Comprehensive Physiology Wiley Online Library

Endocrine Control of Sodium Balance

Full Article on Wiley Online Library



Abstract

The sections in this article are:

1 Components of the Renin–Angiotensin System
2 Prorenin/Renin
2.1 Renin Gene Structure, Evolutionary Function, and Regulation
2.2 Biosynthesis and Chemiosmotic Activation
2.3 Tissue Distribution in Health and Disease
2.4 Kidney
2.5 Heart and Blood Vessels
2.6 Adrenal
2.7 Brain
2.8 Eye, Liver, and Intestine
2.9 Ovary, Uterus, Testis, and Sex Accessory and Subcutaneous Tissue
2.10 Submandibular Gland
2.11 Spontaneously Hypertensive Rat
2.12 Renal Hypertensive Rat
2.13 Growth Retardation
2.14 Relative Renin Plasma Levels and Suggestive Meaning
3 Angiotensinogen
3.1 Biochemical Properties
3.2 Tissue Expression and In Situ Regulation
3.3 Factors Regulating Release
3.4 Mechanism of Action and Physiological Effects
3.5 Summary and Challenges
4 Angiotensin I–Converting Enzyme
4.1 Molecular Structure and Regulation
4.2 Active Sites and Catalytic Properties
4.3 Tissue Distribution
4.4 Summary and Challenges
5 Angiotensin Peptides
6 Aldosterone
6.1 Biosynthesis and Metabolism
6.2 Secretion and Its Regulation
6.3 Angiotensin II
6.4 Potassium
6.5 Corticotropin and Other Proopiomelanocortin Peptides
6.6 Various Stimulators of Aldosterone Secretion
6.7 Various Inhibitors of Aldosterone Secretion
6.8 Cellular Actions
6.9 Disorders of Aldosterone Secretion
7 Systemic Regulation of Sodium Volume Homeostasis
7.1 Regulation of Sodium Volume Homeostasis
7.2 Integrative Regulation of Sodium Volume and Blood Pressure Homeostasis
7.3 Integrative Regulation of Sodium and Potassium Homeostasis
7.4 Integrative Regulation by Potassium and Hydrogen in Volume Homeostasis
8 Polyendocrinopathy Type III: Systemic Dysregulation of Sodium Volume Homeostasis
8.1 Primary and Pseudoprimary Aldosteronism
8.2 Secondary Aldosteronism: Renin Tumors and Edematous States
8.3 High‐Renin States and Low‐Renin Syndromes
9 Summary and Challenges: Defining Functions and Processing Strategies of Renin–Angiotensin System Molecules
Figure 1. Figure 1.

Sensing and response generation in the renin–angiotensin system and the actions of angiotensins. A: Mechanisms mediating renin release and formation of angiotensin I and angiotensin II. Signals are received and integrated by the kidney to cause the release of renin in response to the coordinated messenger regulation and integration by calcium and cAMP. Inside the juxtaglomerular cells, prorenin is processed to renin, most probably by an aspartic protease. Renin and prorenin are trafficked through the cell to the outside. Renin subsequently processes angiotensinogen to angiotensin I, whereas angiotensin‐converting enzyme processes angiotensin I to generate angiotensin II. Angiotensin II achieves its physiological responses by signaling through various subtypes of its receptor. B: Multiplicity of systemic actions of angiotensin II and some metabolites. ECF, extracellular fluid; AT1, AT2, angiotensin II receptor subtypes 1, 2; RBF, renal blood flow; CNS, central nervous system; ADH, antidiuretic hormone. [From Navar et al. 179 with permission.]

Figure 2. Figure 2.

Renin gene organization, activation, and suppression. A: Physiologically relevant functional elements associated with human and mouse renin genes. These include a far upstream enhancer that, together with a Pit‐1‐like sequence in proximal promoter DNA, is required to achieve normal activity of the renin promoter. A classical silencer element also exists in intron 1. Regulatable expression is mediated via an action of cAMP‐response element (CRE)–binding (CREB) protein on the CRE. Part of the cAMP response also involves the Pit‐1‐like site via a CREB protein‐independent mechanism. B: Hypothesis for mechanism by which cAMP and Ca2+ mediate physiological control of the human renin gene. In renin‐synthesizing cells, an increase in cAMP activates protein kinase A (PKA), which in turn phosphorylates CREB protein, causing it to bind to the CRE. This appears to involve a CREB protein/activating transcription factor (ATF)‐1 heterodimer or a CREB protein homodimer. In unstimulated cells, ATF‐1 is phosphorylated and, by binding to the CRE, may suppress the promoter. The fact that the CRE has low binding affinity coupled with the low phosphorylation of CREB protein and its ability to undergo rapid dephosphorylation as well as the capacity for greatly enhanced responsiveness of the promoter under conditions that cause CREB‐1 phosphorylation provides a mechanism for establishing a considerable regulatable response range for transcription of the renin gene under various physiological conditions. Since PKA and Ca2+‐calmodulin (CaM)‐dependent protein kinase can phosphorylate CREB protein and ATF‐1 selectively, the present view includes a possible mechanism for the convergence and integration of multiple extracellular signals that stimulate (via cAMP) or inhibit (via Ca2+–(calmodulin) renin gene expression in renin‐expressing cells in vivo. [From Morris 169 with permission.]

Figure 3. Figure 3.

Processing and granular activity of renin and prorenin. A: Precursor and processing sites in mouse, rat, and human renin. Initial hydrolysis of the N‐terminal signal (pre) peptide occurs prior to completion of synthesis of the nascent protein (site indicated by small arrow nearer left end of polypep tide chain in each case). The question mark at the rat signal peptide cleavage site and the range of values for amino acids in rat pre and pro fragments refer to the uncertainty as to the actual site of hydrolysis. After synthesis of precursor (prorenin), activation in vivo is achieved by hydrolysis at the site indicated by the large vertical arrow. (This differs for rats because rat prorenin has a lysine instead of the arginine at the activation site that is used in the other species). At least a portion of the active renin is later cleaved at the site shown by the small arrow toward the right side of the polypeptide chain, to give twochain renin. The position of this cleavage differs for humans, where the two chains are not held together by disulfide bridges (‐S‐S‐), as is the case for mice and rats. Numbers preceded by + indicate amino acid number with respect to the first amino acid of the mature protein, designated +1. Numbers followed by aa indicate number of amino acids in each polypeptide segment. [From Morris 169 with permission.] B: Chemiosmotic process for renin release from isolated renin‐containing granules. Possible mechanisms of action for some of the drugs used to study renin release from granules. The chemiosmotic process is driven by two major components: the first (component a) is a proton ATPase in the granular membrane which is sensitive to oligomycin and N, N′‐dicyclohexylcarbodiimide (DCCD); the second (component b) is an antiport–symport exchanger which is insensitive to oligomycin and DCCD. Component b exchanges the outflow of proton for the influx of K+ and CI. [From Sigmon and Fray 225 with permission.] C: Effects of changing Ca and K on renin and prorenin secretion. a: Effect of Ca2+ concentration on release of active and inactive (prorenin) renin by rabbit kidney cortex slices. b: Effect of changing K+ concentration in the presence of 2.3 mM Ca2+ (control) and in Ca2+‐depleted media (O Ca2+ + 5 mM EGTA) on release of active and inactive (prorenin) renin by rabbit kidney cortex slices. (pro)Renin, renin plus prorenin. [From King and Fray 132 with permission.]

Figure 4. Figure 4.

Distribution and trafficking pathways and relative values for renin in human plasma. A: Subcellular particulate renin distribution in endoplasmic reticulum (ER) and Golgi transport vesicles a, mature secretory granules (b), and plasma membrane vesicles and light microsomes (c) in three rodent models of low‐renin syndrome. All low‐renin kidneys were removed from rats with low plasma renin activity and renin secretory hyporesponsiveness to acute stimulation. All values are means ± SEM of renin‐specific activities after subfractionation (*P < 0.05; **P < 0.01). [From Fray 79 with permission.] B: Renin content from partially purified subfractions of juxtaglomerular cells from three rodent models of low‐renin syndrome. All low‐renin kidneys were removed from rats with low plasma renin activity and renin secretory hyporesponsiveness to acute stimulation. All values are means ± SEM of renin‐specific activities after subfractionation (*P < 0.05; *P < 0.01). [From Fray 79 with permission.] C: At least three pathways have been identified for renin and prorenin secretion. Pathway 1, conventional regulative degranulation pathway whereby preprocessed and stored renin are liberated from mature secretory granules on demand. Pathway 2, constitutive vesiculation route emanating from post‐Golgi immature vesicles. Pathway 3 begins as pre‐Golgi vesicles bypass Golgi transport to export their contents at the plasma membrane. [From Morris 167 and King and Fray 132 with permission.] D: Relative plasma renin levels in normal subjects under different conditions and impaired patients under pathological conditions. [From Laragh and Sealey 140 with permission.]

Figure 5. Figure 5.

Generation and metabolism of angiotensin peptides and differences in angiotensinogen (renin substrate) concentration and angiotensin I generation. A: Generation and metabolism of angiotensin peptides from the precursor, angiotensinogen. From the generation of angiotensin I by renin processing there are at least five processing enzymes responsible for metabolizing angiotensinogen to “inactive” fragments. [From Ballermann and Onuigbo 9 with permission.] B: Effect of differences in angiotensinogen (renin substrate) concentration on rate of generation of angiotensin. There was no difference in rate when undiluted plasma samples exhibiting various angiotensinogen concentrations were compared with various dilutions of plasma that had endogenously high renin substrate. These samples were obtained at various times during oral contraceptive administration. Normal substrate concentration is ∼1800 ng/ml. Data suggest that concentration of angiotensinogen in human plasma is normally rate‐limiting and that observed differences in rate of angiotensin generation are related to differences in renin substrate concentration and not to variations in concentration of renin activator or inhibitor. [From Newton et al. 181 with permission.]

Figure 6. Figure 6.

Genetic and evolutionary comparisons of elements of angiotensin I–converting enzyme (ACE) from testis and endothelium. A: Relationship between testis ACE and endothelial ACE cDNAs. Open bars, regions of identity; cross‐hatched bar, homologous region; solid and stippled bars, absence of homology. [From Ehlers et al. 67 with permission.] B: Cysteine positions, potential asparagine‐linked glycosylation sites, and positions of putative residues of the active site (one‐letter code). [From Soubrier et al. 235 with permission.] C: Dendrogram showing relationships between core sequences of mammalian N and C domains and Drosophila melanogaster ACE (AnCE). The line below gives estimated time points for the original divergence (450 million years before present), the duplication event (270 million years before present), and the radiation of mammals (50 million years before present) as calculated by the KITSCH algorithm based on these sequences. The table at right summarizes the average relative percent similarities within and between domains and between the N and C domains of the mammalian enzyme and ACE, as calculated using the GCG Distances program. [From Cornell et al. 47 with permission.]

Figure 7. Figure 7.

Summary of kinetic and inhibition constants of angiotensin I–converting enzyme (ACE). A: Kinetic constants for hydrolysis of Hip‐His‐Leu, angiotensin I, and AcSDKP, a specific substrate for the domain active site. B: Inhibition constants using specific ACE inhibitors. C: Structures of specific ACE inhibitors. Enalaprilat, Lisinopril, and trandolaprilat coordinate the active‐site zinc ion of ACE via their carboxyl groups, whereas captopril and fosinoprilat coordinate the active‐site zinc via their sulfydryl and phosphoryl groups, respectively. Captopril does not interact with the theoretical S1 subsite of ACE. [From Williams et al. 272 with permission.]

Figure 8. Figure 8.

Generation and metabolism of angiotensin peptides. A: Classic pathway of angiotensin generation and subsequent cleavage steps. B: Generation of angiotensin III bypassing angiotensin II. C: Generation of angiotensin (1–7) from angiotensin I and subsequent cleavage steps. D: Generation of angiotensin (1–7) from angiotensin II. [From Ballermann and Onuigbo 9 with permission.]

Figure 9. Figure 9.

Biosynthesis and effect of aldosterone on sodium excretion. A: Biosynthetic pathways from cholesterol to mineralocorticoids (aldosterone), glucocorticoids (cortisol), and androgens (androstenedione) and structures of cholesterol, aldosterone, Cortisol, and androstenedione (small amounts of tetosterone and estrogen are also synthesized in adrenal gland). Positions 3, 11, 17, 18, and 21 are marked on the diagram of a steroid molecule (bottom right). Arrows indicate individual biosynthetic conversions. The enzyme specifically mediating each step is indicated at top or left with enzymatic activity in parentheses; note that one protein can mediate more than one step. OH hydroxyl; CMO, corticosterone methyl oxidase. [From White et al. 269 with permission.] B: Relation of renin activity in plasma samples obtained at noon and of corresponding 24 h urinary excretion of aldosterone to concurrent daily rate of sodium excretion. For normal subjects, data describe dynamic hyperbolic relationship between each hormone and sodium excretion. Dynamic fluctuations in renin in response to changes in sodium intake help to maintain blood pressure constant during wide changes in sodium balance. Renin and aldosterone responses work together in kidney to conserve or eliminate sodium in response to changes in dietary sodium intake. Subjects studied on random diets outside the hospital or while on carefully controlled diets in the hospital exhibited similar relationships, validating use of this nomogram in studying outpatients or subjects not receiving constant diets. [From Laragh and Sealey 140 with permission.]

Figure 10. Figure 10.

