Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Neuroscience ; 329: 112-21, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27163380

ABSTRACT

Activation of angiotensinergic pathways by central aldosterone (Aldo)-mineralocorticoid receptor (MR) pathway plays a critical role in angiotensin II (Ang II)-induced hypertension. The subfornical organ (SFO) contains both MR and angiotensin II type 1 receptors (AT1R) and can relay the signals of circulating Ang II to downstream nuclei such as the paraventricular nucleus (PVN), supraoptic nucleus (SON) and rostral ventrolateral medulla (RVLM). In Wistar rats, subcutaneous (sc) infusion of Ang II at 500ng/min/kg for 1 or 2weeks increased reactive oxygen species (ROS) as measured by dihydroethidium (DHE) staining in a nucleus - specific pattern. Intra-SFO infusion of AAV-MR- or AT1aR-siRNA prevented the Ang II-induced increase in AT1R mRNA expression in the SFO and decreased MR mRNA. Both MR- and AT1aR-siRNA prevented increases in ROS in the PVN and RVLM. MR- but not AT1aR-siRNA in the SFO prevented the Ang II-induced ROS in the SON. Both MR- and AT1aR-siRNA in the SFO prevented most of the Ang II-induced hypertension as assessed by telemetry. These results indicate that Aldo-MR signaling in the SFO is needed for the activation of Ang II-AT1R-ROS signaling from the SFO to the PVN and RVLM. Activation of Aldo-MR signaling from the SFO to the SON may enhance AT1R dependent activation of pre-sympathetic neurons in the PVN.


Subject(s)
Angiotensin II/metabolism , Hypothalamus/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, Mineralocorticoid/metabolism , Subfornical Organ/metabolism , Aldosterone/metabolism , Angiotensin II/administration & dosage , Animals , Blood Pressure/physiology , Dependovirus , Gene Knockdown Techniques , Genetic Vectors , Heart Rate/physiology , Hypertension/metabolism , Male , Medulla Oblongata/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering , Random Allocation , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptors, Mineralocorticoid/genetics
2.
PLoS One ; 9(9): e108916, 2014.
Article in English | MEDLINE | ID: mdl-25275393

ABSTRACT

Central nervous system (CNS) administration of angiotensin II (Ang II) raises blood pressure (BP). The rise in BP reflects increased sympathetic outflow and a slower neuromodulatory pressor mechanism mediated by CNS mineralocorticoid receptors (MR). We investigated the hypothesis that the sustained phase of hypertension is associated also with elevated circulating levels of endogenous ouabain (EO), and chronic stimulation of arterial calcium transport proteins including the sodium-calcium exchanger (NCX1), the type 6 canonical transient receptor potential protein (TRPC6), and the sarcoplasmic reticulum calcium ATPase (SERCA2). Wistar rats received a chronic intra-cerebroventricular infusion of vehicle (C) or Ang II (A, 2.5 ng/min, for 14 days) alone or combined with the MR blocker, eplerenone (A+E, 5 µg/day), or the aldosterone synthase inhibitor, FAD286 (A+F, 25 µg/day). Conscious mean BP increased (P<0.05) in A (123 ± 4 mm Hg) vs all other groups. Blood, pituitary and adrenal samples were taken for EO radioimmunoassay (RIA), and aortas for NCX1, TRPC6 and SERCA2 immunoblotting. Central infusion of Ang II raised plasma EO (0.58 ± 0.08 vs C 0.34 ± 0.07 nM (P<0.05), but not in A + E and A + F groups as confirmed by off-line liquid chromatography (LC)-RIA and LC-multistage mass spectrometry. Two novel isomers of EO were elevated by Ang II; the second less polar isomer increased >50-fold in the A+F group. Central Ang II increased arterial expression of NCX1, TRPC6 and SERCA2 (2.6, 1.75 and 3.7-fold, respectively; P<0.01)) but not when co-infused with E or F. Adrenal and pituitary EO were unchanged. We conclude that brain Ang II activates a CNS-humoral axis involving plasma EO. The elevated EO reprograms peripheral ion transport pathways known to control arterial Na(+) and Ca(2+) homeostasis; this increases contractility and augments sympathetic effects. The new axis likely contributes to the chronic pressor effect of brain Ang II.


