51
|
Kopp UC, Jones SY, DiBona GF. Afferent renal denervation impairs baroreflex control of efferent renal sympathetic nerve activity. Am J Physiol Regul Integr Comp Physiol 2008; 295:R1882-90. [PMID: 18945951 DOI: 10.1152/ajpregu.90529.2008] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Increasing efferent renal sympathetic nerve activity (ERSNA) increases afferent renal nerve activity (ARNA), which decreases ERSNA to prevent sodium retention. High-sodium diet enhances ARNA, suggesting an important role for ARNA in suppressing ERSNA during excess sodium intake. Mean arterial pressure (MAP) is elevated in afferent renal denervated by dorsal rhizotomy (DRX) rats fed high-sodium diet. We examined whether the increased MAP in DRX is due to impaired arterial baroreflex function. In DRX and sham DRX rats fed high-sodium diet, arterial baroreflex function was determined in conscious rats by intravenous nitroprusside and phenylephrine or calculation of transfer function gain from arterial pressure to ERSNA (spontaneous baroreflex sensitivity). Increasing MAP did not suppress ERSNA to the same extent in DRX as in sham DRX, -60 +/- 4 vs. -77 +/- 6%. Maximum gain, -4.22 +/- 0.45 vs. -6.04 +/- 0.90% DeltaERSNA/mmHg, and the maximum value of instantaneous gain, -4.19 +/- 0.45 vs. -6.04 +/- 0.81% DeltaERSNA/mmHg, were less in DRX than in sham DRX. Likewise, transfer function gain was lower in DRX than in sham DRX, 3.9 +/- 0.2 vs. 6.1 +/- 0.5 NU/mmHg. Air jet stress produced greater increases in ERSNA in DRX than in sham DRX, 35,000 +/- 4,900 vs. 20,900 +/- 3,410%.s (area under the curve). Likewise, the ERSNA responses to thermal cutaneous stimulation were greater in DRX than in sham DRX. These studies suggest impaired arterial baroreflex suppression of ERSNA in DRX fed high-sodium diet. There were no differences in arterial baroreflex function in DRX and sham DRX fed normal-sodium diet. Impaired arterial baroreflex function contributes to increased ERSNA, which would eventually lead to sodium retention and increased MAP in DRX rats fed high-sodium diet.
Collapse
Affiliation(s)
- Ulla C Kopp
- Department of Internal Medicine, Department of Veterans Affairs Medical Center, Bldg. 41, Rm. 124, Highway 6W, Iowa City, IA 52246, USA.
| | | | | |
Collapse
|
52
|
Gonzalez-Villalobos RA, Seth DM, Satou R, Horton H, Ohashi N, Miyata K, Katsurada A, Tran DV, Kobori H, Navar LG. Intrarenal angiotensin II and angiotensinogen augmentation in chronic angiotensin II-infused mice. Am J Physiol Renal Physiol 2008; 295:F772-F779. [PMID: 18579707 PMCID: PMC2536885 DOI: 10.1152/ajprenal.00019.2008] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Accepted: 06/19/2008] [Indexed: 11/22/2022] Open
Abstract
The objectives of this study were to determine the effects of chronic angiotensin II (ANG II) infusions on ANG II content and angiotensinogen expression in the mouse kidney and the role of the angiotensin II type 1 receptor (AT(1)R) in mediating these changes. C57BL/6J male mice were subjected to ANG II infusions at doses of 400 or 1,000 ng.kg(-1).min(-1) either alone or with an AT(1)R blocker (olmesartan; 3 mg.kg(-1).day(-1)) for 12 days. Systolic and mean arterial pressures were determined by tail-cuff plethysmography and radiotelemetry. On day 13, blood and kidneys were collected for ANG II determinations by radioimmunoanalysis and intrarenal angiotensinogen expression studies by quantitative RT-PCR, Western blotting, and immunohistochemistry. ANG II infusions at the low dose elicited progressive increases in systolic blood pressure (135 +/- 2.5 mmHg). In contrast, the high dose induced a rapid increase (152 +/- 2.5, P < 0.05 vs. controls, 109 +/- 2.8). Renal ANG II content was increased by ANG II infusions at the low dose (1,203 +/- 253 fmol/g) and the high dose (1,258 +/- 173) vs. controls (499 +/- 40, P < 0.05). Kidney angiotensinogen mRNA and protein were increased only by the low dose to 1.13 +/- 0.02 and 1.26 +/- 0.10, respectively, over controls (1.00, P < 0.05). These effects were not observed in mice infused at the high dose and those receiving olmesartan. The results indicate that chronic ANG II infusions augment mouse intrarenal ANG II content with AT(1)R-dependent uptake occurring at both doses, but only the low dose of infusion, which elicited a slow progressive response, causes an AT(1)R-dependent increase in intrarenal angiotensinogen expression.
Collapse
Affiliation(s)
- Romer A Gonzalez-Villalobos
- Department of Physiology, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Kopp UC, Cicha MZ, Yorek MA. Impaired responsiveness of renal sensory nerves in streptozotocin-treated rats and obese Zucker diabetic fatty rats: role of angiotensin. Am J Physiol Regul Integr Comp Physiol 2008; 294:R858-66. [DOI: 10.1152/ajpregu.00830.2007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Increasing afferent renal nerve activity decreases efferent renal nerve activity and increases urinary sodium excretion. Activation of renal pelvic mechanosensory nerves is impaired in streptozotocin (STZ)-treated rats (model of type 1 diabetes). Decreased activation of renal sensory nerves would lead to increased efferent renal nerve activity, sodium retention, and hypertension. We examined whether the reduced activation of renal sensory nerves in STZ rats was due to increased renal angiotensin activity and whether activation of the renal sensory nerves was impaired in obese Zucker diabetic fatty (ZDF) rats (model of type 2 diabetes). In an isolated renal pelvic wall preparation from rats treated with STZ for 2 wk, PGE2failed to increase the release of substance P, from 5 ± 1 to 6 ± 1 pg/min. In pelvises from sham STZ rats, PGE2increased substance P release from 6 ± 1 to 13 ± 2 pg/min. Adding losartan to the incubation bath increased PGE2-mediated release of substance P in STZ rats, from 5 ± 1 to 10 ± 2 pg/min, but had no effect in sham STZ rats. In pelvises from obese ZDF rats (22–46 wk old), PGE2increased substance P release from 12.0 ± 1.2 to 18.3 ± 1.2 pg/min, which was less than that from lean ZDF rats (10.3 ± 1.6 to 22.5 ± 2.4 pg/min). Losartan had no effect on the PGE2-mediated substance P release in obese or lean ZDF rats. We conclude that the mechanisms involved in the decreased responsiveness of the renal sensory nerves in STZ rats involve activation of the renin angiotensin system in STZ but not in obese ZDF rats.
Collapse
|
54
|
Zeng C, Armando I, Luo Y, Eisner GM, Felder RA, Jose PA. Dysregulation of dopamine-dependent mechanisms as a determinant of hypertension: studies in dopamine receptor knockout mice. Am J Physiol Heart Circ Physiol 2008; 294:H551-69. [PMID: 18083900 PMCID: PMC4029502 DOI: 10.1152/ajpheart.01036.2007] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dopamine plays an important role in the pathogenesis of hypertension by regulating epithelial sodium transport and by interacting with vasoactive hormones/humoral factors, such as aldosterone, angiotensin, catecholamines, endothelin, oxytocin, prolactin pro-opiomelancortin, reactive oxygen species, renin, and vasopressin. Dopamine receptors are classified into D(1)-like (D(1) and D(5)) and D(2)-like (D(2), D(3), and D(4)) subtypes based on their structure and pharmacology. In recent years, mice deficient in one or more of the five dopamine receptor subtypes have been generated, leading to a better understanding of the physiological role of each of the dopamine receptor subtypes. This review summarizes the results from studies of various dopamine receptor mutant mice on the role of individual dopamine receptor subtypes and their interactions with other G protein-coupled receptors in the regulation of blood pressure.
Collapse
MESH Headings
- Animals
- Blood Pressure/genetics
- Blood Pressure/physiology
- Dopamine/physiology
- Hypertension/genetics
- Hypertension/physiopathology
- Mice
- Mice, Knockout
- Receptors, Dopamine/genetics
- Receptors, Dopamine/physiology
- Receptors, Dopamine D1/genetics
- Receptors, Dopamine D1/physiology
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/physiology
- Receptors, Dopamine D3/genetics
- Receptors, Dopamine D3/physiology
- Receptors, Dopamine D4/genetics
- Receptors, Dopamine D4/physiology
- Receptors, Dopamine D5/genetics
- Receptors, Dopamine D5/physiology
Collapse
Affiliation(s)
- Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing City 400042, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
55
|
Siragy HM, Huang J. Renal (pro)renin receptor upregulation in diabetic rats through enhanced angiotensin AT1 receptor and NADPH oxidase activity. Exp Physiol 2008; 93:709-14. [PMID: 18192338 DOI: 10.1113/expphysiol.2007.040550] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Recent studies have demonstrated the presence of the (pro)renin receptor (PRR) in the glomerular mesangium and the subendothelial layer of the renal arteries. We hypothesized that diabetes upregulates PRR expression through enhanced angiotensin subtype 1 (AT1) receptor-NADPH oxidase cascade activity. Using real-time polymerase chain reaction, Western blot analysis and immunostaining, we studied renal localization of the PRR in the streptozotocin-induced diabetic rat model and in response to 1 week of treatment with the AT1 receptor blocker valsartan (10 mg kg(-1) day(-1)), the angiotensin AT2 receptor blocker PD123319 (0.5 mg kg(-1) day(-1)) or the NADPH oxidase inhibitor diphenylene iodonium (DPI; 0.5 mg kg(-1) day(-1)) 6 weeks post-induction of diabetes. Both PRR mRNA and protein were expressed constitutively in the kidneys of normal rat renal cortex and medulla, mainly in glomerular mesangium, proximal, distal and collecting tubules. Compared with normal rats (100%), diabetic rats demonstrated an increase in renal PRR mRNA (184%), protein (228%) and immunostaining. Valsartan and DPI prevented the increase in the PRR mRNA (106 and 126%, respectively), protein (97 and 140%, respectively) and immunostaining that was seen in the kidneys of diabetic rats. The AT2 blocker PD123319 did not have significant effects on PRR mRNA (157%) or protein expression (200%) in the kidneys of diabetic rats. These results demonstrate that the PRR is constitutively expressed in renal glomeruli and tubules. Expression of the PRR is upregulated in diabetes via enhancement of AT1 receptor-NADPH oxidase activity.
Collapse
Affiliation(s)
- Helmy M Siragy
- Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908-1409, USA.
| | | |
Collapse
|
56
|
Li XC, Zhuo JL. In vivo regulation of AT1a receptor-mediated intracellular uptake of [125I]Val5-ANG II in the kidneys and adrenals of AT1a receptor-deficient mice. Am J Physiol Renal Physiol 2007; 294:F293-302. [PMID: 18045833 DOI: 10.1152/ajprenal.00398.2007] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Using type 1a angiotensin receptor (AT1a) receptor-deficient (Agtr1a-/-) mice and in vivo autoradiography, we tested the hypothesis that intracellular uptake of ANG II in the kidney and adrenal glands is primarily mediated by AT1a receptors and that the response is regulated by prevailing endogenous ANG II. After pretreatment of wild-type (Agtr1a+/+) and Agtr1a-/- mice (n = 6-9 each group) with or without captopril (25 mg.kg(-1).day(-1)) or losartan (10 mg.kg(-1).day(-1)) for 2 wk, [125I]Val5-ANG II was infused for 60 min. Intracellular uptake of [125I]Val5-ANG II was determined by quantitative in vivo autoradiography after washout of circulating [125I]Val5-ANG II. Basal intracellular ANG II levels were 65% lower in the kidney (P < 0.001), but plasma ANG II levels were threefold higher, in Agtr1a-/- than wild-type mice (P < 0.01). Although plasma [125I]Val5-ANG II levels were similar, urinary excretion of [125I]Val5-ANG II was fourfold higher in Agtr1a-/- mice (P < 0.001). By contrast, intracellular [125I]Val5-ANG II levels were approximately 80% lower in the kidney and adrenal glands of Agtr1a-/- mice (P < 0.01). Captopril decreased endogenous plasma and renal ANG II levels (P < 0.01) but increased intracellular uptake of [125I]Val5-ANG II in the kidney and adrenal glands of wild-type and Agtr1a-/- mice (P < 0.01). Losartan largely blocked renal and adrenal uptake of [125I]Val5-ANG II in wild-type and Agtr1a-/- mice. Thus 80% of intracellular ANG II uptake in the kidney and adrenal glands is mediated by AT1a receptors, whereas AT1b receptor- and other non-receptor-mediated mechanisms account for 20% of the response. Our results suggest that AT1a receptor-mediated uptake of extracellular ANG II may play a physiological role in the kidney and adrenal glands.