Structures of steroid hormone receptor superfamily and transcriptional regulation by mineralocorticoid and glucocorticoid receptors. A: Receptors of steroid and other members of the superfamily along with mineralocorticoid receptor (MR) structure scheme. Solid black area represents DNA‐binding domain and hatched area, ligand‐binding region. [From Funder 89 with permission.] B: Transcriptional regulation by mineralocorticoid and glucocorticiod receptors (GR). Glucocorticoid hormones, such as Cortisol and corticosterone (Cort), and mineralocorticoids, such as aldosterone (Aldo), enter the cell and bind to mineralocorticoid‐glucocorticoid receptors; Cort activates both types of receptor, whereas at physiological concentrations Aldo activates only mineralocorticoid receptors. In cells expressing 11β‐hydroxysteroid dehydrogenase (11‐HSD) Cort is selectively metabolized and inactivated. Activated receptors enter the nucleus and bind to DNA sequences termed glucocorticoid‐response elements (GREs) (the mineralocorticoid receptor is shown here with Aldo bound; Cort is equally effective). Mineralocortoid and glucocorticoid receptors can each bind to simple and composite GREs, and both receptors enhance transcription from simple GREs. In contrast, glucocorticoid, but not mineralocorticoid, receptors alter transcription from the composite GRE; the direction of glucocorticoid receptor regulation depends on the subunit composition of the transcription factor activator protein‐1 (AP‐1), which also binds to the composite element. Hence, response elements are either promiscuous or selective in mediating the activities of different bound receptors. In addition, the pattern of regulation by a given response element is shaped by ligand availability, by 11‐HSD expression, and by the functional composition of nonreceptor factors (such as activator protein‐1) that interact with the receptor. PR, progesterone receptor; AR, androgen receptor; ER, estrogen receptor, T3R, triiodothyronine receptor, RAR, retinoic acid receptor; VDR, vitamin D receptor. [From Funder 88 with permission.]

Figure 11. Figure 11.

Urinary sodium excretion is displayed against plasma renin activity and urinary aldosterone. Data from Figure 9 replotted to illustrated mean urinary sodium excretion for given ranges of plasma renin activity and urinary aldosterone excretion. Differences in sodium excretion are significant at all higher rates, suggesting that angiotensin and aldosterone continue to exert regulatory effects on urinary sodium excretion even when circulating concentrations are low. [From Laragh and Sealey 140 with permission.]

Figure 12. Figure 12.

Periodic fluctuations in urine sodium excretion, sodium balance, plasma renin activity, and urine aldosterone excretion after initiation of high‐sodium diet (270 mEq), preceded by 11 days of sodium deprivation (10 mEq/day). Mean values ± SD from three normal subjects. Each subject exhibited similar periodic fluctuations in urine sodium, renin, and aldosterone. After 4 days of diet, natriuresis occurred for 3 days, and these high levels of sodium excretion were associated with the lowest levels of plasma renin activity and aldosterone excretion. [From Sealey et al. 217 with permission.]

Figure 13. Figure 13.

Angiotensin II infusion as a function of salt intake and angiotensin‐converting enzyme inhibition. A: Prolonged continuous angiotensin infusion in normal subject for 11 days. Dose of angiotensin adjusted to maintain mildly pressor response. Angiotensin II induced a marked and selective increase in adrenal cortical aldosterone secretion. Together with a direct effector of angiotensin II on proximal tubule sodium reabsorption, the rise in aldosterone caused sodium retention. As sodium was retained, angiotensin became more pressor. Because of this increasing pressor sensitivity to angiotensin, the dose of angiotensin II had to be serially reduced to maintain the same level of blood pressure. Consequently, aldosterone secretion fell to near control levels. Thus, pressor sensitivity to angiotensin increases as sodium retention produces and then sustains hypertension in progressively smaller amounts and sodium volume gain turns off the need for angiotensin. [From Laragh and Sealey 140 with permission.] B: When angiotensin II is fixed during changes in sodium intake, blood pressure increases with sodium loading and is at its lowest level with sodium depletion. Changes in mean arterial pressure during chronic changes in sodium intake in control dogs and in dogs during angiotensin II blockade with the converting enzyme inhibitor SQ 14,225 or during angiotensin II infusion (5 mg · kg−1 · min−1) also shown. Values are means ± SE. [From Hall et al. 101 with permission.]

Figure 14. Figure 14.

Integrative aspects of the hormonal double‐cyclic feedback and renal synchronization. A: Interrelationship of the renin–aldosterone hormonal system with changes in sodium and potassium balance in the hormonal double‐cycle feedback loop. B: Coordination of intrarenal physical factors with hormonal factors for sodium and potassium homeostasis to highlight the significance of renal synchronization and the role of distal tubular supply. Rate of excretion of each cation is determined by interaction of aldosterone with distal tubular sodium supply. [From Laragh and Sealey 140 with permission.] C: Effect of sodium depletion on urine aldosterone excretion and potassium balance, combined results from five normal subjects. Despite hyperaldosteronism induced by sodium depletion, potassium, balance became progressively positive. Therefore, other factors counterbalance the kaliuretic action of aldosterone. An increase in the aldosterone: renin ratio most likely reflects a gradual increase in potassium balance; this change was accompanied by only modest increases in plasma potassium. [From Laragh and Sealey 140 with permission.]

Figure 15. Figure 15.

Metabolism balance study of patient with pseudoprimary aldosteronism. Dotted lines indicate intake of potassium and sodium. Normal ranges for aldosterone secretion and for midday serum renin in ambient subjects of each dietary sodium intake are indicated by range bars. Aldosterone secretion was always markedly elevated, and serum renin levels were abnormally low. Responses of aldosterone and renin to manipulations in sodium intake were blunted. Potassium repletion was more easily accomplished when dietary sodium was reduced. [From Laragh and Sealey 140 with permission.]

Figure 16. Figure 16.

Biphasic plasma renin activity levels in three animal models of progressive low‐renin syndrome. A: Sequential changes in systemic (aortic) and renal arterial pressure and plasma renin activity (PRA) in experimental benign hypertension. B: Responses in the conscious dog to thoracic inferior vena caval constriction and development of moderate congestive heart failure. MAP, mean arterial pressure. C: Plasma levels of glucose and renin activity in streptozotocin‐diabetic rats as a function of duration. [From Lush et al. 152 with permission.]

Figure 17. Figure 17.

Biphasic plasma renin time line in generalized pathogenesis of renin‐related clinical disorders. The initial phase of pathogenesis is consistent with the definition of high‐renin state (HRS) because renin secretory expression is elevated at baseline and exaggerated when challenged. The later phase has been termed low‐renin syndrome (LRS) because an identical challenge results in impaired secretory expression. Although renovascular hypertension, congestive heart disease, diabetes mellitus, and essential hypertension have demonstrated the biphasic plasma renin profile, polycystic kidney disease, nephrotic syndrome, and chronic stress remain undetermined. [From King and Fray 132 with permission.]

Figure 18. Figure 18.

Classification of post translational processing systems of regulatory peptides into three classes (A, B, C) and diagrammatic representation of three basic designs for processing of precursors to bioactive peptides. A: Some peptides (For example, renin, neurotensin) appear to be fully processed before secretion. B: Other peptides (for example, angiotensin, bradykinin) are formed primarily via extracellular cleavage of secreted precursors. C: Although speculative at this point, a third possibility involves processing after endocytosis by target or other cells (for example, mineralocorticoid receptors). E, enzyme; S, substrate; P, product; N, nucleus; ANP, atrial natriuretic peptide; ET‐1, endothelin‐I; LTC4, leukotriene C4; EGF, epidermal growth factor; TGFα, transforming growth factor‐α. [From Carraway and Loh 31 with permission.]



Figure 1.

Sensing and response generation in the renin–angiotensin system and the actions of angiotensins. A: Mechanisms mediating renin release and formation of angiotensin I and angiotensin II. Signals are received and integrated by the kidney to cause the release of renin in response to the coordinated messenger regulation and integration by calcium and cAMP. Inside the juxtaglomerular cells, prorenin is processed to renin, most probably by an aspartic protease. Renin and prorenin are trafficked through the cell to the outside. Renin subsequently processes angiotensinogen to angiotensin I, whereas angiotensin‐converting enzyme processes angiotensin I to generate angiotensin II. Angiotensin II achieves its physiological responses by signaling through various subtypes of its receptor. B: Multiplicity of systemic actions of angiotensin II and some metabolites. ECF, extracellular fluid; AT1, AT2, angiotensin II receptor subtypes 1, 2; RBF, renal blood flow; CNS, central nervous system; ADH, antidiuretic hormone. [From Navar et al. 179 with permission.]



Figure 2.

Renin gene organization, activation, and suppression. A: Physiologically relevant functional elements associated with human and mouse renin genes. These include a far upstream enhancer that, together with a Pit‐1‐like sequence in proximal promoter DNA, is required to achieve normal activity of the renin promoter. A classical silencer element also exists in intron 1. Regulatable expression is mediated via an action of cAMP‐response element (CRE)–binding (CREB) protein on the CRE. Part of the cAMP response also involves the Pit‐1‐like site via a CREB protein‐independent mechanism. B: Hypothesis for mechanism by which cAMP and Ca2+ mediate physiological control of the human renin gene. In renin‐synthesizing cells, an increase in cAMP activates protein kinase A (PKA), which in turn phosphorylates CREB protein, causing it to bind to the CRE. This appears to involve a CREB protein/activating transcription factor (ATF)‐1 heterodimer or a CREB protein homodimer. In unstimulated cells, ATF‐1 is phosphorylated and, by binding to the CRE, may suppress the promoter. The fact that the CRE has low binding affinity coupled with the low phosphorylation of CREB protein and its ability to undergo rapid dephosphorylation as well as the capacity for greatly enhanced responsiveness of the promoter under conditions that cause CREB‐1 phosphorylation provides a mechanism for establishing a considerable regulatable response range for transcription of the renin gene under various physiological conditions. Since PKA and Ca2+‐calmodulin (CaM)‐dependent protein kinase can phosphorylate CREB protein and ATF‐1 selectively, the present view includes a possible mechanism for the convergence and integration of multiple extracellular signals that stimulate (via cAMP) or inhibit (via Ca2+–(calmodulin) renin gene expression in renin‐expressing cells in vivo. [From Morris 169 with permission.]



Figure 3.

Processing and granular activity of renin and prorenin. A: Precursor and processing sites in mouse, rat, and human renin. Initial hydrolysis of the N‐terminal signal (pre) peptide occurs prior to completion of synthesis of the nascent protein (site indicated by small arrow nearer left end of polypep tide chain in each case). The question mark at the rat signal peptide cleavage site and the range of values for amino acids in rat pre and pro fragments refer to the uncertainty as to the actual site of hydrolysis. After synthesis of precursor (prorenin), activation in vivo is achieved by hydrolysis at the site indicated by the large vertical arrow. (This differs for rats because rat prorenin has a lysine instead of the arginine at the activation site that is used in the other species). At least a portion of the active renin is later cleaved at the site shown by the small arrow toward the right side of the polypeptide chain, to give twochain renin. The position of this cleavage differs for humans, where the two chains are not held together by disulfide bridges (‐S‐S‐), as is the case for mice and rats. Numbers preceded by + indicate amino acid number with respect to the first amino acid of the mature protein, designated +1. Numbers followed by aa indicate number of amino acids in each polypeptide segment. [From Morris 169 with permission.] B: Chemiosmotic process for renin release from isolated renin‐containing granules. Possible mechanisms of action for some of the drugs used to study renin release from granules. The chemiosmotic process is driven by two major components: the first (component a) is a proton ATPase in the granular membrane which is sensitive to oligomycin and N, N′‐dicyclohexylcarbodiimide (DCCD); the second (component b) is an antiport–symport exchanger which is insensitive to oligomycin and DCCD. Component b exchanges the outflow of proton for the influx of K+ and CI. [From Sigmon and Fray 225 with permission.] C: Effects of changing Ca and K on renin and prorenin secretion. a: Effect of Ca2+ concentration on release of active and inactive (prorenin) renin by rabbit kidney cortex slices. b: Effect of changing K+ concentration in the presence of 2.3 mM Ca2+ (control) and in Ca2+‐depleted media (O Ca2+ + 5 mM EGTA) on release of active and inactive (prorenin) renin by rabbit kidney cortex slices. (pro)Renin, renin plus prorenin. [From King and Fray 132 with permission.]



Figure 4.

Distribution and trafficking pathways and relative values for renin in human plasma. A: Subcellular particulate renin distribution in endoplasmic reticulum (ER) and Golgi transport vesicles a, mature secretory granules (b), and plasma membrane vesicles and light microsomes (c) in three rodent models of low‐renin syndrome. All low‐renin kidneys were removed from rats with low plasma renin activity and renin secretory hyporesponsiveness to acute stimulation. All values are means ± SEM of renin‐specific activities after subfractionation (*P < 0.05; **P < 0.01). [From Fray 79 with permission.] B: Renin content from partially purified subfractions of juxtaglomerular cells from three rodent models of low‐renin syndrome. All low‐renin kidneys were removed from rats with low plasma renin activity and renin secretory hyporesponsiveness to acute stimulation. All values are means ± SEM of renin‐specific activities after subfractionation (*P < 0.05; *P < 0.01). [From Fray 79 with permission.] C: At least three pathways have been identified for renin and prorenin secretion. Pathway 1, conventional regulative degranulation pathway whereby preprocessed and stored renin are liberated from mature secretory granules on demand. Pathway 2, constitutive vesiculation route emanating from post‐Golgi immature vesicles. Pathway 3 begins as pre‐Golgi vesicles bypass Golgi transport to export their contents at the plasma membrane. [From Morris 167 and King and Fray 132 with permission.] D: Relative plasma renin levels in normal subjects under different conditions and impaired patients under pathological conditions. [From Laragh and Sealey 140 with permission.]



