elderly during head-out water immersion. American Journal of Physiology 23: R977-R983.

Takazakura E, Wasabu N, Handa A. Takada A, Shinoda A, and Takeuchi J. (1972) Intrarenal vascular changes with age and disease. Kidney International 2: 224-230.

Tanaka H, Shindo M, Gutkowska J, Kinoshita A, Urata H, Ikeda M, and Arakawa K. (1986) Effect of acute exercise on plasma immunoreactive-atrial natriuretic factor. Life Sciences 39: 1685-1693.

Tang J, Webber RJ, Chang D, Chang JK, Kiang J, and Wei ET. (1984) Depressor and natriuretic activities of several atrial peptides. Regulatory Peptides 9: 53-59.

- 97 - Tauchi H, Tsuboi K, and Okutomi J. (1971) Age changes In the human kidney of the different races. Gerontologia 17: 87-97.

Theiss G. John A. Morich F, Neuser D. Schroder W, Stasch J-P, and Wohlfeil S. (1987) oc-h- ANP is the only form of circulating ANP in humans. FEBS Letters 218: 159-162.

Thibault G. Garcia R Cantin M. and Genest J. (1985) Atrial natriuretic factor: Characterization and partial purification. Hypertension S(Suppl 1): 75-80.

Thibault G, Garcia R Gutkowska J, Lazure C, Seidah NG, Chretien M. Genest J, and Cantin M. (1986) Identification of the released form of atrial natriuretic factor by the perfused rat heart. Proceedings of the Society For Experimental Biology and Medicine 182: 137-141.

Thibault G. Murthy KK, Gutkowska J, Seidah NG, Lazure C, Chretien M and Cantin M.

(1988) NH2-terminal fragment of rat pro-atrial natriuretic factor in the circulation: identification, radioimmunoassay and half-life. Peptides 9: 47-53.

Trippodo NC, NacPhee AA, Cole FE, and Blakesley HL. (1982) Partial chemical characterization of a natriuretic substance in rat atrial tissue. Proceedings of the Society For Experimental Biology and Medicine 170: 502-508.

Tsunoda K. Hodsman GP, Sumithran E, and Johnston CI. (1986) Atrial natriuretic peptide in chronic heart failure in the rat: A correlation with ventricular dysfunction. Circulation Research 59: 256-261.

Tsunoda K, Mendelsohn FAO, Sexton PM, Chai SY, Hodsman GP and Johnston CI. (1988) Decreased atrial natriuretic peptide binding in renal medulla in rats with chronic heart failure. Circulation Research 62: 155-161.

Volpe M, Sosa RE, Muller FB. (1986) Differing hemodynamic responses to atrial natriuretic factor in two models of hypertension. American Journal of Physiology 19: H871-878.

Waldman SA, Rapoport RM, and Murad F. (1984) Atrial natriuretic factor selectively activates particulate guanylate cyclase and elevates cyclic GMP in rat tissues. Journal of Biological Chemistry 259: 14332-14334.

Walford RL. (1981) A speculative proposal about the immemorial ancestry of the MHC. AACHTion News 5; 15-16.

Wei Y, Rodi CP. Day ML, Wiegand RC, Needleman LD, Cole BR and Needleman P. (1987) Developmental changes in the rat atriopeptin hormonal system. Journal of Clinical Investigation 79: 1325-1329.

Weidmann P, DeMyttenaere-Bursztein S, Maxwell MH, and DeLima J. (1975) Effect of aging on plasma renin and aldosterone in normal man. Kidney International 8: 325-333.

Weidmann P, Hasler L, Gnadinger MP. (1986) Blood levels and renal effects of atrial natriuretic peptide in normal man. Journal of Clinical Investigation 77: 734-742.

Weidmann P, Saxenhofer H, Ferrier C, and Shaw SG. (1988) Atrial natriuretic peptide in man. American Journal of 8: 1-14.

- 98 - Weil J, Bidlingmaier F. Dohlemann C. Kuhnle U, Strom T, and Lang RE. (1986) Comparison of plasma atrial natriuretic peptide levels in healthy children from birth to adolescence and in children with cardiac diseases. Paediatric Research 20: 1328-1331.

Windus DW, Stokes TJ, Morgan JR, and Klahr S. (1989) The effects of atrial peptide in humans with chronic renal failure. American Journal of Kidney Diseases 6: 477-484.

Wong NLM. Huang D. Guo NS, Wong EFC, and Hu DCK. (1989) Effects of thyroid status on atrial natriuretic peptide release from isolated rat atria. American Journal of Physiology 256: E64-E67.

Yamaji T, Ishibashi M, and Takaku F. (1985) Atrial natriuretic factor in human blood. Journal of Clinical Investigation 76: 1705-1709.

Yamaji T, Ishibashi M, Nakaoka H, Imataka I, Amano M, and Fujii J. (1985b) Possible role for atrial natriuretic peptide in polyuria associated with paroxysmal atrial arrhythmias. Lancet 1: 1211.

Yamamoto I, Ogura T, and Ota Z. (1987) In vitro macro- and micro-autoradiographic localization of atrial natriuretic peptide In the rat kidney. Research Communications in Chemical Pathology and Pharmacology 56: 185-198.

Yamasaki Y. Nishiuchi T, Kojima A, Saito H, and Saito S. (1988) Effects of an oral water load and intravenous administration of isotonic glucose, hypertonic saline, mannitol and furosemide on the release of atrial natriuretic peptide in man. Acta Endocrinologica 119: 269-276.

Yandle TG. Richards AM, Nicholls MG. (1986) Metabolic clearance rate and plasma half-life of alpha-human atrial natriuretic peptide in man. Life Sciences 38: 1827-1833.

Yandle TG, Espiner EA, Nicholls MG, and Duff H. (1986b) Radio-immunoassay and characterization of atrial natriuretic peptide in human plasma. Journal of Clinical Endocrinology & Metabolism 63: 72.

- 99 -