Collapse
|
57
|
Kopp UC, Cicha MZ, Smith LA, Mulder J, Hökfelt T. Renal sympathetic nerve activity modulates afferent renal nerve activity by PGE2-dependent activation of α1- and α2-adrenoceptors on renal sensory nerve fibers. Am J Physiol Regul Integr Comp Physiol 2007; 293:R1561-72. [PMID: 17699565 DOI: 10.1152/ajpregu.00485.2007] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Increasing efferent renal sympathetic nerve activity (ERSNA) increases afferent renal nerve activity (ARNA). To test whether the ERSNA-induced increases in ARNA involved norepinephrine activating α-adrenoceptors on the renal sensory nerves, we examined the effects of renal pelvic administration of the α1- and α2-adrenoceptor antagonists prazosin and rauwolscine on the ARNA responses to reflex increases in ERSNA (placing the rat's tail in 49°C water) and renal pelvic perfusion with norepinephrine in anesthetized rats. Hot tail increased ERSNA and ARNA, 6,930 ± 900 and 4,870 ± 670%·s (area under the curve ARNA vs. time). Renal pelvic perfusion with norepinephrine increased ARNA 1,870 ± 210%·s. Immunohistochemical studies showed that the sympathetic and sensory nerves were closely related in the pelvic wall. Renal pelvic perfusion with prazosin blocked and rauwolscine enhanced the ARNA responses to reflex increases in ERSNA and norepinephrine. Studies in a denervated renal pelvic wall preparation showed that norepinephrine increased substance P release, from 8 ± 1 to 16 ± 1 pg/min, and PGE2 release, from 77 ± 11 to 161 ± 23 pg/min, suggesting a role for PGE2 in the norepinephrine-induced activation of renal sensory nerves. Prazosin and indomethacin reduced and rauwolscine enhanced the norepinephrine-induced increases in substance P and PGE2. PGE2 enhanced the norepinephrine-induced activation of renal sensory nerves by stimulation of EP4 receptors. Interaction between ERSNA and ARNA is modulated by norepinephrine, which increases and decreases the activation of the renal sensory nerves by stimulating α1- and α2-adrenoceptors, respectively, on the renal pelvic sensory nerve fibers. Norepinephrine-induced activation of the sensory nerves is dependent on renal pelvic synthesis/release of PGE2.
Collapse
MESH Headings
- Adrenergic Fibers/physiology
- Adrenergic alpha-1 Receptor Antagonists
- Adrenergic alpha-2 Receptor Antagonists
- Animals
- Calcitonin Gene-Related Peptide/metabolism
- Dinoprostone/pharmacology
- Indomethacin/pharmacology
- Kidney/innervation
- Male
- Neurons, Afferent/drug effects
- Neurons, Afferent/physiology
- Neurons, Efferent/physiology
- Norepinephrine/metabolism
- Norepinephrine Plasma Membrane Transport Proteins/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Adrenergic, alpha-1/drug effects
- Receptors, Adrenergic, alpha-2/drug effects
- Receptors, Prostaglandin E/antagonists & inhibitors
- Receptors, Prostaglandin E, EP4 Subtype
- Substance P/metabolism
- Thiophenes/pharmacology
- Triazoles/pharmacology
Collapse
Affiliation(s)
- Ulla C Kopp
- Dept. of Internal Medicine, VA Medical Center, Bldg. 41, Rm 124, Highway 6W, Iowa City, IA 52246, USA.
| | | | | | | | | |
Collapse
|
58
|
Liang B, Leenen FHH. Prevention of salt induced hypertension and fibrosis by angiotensin converting enzyme inhibitors in Dahl S rats. Br J Pharmacol 2007; 152:903-14. [PMID: 17906684 PMCID: PMC2078219 DOI: 10.1038/sj.bjp.0707472] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AND PURPOSE In Dahl S rats, high salt increases activity of the tissue renin-angiotensin-aldosterone system (RAAS) in the CNS, heart and kidneys. Here, we assessed the effects of chronic angiotensin converting enzyme (ACE) inhibition on salt-induced hypertension and cardiovascular and renal hypertrophy and fibrosis, relative to the extent of ACE blockade. EXPERIMENTAL APPROACH From 4.5 weeks of age, Dahl S rats received either the lipophilic ACE inhibitor trandolapril (1 or 5 mg kg(-1) day(-1)) or the hydrophilic ACE inhibitor lisinopril (10 or 50 mg kg(-1) day(-1)) and a high salt diet was started 0.5 week later. Treatments ended at 9 weeks of age. KEY RESULTS High salt diet markedly increased blood pressure (BP), decreased plasma angiotensin II and increased ACE binding densities in brain, heart, aorta and kidneys. Trandolapril and lisinopril prevented 50% of the increase in BP in light and dark period of the day. After the last doses, trandolapril decreased ACE densities by approximately 80% in brain nuclei and heart and lisinopril by approximately 60% in the brain and by approximately 70% in the heart. The two ACE inhibitors prevented right ventricular hypertrophy and attenuated left ventricular hypertrophy but did not affect renal hypertrophy caused by high salt. Both drugs prevented high salt-induced fibrosis in heart, kidney and aorta. CONCLUSION AND IMPLICATION As the ACE inhibitors could completely prevent tissue fibrosis and partially prevent tissue hypertrophy and hypertension, the tissue RAAS may play a critical role in salt-induced fibrosis, but a lesser role in the hypertrophy.
Collapse
Affiliation(s)
- B Liang
- Hypertension Unit, University of Ottawa Heart Institute Ottawa, ON, Canada
| | - F H H Leenen
- Hypertension Unit, University of Ottawa Heart Institute Ottawa, ON, Canada
- Author for correspondence:
| |
Collapse
|
59
|
Zhuo JL, Li XC. Novel roles of intracrine angiotensin II and signalling mechanisms in kidney cells. J Renin Angiotensin Aldosterone Syst 2007; 8:23-33. [PMID: 17487823 PMCID: PMC2276849 DOI: 10.3317/jraas.2007.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Angiotensin II (Ang II) has powerful sodium-retaining, growth-promoting and pro- inflammatory properties in addition to its physiological role in maintaining body salt and fluid balance and blood pressure homeostasis. Increased circulating and local tissue Ang II is one of the most important factors contributing to the development of sodium and fluid retention, hypertension and target organ damage. The importance of Ang II in the pathogenesis of hypertension and target organ injury is best demonstrated by the effectiveness of angiotensin- converting enzyme (ACE) inhibitors and AT1-receptor antagonists in treating hypertension and progressive renal disease including diabetic nephropathy. The detrimental effects of Ang II are mediated primarily by the AT1-receptor, while the AT2-receptor may oppose the AT1-receptor. The classical view of the AT1-receptor-mediated effects of Ang II is that the agonist binds its receptors at the cell surface, and following receptor phosphorylation, activates downstream signal transduction pathways and intracellular responses. However, evidence is emerging that binding of Ang II to its cell surface AT1-receptors also activates endocytotic (or internalisation) processes that promote trafficking of both the effector and the receptor into intracellular compartments. Whether internalised Ang II has important intracrine and signalling actions is not well understood. The purpose of this article is to review recent advances in Ang II research with focus on the mechanisms underlying high levels of intracellular Ang II in proximal tubule cells and the contribution of receptor-mediated endocytosis of extracellular Ang II. Further attention is devoted to the question whether intracellular and/or internalised Ang II plays a physiological role by activating cytoplasmic or nuclear receptors in proximal tubule cells. This information may aid future development of drugs to prevent and treat Ang II-induced target organ injury in cardiovascular and renal diseases by blocking intracellular and/or nuclear actions of Ang II.
Collapse
Affiliation(s)
- Jia L Zhuo
- Laboratory of Receptor and Signal Transduction, Henry Ford Hospital, and Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48202, USA.
| | | |
Collapse
|
60
|
Ebrahimian T, He Y, Schiffrin EL, Touyz RM. Differential regulation of thioredoxin and NAD(P)H oxidase by angiotensin II in male and female mice. J Hypertens 2007; 25:1263-71. [PMID: 17563540 DOI: 10.1097/hjh.0b013e3280acac60] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We hypothesized that downregulation of the antioxidant thioredoxin system contributes to oxidative stress in angiotensin II-induced hypertension. As oestrogen may protect against oxidative stress, we also evaluated whether the thioredoxin system, particularly in the heart, is differentially regulated between females and males. RESULTS C57Bl/6 male and intact or ovariectomized female mice were infused with angiotensin II (400 ng/kg per minute for 2 weeks). Systolic blood pressure (SBP) was increased by angiotensin II in both groups week 1 and increased further in males versus females in week 2. Angiotensin II increased SBP from 112 +/- 6 to 143 +/- 9 mmHg in ovariectomized mice. Basal cardiac thioredoxin expression and reductase activity were significantly higher (two to threefold) in females versus males. Angiotensin II increased thioredoxin expression (approximately threefold), thioredoxin reductase activity, nicotinamide adenine dinucleotide phosphate, reduced form (NAD(P)H) oxidase activity and plasma thiobarbituric acid-reducing substances in males but not in females. Angiotensin II increased thioredoxin expression and NAD(P)H oxidase activity in ovariectomized versus control mice. Apurinic/apyrimidinic endonuclease/redox factor 1 (APE/Ref-1) activation, which interacts with thioredoxin to activate inflammatory transcription factors, was increased by angiotensin II only in males. CONCLUSION These results demonstrate sex dimorphism with respect to thioredoxin, oxidative stress and inflammation, and suggest the differential regulation of blood pressure, the cardiac thioredoxin system and NAD(P)H oxidase activity by angiotensin II in male and female mice. Whereas angiotensin II increases the activity of thioredoxin reductase and APE/Ref-1, enhances oxidative stress, and amplifies blood pressure elevation in males, it has little effect in females. Such differences may partly relate to the protective actions of oestrogens.
Collapse
Affiliation(s)
- Talin Ebrahimian
- Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec
| | | | | | | |
Collapse
|
61
|
Grigore D, Ojeda NB, Robertson EB, Dawson AS, Huffman CA, Bourassa EA, Speth RC, Brosnihan KB, Alexander BT. Placental insufficiency results in temporal alterations in the renin angiotensin system in male hypertensive growth restricted offspring. Am J Physiol Regul Integr Comp Physiol 2007; 293:R804-11. [PMID: 17537837 PMCID: PMC2841000 DOI: 10.1152/ajpregu.00725.2006] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Reduced uterine perfusion initiated in late gestation in the rat results in intrauterine growth restriction (IUGR) and development of hypertension by 4 wk of age. We hypothesize that the renin angiotensin system (RAS), a regulatory system important in the long-term control of blood pressure, may be programmed by placental insufficiency and may contribute to the etiology of IUGR hypertension. We previously reported that RAS blockade abolished hypertension in adult IUGR offspring; however, the mechanisms responsible for the early phase of hypertension are unresolved. Therefore, the purpose of this study was to examine RAS involvement in early programmed hypertension and to determine whether temporal changes in RAS expression are observed in IUGR offspring. Renal renin and angiotensinogen mRNA expression were significantly decreased at birth (80 and 60%, respectively); plasma and renal RAS did not differ in conjunction with hypertension (mean increase of 14 mmHg) in young IUGR offspring; however, hypertension (mean increase of 22 mmHg) in adult IUGR offspring was associated with marked increases in renal angiotensin-converting enzyme (ACE) activity (122%) and renal renin and angiotensinogen mRNA (7-fold and 7.4-fold, respectively), but no change in renal ANG II or angiotensin type 1 receptor. ACE inhibition (enalapril, 10 mg x kg(-1) x day(-1), administered from 2 to 4 wk of age) abolished hypertension in IUGR at 4 wk of age (decrease of 15 mmHg, respectively) with no significant depressor effect in control offspring. Therefore, temporal alterations in renal RAS are observed in IUGR offspring and may play a key role in the etiology of IUGR hypertension.