Figure 5.

Generation and metabolism of angiotensin peptides and differences in angiotensinogen (renin substrate) concentration and angiotensin I generation. A: Generation and metabolism of angiotensin peptides from the precursor, angiotensinogen. From the generation of angiotensin I by renin processing there are at least five processing enzymes responsible for metabolizing angiotensinogen to “inactive” fragments. [From Ballermann and Onuigbo 9 with permission.] B: Effect of differences in angiotensinogen (renin substrate) concentration on rate of generation of angiotensin. There was no difference in rate when undiluted plasma samples exhibiting various angiotensinogen concentrations were compared with various dilutions of plasma that had endogenously high renin substrate. These samples were obtained at various times during oral contraceptive administration. Normal substrate concentration is ∼1800 ng/ml. Data suggest that concentration of angiotensinogen in human plasma is normally rate‐limiting and that observed differences in rate of angiotensin generation are related to differences in renin substrate concentration and not to variations in concentration of renin activator or inhibitor. [From Newton et al. 181 with permission.]



Figure 6.

Genetic and evolutionary comparisons of elements of angiotensin I–converting enzyme (ACE) from testis and endothelium. A: Relationship between testis ACE and endothelial ACE cDNAs. Open bars, regions of identity; cross‐hatched bar, homologous region; solid and stippled bars, absence of homology. [From Ehlers et al. 67 with permission.] B: Cysteine positions, potential asparagine‐linked glycosylation sites, and positions of putative residues of the active site (one‐letter code). [From Soubrier et al. 235 with permission.] C: Dendrogram showing relationships between core sequences of mammalian N and C domains and Drosophila melanogaster ACE (AnCE). The line below gives estimated time points for the original divergence (450 million years before present), the duplication event (270 million years before present), and the radiation of mammals (50 million years before present) as calculated by the KITSCH algorithm based on these sequences. The table at right summarizes the average relative percent similarities within and between domains and between the N and C domains of the mammalian enzyme and ACE, as calculated using the GCG Distances program. [From Cornell et al. 47 with permission.]



Figure 7.

Summary of kinetic and inhibition constants of angiotensin I–converting enzyme (ACE). A: Kinetic constants for hydrolysis of Hip‐His‐Leu, angiotensin I, and AcSDKP, a specific substrate for the domain active site. B: Inhibition constants using specific ACE inhibitors. C: Structures of specific ACE inhibitors. Enalaprilat, Lisinopril, and trandolaprilat coordinate the active‐site zinc ion of ACE via their carboxyl groups, whereas captopril and fosinoprilat coordinate the active‐site zinc via their sulfydryl and phosphoryl groups, respectively. Captopril does not interact with the theoretical S1 subsite of ACE. [From Williams et al. 272 with permission.]



Figure 8.

Generation and metabolism of angiotensin peptides. A: Classic pathway of angiotensin generation and subsequent cleavage steps. B: Generation of angiotensin III bypassing angiotensin II. C: Generation of angiotensin (1–7) from angiotensin I and subsequent cleavage steps. D: Generation of angiotensin (1–7) from angiotensin II. [From Ballermann and Onuigbo 9 with permission.]



Figure 9.

Biosynthesis and effect of aldosterone on sodium excretion. A: Biosynthetic pathways from cholesterol to mineralocorticoids (aldosterone), glucocorticoids (cortisol), and androgens (androstenedione) and structures of cholesterol, aldosterone, Cortisol, and androstenedione (small amounts of tetosterone and estrogen are also synthesized in adrenal gland). Positions 3, 11, 17, 18, and 21 are marked on the diagram of a steroid molecule (bottom right). Arrows indicate individual biosynthetic conversions. The enzyme specifically mediating each step is indicated at top or left with enzymatic activity in parentheses; note that one protein can mediate more than one step. OH hydroxyl; CMO, corticosterone methyl oxidase. [From White et al. 269 with permission.] B: Relation of renin activity in plasma samples obtained at noon and of corresponding 24 h urinary excretion of aldosterone to concurrent daily rate of sodium excretion. For normal subjects, data describe dynamic hyperbolic relationship between each hormone and sodium excretion. Dynamic fluctuations in renin in response to changes in sodium intake help to maintain blood pressure constant during wide changes in sodium balance. Renin and aldosterone responses work together in kidney to conserve or eliminate sodium in response to changes in dietary sodium intake. Subjects studied on random diets outside the hospital or while on carefully controlled diets in the hospital exhibited similar relationships, validating use of this nomogram in studying outpatients or subjects not receiving constant diets. [From Laragh and Sealey 140 with permission.]



Figure 10.

Structures of steroid hormone receptor superfamily and transcriptional regulation by mineralocorticoid and glucocorticoid receptors. A: Receptors of steroid and other members of the superfamily along with mineralocorticoid receptor (MR) structure scheme. Solid black area represents DNA‐binding domain and hatched area, ligand‐binding region. [From Funder 89 with permission.] B: Transcriptional regulation by mineralocorticoid and glucocorticiod receptors (GR). Glucocorticoid hormones, such as Cortisol and corticosterone (Cort), and mineralocorticoids, such as aldosterone (Aldo), enter the cell and bind to mineralocorticoid‐glucocorticoid receptors; Cort activates both types of receptor, whereas at physiological concentrations Aldo activates only mineralocorticoid receptors. In cells expressing 11β‐hydroxysteroid dehydrogenase (11‐HSD) Cort is selectively metabolized and inactivated. Activated receptors enter the nucleus and bind to DNA sequences termed glucocorticoid‐response elements (GREs) (the mineralocorticoid receptor is shown here with Aldo bound; Cort is equally effective). Mineralocortoid and glucocorticoid receptors can each bind to simple and composite GREs, and both receptors enhance transcription from simple GREs. In contrast, glucocorticoid, but not mineralocorticoid, receptors alter transcription from the composite GRE; the direction of glucocorticoid receptor regulation depends on the subunit composition of the transcription factor activator protein‐1 (AP‐1), which also binds to the composite element. Hence, response elements are either promiscuous or selective in mediating the activities of different bound receptors. In addition, the pattern of regulation by a given response element is shaped by ligand availability, by 11‐HSD expression, and by the functional composition of nonreceptor factors (such as activator protein‐1) that interact with the receptor. PR, progesterone receptor; AR, androgen receptor; ER, estrogen receptor, T3R, triiodothyronine receptor, RAR, retinoic acid receptor; VDR, vitamin D receptor. [From Funder 88 with permission.]



Figure 11.

Urinary sodium excretion is displayed against plasma renin activity and urinary aldosterone. Data from Figure 9 replotted to illustrated mean urinary sodium excretion for given ranges of plasma renin activity and urinary aldosterone excretion. Differences in sodium excretion are significant at all higher rates, suggesting that angiotensin and aldosterone continue to exert regulatory effects on urinary sodium excretion even when circulating concentrations are low. [From Laragh and Sealey 140 with permission.]



Figure 12.

Periodic fluctuations in urine sodium excretion, sodium balance, plasma renin activity, and urine aldosterone excretion after initiation of high‐sodium diet (270 mEq), preceded by 11 days of sodium deprivation (10 mEq/day). Mean values ± SD from three normal subjects. Each subject exhibited similar periodic fluctuations in urine sodium, renin, and aldosterone. After 4 days of diet, natriuresis occurred for 3 days, and these high levels of sodium excretion were associated with the lowest levels of plasma renin activity and aldosterone excretion. [From Sealey et al. 217 with permission.]



Figure 13.

Angiotensin II infusion as a function of salt intake and angiotensin‐converting enzyme inhibition. A: Prolonged continuous angiotensin infusion in normal subject for 11 days. Dose of angiotensin adjusted to maintain mildly pressor response. Angiotensin II induced a marked and selective increase in adrenal cortical aldosterone secretion. Together with a direct effector of angiotensin II on proximal tubule sodium reabsorption, the rise in aldosterone caused sodium retention. As sodium was retained, angiotensin became more pressor. Because of this increasing pressor sensitivity to angiotensin, the dose of angiotensin II had to be serially reduced to maintain the same level of blood pressure. Consequently, aldosterone secretion fell to near control levels. Thus, pressor sensitivity to angiotensin increases as sodium retention produces and then sustains hypertension in progressively smaller amounts and sodium volume gain turns off the need for angiotensin. [From Laragh and Sealey 140 with permission.] B: When angiotensin II is fixed during changes in sodium intake, blood pressure increases with sodium loading and is at its lowest level with sodium depletion. Changes in mean arterial pressure during chronic changes in sodium intake in control dogs and in dogs during angiotensin II blockade with the converting enzyme inhibitor SQ 14,225 or during angiotensin II infusion (5 mg · kg−1 · min−1) also shown. Values are means ± SE. [From Hall et al. 101 with permission.]



Figure 14.

Integrative aspects of the hormonal double‐cyclic feedback and renal synchronization. A: Interrelationship of the renin–aldosterone hormonal system with changes in sodium and potassium balance in the hormonal double‐cycle feedback loop. B: Coordination of intrarenal physical factors with hormonal factors for sodium and potassium homeostasis to highlight the significance of renal synchronization and the role of distal tubular supply. Rate of excretion of each cation is determined by interaction of aldosterone with distal tubular sodium supply. [From Laragh and Sealey 140 with permission.] C: Effect of sodium depletion on urine aldosterone excretion and potassium balance, combined results from five normal subjects. Despite hyperaldosteronism induced by sodium depletion, potassium, balance became progressively positive. Therefore, other factors counterbalance the kaliuretic action of aldosterone. An increase in the aldosterone: renin ratio most likely reflects a gradual increase in potassium balance; this change was accompanied by only modest increases in plasma potassium. [From Laragh and Sealey 140 with permission.]



Figure 15.

Metabolism balance study of patient with pseudoprimary aldosteronism. Dotted lines indicate intake of potassium and sodium. Normal ranges for aldosterone secretion and for midday serum renin in ambient subjects of each dietary sodium intake are indicated by range bars. Aldosterone secretion was always markedly elevated, and serum renin levels were abnormally low. Responses of aldosterone and renin to manipulations in sodium intake were blunted. Potassium repletion was more easily accomplished when dietary sodium was reduced. [From Laragh and Sealey 140 with permission.]



Figure 16.

Biphasic plasma renin activity levels in three animal models of progressive low‐renin syndrome. A: Sequential changes in systemic (aortic) and renal arterial pressure and plasma renin activity (PRA) in experimental benign hypertension. B: Responses in the conscious dog to thoracic inferior vena caval constriction and development of moderate congestive heart failure. MAP, mean arterial pressure. C: Plasma levels of glucose and renin activity in streptozotocin‐diabetic rats as a function of duration. [From Lush et al. 152 with permission.]



Figure 17.

Biphasic plasma renin time line in generalized pathogenesis of renin‐related clinical disorders. The initial phase of pathogenesis is consistent with the definition of high‐renin state (HRS) because renin secretory expression is elevated at baseline and exaggerated when challenged. The later phase has been termed low‐renin syndrome (LRS) because an identical challenge results in impaired secretory expression. Although renovascular hypertension, congestive heart disease, diabetes mellitus, and essential hypertension have demonstrated the biphasic plasma renin profile, polycystic kidney disease, nephrotic syndrome, and chronic stress remain undetermined. [From King and Fray 132 with permission.]



Figure 18.

Classification of post translational processing systems of regulatory peptides into three classes (A, B, C) and diagrammatic representation of three basic designs for processing of precursors to bioactive peptides. A: Some peptides (For example, renin, neurotensin) appear to be fully processed before secretion. B: Other peptides (for example, angiotensin, bradykinin) are formed primarily via extracellular cleavage of secreted precursors. C: Although speculative at this point, a third possibility involves processing after endocytosis by target or other cells (for example, mineralocorticoid receptors). E, enzyme; S, substrate; P, product; N, nucleus; ANP, atrial natriuretic peptide; ET‐1, endothelin‐I; LTC4, leukotriene C4; EGF, epidermal growth factor; TGFα, transforming growth factor‐α. [From Carraway and Loh 31 with permission.]