Collapse
Affiliation(s)
- Daniela Grigore
- Department of Physiology and the Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Mississippi 39216-4505, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Kopp UC, Cicha MZ, Smith LA. Activation of endothelin-a receptors contributes to angiotensin-induced suppression of renal sensory nerve activation. Hypertension 2006; 49:141-7. [PMID: 17060503 DOI: 10.1161/01.hyp.0000249634.46212.7b] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Activation of renal mechanosensory nerves is enhanced by a high-sodium diet and suppressed by a low-sodium diet. Angiotensin (Ang) II and endothelin (ET)-1 each contributes to the impaired responsiveness of renal mechanosensory nerves in a low-sodium diet. We examined whether stimulation of ETA receptors (Rs) contributes to Ang II-induced suppression of the responsiveness of renal mechanosensory nerves. In anesthetized rats fed a low-sodium diet, renal pelvic administration of the Ang type I receptor (AT1-R) antagonist losartan enhanced the afferent renal nerve activity (ARNA) response to increasing renal pelvic pressure 7.5 mm Hg from 7+/-2% to 15+/-2% and the prostaglandin (PG) E(2)-mediated substance P release from 0+/-1 to 8+/-1 pg/min. Adding the ETA-R antagonist BQ123 to the renal pelvic perfusate containing losartan did not produce any further enhancement of the ARNA response or PGE(2)-mediated release of substance P (17+/-3% and 8+/-1 pg/min). Likewise, renal pelvic administration of BQ123 and BQ123+losartan resulted in similar enhancements of the ARNA responses to increased renal pelvic pressure and PGE(2)-mediated substance P release. In high-sodium-diet rats, pelvic administration of Ang II reduced the ARNA response to increased renal pelvic pressure from 27+/-4% to 8+/-3% and the PGE(2)-mediated substance P release from 9+/-0 to 1+/-1 pg/min. Adding BQ123 to the renal pelvic perfusate containing Ang II restored the increases in ARNA and the PGE(2)-mediated substance P release toward control (27+/-6% and 7+/-1 pg/min). In conclusion, stimulation of ETA-R plays an important contributory role to the Ang II-mediated suppression of the activation of renal mechanosensory nerves in conditions of low-sodium diet.
Collapse
Affiliation(s)
- Ulla C Kopp
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, USA.
| | | | | |
Collapse
|
63
|
Zeng C, Liu Y, Wang Z, He D, Huang L, Yu P, Zheng S, Jones JE, Asico LD, Hopfer U, Eisner GM, Felder RA, Jose PA. Activation of D
3
Dopamine Receptor Decreases Angiotensin II Type 1 Receptor Expression in Rat Renal Proximal Tubule Cells. Circ Res 2006; 99:494-500. [PMID: 16902178 DOI: 10.1161/01.res.0000240500.96746.ec] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The dopaminergic and renin angiotensin systems interact to regulate blood pressure. Disruption of the D
3
dopamine receptor gene in mice produces renin-dependent hypertension. In rats, D
2
-like receptors reduce angiotensin II binding sites in renal proximal tubules (RPTs). Because the major D
2
-like receptor in RPTs is the D
3
receptor, we examined whether D
3
receptors regulate angiotensin II type 1 (AT
1
) receptors in rat RPT cells. The effect of D
3
receptors on AT
1
receptors was studied in vitro and in vivo. The D
3
receptor agonist PD128907 decreased AT
1
receptor protein and mRNA in WKY RPT cells and increased it in SHR cells. PD128907 increased D
3
receptors in WKY cells but had no effect in SHR cells. D
3
/AT
1
receptors colocalized in RPT cells; D
3
receptor stimulation decreased the percent amount of D
3
receptors that coimmunoprecipitated with AT
1
receptors to a greater extent in WKY than in SHR cells. However, D
3
receptor stimulation did not change the percent amount of AT
1
receptors that coimmunoprecipitated with D
3
receptors in WKY cells and markedly decreased the coimmunoprecipitation in SHR cells. The D
3
receptor also regulated the AT
1
receptor in vivo because AT
1
receptor expression was increased in kidneys of D
3
receptor–null mice compared with wild type littermates. D
3
receptors may regulate AT
1
receptor function by direct interaction with and regulation of AT
1
receptor expression. One mechanism of hypertension may be related to increased renal expression of AT
1
receptors due decreased D
3
receptor regulation.
Collapse
MESH Headings
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Benzopyrans/pharmacology
- Cells, Cultured
- Dopamine Agonists/pharmacology
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Mice
- Mice, Knockout
- Oxazines/pharmacology
- Rats
- Rats, Inbred SHR
- Rats, Inbred WKY
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, Dopamine D3/agonists
- Receptors, Dopamine D3/deficiency
- Receptors, Dopamine D3/metabolism
- Tissue Distribution
Collapse
Affiliation(s)
- Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing 400042, People's Republic of China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Kopp UC, Cicha MZ, Smith LA. Differential effects of endothelin on activation of renal mechanosensory nerves: stimulatory in high-sodium diet and inhibitory in low-sodium diet. Am J Physiol Regul Integr Comp Physiol 2006; 291:R1545-56. [PMID: 16763077 DOI: 10.1152/ajpregu.00878.2005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of renal mechanosensory nerves is enhanced by high and suppressed by low sodium dietary intake. Afferent renal denervation results in salt-sensitive hypertension, suggesting that activation of the afferent renal nerves contributes to water and sodium balance. Another model of salt-sensitive hypertension is the endothelin B receptor (ETBR)-deficient rat. ET and its receptors are present in sensory nerves. Therefore, we examined whether ET receptor blockade altered the responsiveness of the renal sensory nerves. In anesthetized rats fed high-sodium diet, renal pelvic administration of the ETBR antagonist BQ-788 reduced the afferent renal nerve activity (ARNA) response to increasing renal pelvic pressure 7.5 mmHg from 26+/-3 to 9+/-3% and the PGE2-mediated renal pelvic release of substance P from 9+/-1 to 3+/-1 pg/min. Conversely, in rats fed low-sodium diet, renal pelvic administration of the ETAR antagonist BQ-123 enhanced the ARNA response to increased renal pelvic pressure from 9+/-2 to 23+/-6% and the PGE2-mediated renal pelvic release of substance P from 0+/-0 to 6+/-1 pg/min. Adding the ETAR antagonist to ETBR-blocked renal pelvises restored the responsiveness of renal sensory nerves in rats fed a high-sodium diet. Adding the ETBR antagonist to ETAR-blocked pelvises suppressed the responsiveness of the renal sensory nerves in rats fed a low-sodium diet. In conclusion, activation of ETBR and ETAR contributes to the enhanced and suppressed responsiveness of renal sensory nerves in conditions of high- and low-sodium dietary intake, respectively. Impaired renorenal reflexes may contribute to the salt-sensitive hypertension in the ETBR-deficient rat.
Collapse
MESH Headings
- Animals
- Antihypertensive Agents/pharmacology
- Diet, Sodium-Restricted
- Endothelin A Receptor Antagonists
- Endothelin B Receptor Antagonists
- Endothelins/genetics
- Endothelins/physiology
- Ganglia, Spinal/metabolism
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/physiology
- Hypertension/genetics
- Hypertension/metabolism
- Hypertension/physiopathology
- Kidney/innervation
- Kidney/metabolism
- Male
- Mechanotransduction, Cellular/drug effects
- Mechanotransduction, Cellular/physiology
- Neurons, Afferent/drug effects
- Neurons, Afferent/metabolism
- Oligopeptides/pharmacology
- Peptides, Cyclic/pharmacology
- Piperidines/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptor, Endothelin A/drug effects
- Receptor, Endothelin A/genetics
- Receptor, Endothelin A/metabolism
- Receptor, Endothelin B/drug effects
- Receptor, Endothelin B/genetics
- Receptor, Endothelin B/metabolism
- Sodium, Dietary/pharmacology
- Substance P/metabolism
Collapse
Affiliation(s)
- Ulla C Kopp
- Dept. of Internal Medicine, VA Medical Center, University of Iowa Carver College of Medicine, Bldg. 3, Rm. 226, Highway 6W, Iowa City, IA 52246, USA.
| | | | | |
Collapse
|
65
|
Cheung WJ, Kent MAH, El-Shahat E, Wang H, Tan J, White R, Leenen FHH. Central and peripheral renin-angiotensin systems in ouabain-induced hypertension. Am J Physiol Heart Circ Physiol 2006; 291:H624-30. [PMID: 16565308 DOI: 10.1152/ajpheart.01148.2005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic subcutaneous infusion of ouabain causes hypertension via central pathways involving angiotensin type 1 (AT(1)) receptor stimulation. The present study assessed plasma and tissue ANG I and II levels as well as AT1 receptor and angiotensin-converting enzyme (ACE) mRNA levels and binding densities by real-time PCR and in vitro autoradiography in relevant brain nuclei and peripheral tissues (heart and kidney) in rats at 1 and/or 2 wk after start of ouabain infusion at 50 microg/day. After 2 wk (but not after 1 wk), blood pressures significantly increased (+15 mmHg). At 2 wk, plasma ANG I and II levels were markedly suppressed by ouabain. In contrast, in the heart and kidneys, ANG I levels were not affected, and ANG II levels tended to decrease, whereas in the hypothalamus ANG II content clearly increased. At 1 wk, no changes in ACE and AT1 receptor densities were seen. After 2 wk, there were significant decreases in AT(1) receptor mRNA and densities in the organum vasculosum of the lamina terminalis (OVLT), subfornical organ (SFO), and paraventricular nucleus (PVN). ACE densities decreased only in the OVLT and SFO, but ACE mRNA showed more variable responses (decrease in OVLT vs. increase in PVN). In the kidneys, at 2 wk both AT1 receptor and ACE densities were decreased, but mRNA abundance did not change. The heart showed no significant changes. The increase in hypothalamic ANG II content and associated decreases in central AT1 receptor and ACE densities support the involvement of the brain renin-angiotensin system in the central hypertensive mechanism of action of ouabain.
Collapse
Affiliation(s)
- Warren J Cheung
- Hypertension Unit, Univ. of Ottawa Heart Institute, Ottawa, Ontario, Canada K1Y 4W7
| | | | | | | | | | | | | |
Collapse
|
66
|
Li XC, Carretero OA, Navar LG, Zhuo JL. AT1 receptor-mediated accumulation of extracellular angiotensin II in proximal tubule cells: role of cytoskeleton microtubules and tyrosine phosphatases. Am J Physiol Renal Physiol 2006; 291:F375-83. [PMID: 16478976 PMCID: PMC2276851 DOI: 10.1152/ajprenal.00405.2005] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Long-term angiotensin II (ANG II) administration is associated with increased ANG II accumulation in the kidney, but intrarenal compartment(s) involved in this response remains to be determined. We tested the hypothesis that 1) extracellular ANG II is taken up by proximal tubule cells (PTCs) through AT(1) receptor-mediated endocytosis, 2) this process is regulated by cytoskeleton microtubule- and tyrosine phosphatase-dependent mechanisms, and 3) AT(1) receptor-mediated endocytosis of ANG II has a functional relevance by modulating intracellular cAMP signaling. In cultured PTCs, [(125)I]Tyr-labeled ANG II and fluorescein labeled-ANG II were internalized in a time-dependent manner and colocalized with the endosome marker Alexa Fluor 594-transferrin. Endocytosis of extracellular ANG II was inhibited by the AT(1) receptor blocker losartan (16.5 +/- 4.6%, P < 0.01 vs. ANG II, 78.3 +/- 6.2%) and by the tyrosine phosphatase inhibitor phenylarsine oxide (PAO; 30.0 +/- 3.5%, P < 0.05 vs. ANG II). Intracellular ANG II levels were increased by approximately 58% (basal, 229.8 +/- 11.4 vs. ANG II, 361.3 +/- 11.8 pg ANG II/mg protein, P < 0.01), and the responses were blocked by losartan (P < 0.01), the cytoskeleton microtubule inhibitor colchicine (P < 0.05), and PAO (P < 0.01), whereas depletion of clathrin-coated pits with hyperosmotic sucrose had no effect (356.1 +/- 25.5 pg ANG II/mg protein, not significant). ANG II accumulation was associated with significant inhibition of both basal (control, 15.5 +/- 2.8 vs. ANG II, 9.1 +/- 2.4 pmol/mg protein, P < 0.05) and forskolin-stimulated cAMP signaling (forskolin, 68.7 +/- 8.6 vs. forskolin + ANG II, 42.8 +/- 13.8 pmol/mg protein, P < 0.01). These effects were blocked by losartan and PAO. We conclude that extracellular ANG II is internalized in PTCs through AT(1) receptor-mediated endocytosis and that internalized ANG II may play a functional role in proximal tubule cells by inhibiting intracellular cAMP signaling.
Collapse
MESH Headings
- Angiotensin II/metabolism
- Angiotensin II/pharmacology
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Arsenicals/pharmacology
- Cells, Cultured
- Colchicine/pharmacology
- Colforsin/pharmacology
- Cyclic AMP/physiology
- Cytoskeleton/drug effects
- Cytoskeleton/metabolism
- Endocytosis/physiology
- Enzyme Inhibitors/pharmacology
- Gene Expression Regulation/physiology
- Imidazoles/pharmacology
- Kidney Tubules, Proximal/cytology
- Kidney Tubules, Proximal/metabolism
- Losartan/pharmacology
- Microtubules/drug effects
- Microtubules/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 1
- Protein Tyrosine Phosphatases/antagonists & inhibitors
- Protein Tyrosine Phosphatases/physiology
- Pyridines/pharmacology
- Rabbits
- Receptor, Angiotensin, Type 1/drug effects
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/physiology
- Receptor, Angiotensin, Type 2/analysis
- Receptor, Angiotensin, Type 2/drug effects
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/physiology
- Signal Transduction/physiology
- Sucrose/pharmacology
Collapse
Affiliation(s)
- Xiao C Li
- Laboratory of Receptor and Signal Transduction, Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit 48202, USA
| | | | | | | |
Collapse
|
67
|
Pendergrass KD, Averill DB, Ferrario CM, Diz DI, Chappell MC. Differential expression of nuclear AT1 receptors and angiotensin II within the kidney of the male congenic mRen2. Lewis rat. Am J Physiol Renal Physiol 2006; 290:F1497-506. [PMID: 16403834 DOI: 10.1152/ajprenal.00317.2005] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We established a new congenic model of hypertension, the mRen(2). Lewis rat and assessed the intracellular expression of angiotensin peptides and receptors in the kidney. The congenic strain was established from the backcross of the (mRen2)27 transgenic rat that expresses the mouse renin 2 gene onto the Lewis strain. The 20-wk-old male congenic rats were markedly hypertensive compared with the Lewis controls (systolic blood pressure: 195 +/- 2 vs. 107 +/- 2 mmHg, P < 0.01). Although plasma ANG II levels were not different between strains, circulating levels of ANG-(1-7) were 270% higher and ANG I concentrations were 40% lower in the mRen2. Lewis rats. In contrast, both cortical (CORT) and medullary (MED) ANG II concentrations were 60% higher in the mRen2. Lewis rats, whereas tissue ANG I was 66 and 84% lower in CORT and MED. For both strains, MED ANG II, ANG I, and ANG-(1-7) were significantly higher than CORT levels. Intracellular ANG II binding distinguished nuclear (NUC) and plasma membrane (PM) receptor using the ANG II radioligand 125I-sarthran. Isolated CORT nuclei exhibited a high density (Bmax >200 fmol/mg protein) and affinity for the sarthran ligand (KD<0.5 nM); the majority of these sites (>95%) were the AT1 receptor subtype. CORT ANG II receptor Bmax and KD values in nuclei were 75 and 50% lower, respectively, for the mRen2. Lewis vs. the Lewis rats. In the MED, the PM receptor density (Lewis: 50 +/- 4 vs. mRen2. Lewis: 21 +/- 5 fmol/mg protein) and affinity (Lewis: 0.31 +/- 0.1 vs. 0.69 +/- 0.1 nM) were lower in the mRen2. Lewis rats. In summary, the hypertensive mRen2. Lewis rats exhibit higher ANG II in both CORT and MED regions of the kidney. Evaluation of intracellular ANG II receptors revealed lower CORT NUC and MED PM AT1 sites in the mRen2. Lewis. The downregulation of AT1 sites in the mRen2. Lewis rats may reflect a compensatory response to dampen the elevated levels of intrarenal ANG II.
Collapse
Affiliation(s)
- Karl D Pendergrass
- Hypertension and Vascular Disease Ctr., Wake Forest Univ. Health Sciences, Medical Center Blvd., Winston-Salem, NC 27157-1095, USA
| | | | | | | | | |
Collapse
|
68
|
Linz P, Amann K, Freisinger W, Ditting T, Hilgers KF, Veelken R. Sensory neurons with afferents from hind limbs: enhanced sensitivity in secondary hypertension. Hypertension 2006; 47:527-31. [PMID: 16401763 DOI: 10.1161/01.hyp.0000199984.78039.36] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sensory nerve fibers from the dorsal root ganglia (DRG) may contribute to the regulation of peripheral vascular resistance. Axons of DRG neurons of the lower thoracic cord project mainly to resistance vessels in the lower limbs, likely opposing the vasoconstrictor effects of the sympathetic activity. This mechanism might be of importance in hypertension with increased sympathetic activity. We tested the hypothesis that sensory neurons of the DRG in the lower thoracic cord show an altered sensitivity to mechanical stimuli in hypertension. Neurons from DRG (T11 to L1) of rats with hypertension (2 kidney-1 clip hypertensive rats and 5 of 6 nephrectomized rats) were cultured on coverslips. Current time relationships were established with whole-cell patch recordings. Cells were characterized under control conditions and after exposure to hypoosmotic solutions to induce mechanical stress. Neurons with projections to the kidney were studied for comparison. The hypoosmotic extracellular medium induced a significant change in conductance of the cells in all of the groups of rats. In hypertensive rats, responses of cells with hindlimb axons were significantly different from controls: (2 kidney-1 clip hypertensives: delta-351+/-52 pA and 5 of 6 nephrectomized rats: delta-372+/-43 pA versus controls: delta-190+/-25 pA; P<0.05). Responses of DRG cells with renal afferents to mechanical stress were unaffected. Neurons from DRG in the lower thoracic cord with projections to the lower limbs exhibited an increased sensitivity to mechanical stress. We speculate that this observation may indicate an increased activity of these neurons, their axons, and neurotransmitters in the control of resistance vessels in hypertension.
Collapse
Affiliation(s)
- Peter Linz
- Department of Internal Medicine 4/Nephrology and Hypertension, University of Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
69
|
Zhuo JL, Li XC, Garvin JL, Navar LG, Carretero OA. Intracellular ANG II induces cytosolic Ca2+ mobilization by stimulating intracellular AT1 receptors in proximal tubule cells. Am J Physiol Renal Physiol 2005; 290:F1382-90. [PMID: 16380461 PMCID: PMC2276850 DOI: 10.1152/ajprenal.00269.2005] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Intracellular ANG II induces biological effects in nonrenal cells, but it is not known whether it plays a physiological role in renal proximal tubule cells (PTCs). PTCs express angiotensinogen, renin, and angiotensin-converting enzyme mRNAs, suggesting the presence of high levels of intracellular ANG II. We determined if microinjection of ANG II directly in single PTCs increases intracellular calcium concentration ([Ca2+]i) and, if so, elucidated the cellular mechanisms involved. Changes in [Ca2+]i responses were studied by fluorescence imaging using the Ca2+ indicator fluo 3. ANG II (1 nM) was microinjected directly in the cells, whereas cell-surface angiotensin type 1 (AT1) receptors were blocked by losartan (10 microM). When ANG II (1 nM) was added to the perfusate, there was a marked increase in [Ca2+]i that was blocked by extracellular losartan. With losartan in the perfusate, intracellular microinjection of ANG II elicited a robust increase in cytoplasmic [Ca2+]i that peaked at 30 s (basal: 2.2 +/- 0.3 vs. ANG II: 14.9 +/- 0.4 relative fluorescence units; P < 0.01). Chelation of extracellular Ca2+ with EGTA (2 mM) did not alter microinjected ANG II-induced [Ca2+]i responses (Ca2+ free + ANG II: 12.3 +/- 2.6 relative fluorescence units, not significant vs. ANG II); however, pretreatment with thapsigargin to deplete intracellular Ca2+ stores or with U-73122 to inhibit phospholipase C (1 microM each) markedly attenuated microinjected ANG II-induced [Ca2+]i responses. Combined microinjection of ANG II and losartan abolished [Ca2+]i responses, whereas a combination of ANG II and PD-123319 had no effect. These data demonstrate for the first time that direct microinjection of ANG II in single PTCs increases [Ca2+]i by stimulating intracellular AT1 receptors and releases Ca2+ from intracellular stores, suggesting that intracellular ANG II may play a physiological role in PTC function.
Collapse
MESH Headings
- Angiotensin II/analysis
- Angiotensin II/pharmacology
- Animals
- Blotting, Western
- Calcium/metabolism
- Calcium/pharmacology
- Cells, Cultured
- Cytosol/metabolism
- Enzyme Activation/drug effects
- Kidney Tubules, Proximal/chemistry
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/ultrastructure
- Microinjections
- Rabbits
- Receptor, Angiotensin, Type 1/analysis
- Receptor, Angiotensin, Type 1/drug effects
- Receptor, Angiotensin, Type 1/physiology
- Receptor, Angiotensin, Type 2/drug effects
- Receptor, Angiotensin, Type 2/physiology
- Thapsigargin/pharmacology
- Type C Phospholipases/metabolism
Collapse
Affiliation(s)
- Jia L Zhuo
- Division of Hypertension and Vascular Research, Henry Ford Hospital, 2799 West Grand Blvd., Detroit, MI 48202, USA.
| | | | | | | | | |
Collapse
|
70
|
Sadjadi J, Kramer GL, Yu CH, Welborn MB, Modrall JG. Angiotensin II Exerts Positive Feedback on the Intrarenal Renin-Angiotensin System by an Angiotensin Converting Enzyme-Dependent Mechanism1. J Surg Res 2005; 129:272-7. [PMID: 15992826 DOI: 10.1016/j.jss.2005.04.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2005] [Revised: 04/25/2005] [Accepted: 04/26/2005] [Indexed: 10/25/2022]
Abstract
BACKGROUND Plasma angiotensin II (ANG II) is not increased significantly in renovascular hypertension (RVH), but tissue ANG II levels are elevated in both kidneys of renovascular rats. Because the contralateral, non-ischemic kidney is critical for maintenance of hypertension in RVH, this study sought to understand the mechanism by which intrarenal ANG II levels are augmented in the non-ischemic kidney. This study tested the hypothesis that an incremental increase in plasma ANG II induces the intrarenal renin-angiotensin system (RAS) in the non-ischemic kidney by an angiotensin converting enzyme (ACE) dependent mechanism. METHODS To simulate the incremental increase in plasma ANG II induced by the ischemic kidney in RVH, an ANG II infusion model was used. This model used a chronic infusion of ANG II (40 ng/min) or vehicle by osmotic minipump into uninephrectomized rats. Parallel groups were treated with the ACE inhibitor Enalaprilat (200 mg/kg/day). Intrarenal ACE activity was measured by radioenzymatic assay. ANG II levels were quantified by radioimmunoassay. RESULTS Hypertension was evident in ANG II-infused rats, compared to control rats (155 +/- 4 versus 112 +/- 1 mmHg; P < 0.001). Concurrent treatment with Enalaprilat reversed the hypertension induced by ANG II infusion (98 +/- 3 versus 155 +/- 4 mmHg; P < 0.001). ANG II up-regulated intrarenal ACE activity in the non-ischemic kidney (59.2 +/- 11.9 versus 25.2 +/- 6.8 units/mg protein; P < 0.01). Enalaprilat significantly decreased renal ACE activity in ANG II-treated rats, compared to ANG II alone (11.4 +/- 1.0 versus 59.2 +/- 11.9 units/mg protein; P < 0.001). Intrarenal ANG II was increased in ANG II-infused rats, compared to control animals (52.9 +/- 7.1 versus 23.0 +/- 3.2 fmol/mg tissue; P < 0.001), and Enalaprilat prevented ANG II-induced increases in intrarenal ANG II (29.9 +/- 2.6 versus 52.9 +/- 7.1 fmol/mg tissue; P < 0.05). CONCLUSION Incremental changes in plasma ANG II induce de novo production of ANG II in the non-ischemic kidney to augment intrarenal ANG II content. ACE inhibition blocks this positive feedback loop, suggesting that ANG II activates the intrarenal RAS by an ACE-dependent mechanism. The impact of ACE inhibition on blood pressure suggests that this feedback loop may be an important mechanism for maintenance of hypertension in RVH.