References
 1. Abiko, M., K. E., Rodgers, J. D. Compeau, R. M. Nakamura, and G. S. Dizerega. Alterations of angiotensin II receptor levels in sutured wounds in rat skin. J. Invest. Surg. 9: 447–453, 1996.
 2. Addison, R.. Primary structure of the Neurospora plasma membrane H+‐ATPase deduced from the gene sequence. J. Biol. Chem. 261: 14896–14901, 1986.
 3. Alderman, M. H., S., Madhavan, W. L. Ooi, H. Cohen, J. E. Sealey, and J. H. Laragh. Association of the renin–sodium profile with the risk of myocardial infarction in patients with hypertension. N. Engl. J. Med. 324: 1098–1104, 1991.
 4. Alhenc‐Gelas, F., and P. Corvol, Molecular and physiological aspects of angiotensin I converting enzyme. In: Handbook of Physiology. The Endocrine System. Endocrine Regulation of Water and Electrolyte Balance, edited by J. Fray. Bethesda, MD: Am. Physiol. Soc., 1999, sect. 7, vol. III, p. 81–103.
 5. Ando, T., K., Shimamato, Y. Nakahashi, T. Nishitani, S. Hosoda, H. Ishida, T. Yokoyama, S. Tanaka, and O. Iimura. Plasma antidiuretic hormone levels in patients with normal and low renin essential hypertension, and secondary hypertension. Endocrinol. Jpn. 30: 567–570, 1983.
 6. Arakawa, K.. Serine protease angiotensin II systems. J. Hypertens. Suppl. 14: S3–S7, 1996.
 7. Ardaillou, R.. Active fragments of angiotensin II: enzymatic pathways of synthesis and biological effects. Curr. Opin. Nephrol. Hypertens. 6: 28–34, 1997.
 8. Aubert, J., C., Darimont, I. Safonova, G. Ailhand, and R. Negrel. Regulation by glucocorticoids of angiotensinogen gene expression and secretion in adipose cells. Biochem. J. 328: 701–706, 1997.
 9. Ballermann, B. J., and M. A. Onuigbo, Angiotensins. In: Handbook of Physiology. The Endocrine System. Endocrine Regulation of Water and Electrolyte Balance, edited by J. Fray. Bethesda, MD: Am. Physiol. Soc., 1999, sect. 7, vol. III, p. 104–155.
 10. Barger, A. C.. The Goldblatt Memorial Lecture, Part I. Experimental renovascular hypertension. Hypertension 1: 447–455, 1979.
 11. Barker, D. J., C., Osmond, J. Golding, D. Kuh, and M. E. Wadsworth. Growth in utero, blood pressure in childhood and adult life, and mortality from cardiovascular disease. BMJ 298: 564–567, 1989.
 12. Behar, S., M. Zion H. Reicher‐Reiss et al, Short‐ and long‐term prognosis of patients with a first acute myocardial infarction with concomitant peripheral vascular disease. SPRINT Study Group. Am. J. Med. 96: 15–19, 1994.
 13. Berka, J. L., D., Alcorn, J. P. Coghlan, R. T. Fernley, T. O. Morgan, G. B. Ryan, S. L. Skinner, and D. A. Weaver. Granular juxtaglomerular cells and prorenin synthesis in mice treated with enalapril. J. Hypertens. 8: 229–238, 1990.
 14. Bernstein, K. E.. The renin–angiotensin system: a biological machine. Ann. Med. 24: 113–115, 1992.
 15. Bernstein, K. E., B. M., Martin, A. S. Edwards, and E. A. Bernstein. Mouse angiotensin‐converting‐enzyme is a protein composed of two homologous domains. J. Biol. Chem. 264: 11945–51, 1989.
 16. Bird, I. M., D. S., Millican, and R. R. Magness. Specific pregnancy‐induced angiotensin II type‐1 receptor expression in ovine uterine artery does not involve formation of alternate splice variants or alternate promoter usage. Biol. Reprod. 59: 219–224, 1998.
 17. Bird, I. M., J., Zheng, J. M. Cale, and R. R. Magness. Pregnancy induces an increase in angiotensin II type‐1 receptor expression in uterine but not systemic artery endothelium. Endocrinology 138: 490–498, 1997.
 18. Boller, T., and A. Wiemken. Dynamics of vacuolar compartmentation. Annu. Rev. Plant Physiol. Plant Mol. Biol. 37: 137–164, 1986.
 19. Borensztein, P., S., Germain, S. Fuchs, J. Philippe, P. Corvol, and F. Pinet. Cis‐regulatory elements and trans‐acting factors directing basal and cAMP‐stimulated human renin gene expression in chorionic cells. Circ. Res. 74: 764–773, 1994.
 20. Boulay, G., L., Chretien, D. E. Richard, and G. Guillemette. Short‐term desensitization of the angiotensin II receptor of bovine adrenal glomerulosa cells corresponds to a shift from a high to a low affinity state. Endocrinology 135: 2130–2136, 1994.
 21. Bouley, R., M., Gosselin, H. Plante, G. Servant, J. Perodin, M. Arcand, G. Guillemette, and E. Escher. Characterization of a specific binding site for angotensin II in chicken liver. Can. J. Physiol. Pharmacol. 75: 568–575, 1997.
 22. Bramucci, M., A., Miano, A. Gobbetti, M. Zerani, L. Quassinti, E. Maccari, O. Murri, and D. Amici. Different modulation of steroidogenesis and prostaglandin production in frog ovary in vitro by ACE and ANG II. Am. J. Physiol. 273 (Regulatory Integrative Comp. Physiol. 42): R2089–R2096, 1997.
 23. Brandt, C. R., A. M., Pumfery, B. Micales, C. D. Bindley, G. E. Lyons, S. J. Sramek, and I. H. Willow. Renin mRNA is synthesized locally in rat ocular tissues. Curr. Eye Res. 13: 755–763, 1994.
 24. Brown, L., D., Wall, C. Marchant, and C. Sernia. Tissue‐specific changes in angiotensin II receptors in straptozotocin‐diabetic rats. J. Endocrinol. 154: 355–362, 1997.
 25. Buikema, H., Y. M. Pinto, P. P. Van Geel, G. Rooks, C. D. de Langen, P. A. de Graeff, and W. H. Van Gilst. Differential inhibition of plasma versus tissue ACE by utibapril: biochemical and functional evidence of inhibition of vascular ACE activity. J. Cardiovasc. Pharmacol. 29: 684–691, 1997.
 26. Burt, D. W., N., Nakamura, P. Kelley, and V. J. Dzau. Identification of negative and positive regulatory elements in the human renin gene. J. Biol. Chem. 264: 7357–7362, 1989.
 27. Burrell, J. H., and E. R. Lumbers. Angiotensin receptor subtypes in the uterine artery during ovine pregnancy. Eur. J. Pharmacol. 330: 257–267, 1997.
 28. Campbell, D. J., and J. F. Habener. Cellular localization of angiotensinogen gene expression in brown adipose tissue and mesentery: quantification of messenger ribonucleic acid abundance using hybridization in situ. Endocrinology 121: 1616–1626, 1987.
 29. Campbell, D. J., and J. F. Habener. Hybridization in situ studies of angiotensinogen gene expression in rat adrenal and lung. Endocrinology 124: 218–222, 1989.
 30. Caroff, N., R. Della Bruna, J. Philippe, P. Corvol, and F. Pinet. Regulation of human renin secretion and renin transcription by quantitative PCR in cultured chorionic cells: synergistic effect of cyclic AMP and protein kinase C. Biochem. Biophys. Res. Commun. 193: 1332–1338, 1993.
 31. Carraway, R. E., and Y. P. Loh, Post‐translational processing of regulatory peptides. In: Handbook of Experimental Pharmacology, edited by Berlin: D. R. Brown. Berlin: Springer‐Verlag, 1993, vol. 106, p. 70–104.
 32. Cassis, L. A., K. R., Lynch, and M. J. Peach. Localization of angiotensinogen messenger RNA in rat aorta. Circ. Res. 62: 1259–1262, 1988.
 33. Cassis, L. A., J., Saye, and M. J. Peach. Location and regulation of rat angiotensinogen messenger RNA. Hypertension 11: 591–596, 1988.
 34. Catanzaro, D. F., N., Mesterovic, and B. J. Morris. Studies of the regulation of mouse renin genes by measurement of renin messenger ribonucleic acid. Endocrinology 117: 872–878, 1985.
 35. Catanzaro, D. F., J. J., Mullins, and B. J. Morris. The bio‐synthetic pathway of renin in mouse submandibular gland. J. Biol. Chem. 258: 7364–7368, 1983.
 36. Chai, C. Y., W., Hellmann, C. J. Tseng, F. C. Luft, and D. Ganten. Angiotensinogen mRNA and pressor reactions to angiotensin in brain stem areas of spontaneously hypertensive rats. Clin. Exp. Hypertens. 15: 709–725, 1993.
 37. Chen, M., M. P., Harris, D. Rose, A. Smart, X.‐R. He, M. Kretzler, J. P. Briggs, and J. Schnermann. Renin and renin mRNA in proximal tubules of the rat kidney. J. Clin. Invest. 94: 237–243, 1994.
 38. Chen, M., J., Schnermann, R. L. Malvin, P. D. Killen, and J. P. Briggs. Time course of stimulation of renal renin messenger RNA by furosemide. Hypertension 21: 36–41, 1993.
 39. Chen, M., J., Schnermann, A. M. Smart, F. C. Brosius, P. D. Killen, and J. P. Briggs. Cyclic AMP selectively increases renin mRNA stability in cultured juxtaglomerular granular cells. J. Biol. Chem. 268: 24138–24144, 1993.
 40. Chen, X., W., Li, H. Yoshida, S. Tsuchida, H. Nishimura, F. Takemoto, S. Okubo, A. Fogo, T. Matsusaka, and I. Ichikawa. Targeting deletion of angiotensin type 1β receptor gene in the mouse. Am. J. Physiol. 272 (Renal Physiol. 41): F299–F304, 1997.
 41. Chidgey, M. A., and T. M. Harrison. Renin is sorted to the regulated secretory pathway in transfected PC12 cells by a mechanism which does not require expression of the propeptide. Eur. J. Biochem. 190: 139–144, 1990.
 42. Christlieb, A. R.. Diabetes and hypertensive vascular disease. Am. J. Cardiol. 32: 592–606, 1973.
 43. Cohen, A. J., P., Laurens, and J. C. S. Fray. Suppression of renin secretion by insulin: dependence on extracellular calcium. Am. J. Physiol. 245 (Endocrinol. Metab. 8): E531–E534, 1983.
 44. Cohen‐Haguenauer, O., F. Soubrier, N. Van Cong, S. Serero, C. Tuereau, C. Jegou, M. S. Gross, P. Corvol, and J. Frézal. Regional mapping of the human renin gene to 1q32 by in situ hybridization. Ann. Genet. 32: 16–20, 1989.
 45. Chu, W. N., J. D., Baxter, and T. L. Reudelhuber. A targeting sequence for dense secretory granules resides in the active renin protein moiety of human preprorenin. Mol. Endocrinol. 4: 1905–1913, 1990.
 46. Churchill, P. C.. Second messengers in renin secretion. Am. J. Physiol. 249 (Renal Fluid Electrolyte Physiol. 18): F125–F184, 1985.
 47. Cornell, M. J., T. A., Williams, N. S. Lamango, D. Coates, P. Corvol, F. Soubrier, J. Hoheisel, H. Lebrach, and R. E. Isaac. Cloning and expression of an evolutionarily conserved single‐domain angiotensin converting enzyme from Drosophila melanogaster. J. Biol. Chem. 270: 13613–13619, 1995.
 48. Cox, B. E., R. A., Word, and C. R. Rosenfeld. Angiotensin II receptor characteristics and subtype expression in uterine arteries and myometrium during pregnancy. J. Clin. Endocrinol. Metab. 81: 49–58, 1996.
 49. Danser, A. H., F. H., Derkx, P. J. Admiraal, J. Deinum, P. T. de Jong, and M. A. Schalekamp. Angiotensinogen levels in the eye. Invest. Opthalmol. Vis. Sci. 35: 1008–1018, 1994.
 50. Darby, I. A., M., Congiu, R. T. Fernley, C. Sernia, and J. P. Coghlan. Cellular and ultrastructural location of angiotensinogen in rat and sheep kidney. Kidney Int. 46: 1557–1560, 1994.
 51. Darby, I. A., and C. Sernia. In situ hybridization and immunohistochemistry of renal angiotensinogen in neonatal and adult rat kidneys. Cell Tissue Res. 281: 197–206, 1995.
 52. Del Carmen Caro, M., M. Montiel, and E. Jimenez. Characterization of the functional angiotensin II‐receptor complex isoform in rat liver plasma membrane. Life Sci. 62: 51–57, 1998.
 53. Derkx, F. H. M., M. P. A., Schalekamp, and M. A. D. H. Schalekamp. Two‐step prorenin–renin conversion: isolation of an intermediary form of activated prorenin. J. Biol. Chem. 262: 2472–2477, 1987.
 54. Deschepper, C. F., and L. Q. Hong‐Brown, Hormonal regulation of the angiotensinogen gene in liver and other tissues. In: Cellular and Molecular Biology of the Renin–Angiotensin System, edited by A. K. Raizada, A. I. Phillips, and C. Summers. Boca Raton, FL: CRC, 1993, p. 149–166.
 55. Deschepper, C. F., S. H., Mellon, F. Cumin, J. D. Baxter, and W. F. Ganong. Analysis by immunocytochemisty and in situ hybridization of renin and its mRNA in kidney, testis, adrenal, and pituitary of the rat. Proc. Natl. Acad. Sci. U.S.A. 83: 7552–7556, 1986.
 56. Do, Y.‐S., T., Shinagawa, H. Tam, T. Inagami, and W. A. Hsueh. Characterization of pure human renal renin. J. Biol. Chem. 262: 1037–1043, 1987.
 57. Doolittle, R. F.. Angiotensinogen is related to the antitrypsin–antithrombin–ovalbumin family. Science 222: 417–419, 1983.
 58. Dostal, D. E., K. N., Rothblum, and K. M. Baker. An improved method for absolute quantification of mRNA using multiplex polymerase chain reaction: determination of renin and angiotensinogen mRNA levels in various tissues. Anal. Biochem. 223: 239–250, 1994.
 59. Doughty, S. E., R. K., Ferrier, K. J. Hillan, and D. G. Jackson. The effects of ZENECA ZD8731, an angiotensin II antagonist, on renin expression by juxtaglomerular cells in the rat: comparison of protein and mRNA expression as detected by immunohistochemistry and in situ hybridization. Toxicol. Pathol. 23: 256–261, 1995.
 60. Dubin, D., R. E., Pratt, K. Y. Hui, and V. J. Dzau. Characterization of prorenin activation using a synthetic peptide substrate. J. Hypertens. 9: 483–486, 1991.
 61. Duggan, K. A., and V. Z. Ye. Stimulation of the gastric sodium monitor reduces hepatic angiotensin‐converting enzyme activity. Clin. Exp. Pharmacol. Physiol. 24: 449–450, 1997.
 62. Dzau, V. J., D. W., Burt, and R. E. Pratt. Molecular biology of the renin‐angiotensin system. Am. J. Physiol. 255 (Renal Fluid Electrolyte Physiol. 24): F563–F573, 1988.
 63. Dzau, V. J., K. E., Ellison, T. Brody, J. Ingelfinger, and R. E. Pratt. A comparative study of the distributions of renin and angiotensinogen messenger ribonucleic acids in rat and mouse tissues. Endocrinology 120: 2334–2338, 1987.
 64. Egel‐Mitani, M., H. P., Flygenring, and M. T. Hansen. A novel aspartyl protease allowing KEX2‐independent MF alpha propheromone processing in yeast. Yeast 6: 127–137, 1990.
 65. Eggena, P., J. H., Zhu, S. Sereevinyayut, M. Giordani, K. Clegg, P. C. Andersen, P. Hyun, and J. D. Barrett. Hepatic angiotensin II nuclear receptors and transcription of growth related factors. J. Hypertens. 14: 961–968, 1996.
 66. Ehlers, M., E., Fox, D. Strydom, and J. Riordan. Molecular cloning of human testicular angiotensin‐converting‐enzyme: the testis isozyme, is identical to the C‐terminal half of endothelial angiotensin‐converting‐enzyme. Proc. Natl. Acad. Sci. U.S.A. 86: 7741–7745, 1989.
 67. Ehlers, M. R. W., and J. F. Riordan. Angiotensin‐converting enzyme: new concepts concerning its biological role. Biochemistry 28: 5311–5318, 1989.
 68. Eisenbarth, G. S., and R. A. Jackson, The immunoendocrinopathy syndromes. In: William's Textbook of Endocrinology (8th ed.), edited by J. D. Wilson and D. W. Foster. Philadelphia: Saunders, 1992, p. 1555–1566.
 69. Ekker, M., C., Sola, and F. Rougeon. The activity of the mouse renin promoter in cells that do not normally produce renin is dependent upon the presence of a functional enhancer. FEBS Lett. 255: 241–247, 1989.
 70. Ekker, M., D., Tronik, and F. Rougeon. Extra‐renal transcription of the renin genes in multiple tissues of mice and rats. Proc. Natl. Acad. Sci. U.S.A. 86: 5155–5158, 1989.
 71. El‐Dahr, S. S., R. A., Gomez, M. S. Gray, M. J. Peach, R. M. Carey, and R. L. Chevalier. In situ localization of renin and its mRNA in neonatal ureteral obstruction. Am. J. Physiol. 258 (Renal Fluid Electrolyte Physiol. 27): F854–F862, 1990.
 72. Erman, A., B. Chen‐Gal, D. J. Van Dijk, J. Sulkes, B. Kaplan, G. Boner, and A. Neri. Ovarian angiotensin‐converting enzyme activity in humans: relationship to estradiol, age, uterine pathology. J. Clin. Endocrinol. Metab. 81: 1104–1107, 1996.
 73. Fabian, J. R., L. J., Field, R. A. McGowan, J. J. Mullins, C. D. Sigmund, and K. W. Gross. Allele‐specific expression of the murine Ren‐1 genes. J. Biol. Chem. 264: 17589–17594, 1989.
 74. Fabian, J. R., C. M., Kane, K. J. Abel, and K. W. Gross. Expression of the mouse Ren‐1 gene in the co‐agulating gland: localization and regulation. Biol. Reprod. 48: 1383–1394, 1993.
 75. Forgac, M.. Structure and function of vacuolar class of ATP‐driven proton pumps. Physiol. Rev. 69: 765–796, 1989.
 76. Fray, J. C. S., Hypertension in blacks: physiological, psychosocial, theoretical and therapeutic challenges. In: Pathophysiology of Hypertension in Blacks, edited by J. C. S. Fray and J. D. Douglas. New York: Oxford University Press, 1993, p. 3–22.
 77. Fray, J. C. S., (pro)Renin processing and secretion in black essential hypertension and other low‐renin syndromes. In: Pathophysiology of Hypertension in Blacks, edited by J. C. S. Fray and J. G. Douglas. New York: Oxford University Press, 1993, p. 214–236.
 78. Fray, J. C. S.. Regulation of renin secretion by calcium and chemiosmotic forces: (patho)physiological considerations. Biochim. Biophys. Acta 1097: 243–262, 1991.
 79. Fray, J. C. S.. Subcellular dysregulation of renin and prorenin expression in black essential and diabetic hypertensives. Ethn. Dis. 2: 142–157, 1992.
 80. Fray, J. C. S., and J. G. Douglas (Eds.), Pathophysiology of Hypertension in Blacks. New York: Oxford University Press, 1993.
 81. Fray, J. C. S., and S. M. Russo. Mechanism for low renin in blacks: studies in hypophysectomised rat model. J. Hum. Hypertens. 4: 160–162, 1990.
 82. Frederich, R. C., Jr., B. B. Kahn, M. J. Peach, and J. S. Flier. Tissue‐specific nutritional regulation of angiotensinogen in adipose tissue. Hypertension 19: 339–344, 1992.
 83. Fritz, L. C., A. E., Arfsten, V. J. Dzau, S. A. Atlas, J. D. Baxter, J. C. Fiddes, J. Shine, C. L. Cofer, P. Kushner, and P. A. Ponte. Characterization of human prorenin expressed in mammalian cells from cloned cDNA. Proc. Natl. Acad. Sci. U.S.A. 83: 4114–4118, 1986.
 84. Fukamizu, A., K., Sugimura, E. Takimoto, F. Sugiyama, M.‐S. Seo, S. Takahashi, T. Hatae, N. Kajiwara, K. Yagami, and K. Murakami. Chimeric renin‐angiotensin system demonstrates sustained increase in blood pressure of transgenic mice carrying both human renin and human angiotensinogen genes. J. Biol. Chem. 268: 11617–11621, 1993.
 85. Fukamizu, A., E., Takimoto, K. Sugimura, T. Hatae, M.‐S. Seo, S. Takahashi, F. Sugiyama, N. Kajiwara, K. Yagami, and K. Murakami. Dependence of angiotensin production in transgenic mice carrying either the human renin or human angiotensinogen genes on species specific kinetics of the renin‐angiotensin system. Arzneimittelforschung 43: 222–225, 1991.
 86. Funder, J. W.. Mineralocorticoids, glucocorticoids, receptors and response elements. Science 259: 1132–1133, 1993.
 87. Funder, J. W.. Steroid receptors. Semin. Nephrol. 12: 6–11, 1992.
 88. Gaillard, I., E., Clauser, and P. Corvol. Structure of human angiotensinogen gene. DNA 8: 87–99, 1989.
 89. Gal, S., and N. V. Raikhel. Protein sorting in the endomembrane system of plant cells. Curr. Opin. Cell Biol. 5: 636–640, 1993.
 90. Ganguly, A., Aldosterone. In: Handbook of Physiology. The Endocrine System. Endocrine Regulation of Water and Electrolyte Balance. edited by J. Fray. Bechesda, MD: Am. Physiol. Soc. 1999, p. 156–227.
 91. Ganguly, A., and T. Hampton. Calcium‐dependence of serotonin‐mediated aldosterone secretion and differential effects of calcium antagonists. Life Sci. 36: 1459–1464, 1985.
 92. Garcia‐Sainz, J. A., M., Martinez‐Alfaro, M. T. Romero‐Avila, and C. Gonzalez‐Espinosa. Characterization of the AT1 receptor expressed in guinea pig liver. J. Endocrinol. 154: 133–138, 1997.
 93. Ginesi, L. M., K. A., Munday, and A. R. Noble. Secretion control for active and inactive renin: effects of calcium and potassium on rabbit kidney cortex slices. J. Physiol. (Land.) 344: 453–463, 1983.
 94. Gomez, R. A., K. R., Lynch, B. C. Sturgill, J. P. Elwood, R. L. Chevalier, R. M. Carey, and M. J. Peach. Distribution of renin mRNA and its protein in the developing kidney. Am. J. Physiol. 257 (Renal Fluid Electrolyte Physiol. 26): F850–F858, 1989.
 95. Gotoh, E., K., Murakami, T. D. Bahnson, and W. F. Ganong. Role of brain serotonergic pathways and hypothalamus in regulation of renin secretion. Am. J. Physiol. 253 (Regulatory Integrative Comp. Physiol. 22): R179–R185, 1987.
 96. Grandbastien, M. A., A., Spilmanna and M. Caboche. Tnt1, a mobile retroviral‐like transposable element of tobacco isolated by plant cell genetics. Nature 337: 376, 1989.
 97. Griffin, C. A., G., Giacchetti, M. Schambelan, and L. A. Sechi. Ontogenic expression of renal and hepatic angiotensin II receptor genes in the rat. Nephron 76: 103–110, 1997.
 98. Gyurko, R., D., Wielbo, and M. L Phillips. Antisense inhibition of AT1 receptor mRNA and angiotensinogen mRNA in the brain of spontaneously hypertensive rats reduces hypertension of neurogenic origin. Regul. Pept. 49: 167–174, 1993.
 99. Hall, J. E. A. C. Guyton, M. J. Smith, and T. G. Coleman. Blood pressure and renal function during chronic changes in sodium intake: role of angiotensin. Am. J. Physiol. 239 (Renal Fluid Electrolyte Physiol. 8): F271–F280, 1980.
 100. Hardman, J. A., Y. J., Hort, D. F. Catanzaro, J. T. Tellam, J. D. Baxter, B. J. Morris, and J. Shine. Primary structure of the human renin gene. DNA 3: 457–468, 1984.
 101. Harp, J. B., and M. DiGirolamo. Components of the rennin–angiotensin system in adipose tissue: changes with maturation and adipose mass enlargement. J. Gerontol. A Biol. Sci. Med. Sci. 50: B270–B276, 1995.
 102. Harrison, T. M., M. A. J., Chidgey, W. J. Brammar, and G. J. Adams. The pro‐peptide is not necessary for active renin secretion from transfected mammalian cells. Protein Struct. Funct. Genet. 5: 259–265, 1989.
 103. Hatsuzawa, K., M. Hosaka, T. Nakagawa, M. Nagase, A. Shoda, K. Murakami, and K. Nakayama. Structure and expression of mouse furin a yeast Kex2–related protease: lack of processing of coexpressed prorenin in GH4.C1 cells. J. Biol. Ghent. 265: 22075–22078, 1990.
 104. Heinrikson, R. L., and R. A. Poorman, Biochemistry and molecular biology of recombinant human renin and proprenin. In: Hypertension. Pathophysiology, Diagnosis, and Management. New York: Raven, 1990, p. 1179–1196.
 105. Helmer, O. M.. The renin–angiotensin system and its relation to hypertension. Prog. Cardiovasc. Dis. 8: 117–128, 1965.
 106. Holmer, S., K.‐U., Eckardt, O. Aedtner, M. LeHir, K. Schricker, M. Hamann, K. Götz, G. Riegger, W. Moll, and A. Kurtz. Which factor mediates reno‐renal control of renin gene expression? J. Hypertens. 11: 1011–1019, 1993.
 107. Holmer, S., K.‐U., Eckardt, M. LeHir, K. Schricker, G. Riegger, and A. Kurtz. Influence of dietary NaCl intake on renin gene expression in the kidneys and adrenal glands of rats. Pflugers Arch. 425: 62–67, 1993.
 108. Honeyman, T. W., H. M., Goodman, and J. C. S. Fray. The effects of growth hormone on blood pressure and renin secretion in hypophysectomized rats. Endocrinology 112: 1613–1617, 1983.
 109. Hong‐Brown, L. Q., and C. F. Deschepper. Effects of thyroid hormones on angiotensinogen gene expression in rat liver, brain, and cultured cells. Endocrinology 130: 1231–1237, 1992.
 110. Hsueh, W. A., and J. D. Baxter. Human prorenin. Hypertension 17: 469–479, 1991.
 111. Huang, W., F., Alhenc‐Gelas, and M. J. Osborne‐Pellegrin. Protection of the arterial internal elastic lamina by inhibition of the renin‐angiotensin system in the rat. Circ. Res. 82: 879–890, 1998.
 112. Hunt, R. A., G. M., Ciuffo, J. M. Saavedra, and D. C. Tucker. Sympathetic innervation modulates the expression of angiotensin II receptors in embryonic rat heart grafted in oculo. J. Mol. Cell. Cardiol. 27: 2445–2452, 1995.