Collapse
Affiliation(s)
- Javid Sadjadi
- Department of Surgery, Division of Vascular Surgery, Dallas Veterans Affairs Medical Center and the University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, USA
| | | | | | | | | |
Collapse
|
71
|
Sadjadi J, Kramer GL, Yu CH, Burress Welborn M, Chappell MC, Gregory Modrall J. Angiotensin converting enzyme-independent angiotensin ii production by chymase is up-regulated in the ischemic kidney in renovascular hypertension. J Surg Res 2005; 127:65-9. [PMID: 15869764 DOI: 10.1016/j.jss.2005.02.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2003] [Revised: 02/27/2005] [Accepted: 02/28/2005] [Indexed: 11/19/2022]
Abstract
BACKGROUND Tissue angiotensin II (ANG II) levels are elevated in both kidneys in renovascular hypertension (RVH). It has been demonstrated previously that intrarenal ANG II is augmented by an angiotensin converting enzyme (ACE) dependent mechanism in the non-ischemic kidney, but the role of ACE-independent production of ANG II in the kidney by the enzyme chymase is unknown. This study tested the hypothesis that intrarenal chymase activity is up-regulated in RVH. METHODS A two-kidney, one-clip (2K1C) rat model was used to induce RVH (n = 6 rats/group). Regulation of intrarenal chymase activity by plasma ANG II was investigated using an ANG II-infusion model. At sacrifice 14 days post-operatively, steady-state ANG II levels in plasma and kidney were quantified by radioimmunoassay. ANG II production was quantified in kidney homogenates by incubating at 37 degrees C for 60 min with enzyme substrate (200 microm ANG I) alone or substrate containing the chymase inhibitor chymostatin. ANG II was separated and quantitated by HPLC. Chymase activity was defined as the fraction of ANG II production inhibited by Chymostatin. RESULTS 2K1C and ANG II-infused rats developed significant hypertension, compared to control rats (P = 0.0001 and P = 0.001, respectively). Chymase-dependent ANG II production was increased in the ischemic kidney, but not the non-ischemic kidney, of 2K1C rats compared to control animals (*P < 0.05). Intrarenal chymase activity was unchanged by ANG II infusion (P = NS). CONCLUSIONS Chymase activity is up-regulated in the ischemic kidney of 2K1C rats. Plasma ANG II does not appear to regulate intrarenal chymase activity, suggesting that ischemia per se up-regulates chymase activity in the kidney. ACE-independent ANG II production by chymase may provide a mechanism for augmenting intrarenal ANG II in the ischemic kidney in RVH.
Collapse
Affiliation(s)
- Javid Sadjadi
- Department of Surgery, Division of Vascular Surgery, Dallas Veterans Affairs Medical Center and the University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | | | | | |
Collapse
|
72
|
Banday AA, Siddiqui AH, Menezes MM, Hussain T. Insulin treatment enhances AT1receptor function in OK cells. Am J Physiol Renal Physiol 2005; 288:F1213-9. [PMID: 15713908 DOI: 10.1152/ajprenal.00361.2003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increased renal sodium retention is considered a major risk factor contributing to hypertension associated with chronic hyperinsulinemia and obesity. However, the molecular mechanism involved is not understood. The present study investigates the effect of insulin treatment on AT1receptor expression and ANG II-induced stimulation of Na/H exchanger (NHE) and Na-K-ATPase (NKA) in opossum kidney (OK) cells, a proximal tubule cell line. The presence of the AT1receptors in OK cells was confirmed by the specific binding of125I-sar-ANG II and by detecting ∼43-kDa protein on Western blot analysis with AT1receptor antibody and blocking peptide as well as by expression of AT1receptor mRNA as determined by RT-PCR. Insulin treatment (100 nM for 24 h) caused an increase in125I-sar-ANG II binding, AT1receptor protein content, and mRNA levels. The whole cell lysate and membrane showed similar insulin-induced increase in the AT1receptor protein expression, which was blocked by genistein (100 nM), a tyrosine kinase inhibitor, and cycloheximide (1.5 μg/ml), a protein synthesis inhibitor. Determination of ethyl isopropyl amiloride-sensitive22Na+uptake, a measure of the NHE activity, revealed that ANG II (1–100 pM)-induced stimulation of NHE in insulin-treated cells was significantly greater than in the control cells. Similarly, ANG II (1–100 pM)-induced stimulation of ouabain-sensitive86Rb+uptake, a measure of NKA activity in insulin-treated cells, was significantly greater than in the control cells. ANG II stimulation of both the transporters was blocked by AT1receptor antagonist losartan, suggesting the involvement of AT1receptors. Thus chronic insulin treatment causes upregulation of AT1receptors, which evoked ANG II-induced stimulation of NHE and NKA. We propose that insulin-induced increase in the renal AT1receptor function serves as a mechanism responsible for the increased renal sodium reabsorption and thus may contribute to development of hypertension in conditions associated with hyperinsulinemia.
Collapse
Affiliation(s)
- Anees Ahmad Banday
- Heart and Kidney Institute, College of Pharmacy, University of Houston, TX 77204, USA
| | | | | | | |
Collapse
|
73
|
Zeng C, Yang Z, Wang Z, Jones J, Wang X, Altea J, Mangrum AJ, Hopfer U, Sibley DR, Eisner GM, Felder RA, Jose PA. Interaction of Angiotensin II Type 1 and D
5
Dopamine Receptors in Renal Proximal Tubule Cells. Hypertension 2005; 45:804-10. [PMID: 15699451 DOI: 10.1161/01.hyp.0000155212.33212.99] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Angiotensin II type 1 (AT
1
) receptor and D
1
and D
3
dopamine receptors directly interact in renal proximal tubule (RPT) cells from normotensive Wistar-Kyoto rats (WKY). There is indirect evidence for a D
5
and AT
1
receptor interaction in WKY and spontaneously hypertensive rats (SHR). Therefore, we sought direct evidence of an interaction between AT
1
and D
5
receptors in RPT cells. D
5
and AT
1
receptors colocalized in WKY cells. Angiotensin II decreased D
5
receptors in WKY cells in a time- and concentration-dependent manner (EC
50
=2.7×10
−9
M; t
1/2
=4.9 hours), effects that were blocked by an AT
1
receptor antagonist (losartan). In SHR, angiotensin II (10
−8
M/24 hours) also decreased D
5
receptors (0.96±0.08 versus 0.72±0.08; n=12) and to the same degree as in WKY cells (1.44±0.07 versus 0.92±0.08). However, basal D
5
receptors were decreased in SHR RPT cells (SHR 0.96±0.08; WKY 1.44±0.07; n=12 per strain;
P
<0.05) and renal brush border membranes of SHR compared with WKY (SHR 0.54±0.16 versus WKY 1.46±0.10; n=5 per strain;
P
<0.05). Angiotensin II decreased AT
1
receptor expression in WKY (1.00±0.04 versus 0.72±0.08; n=8;
P
<0.05) but increased it in SHR (0.96±0.04 versus 1.32±0.08; n=8;
P
<0.05). AT
1
and D
5
receptors also interacted in vivo; renal D
5
receptor protein was higher in mice lacking the AT
1A
receptor (AT
1A
−/−; 1.61±0.31; n=6) than in wild-type littermates used as controls (AT
1A
+/+; 0.81±0.08; n=6;
P
<0.05), and renal cortical AT
1
receptor protein was higher in D
5
receptor null mice than in wild-type littermates (1.18±0.08 versus 0.84±0.07; n=4;
P
<0.05). We conclude that D
5
and AT
1
receptors interact with each other. Altered interactions between AT
1
and dopamine receptors may play a role in the pathogenesis of hypertension.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Cells, Cultured
- Drug Interactions
- Kidney Tubules, Proximal/cytology
- Kidney Tubules, Proximal/metabolism
- Mice
- Mice, Inbred Strains
- Mice, Knockout
- Rats
- Rats, Inbred SHR
- Rats, Inbred WKY
- Receptor, Angiotensin, Type 1/deficiency
- Receptor, Angiotensin, Type 1/drug effects
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, Dopamine D1/antagonists & inhibitors
- Receptors, Dopamine D1/deficiency
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D5
- Tissue Distribution
Collapse
Affiliation(s)
- Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, P.R. China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Gilbert JS, Lang AL, Grant AR, Nijland MJ. Maternal nutrient restriction in sheep: hypertension and decreased nephron number in offspring at 9 months of age. J Physiol 2005; 565:137-47. [PMID: 15790663 PMCID: PMC1464480 DOI: 10.1113/jphysiol.2005.084202] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Pregnant ewes were fed either a 50% nutrient-restricted (NR; n= 8) or a control 100% (C; n= 8) diet from day 28 to day 78 of gestation (dGA; term = 150 dGA). Lambs were born naturally, and fed to appetite throughout the study period. At 245 +/- 1 days postnatal age (DPNA), offspring were instrumented for blood pressure measurements, with tissue collection at 270 DPNA. Protein expression was assessed using Western blot, glomerulus number determined via acid maceration and hormone changes by radioimmunoassay (RIA) or enzyme-linked immunosorbent assay (ELISA). NR lambs had higher mean arterial pressure (MAP; 89.0 +/- 6.6 versus 73.4 +/- 1.6 mmHg; P < 0.05), fewer renal glomeruli (57.8 +/- 23.8 versus 64.6 +/- 19.3 x 10(4); P < 0.05), increased expression of angiotensin converting enzyme (ACE) in the renal cortex (942 +/- 130 versus 464 +/- 60 arbitrary pixel units (apu); P < 0.03), and increased angiotensin II receptor AT2 expression in the renal medulla (63.3 +/- 12.1 versus 19.5 +/- 44.2 x 10(4) apu; P < 0.03). All data are presented as mean +/-S.E.M. The present data indicate that global maternal nutrient restriction (50%) during early to mid-gestation impairs renal nephrogenesis, increases MAP, and alters expression of AT2 and ACE without an associated change in birth weight. These data demonstrate the existence of a critical window of fetal susceptibility during early to mid-gestation that alters kidney development and blood pressure regulation in later life.
Collapse
Affiliation(s)
- Jeffrey S Gilbert
- Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, TX 78229-3900, USA
| | | | | | | |
Collapse
|
75
|
Abstract
PURPOSE OF REVIEW Recent studies have reported that intrarenal angiotensin II content and angiotensin II concentrations in the proximal tubular fluid and renal interstitial fluid are much greater than the circulating angiotensin II levels. These high intrarenal angiotensin II levels are responsible for regulating renal hemodynamics and tubular transport. RECENT FINDINGS Intrarenal angiotensin II levels have been assessed from total tissue contents as well as renal interstitial fluid and proximal tubular fluid concentrations. Total tissue contents expressed per gram of tissue weight are greater than plasma angiotensin II concentrations; tubular fluid concentrations and renal interstitial fluid concentrations are even greater in the range of 3-10 pmoles/ml. In hypertensive states, there is also an increased intracellular accumulation of angiotensin II mediated by angiotensin type 1 receptor-dependent endocytosis. The high intrarenal angiotensin II levels are also caused by the presence of angiotensinogen messenger RNA and protein in the proximal tubule cells. Furthermore, there is positive amplification by which increases in circulating angiotensin II stimulate increased production and secretion of angiotensinogen, which is also manifested as an increased urinary excretion rate. SUMMARY The ability of the kidney to generate high intratubular and interstitial concentrations allows the kidney to regulate intrarenal levels in accord with the homeostatic needs for the regulation of renal hemodynamics and tubular reabsorption and the regulation of sodium balance. When inappropriately stimulated, high intrarenal angiotensin II levels contribute to excessive salt and water retention, the development of hypertension, and long-term proliferative effects leading to renal injury.
Collapse
Affiliation(s)
- L Gabriel Navar
- Department of Physiology and Hypertension, Renal Center of Excellence, Tulane University Health Scences Center, New Orleans, Louisiana 70112, USA.
| | | |
Collapse
|
76
|
Nishiyama A, Yoshizumi M, Rahman M, Kobori H, Seth DM, Miyatake A, Zhang GX, Yao L, Hitomi H, Shokoji T, Kiyomoto H, Kimura S, Tamaki T, Kohno M, Abe Y. Effects of AT1 receptor blockade on renal injury and mitogen-activated protein activity in Dahl salt-sensitive rats. Kidney Int 2004; 65:972-81. [PMID: 14871417 PMCID: PMC2573027 DOI: 10.1111/j.1523-1755.2004.00476.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND The mitogen-activated protein kinase (MAPK) cascade is an important intracellular mediator of angiotensin II (Ang II)-induced cell growth and differentiation. Here, we examined the effect of angiotensin II type 1 receptor (AT1) receptor blockade on renal injury and MAPK activity in Dahl salt-sensitive (DS) rats. METHODS DS rats were maintained on a high (H: 8.0%NaCl, N= 8) or low (L: 0.3%NaCl, N= 7) salt diet, or H + candesartan cilexetil (10 to 15 mg/kg/day, N= 8). Urinary protein excretion (UproteinV), renal cortical collagen content, and glomerular injury (assessed by semiquantitative morphometric analysis) were determined after 4-week treatments. Plasma and kidney Ang II levels were measured by radioimmunoassay. Protein levels of AT1 and AT2 receptors in the renal cortical tissues were analyzed by Western-blotting analyses. MAPKs activities, including extracellular signal-regulated kinases (ERK)1/2, c-Jun NH2-terminal kinases (JNK), p38 MAPK, and Big-MAPK-1 (BMK1), were measured by Western-blotting analyses or in vitro kinase assays. RESULTS DS/H rats showed higher mean blood pressure (MBP), UproteinV, and renal cortical collagen content than DS/L rats. Increased ERK1/2, JNK, and BMK1 activities were observed in renal cortical tissues of DS/H rats (approximately 6.3-, 4.5-, and 2.5-fold, respectively), whereas p38 MAPK activity was unchanged. Plasma Ang II levels were significantly reduced in DS/H rats compared with DS/L rats, whereas kidney Ang II contents and AT1 receptor protein levels were similar. Candesartan did not alter MBP, but significantly reduced UproteinV and collagen content, and ameliorated progressive sclerotic and proliferative glomerular changes. Furthermore, candesartan decreased renal tissue Ang II contents (from 216 +/- 19 to 46 +/- 3 fmol/mL) and ERK1/2, JNK, and BMK1 activities (-45%, -60%, and -70%, respectively) in DS/H rats. CONCLUSION In DS hypertensive rats, some of the renoprotective effects of AT1 receptor blockade are accompanied by reductions in intrarenal Ang II contents and MAPK activity, which might not be mediated through arterial pressure changes.