 113. Ice, K. S., K. M., Geary, R. A. Gomez, D. W. Johns, M. J. Peach, and R. M. Carey. Cell and molecular studies of renin secretion. Clin. Exp. Hypertens. A10: 1169–1187, 1988.
 114. Ishizuka, Y., S., Iizumi, F. Akiyama, H. Hori, R. A. Poorman, T. J. Lobl, and K. Murakami. Characterization of monoclonal antibodies against human prorenin profragment and identification of active prorenins in plasma. J. Biochem. 106: 430–435, 1989.
 115. Iwai, N., and T. Inagami. Quantitative analysis of renin gene expression in extrarenal tissues by polymerase chain reaction method.;. Hypertens. 10: 717–724, 1992.
 116. Iwai, N., T., Inagami, N. Ohmichi, and M. Kinoshita. Renin is expressed in rat macrophage/monocyte cells. Hypertension 27: 399–403, 1996.
 117. Iwai, N., H., Shimoike, and M. Kinoshita. Cardiac renin–angiotensin system in the hypertrophied heart. Circulation 92: 2690–2696, 1995.
 118. Iwai, N., H., Shimoike, and M. Kinoshita. Genetic analysis of renin gene expression in rat adrenal gland. Hypertension 27: 975–978, 1996.
 119. Jikihara, H., A. M., Poisner, R. Hirsch, and S. Handwerger. Human uterine decidual macrophages express renin. J. Clin. Endocrinol. Metab. 80: 1273–1277, 1995.
 120. Jimenez, E., M., Munoz, J. Pavia, and M. Montiel. Angiotensin II receptor in human placental syncytiotrophoblast plasma membranes. Life Sci. 58: 877–882, 1996.
 121. Jo, H., E. K., Yang, W. J. Lee, K. Y. Park, H. J. Kim, and J. S. Park. Gene expression of central and peripheral rennin–angiotensin system components upon dietary sodium intake in rats. Regul. Pept. 67: 115–121, 1996.
 122. Jones, C. A., C. D., Sigmund, R. A. McGowan, C. Kane‐Haas, and K. W. Gross. Expression of murine renin genes during fetal development. Mol. Endocrinol. 4: 375–383, 1990.
 123. Jonsson, J. R., P. A., Game, R. J. Head, and D. B. Frewin. The expression and localisation of the angiotensin‐converting enzyme mRNA in human adipose tissue. Blood Press. 3: 72–75, 1994.
 124. Juillerat‐Jeanneret, L., P. Dessous L'Eglise Manage, A. C. Eskenasy‐Cottier, and R. C. Janzer. Direct and astrocyte‐mediated effects of ethanol on brain‐derived endothelial cells. Life Sci. 56: 1499–1509, 1995.
 125. Kalenga, M. K., K. Thomas, M. De Gaspara, and R. De Hertogh. Determination of renin, angiotensin converting enzyme and angiotensin II levels in human placenta, chorion and amnion from women with pregnancy induced hypertension. Clin. Endocrinol. (Oxf.) 44: 429–433, 1996.
 126. Karen, P., and B. J. Morris. Stimulation by thyroid hormone of renin mRNA in mouse submandibular gland. Am. J. Physiol. 251 (Endocrinol. Metab. 14): E290–E293, 1986.
 127. Kervinen, J., K., Tormakangas, P. Runeberg‐Roos, K. Guruprasad, T. Blundell, and T. H. Teeri, Structure and possible function of aspartic proteinases in barley and other plants. In: Aspartic Proteinases, edited by K. Takahashi. New York: Plenum, 1995, p. 241–254.
 128. Kettani, S., V., Beldent, M.‐C. Rousselet, P. Ronco, P. Verroust, and J.‐P. Saint‐Andre. Presence of renin, angiotensinogen, angiotensin II in the lamb anterior pituitary gland: immunocytochemical study after cryoultramicrotomy. Histochemistry 95: 561–566, 1991.
 129. Kikkawa, R., E., Kitamura, Y. Fujiwara, M. Haneda, and Y. Shigeta. Biophasic alteration of rennin–angiotensin–aldosterone system in streptozotocin‐diabetic rats. Ren. Physiol. 9: 187–192, 1986.
 130. King, J. A., and J. C. S. Fray. Hydrogen and potassium regulation of (pro)renin processing and secretion. Am. J. Physiol. 267 (Renal Fluid Electrolyte Physiol. 36): F1–F12, 1994.
 131. King, J. A., D. J., Lush, and J. C. S. Fray. Regulation of renin processing and secretion: chemiosmotic control and novel secretory pathway. Am. J. Physiol. 265 (Cell Physiol. 34): C305–320, 1993.
 132. Kingdom, J. C. P., M., Hayes, J. McQueen, A. Howatson, and G. B. M. Lindop. Abnormal renin gene expression in the fetal kidney in intrauterine growth restriction [abstract]. J. Hypertens. 13: 1489, 1995.
 133. Kitami, Y., K., Hiwada, and T. Kokubu. Kidney renin gene expression in spontaneously hypertensive rats. J. Hypertens. 7: 727–731, 1989.
 134. Kon, Y., D., Endoh, A. Fukamizu, K. Murakami, Y. Hashimoto, and M. Sugimura. Comparative study of renin expression in the coagulating glands of C57BL/6 and BALB/c mice. J. Anat. 181: 335–343, 1992.
 135. Lacasse, J., M., Ballak, C. Mercure, J. Gutkowska, C. Chapeau, S. Foote, J. Menard, P. Corvol, M. Cantin, and J. Genest. Immunocytochemical localization of renin in juxtaglomerular cells. J. Histochem. Cytochem. 33: 323–332, 1985.
 136. Lamango, N., and R. E. Isaac. Identification of an ACE‐like peptidyl dipeptidase activity in the housefly, Musca demestica. Biochem. Soc. Trans. 21: 245S, 1993.
 137. Lang, J. A., L. H., Ying, B. J. Morris, and C. D. Sigmund. Transcriptional and post‐transcriptional mechanisms regulate human renin gene expression in Calu‐6 cells. Am. J. Physiol. 271 (Renal Fluid Electrolyte Physiol. 40): F94–F100, 1996.
 138. Laragh, J. H., and Sealey, J. E., Renin‐angiotensin‐aldosterone system and the renal regulation of sodium, potassium, and blood pressure homeostasis. In: Handbook of Physiology. Renal Physiology. edited by E. E. Windhager. Bethesda, MD: Am. Physiol. Soc., 1992, sect. 8, vol. II, p. 1409–1541.
 139. Laragh, J. H., and J. E. Sealey, The renin‐angiotensin‐aldosterone hormonal system and regulation of sodium, potassium, and blood pressure homeostasis. In: Handbook of Physiology. Renal Physiology, edited by J. Orloff and R. W. Berliner. Washington, DC: Am. Physiol. Soc., 1973, sect. 8, p. 831–908.
 140. Lattion, A. L, F., Soubrier, J. Allegrini, C. Hubert, P. Corvol, and F. Alhenc‐Gelas. The testicular transcript of the angiotensin I‐converting enzyme encodes for the ancestral, nonduplicated form of the enzyme. FEBS Lett. 252: 99–104, 1989.
 141. Lee, K. A. W., and N. Masson. Transcriptional regulation by CREB and its relatives. Biochim. Biphys. Acta 1174: 221–233, 1993.
 142. Lenz, T., J. E., Sealey, and D. A. Tewksbury. Regional distribution of the angiotensinogens in human placentae. Placenta 14: 695–699, 1993.
 143. Leung, P. S., X. Q., Yao, H. C. Chan, L. X. Fu, and P. Y. Wong. Differential gene expression of angiotensin II receptor subtypes in the epididymides of mature and immature rats. Life Sci. 62: 461–468, 1998.
 144. Li, X., M., Shams, J. Zhu, A. Khalig, M. Wilkes, M. Whittle, N. Barnes, and A. Ahmed. Cellular localization of AT1 receptor mRNA and protein in normal placenta and its reduced expression in intrauterine growth restriction. Angiotensin II stimulates the release of vasorelaxants. J. Clin. Invest. 101: 442–454, 1998.
 145. Lindop, G. B. M., and A. F. Lever. Anatomy of the renin–angiotensin system in the normal and pathological kidney. Histopathology 76: 61–69, 1986.
 146. Lou, Y.‐K., D. T., Liu, J. A. Whitworth, and B. J. Morris. Renin mRNA concentration in hypothalamus is decreased by enalapril. Clin. Exp. Pharmacol. Physiol. 22: 493–495, 1995.
 147. Lou, Y.‐K., D. T., Liu, J. A. Whitworth, and B. J. Morris. Renin mRNA, quantified by polymerase chain reaction, in renal hypertensive rat tissues. Hypertension 26: 656–664, 1995.
 148. Lou, Y.‐K., B. G., Robinson, and B. J. Morris. Renin messenger RNA, detected by polymerase chain reaction, can be switched on in rat atrium. J. Hypertens. 11: 237–243, 1993.
 149. Lou, Y.‐K., D. L., Smith, B. G. Robinson, and B. J. Morris. Renin gene expression in various tissues determined by single‐step polymerase chain reaction. Clin. Exp. Pharmacol. Physiol. 18: 357–362, 1991.
 150. Lush, D. J., J. A., King, and J. C. S. Fray. Pathophysiology of low renin syndromes: sites of renal renin secretory impairment and prorenin overexpression. Kidney Int. 43: 983–999, 1993.
 151. Makrides, S. C., R., Mulinari, V. I. Zannis, and H. Gavras. Regulation of renin gene expression in hypertensive rats. Hypertension 12: 405–410, 1988.
 152. Manninen, I., and A. H. Schulman. BARE‐1 a copia‐like retro element in barley (Hordeum vulgare L.). Plant Mol. Biol. 22: 829–846, 1993.
 153. Matsuba, H., T., Watanabe, M. Watanabe, Y. Ishii, S. Waguri, E. Kominami, and Y. Uchiyama. Immunocytochemical locali zation of prorenin, renin, and cathepsins B, H, and L in jux taglomerular cells of rat kidney. J. Histochem. Cytochem. 37: 1689–1697, 1989.
 154. Mellman, I., R., Fuchs, and A. Helenius. Acidification of the endocytic and exocytic pathways. Annu. Rev. Biochem. 55: 663–700, 1986.
 155. Miller, M., M. Jaskolski, J. K. M. Rao, J. Leis, and A. Wlodawer. Crystal structure of a retroviral protease proves relationship to aspartic protease family. Nature 337: 576–579, 1989.
 156. Moeller, I., S. Y., Chai, D. P. MacGregor, G. P. Aldred, and F. A. Mendelsohn. Localization and quantitation of angioten sin AT1 and A2 receptors in the pregnant and non‐pregnant sheep uterus. Regul. Pep. 61: 213–218, 1996.
 157. Moffett, R. B., R. A., McGowan, and K. W. Gross. Modulation of kidney renin messenger RNA levels during experimentally induced hypertension. Hypertension 8: 874–882, 1986.
 158. Molinari, A. M., C., Abbondanza, I. Armetta, N. Medici, C. Minucci, B. Moncharmont, V. Nigro, and G. A. Puca. Proteolytic activity of the purified hormone‐binding subunit in the estrogen receptor. Proc. Natl. Acad. Sci. U.S.A. 88: 4463–4467, 1991.
 159. Montgomery, H. E., R., Marshall, H. Hemingway, et al, Human gene for physical performance. Nature 393: 221–222, 1998.
 160. Montiel, M., M. C., Caro, and E. Jimenez. Agonist‐induced internalisation of the angiotensin II‐binding sites from plasma membranes of isolated rat hepatocytes. J. Endocrinol. 152: 407–412, 1997.
 161. Morgan, T., C., Craven, L. Nelson, J. M. Lalouel, and K. Ward. Angiotensinogen T235 expression is elevated in decidual spiral arteries. J. Clin. Invest. 100: 1406–1415, 1997.
 162. Moriguchi, A., K. B., Brosnihan, H. Kumagai, D. Ganten, and C. M. Ferrario. Mechanisms of hypertension in transgenic rats expressing the mouse Ren‐2 gene. Am. J. Physiol. 266 (Regulatory Integrative Comp. Physiol. 35): R1273–R1279, 1994.
 163. Morishita, R., J., Higaki, H. Okunishi, T. Tanaka, K. Ishii, M. Nagano, H. Mikami, T. Ogihara, K. Murakami, and M. Miyazaki. Changes in gene expression of the renin‐angiotensin system in two‐kidney, one clip hypertensive rats. J. Hypertens. 9: 187–192, 1991.
 164. Morrell, N. W., K. G., Morris, and K. R. Stenmark. Role of angiotensin‐converting enzyme and angiotensin II in development of hypoxic pulmonary hypertension. Am. J. Physiol. 269 (Heart Circ. Physiol. 38): H1186–H1194, 1995.
 165. Morris, B. J.. New possibilities for intracellular renin and inactive renin now that the structure of the human renin gene has been elucidated. Clin. Sci. (Colch.) 71: 345–355, 1986.
 166. Morris, B. J., and D. F. Catanzaro. Biosynthesis of preprorenin and intracellular conversion of prorenin to renin. Clin. Exp. Pharmacol. Physiol. 8: 441–445, 1981.
 167. Morris, B. J. Renin, In: Handbook of Physiology. The Endocrine System. Endocrine Regulation of Water and Electrolyte Balance. Edited by J. Fray. Bethesda, MD: Am. Physiol. Soc., 1999, Sec. 7, vol. III, P. 3–58.
 168. Morris, B. J., and Y.‐k. Lou, Cardiac renin, gene expression, and regulation. In: The Cardiac Renin‐Angiotensin System, edited by K. Lindpaintner and D. Ganten. Armonk, NY: Futura, p. 21–45, 1994.
 169. Morris, B. J., D. L., Smith, R. E. Law, Y. S. Do, K. J. Shaw, and W. A. Hsueh. Function of human renin proximal promoter DNA. Kidney Int. 46: 1516–1521, 1994.