Collapse
Affiliation(s)
- Akira Nishiyama
- Department of Pharmacology, Research Equipment Center, Kagawa Medical University, Kagawa, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Kopp UC, Cicha MZ, Nakamura K, Nüsing RM, Smith LA, Hökfelt T. Activation of EP4 receptors contributes to prostaglandin E2-mediated stimulation of renal sensory nerves. Am J Physiol Renal Physiol 2004; 287:F1269-82. [PMID: 15292051 DOI: 10.1152/ajprenal.00230.2004] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Induction of cyclooxygenase-2 (COX-2) in the renal pelvic wall increases prostaglandin E(2) (PGE(2)) leading to stimulation of cAMP production, which results in substance P (SP) release and activation of renal mechanosensory nerves. The subtype of PGE receptors involved, EP2 and/or EP4, was studied by immunohistochemistry and renal pelvic administration of agonists and antagonists of EP2 and EP4 receptors. EP4 receptor-like immunoreactivity (LI) was colocalized with calcitonin gene-related peptide (CGRP)-LI in dorsal root ganglia (DRGs) at Th(9)-L(1) and in nerve terminals in the renal pelvic wall. Th(9)-L(1) DRG neurons also contained EP3 receptor-LI and COX-2-LI, each of which was colocalized with CGRP-LI in some neurons. No renal pelvic nerves contained EP3 receptor-LI and only very few nerves COX-2-LI. The EP1/EP2 receptor antagonist AH-6809 (20 microM) had no effect on SP release produced by PGE(2) (0.14 microM) from an isolated rat renal pelvic wall preparation. However, the EP4 receptor antagonist L-161,982 (10 microM) blocked the SP release produced by the EP2/EP4 receptor agonist butaprost (10 microM) 12 +/- 2 vs. 2 +/- 1 and PGE(2), 9 +/- 1 vs. 1 +/- 0 pg/min. The SP release by butaprost and PGE(2) was similarly blocked by the EP4 receptor antagonist AH-23848 (30 microM). In anesthetized rats, the afferent renal nerve activity (ARNA) responses to butaprost 700 +/- 100 and PGE(2).780 +/- 100%.s (area under the curve of ARNA vs. time) were unaffected by renal pelvic perfusion with AH-6809. However, 1 microM L-161,982 and 10 microM AH-23848 blocked the ARNA responses to butaprost by 94 +/- 5 and 78 +/- 10%, respectively, and to PGE(2) by 74 +/- 16 and 74 +/- 11%, respectively. L-161,982 also blocked the ARNA response to increasing renal pelvic pressure 10 mmHg, 85 +/- 5%. In conclusion, PGE(2) increases renal pelvic release of SP and ARNA by activating EP4 receptors on renal sensory nerve fibers.
Collapse
MESH Headings
- Afferent Pathways/drug effects
- Afferent Pathways/physiology
- Alprostadil/analogs & derivatives
- Alprostadil/pharmacology
- Animals
- Calcitonin Gene-Related Peptide/analysis
- Cyclic AMP/biosynthesis
- Cyclooxygenase 2
- Dinoprostone/pharmacology
- Fluorescent Antibody Technique
- Ganglia, Spinal/chemistry
- Immunohistochemistry
- Isoenzymes/analysis
- Kidney/chemistry
- Kidney/innervation
- Male
- Prostaglandin-Endoperoxide Synthases/analysis
- Prostaglandins E, Synthetic
- Rats
- Rats, Sprague-Dawley
- Receptors, Prostaglandin E/analysis
- Receptors, Prostaglandin E/antagonists & inhibitors
- Receptors, Prostaglandin E/physiology
- Receptors, Prostaglandin E, EP1 Subtype
- Receptors, Prostaglandin E, EP2 Subtype
- Receptors, Prostaglandin E, EP4 Subtype
- Substance P/metabolism
Collapse
Affiliation(s)
- Ulla C Kopp
- Department of Internal Medicine, Department of Veterans Affairs Medical Center and University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | | | | | | | | | | |
Collapse
|
78
|
Prieto-Carrasquero MC, Harrison-Bernard LM, Kobori H, Ozawa Y, Hering-Smith KS, Hamm LL, Navar LG. Enhancement of collecting duct renin in angiotensin II-dependent hypertensive rats. Hypertension 2004; 44:223-9. [PMID: 15226276 PMCID: PMC2628717 DOI: 10.1161/01.hyp.0000135678.20725.54] [Citation(s) in RCA: 185] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Distal nephron renin may provide a possible pathway for angiotensin (Ang) I generation from proximally delivered angiotensinogen. To examine the effects of Ang II on distal nephron renin, we compared renin protein and mRNA expression in control and Ang II-infused rats. Kidneys from sham (n=9) and Ang II-infused (80 ng/kg per minute, 13 days, n=10) Sprague-Dawley rats were processed by immunohistochemistry, Western blot, reverse transcriptase-polymerase chain reaction (RT-PCR), and quantitative real-time RT-PCR. Ang II infusion increased systolic blood pressure (181+/-4 versus 115+/-5 mm Hg) and suppressed plasma and kidney cortex renin activity. Renin immunoreactivity was suppressed in juxtaglomerular apparatus (JGA) cells in Ang II-infused rats compared with sham (0.1+/-0.1 versus 1.0+/-0.1 relative ratio) but increased in distal nephron segments (6.4+/-1.4 versus 1.0+/-0.1 cortex; 2.5+/-0.3 versus 1.0+/-0.2 medulla). Tubular renin immunostaining was apically distributed in principal cells colocalizing with aquaporin-2 in connecting tubules and cortical and medullary collecting ducts. Renin protein levels were decreased in the kidney cortex of Ang II-infused rats compared with that of sham (0.4+/-0.2 versus 1.0+/-0.4) rats but higher in the kidney medulla (1.2+/-0.4 versus 1.0+/-0.1). In kidney medulla, RT-PCR and quantitative real-time PCR showed similar levels of renin transcript in both groups. In summary, the detection of renin mRNA in the renal medulla, which is devoid of JGA, indicates local synthesis rather than an uptake of JGA renin. In contrast to the inhibitory effect of Ang II on JGA renin, Ang II infusion stimulates renin protein expression in collecting ducts and maintains renin transcriptional levels in the medulla, which may contribute to the increased intrarenal Ang II levels in Ang II-dependent hypertension.
Collapse
Affiliation(s)
- Minolfa C Prieto-Carrasquero
- Department of Physiology (SL39), Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112-2699, USA.
| | | | | | | | | | | | | |
Collapse
|
79
|
Nishiyama A, Seth DM, Navar LG. Angiotensin II type 1 receptor-mediated augmentation of renal interstitial fluid angiotensin II in angiotensin II-induced hypertension. J Hypertens 2004; 21:1897-903. [PMID: 14508196 DOI: 10.1097/00004872-200310000-00017] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Angiotensin II (Ang II)-dependent hypertension is associated with augmented intrarenal concentrations of Ang II; however, the distribution of the increased intrarenal Ang II has not been fully established. OBJECTIVE To determine the changes in renal interstitial fluid Ang II concentrations in Ang II-induced hypertension and the consequences of treatment with an angiotensin II type 1 (AT1) receptor blocker. DESIGN AND METHODS Rats were selected to receive vehicle (5% acetic acid subcutaneously; n = 6), Ang II (80 ng/min subcutaneously, via osmotic minipump; n = 7) or Ang II plus an AT1 receptor antagonist, candesartan cilexetil (10 mg/kg per day, in drinking water; n = 6) for 13-14 days, at which time, experiments were performed on anesthetized rats. Microdialysis probes were implanted in the renal cortex and were perfused at 2 microl/min. The effluent dialysate concentrations of Ang I and Ang II were measured by radioimmunoassay and reported values were corrected for the equilibrium rates at this perfusion rate. RESULTS Ang II-infused rats developed greater mean arterial pressures (155 +/- 7 mmHg) than vehicle-infused rats (108 +/- 3 mmHg). Ang II-infused rats showed greater plasma (181 +/- 30 fmol/ml) and kidney (330 +/- 38 fmol/g) Ang II concentrations than vehicle-infused rats (98 +/- 14 fmol/ml and 157 +/- 22 fmol/g, respectively). Renal interstitial fluid Ang II concentrations were much greater than plasma concentrations, averaging 5.74 +/- 0.26 pmol/ml in Ang II-infused rats - significantly greater than those in vehicle-infused rats (2.86 +/- 0.23 pmol/ml). Candesartan treatment prevented the hypertension (87 +/- 3 mmHg) and led to increased plasma Ang II concentrations (441 +/- 27 fmol/ml), but prevented increases in kidney (120 +/- 15 fmol/g) and renal interstitial fluid (2.15 +/- 0.12 pmol/ml) Ang II concentrations. CONCLUSIONS These data indicate that Ang II-infused rats develop increased renal interstitial fluid concentrations of Ang II, which may contribute to the increased vascular resistance and reduced sodium excretion. Furthermore, the augmentation of renal interstitial fluid Ang II is the result of an AT1 receptor-mediated process and can be dissociated from the plasma concentrations.
Collapse
Affiliation(s)
- Akira Nishiyama
- Department of Pharmacology, Kagawa Medical University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | | | | |
Collapse
|
80
|
Pueyo ME, Challah M, Gauguier D, Louedec L, Philippe M, Gaertner R, Marre M, Michel JB, Jacob MP. Transforming growth factor-beta 1 production is correlated with genetically determined ACE expression in congenic rats: a possible link between ACE genotype and diabetic nephropathy. Diabetes 2004; 53:1111-8. [PMID: 15047629 DOI: 10.2337/diabetes.53.4.1111] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Genetic background appears to modulate the development of diabetic vascular complications. In particular, polymorphisms in the ACE gene have been associated with diabetic nephropathy and, in some studies, macrovascular complications. However, the links between ACE gene polymorphism and factors implicated in diabetes complications remain unknown. The aim of this study was to determine whether the ACE genotype could modify factors, such as transforming growth factor (TGF)-beta 1, involved in the complications of diabetes. For this purpose, congenic rats (L.BNAce10), differing from the LOU strain in only a small segment of chromosome 10 containing the ACE locus, were generated. These congenic rats have plasma ACE levels twice as high as the donor strain. Diabetes was induced in rats of both strains, and its effects on ACE and TGF-beta 1 expressions were evaluated in lungs and kidneys. In lung, the main source of ACE production, ACE mRNA levels and activity were higher in L.BNAce10 rats than in LOU rats. Diabetes increased ACE lung expression in rats of both strains in a similar manner. TGF-beta 1 expression was also higher in lungs of L.BNAce10 compared with LOU rats and was also increased by diabetes. Furthermore, a strong correlation was found between TGF-beta 1 and ACE expressions. In renal arterioles, ACE and TGF-beta mRNA expressions were higher in L.BNAce10 rats than LOU rats (both diabetic and nondiabetic). In these vessels, there was also a correlation between ACE and TGF-beta 1 expressions. Urine TGF-beta 1 concentration depended on the genotype and was further increased by diabetes. These results show that TGF-beta 1 expression is correlated with ACE expression and suggest that this growth factor could be a link between ACE gene polymorphism and diabetic vascular complications.