 170. Morrissey, J. J., and S. Klahr. Differential effects of ACE and AT1 receptor inhibition on chemoattractant and adhesion molecule synthesis. Am. J. Physiol. 274 (Renal Physiol. 43): F580–F586, 1998.
 171. Mullins, J. J., D. W., Burt, J. D. Windass, P. McTurk, H. George, and W. J. Brammer. Molecular cloning of two distinct renin genes from the DBA/2 mouse. EMBO J. 1: 1461–1466, 1982.
 172. Murata, M., M., Nakagawa, and S. Takahashi. Angiotensinogen mRNA is synthesized locally in rat ocular tissues. Opthalmologia 211: 301–304, 1997.
 173. Nagahama, M., K., Nakayama, H. Hori, and K. Murakami. Expression of a deletion mutant of the prosegment of human prorenin in Chinese hamster ovary cells. FEBS Lett. 259: 202–204, 1989.
 174. Nagahama, M., K., Nakayama, and K. Murakami. Effects of propeptide deletion on human renin secretion from mouse pituitary AtT‐20 cells. FEBS Lett. 264: 67–70, 1990.
 175. Nagahama, M., K., Nakayama, and K. Murakami. Sequence requirements for prohormone processing in mouse pituitary AtT‐20 cells. Eur. J. Biochem. 197: 135–140, 1991.
 176. Nakamura, A., and E. J. Johns. Influence of the renal sympathetic nerves on renal renin and angiotensinogen gene expression in spontaneously hypertensive rats during development. J. Hypertens. 13: 301–309, 1995.
 177. Navar, L. G., L., Zou, A. V. Thun, C. T. Wang, J. D. Imig, and K. D. Mitchell. Unraveling the mystery of Goldblatt hypertension. News Physiol. Sci. 13: 170–176, 1998.
 178. Neves, F. A. R., K. G., Duncan, and J. D. Baxter. Cathepsin B is a prorenin processing enzyme. Hypertension 27 (pt. 2): 514–517, 1996.
 179. Newton, M. A., J. E., Sealey, J. G. Ledingham, and J. H. Laragh. High blood pressure and oral contraceptives: changes in plasma renin and renin substrates and in aldosterone excretion. Am. J. Obstet. Gynecol. 101: 1037–1045, 1968.
 180. Nichols, M., F., Weih, W. Schmid, C. DeVack, E. Kowenz‐Leutz, B. Luckow, M. Boshart, and G. Schütz. Phosphorylation of CREB affects its binding to high and low affinity sites: implications for cAMP‐induced gene transcription. EMBO J. 11: 3337–3346, 1992.
 181. Nickenig, G., G., Geisen, H. Vetter, and A. Sachinidis. Characterization of angiotensin receptors on human skin fibroblasts. J. Mol. Med. 75: 217–222, 1997.
 182. Nielsen, A. H., H., Winther, V. Dantzer, and K. Poulsen. High densities of angiotensin II subtype 2 (AT2) receptors in the porcine placenta and fetal membranes. Placenta 17: 147–153, 1996.
 183. Ohkubo, H., H., Kawakami, Y. Kakehi, T. Takumi, H. Arai, Y. Yokota, M. Iwai, Y. Tanabe, M. Masu, J. Hata, H. Iwao, H. Okamoto, M. Yokoyama, T. Nomura, M. Katsuki, and S. Nakanishi. Generation of transgenic mice with elevated blood pressure by introduction of the rat renin and angiotensinogen genes. Proc. Natl. Acad. Sci. U.S.A. 87: 5153–5157, 1990.
 184. Panthier, J.‐J., S., Foote, B. Chambraud, A. D. Strosberg, P. Corvol, and F. Rougeon. Complete amino acid sequence and maturation of the mouse submaxillary gland renin precursor. Nature 298: 90–92, 1982.
 185. Park, C. S., and J. C. S. Fray. Driving force for renin secretion [abstract]. Federation Proc. 45: 1032, 1986.
 186. Park, C. S., D. S., Han, and J. C. S. Fray. Calcium in the control of renin secretion: Ca2+ infulx as an inhibitory signal. Am. J. Physiol. 240 (Renal Fluid Electrolyte Physiol. 9): F70–F74, 1981.
 187. Park, C. S., T. W., Honeyman, E. S. Chung, J. S. Lee, D. H. Sigmon, and J. C. S. Fray. Involvement of calmodulin in mediating inhibitory action of intracellular Ca2+ on renin secretion. Am. J. Physiol. 251 (Renal Fluid Electrolyte Physiol. 20): F1055–F1062, 1986.
 188. Park, C. S., C. D., Hong, and T. W. Honeyman. Calcium‐dependent inhibitory step in control of renin secretion. Am. J. Physiol. 262 (Renal Fluid Electrolyte Physiol. 31): F793–F798, 1992.
 189. Park, C. S., D. H., Sigmon, D. S. Han, T. W. Honeyman, and J. C. S. Fray. Control of renin secretion by Ca++ and cyclic AMP through two parallel mechanisms. Am. J. Physiol. 251 (Regulatory Integrative Comp. Physiol. 20): R531–R536, 1986.
 190. Paulsen, E. P., R. L., Seip, C. R. Ayers, B. Y. Croft, and D. L. Kaiser. Plasma renin activity and albumin excretion in teenage type 1 diabetic subjects. Hypertension 13: 781–788, 1989.
 191. Petit, A., P., Geoffroy, and S. Belisle. Expression of angiotensin II type‐1 receptor and phospholipase C‐linked G alpha4 11 protein in human plcenta. J. Soc. Gynecol. Invest. 3: 316–321, 1996.
 192. Piccini, N., J. L., Knopf, and K. W. Gross. A DNA polymorphism, consistent with gene duplication, correlates with high renin levels in the mouse submaxillary gland. Cell 30: 205–213, 1982.
 193. Pratt, R. E., J. E., Carleton, J. P. Richie, C. Heusser, and V. J. Dzau. Human renin biosynthesis and secretion in normal and ischaemic kidneys. Proc. Natl. Acad. Sci. U.S.A. 84: 7837–7840, 1987.
 194. Puca, G. A., C., Abbondaza, V. Nigro, I. Aarmetta, N. Medici, and A. M. Molonari. Estradiol receptor has proteolytic activity that is responsible for its own transformation. Proc. Natl. Acad. Sci. U.S.A. 83: 5367–5371, 1986.
 195. Qin, H., Y.‐H., Chen, M.‐Y. Yip, P. R. L. Lam‐Po‐Tang, and B. J. Morris. Reassignment of human renin gene to chromosome 1q32 in studies of a (1;4)(q42;p16) translocation. Hum. Hered. 43: 261–264, 1993.
 196. Ramirez, M., E. A., Davidson, L. Luttenauer, P. P. Elena, F. Cumin, G. A. Mathis, and M. De Gasparo. The renin‐angiotensin system in the rabbit eye. J. Ocul. Pharmacol. Ther. 12: 299–312, 1996.
 197. Rao, R. H.. Pressor doses of angiotensin II increase hepatic glucose output and decrease insulin sensitivity in rats. J. Endocrinol. 148: 311–318, 1996.
 198. Reisenleiter, F., N., Katz, and A. Gardemann. Control of hepatic carbohydrate metabolism and haemodynamics in perfused rat liver by arterial and portal angiotensin II. Eur. J. Gastroenterol. Hepatol. 8: 279–286, 1996.
 199. Rocco, S., C., Ambroz, and G. Aguilera. Interaction between serotonin and other regulators of aldosterone secretion in rat adrenal glomerulosa cells. Endocrinology 127: 3103–3110, 1990.
 200. Roland, B. L., K. X. Z., Li, and J. W. Funder. Hybridization histochemical localization of 11β hydroxysteroid dehydrogenase type 2 in rat brain. Endocrinology 136: 4697–4699, 1995.
 201. Runenberg‐Roos, P., J., Kervinen, V. Kovalevva, S. Gal, and N. V. Raikhel. The aspartic proteinase of barley is a vacuolar enzyme that processes probarley lectin in vitro. Plant Physiol. 105: 321–329, 1994.
 202. Saint‐Andri, J. P., V., Rohmer, F. Alhenc‐Gelas, J. Menard, J. C. Bigorgne, and P. Corvoe. Presence of renin, angiotensinogen and converting enzyme in human pituitary lactotroph cells and prolactin adenomas. J. Clin. Endocrinol. Metab. 63: 231–237, 1986.
 203. Samani, N. J., W. J., Brammar, and J. D. Swales. Renal and extra‐renal levels of renin mRNA in experimental hypertension. Clin. Sci. (Colch.) 80: 339–344, 1991.
 204. Samani, N. J., F., Cumin, M. Kelly, and J. M. Wood. Expression of components of the RAS during prolonged blockade at different levels in primates. Am. J. Physiol. 267 (Endocrinol. Metab. 30): E612–E619, 1994.
 205. Samani, N. J., N. P., Godfrey, J. S. Major, W. J. Brammar, and J. D. Swales. Kidney renin mRNA levels in the early and chronic phases of two‐kidney, one clip hypertension in the rat. J. Hypertens. 7: 105–112, 1989.
 206. Samani, N. J., J. D., Swales, and W. J. Brammar. Expression of the renin gene in extra‐renal tissues of the rat. Biochem. J. 253: 907–910, 1988.
 207. Saye, J. A., N. V., Ragsdale, R. M. Carey, and M. J. Peach. Localization of angiotensin peptide‐forming enzymes of 3T3–F442A adipocytes. Am. J. Physiol. 264: (Cell Physiol. 33): C1570–C1576, 1993.
 208. Schauser, K. H., A. H., Nielsen, H. Windier, V. Dantzer, and K. Poulsen. Autoradiographic localization and characterization of angiotensin II receptors in the bovine placenta and fetal membranes. Biol. Reprod. 59: 684–692, 1998.
 209. Scheidegger, K. J., S., Butler, and J. L. Witztum. Angiotensin II increases macrophage‐mediated modification of low density lipoprotein via a lipoxygenase‐dependent pathway. J. Biol. Chem. 272: 21609–21615, 1997.
 210. Schmid, C., H., Castrop, J. Reitbauer, R. Della Bruna, and A. Kurtz. Dietary salt intake modulates angiotensin II type 1 receptor gene expression. Hypertension 29: 923–929, 1997.
 211. Schricker, K., R. Della Bruna, M. Hamann, and A. Kurtz. Endothelium derived relaxing factor is involved in the pressure control of renin gene expression in the kidney. Pflugers Arch. 428: 261–268, 1994.
 212. Schriker, K., S., Holmer, M. Hamann, G. Riegger, and A. Kurtz. Interrelation between renin mRNA levels, renin secretion, and blood pressure in two‐kidney, one clip rats. Hypertension 24: 157–162, 1994.
 213. Schricker, K., S., Holmer, B. K. Krämer, G. Riegger, and A. Kurtz. Control of renin gene expression in 2 kidney‐1 clip rats. Kidney Int. 46: 1539–1541, 1994.
 214. Sealey, J. E., S. A., Atlas, and J. H. Laragh. Prorenin and other large molecular weight forms of renin. Endocr. Rev. 1: 365–391, 1980.
 215. Sealey, J. E., F. R., Buhler, J. H. Laragh, E. L. Manning, and H. R. Brunner. Aldosterone excretion: physiological variations in man measured by radioimmunoassay or double‐isotope dilution. Circ. Res. 31: 367–378, 1972.
 216. Sechi, L. A., C. A., Griffin, G. Giacchetti, J. P. Valentin, C. Llorens‐Cortes, P. Corvol, and M. Schambelan. Tissue‐specific regulation of type I angiotensin II receptor mRNA levels in the rat. Hypertension 28: 403–408, 1996.
 217. Seo, M.‐S., A., Fukamizu, T. Saito, and K. Murakami. Identification of a previously unrecognized production site of human renin. Biochim. Biophys. Acta 1129: 87–89, 1991.
 218. Sernia, C., T. A., Shinkel, W. G. Thomas, K. K. Y. Ho, and D. Lincoln. Angiotensinogen secretion by single rat pituitary cells: detection by a reverse haemolytic plaque assay and cell identification by immunocytochemistry. Neuroendocrinology 55: 308–316, 1992.
 219. Shenoy, U., and L. Cassis. Characterization of renin activity in brown adipose tissue. Am. J. Physiol. 272 (Cell Physiol. 41): C989–C999, 1997.
 220. Shinagawa, T., Y.‐S., Do, H. Tam, and W. A. Hsueh. Complete purification of human renin and sequence of the amino terminus. Biochem. Biophys. Res. Commun. 139: 446–454, 1986.
 221. Shinagawa, T., N., Nakayama, Y. Uchiyama, E. Kominami, Y. Doi, K. Hashiba, K. Yano, W. A. Hsueh, and K. Murakami. Role of cathepsin B as prorenin processing enzyme in human kidney. Hypertens. Res. 18: 131–136, 1995.
 222. Shiota, N., Y., Saegusa, K. Nishimura, and M. Miyazaki. Angiotensin II–generating system in dog and monkey ocular tissues. Clin. Exp. Pharmacol. Physiol. 24: 243–248, 1997.
 223. Sigmon, D. H., and J. C. S. Fray. Chemiosmotic control of renin release from isolated renin granules of rat kidneys. J. Physiol. (Lond.) 436: 237–256, 1991.
 224. Sigmund, C. D., and K. W. Gross. Differential expression of the murine and rat renin genes in peripheral subcutaneous tissue. Biochem. Biophys. Res. Commun. 173: 218–223, 1990.
 225. Sigmund, C. D., C. A., Jones, J. R. Fabian, J. J. Mullins, and K. W. Gross. Tissue and cell specific expression of a renin promoter‐reporter gene construct in transgenic mice. Biochem. Biophys. Res. Commun. 170: 344–350, 1990.