Collapse
Affiliation(s)
- Maria E Pueyo
- INSERM U460,and Service de Diabetologie, CHU Bichat, Paris, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Kobori H, Prieto-Carrasquero MC, Ozawa Y, Navar LG. AT1 receptor mediated augmentation of intrarenal angiotensinogen in angiotensin II-dependent hypertension. Hypertension 2004; 43:1126-32. [PMID: 15037565 PMCID: PMC2637079 DOI: 10.1161/01.hyp.0000122875.91100.28] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Angiotensin (Ang) II-infused hypertensive rats exhibit increases in renal angiotensinogen mRNA and protein, as well as urinary angiotensinogen excretion in association with increased intrarenal Ang II content. The present study was performed to determine if the augmentation of intrarenal angiotensinogen requires activation of Ang II type 1 (AT1) receptors. Male Sprague-Dawley rats (200 to 220 g) were divided into 3 groups: sham surgery (n=10), subcutaneous infusion of Ang II (80 ng/min, n=11), and Ang II infusion plus AT1 blocker (ARB), olmesartan (5 mg/d, n=12). Ang II infusion progressively increased systolic blood pressure (SBP) compared with sham (178+/-8 mm Hg versus 119+/-4 at day 11). ARB treatment prevented hypertension (113+/-6 at day 11). Twenty-four-hour urine collections were taken at day 12, and plasma and tissue samples were harvested at day 13. The Ang II+ARB group had a significant increase in plasma Ang II compared with Ang II and sham groups (365+/-46 fmol/mL versus 76+/-9 and 45+/-14, respectively). Nevertheless, ARB treatment markedly limited the enhancement of kidney Ang II by Ang II infusion (65+/-17 fmol/g in sham, 606+/-147 in Ang II group, and 288+/-28 in Ang II+ARB group). Ang II infusion significantly increased kidney angiotensinogen compared with sham (1.69+/-0.21 densitometric units versus 1.00+/-0.17). This change was reflected by increased angiotensinogen immunostaining in proximal tubules. ARB treatment prevented this increase (1.14+/-0.12). Urinary angiotensinogen excretion rates were enhanced 4.7x in Ang II group (4.67+/-0.41 densitometric units versus 1.00+/-0.21) but ARB treatment prevented the augmentation of urinary angiotensinogen (0.96+/-0.23). These data demonstrate that augmentation of intrarenal angiotensinogen in Ang II-infused rats is AT1-dependent and provide further evidence that urinary angiotensinogen is closely linked to intrarenal Ang II in Ang II-dependent hypertension.
Collapse
Affiliation(s)
- Hiroyuki Kobori
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, 1430 Tulane Avenue, #SL39, New Orleans, LA 70112-2699, USA.
| | | | | | | |
Collapse
|
82
|
Kopp UC, Cicha MZ. Impaired substance P release from renal sensory nerves in SHR involves a pertussis toxin-sensitive mechanism. Am J Physiol Regul Integr Comp Physiol 2004; 286:R326-33. [PMID: 14578115 DOI: 10.1152/ajpregu.00493.2003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Stretching the renal pelvic wall activates renal mechanosensory nerves by a PGE2-mediated release of substance P via activation of the cAMP-PKA pathway. Renal pelvic ANG II modulates the responsiveness of renal sensory nerves by suppressing the PGE2-mediated activation of adenylyl cyclase via a pertussis toxin (PTX)-sensitive mechanism. In SHR, activation of renal mechanosensory nerves is impaired. This is due to suppressed release of substance P in response to increased pelvic pressure. The present study was performed to investigate whether the PGE2-mediated release of substance P was suppressed in SHR vs. WKY and, if so, whether the impaired PGE2-mediated release of substance P was due to ANG II activating a PTX-sensitive mechanism. In an isolated renal pelvic wall preparation, PGE2, 0.14 μM, increased substance P release from 9 ± 3 to 22 ± 3 pg/min ( P < 0.01) in Wistar-Kyoto rats (WKY), but had no effect in spontaneously hypertensive rats (SHR). A tenfold higher concentration of PGE2, 1.4 μM, was required to increase substance P release in SHR, from 7 ± 1 to 22 ± 3 pg/min ( P < 0.01). In SHR, treating renal pelvises with losartan enhanced the release of substance P produced by subthreshold concentration of PGE2, 0.3 μM, from 16 ± 2 to 26 ± 3 pg/min ( P < 0.01). Likewise, treating renal pelvises with PTX enhanced the PGE2-mediated release of substance P from 10 ± 1 to 33 ± 3 pg/min ( P < 0.01) in SHR. In WKY, neither losartan nor PTX had an effect on the release of substance P produced by subthreshold concentrations of PGE2, 0.03 μM. In conclusion, the impaired responsiveness of renal sensory nerves in SHR involves endogenous ANG II suppressing the PGE2-mediated release of substance P via a PTX-sensitive mechanism.
Collapse
Affiliation(s)
- Ulla C Kopp
- Dept. of Internal Medicine, VA Medical Center, Bldg. 3, Rm 226, Highway 6W, Iowa City, IA 52246, USA.
| | | |
Collapse
|
83
|
Tan J, Wang H, Leenen FHH. Increases in brain and cardiac AT1 receptor and ACE densities after myocardial infarct in rats. Am J Physiol Heart Circ Physiol 2003; 286:H1665-71. [PMID: 14693687 DOI: 10.1152/ajpheart.00858.2003] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the brain, ouabain-like compounds (OLC) and the reninangiotensin system (RAS) contribute to sympathetic hyperactivity in rats after myocardial infarction (MI). This study aimed to evaluate changes in components of the central vs. the peripheral RAS. Angiotensin-converting enzyme (ACE) and angiotensin type 1 (AT1) receptor binding densities were determined by measuring 125I-labeled 351A and 125I-labeled ANG II binding 4 and 8 wk after MI. In the brain, ACE and AT1 receptor binding increased 8-15% in the subfornical organ, 14-22% in the organum vasculosum laminae terminalis, 20-34% in the paraventricular nucleus, and 13-15% in the median preoptic nucleus. In the heart, the greatest increase in ACE and AT1 receptor binding occurred at the infarct scar (approximately 10-fold) and the least in the right ventricle (2-fold). In kidneys, ACE and AT1 receptor binding decreased 10-15%. After intracerebroventricular infusion of Fab fragments to block brain OLC from 0.5 to 4 wk after MI, increases in ACE and AT1 receptors in the subfornical organ, organum vasculosum laminae terminalis, paraventricular nucleus, and medial preoptic nucleus were markedly inhibited, and ACE and AT1 receptor densities in the heart increased less (6-fold in the infarct scar). In kidneys, decreases in ACE and AT1 receptor binding were absent after treatment with Fab fragments. These results demonstrate that ACE and AT1 receptor binding densities increase not only in the heart but also in relevant areas of the brain of rats after MI. Brain OLC appears to play a major role in activation of brain RAS in rats after MI and, to a modest degree, in activation of the cardiac RAS.
Collapse
Affiliation(s)
- Junhui Tan
- Hypertension Unit, University of Ottawa Heart Institute, Ottawa, Ontario, Canada K1Y 4W7
| | | | | |
Collapse
|
84
|
Harrison-Bernard LM, Schulman IH, Raij L. Postovariectomy Hypertension Is Linked to Increased Renal AT1Receptor and Salt Sensitivity. Hypertension 2003; 42:1157-63. [PMID: 14610098 DOI: 10.1161/01.hyp.0000102180.13341.50] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The functional balance between angiotensin II (Ang II) and nitric oxide (NO) plays a key role in modulating salt sensitivity. Estrogen has been shown to downregulate angiotensin type 1 (AT1) receptor expression and to increase the bioavailability of endothelium-derived NO, which decreases AT1 receptor expression. The present study tests the hypothesis that in the presence of genetic salt sensitivity, deficiency of endogenous estrogens after ovariectomy (OVX) fosters an upregulation of Ang II. Female Dahl salt-resistant (DR), Dahl salt-sensitive (DS), Wistar-Kyoto (WKY), and spontaneously hypertensive (SHR) rats underwent bilateral OVX or sham surgery (SHX) and were fed a normal salt diet (0.5% NaCl) for 14 weeks. Systolic blood pressures were measured every 2 weeks and were not significantly different between OVX and SHX for DR, WKY, and SHR groups. However, at the end of 14 weeks of normal salt diet, hypertension developed in DS OVX but not SHX rats (160+/-3 versus 136+/-3 mm Hg; P<0.05). Hypertension also developed in DS OVX rats pair-fed a normal salt diet (166+/-7 mm Hg). Development of hypertension in DS OVX rats was prevented by estrogen replacement (132+/-3 mm Hg), AT1 receptor blockade (119+/-3 mm Hg), or feeding a very low salt diet (0.1% NaCl; 129+/-4 mm Hg). Renal AT1 receptor protein expression was significantly elevated 2-fold in DS OVX relative to SHX rats and was prevented by estrogen replacement. These data strongly suggest that after OVX in salt-sensitive rats there is a lower threshold for the hypertensinogenic effect of salt that is linked to an activation of Ang II.
Collapse
Affiliation(s)
- Lisa M Harrison-Bernard
- Department of Physiology, SL39 Tulane University Health Sciences Center, 1430 Tulane Ave, New Orleans, LA 70112-2699, USA.
| | | | | |
Collapse
|
85
|
Matsuo T, Ikura Y, Ohsawa M, Ogami M, Kayo S, Yoshimi N, Hai E, Naruko T, Ohishi M, Higuchi K, Arakawa T, Ueda M. Mast cell chymase expression in Helicobacter pylori-associated gastritis. Histopathology 2003; 43:538-549. [PMID: 14636254 DOI: 10.1111/j.1365-2559.2003.01731.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIMS To study the role of mast cell chymase in the inflammatory processes of human chronic gastritis. Experimental studies have shown that mast cell chymase stimulates inflammatory cell accumulation, and contributes to angiotensin II formation. METHODS AND RESULTS Tissue sections from human stomachs with Helicobacter pylori-associated gastritis (surgery/autopsy n = 20; biopsy n = 16) and normal stomachs (n = 10) were studied using immunohistochemical single and double labelling techniques. Monoclonal antibodies used were directed against mast cell chymase, tryptase, neutrophils (CD66b, elastase, and myeloperoxidase), macrophages, T-lymphocytes, and interleukin (IL)-4. The expression of angiotensin-converting enzyme and angiotensin II type 1 receptor was investigated using immunohistochemical analysis and the reverse transcription-polymerase chain reaction. The number of chymase-positive mast cells was significantly higher (P < 0.0001) in H. pylori-associated gastritis than in normal stomachs. Increased expression of chymase in inflamed mucosa was closely related to an increase in the accumulation of neutrophils, macrophages, T-lymphocytes, and IL-4-positive cells. The expression of angiotensin-converting enzyme and angiotensin II type 1 receptor was not altered in gastritis specimens. CONCLUSIONS These observations suggest that mast cell chymase may be an important mediator in the inflammatory processes of human H. pylori-associated gastritis.
Collapse
Affiliation(s)
- T Matsuo
- Department of Pathology, Osaka City University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Kopp UC, Cicha MZ, Smith LA. Dietary sodium loading increases arterial pressure in afferent renal-denervated rats. Hypertension 2003; 42:968-73. [PMID: 14568995 DOI: 10.1161/01.hyp.0000097549.70134.d8] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In rats fed high sodium diet, increasing renal pelvic pressure > or =3 mm Hg activates renal mechanosensory nerves, resulting in a renorenal reflex-induced increase in urinary sodium excretion. The low activation threshold of the renal mechanosensory nerves suggests a role for natriuretic renorenal reflexes in the regulation of arterial pressure and sodium balance. If so, interruption of the afferent renal innervation by dorsal rhizotomy (DRX) at T9-L1 would impair urinary sodium excretion and/or increase arterial pressure during high dietary sodium intake. DRX and sham-DRX rats were fed either a high or a normal sodium diet for 3 weeks. Mean arterial pressure measured in conscious rats was higher in DRX than in sham-DRX rats fed a high sodium diet, 130+/-2 vs 100+/-3 mm Hg (P<0.01). However, mean arterial pressure was similar in DRX and sham-DRX rats fed a normal sodium diet, 115+/-1 and 113+/-1 mm Hg, respectively. Steady-state urinary sodium excretion was similar in DRX and sham-DRX rats on high (17.9+/-2.2 and 16.4+/-1.8 mmol/24 h, respectively) and normal (4.8+/-0.3 and 5.0+/-0.4 mmol/24 h, respectively) sodium diets. Studies in anesthetized rats showed a lack of an increase in afferent renal nerve activity in response to increased renal pelvic pressure and impaired prostaglandin E2-mediated release of substance P from the renal pelvic nerves in DRX rats fed either a high or a normal sodium diet, suggesting that DRX resulted in decreased responsiveness of peripheral renal sensory nerves. In conclusion, when the afferent limb of the renorenal reflex is interrupted, a high sodium diet results in increased arterial pressure to facilitate the natriuresis and maintenance of sodium balance.
Collapse
Affiliation(s)
- Ulla C Kopp
- Department of Internal Medicine, Veterans Affairs Medical Center, and the Roy J. and Lucille Carver College of Medicine, University of Iowa, Bldg 3, Room 226, Highway 6W, Iowa City, IA 52246, USA.
| | | | | |
Collapse
|
87
|
Nishiyama A, Kobori H, Fukui T, Zhang GX, Yao L, Rahman M, Hitomi H, Kiyomoto H, Shokoji T, Kimura S, Kohno M, Abe Y. Role of angiotensin II and reactive oxygen species in cyclosporine A-dependent hypertension. Hypertension 2003; 42:754-60. [PMID: 12874088 PMCID: PMC2572573 DOI: 10.1161/01.hyp.0000085195.38870.44] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Treatment with cyclosporine A (CysA), a potent immunosuppressive agent, is associated with systemic and renal vasoconstriction, leading to hypertension. The present study was conducted to elucidate the contribution of angiotensin II (Ang II) to CysA-induced hypertension and reactive oxygen species (ROS) generation. CysA (30 mg/kg per day SC), given for 3 weeks in rats, increased systolic blood pressure (SBP) from 119+/-2 to 145+/-3 mm Hg (n=7). Plasma and kidney Ang II levels were significantly higher in CysA-treated rats (136+/-10 fmol/mL and 516+/-70 fmol/g) than in vehicle-treated (1 mL olive oil) rats (76+/-10 fmol/mL and 222+/-21 fmol/g, n=7). CysA treatment increased AT1 receptor protein expression in the aorta (by 251+/-35%), whereas it was reduced in the kidney (by -32+/-4%). Superoxide anion production in aortic segments and kidney thiobarbituric acid-reactive substance (TBARS) contents were higher in CysA-treated rats (26+/-2 counts/min per milligram and 37+/-3 nmol/g) than in vehicle-treated rats (17+/-1 counts/min per milligram and 24+/-3 nmol/g). Concurrent administration of an AT1 receptor antagonist, valsartan (30 mg/kg per day, in drinking water), to CysA-treated rats (n=7) significantly decreased SBP (113+/-4 mm Hg) and prevented increases in vascular superoxide (16+/-2 counts/min per milligram) and kidney TBARS contents (21+/-3 nmol/g). Similarly, treatment with a superoxide dismutase mimetic, 4-hydroxy-2,2,6,6,-tetramethylpiperidine-N-oxyl (Tempol; 3 mmol/L in drinking water, n=7), prevented CysA-induced increases in SBP (115+/-3 mm Hg), vascular superoxide (16+/-1 counts/min per milligram), and kidney TBARS contents (19+/-2 nmol/g). These data suggest that ROS generation induced by augmented Ang II levels contributes to the development of CysA-induced hypertension.
Collapse
Affiliation(s)
- Akira Nishiyama
- Department of Pharmacology, Kagawa Medical University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Vío CP, Jeanneret VA. Local induction of angiotensin-converting enzyme in the kidney as a mechanism of progressive renal diseases. KIDNEY INTERNATIONAL. SUPPLEMENT 2003:S57-63. [PMID: 12969129 DOI: 10.1046/j.1523-1755.64.s86.11.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Angiotensin Converting Enzyme (ACE) or Kininase II has a pivotal role determining the local activity of the renin angiotensin and kallikrein kinin systems. Angiotensin II (Ang II), a main hormone of the renin system, has a well established participation as a renal injury agent in models of renal disease with tubulointerstitial fibrosis. Although, since its discovery, ACE has been known to convert Ang I to Ang II, and to inactivate bradykinin (BK), only recently has been emerged evidence for a role of BK with renal protective and antifibrotic effects opposing Ang II. Pertinent to the tubulointerstitial injury, where infiltration and proliferation of macrophages and fibroblast occur, ACE also regulates the levels of the natural hemoregulatory peptide, N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP). Owing the importance of tissue ACE, its distribution was studied in several models of renal injury. The results showed increased ACE in proximal tubules and ACE induction in the cell infiltrated tubulointerstitium (macrophages and myofibroblasts) of injured kidneys from hypokalemic, Goldblatt hypertensive, Ang II and phenylefrine infused rats, and in both human diabetic and membranous nephropathies. ACE, in addition to Ang II generation, may play a pathogenic role through the hydrolysis of BK and Ac-SDKP. Thus, local increase in ACE can be a novel mechanism involved in tubulointerstitial renal injury, providing, from an anatomical ground, a pathological basis for the putative deleterious effect of ACE in the diseased kidneys, and the beneficial effect of ACE inhibition.
Collapse
Affiliation(s)
- Carlos P Vío
- Department of Physiology, Pontificia Universidad Catolica de Chile, Santiago, Chile.
| | | |
Collapse
|
89
|
Kopp UC, Cicha MZ, Smith LA. Angiotensin blocks substance P release from renal sensory nerves by inhibiting PGE2-mediated activation of cAMP. Am J Physiol Renal Physiol 2003; 285:F472-83. [PMID: 12746258 DOI: 10.1152/ajprenal.00399.2002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Activation of renal sensory nerves involves PGE2-mediated release of substance P (SP) via activation of the cAMP-PKA pathway. The PGE2-mediated SP release is suppressed by a low- and enhanced by a high-sodium (Na+) diet, suggesting an inhibitory effect of ANG. We now examined whether ANG II is present in the pelvic wall and inhibits PGE2-mediated SP release by blocking PGE2-mediated increases in cAMP. ANG II levels in renal pelvic tissue were 710 +/- 95 and 260 +/- 30 fmol/g tissue in rats fed a low- and high-Na+ diet, respectively. In a renal pelvic preparation from high-Na+-diet rats, 0.14 microM PGE2 produced an increase in SP release from 7 +/- 1 to 19 +/- 3 pg/min that was blocked by 15 nM ANG II. Treating pelvises with pertussis toxin (PTX) abolished the effects of ANG II. In pelvises from low-Na+ rats, neither basal nor bradykinin-mediated SP release was altered by PGE2. However, the bradykinin-mediated release of SP was enhanced by the permeable cAMP analog CPT-cAMP, from 4 +/- 1 to 11 +/- 2 pg/min, a response similar to that in normal-Na+-diet rats. In vivo, renal pelvic administration of PGE2 enhanced the afferent renal nerve activity (ARNA) response to bradykinin in normal- but not in low-Na+ diet rats. CPT-cAMP produced similar enhancement of the ARNA responses to bradykinin in normal- and low-Na+-diet rats, 1,670 +/- 490 and 1,760 +/- 400%.s (area under the curve of ARNA vs. time). Similarly, the ARNA responses to increases in renal pelvic pressure were similarly enhanced by CPT-cAMP in normal- and low-Na+-diet rats. In conclusion, renal pelvic ANG II modulates the responsiveness of renal sensory nerves by suppressing PGE2-mediated activation of adenylyl cyclase via a PTX-sensitive mechanism.
Collapse
Affiliation(s)
- Ulla C Kopp
- Dept. of Internal Medicine, Medical Center, University of Iowa Roy J. and Lucille Carver College of Medicine, Iowa City 52242, USA.
| | | | | |
Collapse
|
90
|
Affiliation(s)
- Holger Scholz
- Johannes-Müller-Institut für Physiologie, Humboldt-Universität, Charité, Tucholskystrasse 2, 10117 Berlin, Germany.
| |
Collapse
|
91
|
Abstract
Elevations in intrarenal angiotensin II (Ang II) cause reductions in renal function and sodium excretion that contribute to progressive hypertension and lead to renal and vascular injury. Augmentation of intrarenal Ang II occurs by several processes, leading to levels much greater than can be explained from the circulating levels. In Ang II-dependent hypertension, Ang II is internalized via an AT1 receptor mechanism, but there is also sustained intrarenal production of Ang II. Ang II exerts a positive feedback action on intrarenal angiotensinogen (AGT) mRNA and protein. The increased intrarenal AGT production is associated with increased intrarenal and intracellular Ang II contents and urinary AGT excretion rates. The increased urinary AGT indicates spillover of AGT into distal nephron segments supporting enhanced distal Ang II formation and sodium reabsorption. The augmentation of intrarenal Ang II provides the basis for sustained actions on renal function, sodium excretion, and maintenance of hypertension.
Collapse
Affiliation(s)
- L Gabriel Navar
- Department of Physiology SL39, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA.
| | | | | |
Collapse
|
92
|
Zeng C, Asico LD, Wang X, Hopfer U, Eisner GM, Felder RA, Jose PA. Angiotensin II regulation of AT1 and D3 dopamine receptors in renal proximal tubule cells of SHR. Hypertension 2003; 41:724-9. [PMID: 12623987 DOI: 10.1161/01.hyp.0000047880.78462.0e] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Dopamine and angiotensin II negatively interact to regulate sodium excretion and blood pressure. D3 dopamine receptors downregulate angiotensin type 1 (AT1) receptors in renal proximal tubule cells from normotensive Wistar-Kyoto rats. We determined whether AT1 receptors regulate D3 receptors and whether the regulation is different in cultured renal proximal tubule cells from normotensive and spontaneously hypertensive rats. Angiotensin II (10(-8)M/24 hours) decreased D3 receptors in both normotensive (control, 36+/-3; angiotensin II, 24+/-3 U) and hypertensive (control, 30+/-3; angiotensin II, 11+/-3 U; n=9 per group) rats; effects that were blocked by the AT1 receptor antagonist, losartan (10(-8)M/24 hours). However, the reduction in D3 expression was greater in hypertensive (60+/-10%) than in normotensive rats (32+/-9%). In normotensive rats, angiotensin II (10(-8)M/24hr) also decreased AT1 receptors. In contrast, in cells from hypertensive rats, angiotensin II increased AT1 receptors. AT1 and D3 receptors co-immunoprecipitated in renal proximal tubule cells from both strains. Angiotensin II decreased D3/AT1 receptor co-immunoprecipitation similarly in both rat strains, but basal D3/AT1 co-immunoprecipitation was 6 times higher in normotensive than in hypertensive rats. Therefore, AT1 and D3 receptor interaction is qualitatively and quantitatively different between normotensive and hypertensive rats; angiotensin II decreases AT1 expression in normotensive but increases it in hypertensive rats. In addition, angiotensin II decreases D3 expression to a greater extent in hypertensive than in normotensive rats. Aberrant interactions between D3 and AT1 receptors may play a role in the pathogenesis of hypertension.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Antibody Specificity
- Cells, Cultured
- Hypertension/metabolism
- Immunoblotting
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/metabolism
- Precipitin Tests
- Rats
- Rats, Inbred SHR
- Rats, Inbred WKY
- Receptor, Angiotensin, Type 1
- Receptors, Angiotensin/immunology
- Receptors, Angiotensin/metabolism
- Receptors, Dopamine D2/immunology
- Receptors, Dopamine D2/metabolism
- Receptors, Dopamine D3
Collapse
Affiliation(s)
- Chunyu Zeng
- Department of Pediatrics, Georgetown University Medical Center, Washington, DC 20007, USA.
| | | | | | | | | | | | | |
Collapse
|
93
|
Navar LG, Harrison-Bernard LM, Nishiyama A, Kobori H. Regulation of intrarenal angiotensin II in hypertension. Hypertension 2002; 39:316-22. [PMID: 11882566 PMCID: PMC2575645 DOI: 10.1161/hy0202.103821] [Citation(s) in RCA: 280] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Intrarenal angiotensin II (Ang II) is regulated by several complex processes involving formation from both systemically delivered and intrarenally formed substrate, as well as receptor-mediated internalization. There is substantial compartmentalization of intrarenal Ang II, with levels in the renal interstitial fluid and in proximal tubule fluid being much greater than can be explained from the circulating levels. In Ang II--dependent hypertension, elevated intrarenal Ang II levels occur even when intrarenal renin expression and content are suppressed. Studies in Ang II--infused rats have demonstrated that augmentation of intrarenal Ang II is due, in part, to uptake of circulating Ang II via an Ang II type 1 (AT(1)) receptor mechanism and also to sustained endogenous production of Ang II. Some of the internalized Ang II accumulates in the light and heavy endosomes and is therefore potentially available for intracellular actions. The enhanced intrarenal Ang II also exerts a positive feedback action to augment intrarenal levels of angiotensinogen (AGT) mRNA and protein, which contribute further to the increased intrarenal Ang II in hypertensive states. In addition, renal AT(1) receptor protein and mRNA levels are maintained, allowing increased Ang II levels to elicit progressive effects. The increased intrarenal Ang II activity and AGT production are associated with increased urinary AGT excretion rates. The urinary AGT excretion rates show a clear relationship to kidney Ang II content, suggesting that urinary AGT may serve as an index of Ang II--dependent hypertension. Collectively, the data support a powerful role for intrarenal Ang II in the pathogenesis of hypertension.
Collapse
Affiliation(s)
- L Gabriel Navar
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | | | |
Collapse
|