 226. Sigmund, C. D., C. A., Jones, H. J. Jacob, J. Ingelfinger, U. Kim, D. Gamble, V. J. Dzau, and K. W. Gross. Pathophysiology of vascular smooth muscle in renin promoter‐T‐antigen transgenic mice. Am. J. Physiol. 260 (Renal Fluid Electrolyte Physiol. 29): F249–F257, 1991.
 227. Simon, C. D., T. W., Honeyman, and J. C. S. Fray. Renin–angiotensin system in hypophysectomized rats. I. Control of blood pressure. Am. J. Physiol. 246 (Endocrinol. Metab. 9): E84–E88, 1984.
 228. Skott, O.. Calcium and osmotic stimulation in renin release from isolated rat glomeruli. Pflugers Arch. 406: 485–491, 1986.
 229. Skott, O.. Do osmotic forces play a role in renin secretion? Am. J. Physiol. 255 (Renal Fluid Electrolyte Physiol. 24): F1–F10, 1988.
 230. Skott, O.. Effects of amines, monensin and nigericin on the renin release from isolated superfused rat glomeruli. Pflugers Arch. 409: 93–99, 1987.
 231. Smith, D. L., R. E., Law, K. J. Shaw, Y. S. Do, W. A. Hsueh, and B. J. Morris. Proximal 2.6 kb of 5'‐flanking DNA is insufficient for human renin promoter activity in renin‐synthesizing chorio‐decidual cells. Biochim. Biophys. Acta 1219: 465–474, 1994.
 232. Smith, D. L., and B. J. Morris. Transient expression analyses of DNA extending 2.4 kb upstream of the human renin gene. Mol. Cell. Endocrinol. 80: 139–146, 1991.
 233. Soubrier, F., F. Alhenc‐Gelas, C. Hubert et al, Two putative active centers in human angiotensin I‐converting enzyme revealed by molecular cloning. Proc. Natl. Acad. Sci. U.S.A. 85: 9386–9390, 1988.
 234. Soubrier, F., J.‐J., Panthier, A.‐M. Houot, F. Rougeon, and P. Corvol. Segmental homology between the promoter region of the human renin gene and the mouse ren1 and ren2 promoter regions. Gene 41: 85–92, 1986.
 235. Steckelings, U. M., M., Artuc, M. Paul, M. Stoll, and B. M. Henz. Angiotensin II stimulates proliferation of primary human keratinocytes via a non‐AT1, non AT2 receptor. Biochem. Biophys. Res. Commun. 229: 329–333, 1996.
 236. Szymura, J. M., B. A., Taylor, and J. Klein. Upg‐2: a urinary pepsinogen variant located on chromosome 1 of the mouse. Biochem. Genet. 20: 1211–1219, 1982.
 237. Tada, M., A., Fukamizu, M. S. Seo, S. Takahashi, and K. Murakami. Renin expression in the kidney and brain is reciprocally controlled by captopril. Biochem. Biophys. Res. Commun. 159: 1065–1071, 1989.
 238. Takahashi, S., A., Fukamizu, T. Hasegawa, M. Yokoyama, T. Nomura, M. Katsuki, and K. Murakami. Expression of the human angiotensinogen gene in transgenic mice and transfected cells. Biochem. Biophys. Res. Commun. 180: 1103–1109, 1991.
 239. Takemura, T., T., Shishiba, O. Akiyama, M. Oritsu, Y. Matsui, and Y. Eishi. Vascular involvement in cutaneous sarcoidosis. Pathol. Int. 47: 84–89, 1997.
 240. Tamura, K., S. Umemura, T. Yamakawa et al, Modulation of tissue angiotensinogen gene expression in genetically obese hypertensive rats. Am. J. Physiol. 272 (Regulatory Integrative Comp. Physiol. 41): R1704–R1711, 1997.
 241. Tang, J., and R. N. S. Wong. Evolution in the structure and function of aspartic proteases. J. Cell. Biochem. 33: 53–63, 1987.
 242. Tatei, K., H., Cai, Y. T. Ip, and M. Livine. Race: a Drosophila homologue of the angiotensin converting enzyme. Mech. Dev. 51: 157–168, 1995.
 243. Taugner, R., C. P., Bührle, R. Nobiling, and H. Kirschke. Coexistence of renin and cathepsin B in epitheloid cell secretory granules. Histochemistry 83: 103–108, 1985.
 244. Taugner, R., S.‐J., Kim, K. Murakami, and R. Waldherr. The fate of prorenin during granulopoiesis in epithelioid cells: immunocytochemical experiments with antisera against renin and different portions of the renin prosegment. Histochemistry 86: 249–253, 1987.
 245. Terada, Y., K., Tomita, H. Nonoguchi, and F. Marumo. PCR localization of angiotensin II receptor and angiotensinogen mRNAs in rat kidney. Kidney Int. 43: 1251–1259, 1993.
 246. Tewksbury, D. A., Angiotensinogen. In: Handbook of Physiology. The Endocrine System. Endocrine Regulation of Water and Electrolyte Balance, edited by J. Fray. Bethesda, MD: Am. Physiol. Soc., 1999, Sect. 7, vol. III, p. 59–80.
 247. Tewksbury, D. A. Angiotensinogen, Biochemistry and molecular biology. In: Hypertension. Pathophysiology, Diagnosis, and Management, edited by J. H. Laragh and B. M. Brenner. New York: Raven, 1990, p. 1197–1216.
 248. Tewksbury, D. A., and R. A. Dart. High molecular weight angiotensinogen levels in hypertensive pregnant women. Hypertension 4: 729–734, 1982.
 249. Tewksbury, D. A., R. A., Dart, and J. Travis. The amino terminal amino acid sequence of human angiotensinogen. Biochem. Biophys. Res. Commun. 99: 1311–1315, 1981.
 250. Thomas, S. E., T. F. Andoh, et al, Accelerated apoptosis characterizes cyclosporine‐associated interstitial fibrosis. Kidney Int. 53: 897–908, 1998.
 251. Tigerstedt, R., and P. G. Bergman. Niere und kreislauf. Skand. Arch. Physiol. 8: 223–271, 1898.
 252. Tomita, N., R., Morishita, J. Higaki, M. Aoki, Y. Nakamura, H. Mikami, A. Fukamizu, K. Murakami, Y. Kaneda, and T. Ogihara. Transient decrease in high blood pressure by in vivo transfer of antisense oligodeoxynucleotides against rat angiotensinogen. Hypertension 26: 131–136, 1995.
 253. Torruella, M., K., Gordon, and T. Hohn. Cauliflower mosaic virus produces an aspartic proteinase to cleave its polyproteins. EMBO J. 8: 2819–2825, 1989.
 254. Tronik, D., M., Dreyfus, C. Babinet, and F. Rougeon. Regulated expression of the Ren‐2 gene in transgenic mice derived from parental strains carrying only the Ren‐1 gene. EMBO J. 6: 983–987, 1987.
 255. Ubeda, M., M. M., Matzilevich, N. M. Atucha, J. García‐Estan, T. Quesada, S.‐S. Tang, and J. R. Ingelfinger. Renin and angiotensinogen mRNA expression in the kidneys of rats subjected to long‐term bile duct ligation. Hepatology 19: 1431–1436, 1994.
 256. Urata, H., K. D., Boehm, A. Philip, A. Kinoshita, J. Gabrovsek, F. M. Bumpus, and A. Husain. Cellular localization and regional distribution of an angiotensin II–forming chymase in the heart. J. Clin. Invest. 91: 1269–1281, 1993.
 257. Urata, H., A., Kinoshita, K. S. Misono, F. M. Bumpus, and A. Husain. Identification of a highly specific chymase as the major angiotensin II–forming enzyme in the human heart [published erratum appears in J. Biol. Chem. 266: 12114, 1991]. J. Biol. Chem. 265: 22348–22357, 1990.
 258. Urata, H., H., Nishimura, and D. Ganten. Chymase‐dependent angiotensin II forming systems in humans. Am. J. Hypertens. 9: 277–284, 1996.
 259. Usui, M., T., Ichiki, M. Katoh, K. Egashira, and A. Takeshita. Regulation of angiotensin II receptor expression by nitric oxide in rat adrenal gland. Hypertension 32: 527–533, 1998.
 260. Vonthun, A. M., S. S., Eldahr, R. C. Vari, and L. G. Navar. Modulation of renin–angiotesnin and kallikrein gene expression in experimental hypertension. Hypertension 23 (Suppl. I): I‐131–I‐136, 1994.
 261. Wagner, D., R., Metzger, M. Paul, G. Ludwig, F. Suzuki, S. Takahashi, K. Murakami, and D. Ganten. Androgen dependence and tissue specificity of renin messenger RNA expression in mice. J. Hypertens. 8: 45–52, 1990.
 262. Wagner, J., A. H. J., Danser, F. H. M. Derkx, P. T. V. M. de Jong, M. Paul, M. A. D. H. Schalekamp, and D. Ganten. Demonstration of renin mRNA, angiotensinogen mRNA, and angiotensin converting enzyme mRNA expression in the human eye: evidence for an intraocular renin‐angiotensin system. Br. J. Ophthalmol. 80: 159–163, 1996.
 263. Wang, P. H., Y. S., Do, L. Macaulay, T. Shinagawa, P. W. Anderson, J. D. Baxter, and W. A. Hseuh. Identification of renal cathepsin‐B as a human prorenin‐processing enzyme. J. Biol. Chem. 266: 12633–12638, 1991.
 264. Watanabe, T., T., Nakagawa, J. Ikemizu, M. Nagahama, K. Murakami, and K. Nakayama. Sequence requirements for precursor cleavage within the constitutive secretory pathway. J. Biol. Chem. 267: 8270–8274, 1992.
 265. Watkins, L., Jr., J. A. Burton, E. Haber, J. R. Cant, F. W. Smith, and A. C. Barger. The renin‐angiotensin‐aldosterone system in congestive heart failure in conscious dogs. J. Clin. Invest. 57: 1606–1617, 1976.
 266. Weaver, D., S., Skinner, L. Walker, and M. Sangster. Phenotypic inhibition of the renin‐angiotensin system, emergence of the Ren‐2 gene, and adaptive radiation of mice. Gen. Comp. Endocrinol. 83: 306–315, 1991.
 267. White, P. C., M. I., New, and B. Dupont. Congenital adrenal hyperplasia. N. Engl. J. Med. 316: 1519–1524, 1987.
 268. Wielbo, D., C., Sernia, R. Gyurko, and M. I. Phillips. Antisense inhibition of hypertension in the spontaneously hypertensive rat. Hypertension 25: 314–319, 1995.
 269. Williams, T. A., P., Corvol, and F. Soubrier. Identification of two active site residues in human angiotensin I‐converting enzyme. J. Biol. Chem. 269: 29430–29434, 1994.
 270. Williams, T. A., A., Michaud, X. Houard, M.‐T. Chauvet, F. Soubrier, and P. Corvol. Drosophila melanogaster angiotensin I‐converting enzyme expressed in Pichia pastoris resembles the C domain of the mammalian homologue and does not require glycosylation for secretion and enzymic activity. Biochem. J. 318: 125–131, 1996.
 271. Wilson, C. M., E. G., Erdös, J. D. Wilson, and B. A. Taylor. Location on chromosome 1 of Rnr, a gene that regulates renin in the submaxillary gland of the mouse. Proc. Natl. Acad. Sci. U.S.A. 75: 5623–5626, 1978.
 272. Wilson, D. M., and J. A. Luetscher. Plasma prorenin activity and complications in children with insulin‐dependent diabetes mellitus. N. Engl. J. Med. 323: 1101–1106, 1990.
 273. Wilson, H. E., and A. White. Prohormones: their clinical relevance. Trends Endocrinol. Metab. 9: 396–402, 1998.
 274. Wu, Y., P. A., Cahill, and J. V. Sitzmann. Decreased angiotensin II receptors mediate decreased vascular response in hepatocellular cancer. Ann. Surg. 223: 225–231, 1996.
 275. Wyse, B., and C. Sernia. Growth hormone regulates AT‐1a angiotensin receptors in astrocytes. Endocrinology 138: 4176–4180, 1997.
 276. Yang, G., D. C., Merrill, M. W. Thompson, J. E. Robillard, and C. D. Sigmund. Functional expression of the human angiotensinogen gene in transgenic mice. J. Biol. Chem. 269: 32497–32502, 1994.
 277. Yu, H., and R. Di Nicolantonio. Altered age‐dependent modulation of tissue renin messenger RNA levels in the spontaneously hypertensive rat. J. Hypertens. 14: 871–880, 1996.
 278. Yu, X. C., E. D., Sturrock, Z. Wu, K. Biemann, M. R. Ehlers, and J. F. Riordan. Identification of N‐linked glycosylation sites in human testis angiotensin‐converting enzyme and expression of an active deglycosylated form. J. Biol. Chem. 272: 3511–3519, 1997.
 279. Zhang, D. Y., E. R., Lumbers, and G. Simonetta. Changes in renal renin gene expression in fetal sheep. Clin. Exp. Pharmacol. Physiol. 23: 682–684, 1996.
 280. Zhao, Y., J., Peters, D. Ganten, and M. Bader. Expression of the mouse ren‐2 gene in the small intestine is regulated by food intake. Pflügers Arch. Eur. J. Physiol. 424: 199–202, 1993.
 281. Zheng, J., K. E., Vagnoni, I. M. Bird, and R. R. Magness. Expression of basic fibroblast growth factor endothelial mitogenic activity, and angotensin II type‐1 receptors in the ovine placenta during the third trimester of pregnancy. Biol. Reprod. 56: 1189–1197, 1997.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

John Fray. Endocrine Control of Sodium Balance. Compr Physiol 2011, Supplement 22: Handbook of Physiology, The Endocrine System, Endocrine Regulation of Water and Electrolyte Balance: 250-305. First published in print 2000. doi: 10.1002/cphy.cp070307