Subject(s)
Angiotensin II/pharmacology , Blood Pressure/drug effects , Brain/metabolism , Neurosecretory Systems/blood supply , Neurosecretory Systems/metabolism , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Angiotensin II/administration & dosage , Animals , Brain/drug effects , Calcium/metabolism , Cation Transport Proteins/metabolism , Chromatography, Liquid , Infusions, Intraventricular , Isomerism , Male , Models, Biological , Muscle Cells/drug effects , Muscle Cells/metabolism , Ouabain/blood , Ouabain/chemistry , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Radioimmunoassay , Rats, Wistar , Solid Phase Extraction
3.
J Physiol ; 592(16): 3523-36, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24973408

ABSTRACT

Circulating Ang II activates an aldosterone-mineralocorticoid receptor (MR) - angiotensin II (Ang II) - angiotensin type 1 receptor (AT1R) pathway in the hypothalamus. To obtain insights into the actual neuronal projections involved, adeno-associated virus carrying small interfering RNA against either AT1aR (AAV-AT1aR-siRNA) or MR (AAV-MR-siRNA) were infused into the paraventricular nucleus (PVN) in Wistar rats. Intra-PVN infusion of AAV-AT1aR-siRNA or AAV-MR-siRNA decreased AT1R or MR expression in the PVN but not in the subfornical organ (SFO) or supraoptic nucleus (SON). Subcutaneous infusion of Ang II at 500 ng kg(-1) min(-1) for 2 weeks increased mean arterial pressure by 60-70 mmHg, and increased AT1R and MR expression in the SFO, SON and PVN. Intra-PVN AT1aR-siRNA prevented the Ang II-induced increase in AT1R but not MR expression in the PVN, and MR-siRNA prevented MR but not AT1R expression in the PVN. The increases in AT1R and MR expression in both the SFO and the SON were not changed by the two AAV-siRNAs. Specific knockdown of AT1R or MR in the PVN by AAV-siRNA each prevented most of the Ang II-induced hypertension. Prevention of the subcutaneous Ang II-induced increase in MR but not the increase in AT1R by knockdown of MR and vice versa suggests an independent regulation of MR and AT1R expression in the PVN. Both AT1R and MR activation in the PVN play a critical role in Ang II-induced hypertension in rats.


Subject(s)
Angiotensin II/metabolism , Hypertension/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Receptors, Mineralocorticoid/metabolism , Animals , Male , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 2/genetics , Receptors, Mineralocorticoid/genetics
4.
J Physiol ; 592(15): 3273-86, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24951624

ABSTRACT

Intracerebroventricular infusion of a mineralocorticoid receptor (MR) or angiotensin II type 1 receptor (AT1R) blocker in rats attenuates sympathetic hyperactivity and progressive left ventricular (LV) dysfunction post myocardial infarction (MI). The present study examined whether knockdown of MRs or AT1Rs specifically in the paraventricular nucleus (PVN) contributes to these effects, and compared cardiac effects with those of systemic treatment with the ß1-adrenergic receptor blocker metoprolol. The PVN of rats was infused with adeno-associated virus carrying small interfering RNA against either MR (AAV-MR-siRNA) or AT1R (AAV-AT1R-siRNA), or as control scrambled siRNA. At 4 weeks post MI, AT1R but not MR expression was increased in the PVN, excitatory renal sympathetic nerve activity and pressor responses to air stress were enhanced, and arterial baroreflex function was impaired; LV end-diastolic pressure (LVEDP) was increased and LV peak systolic pressure (LVPSP), ejection fraction (EF) and dP/dtmax decreased. AAV-MR-siRNA and AAV-AT1R-siRNA both normalized AT1R expression in the PVN, similarly ameliorated sympathetic and pressor responses to air stress, largely prevented baroreflex desensitization, and improved LVEDP, EF and dP/dtmax as well as cardiac interstitial (but not perivascular) fibrosis. In a second set of rats, metoprolol at 70 or 250 mg kg(-1) day(-1) in the drinking water for 4 weeks post MI did not improve LV function except for a decrease in LVEDP at the lower dose. These results suggest that in rats MR-dependent upregulation of AT1Rs in the PVN contributes to sympathetic hyperactivity, and LV dysfunction and remodelling post MI. In rats, normalizing MR-AT1R signalling in the PVN is a more effective strategy to improve LV dysfunction post MI than systemic ß1 blockade.


Subject(s)
Myocardial Infarction/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, Mineralocorticoid/metabolism , Sympathetic Nervous System/physiopathology , Ventricular Dysfunction/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Baroreflex , Male , Metoprolol/pharmacology , Myocardial Infarction/physiopathology , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/genetics , Receptors, Mineralocorticoid/genetics , Sympathetic Nervous System/metabolism , Up-Regulation , Ventricular Dysfunction/physiopathology
5.
Brain Res ; 1527: 79-86, 2013 Aug 21.
Article in English | MEDLINE | ID: mdl-23816375

ABSTRACT

Central infusion of Na(+)-rich artificial cerebro-spinal fluid (aCSF) activates the brain renin-angiotensin system and causes sympatho-excitatory and pressor responses. We evaluated the role of the subfornical organ (SFO) and angiotensin II type 1 (AT1) receptors in the SFO in mediating the central Na(+)-induced pressor response. In conscious Wistar rats, intra SFO infusions of Na(+)-rich aCSF containing 0.45 and 0.6M Na(+) at 10 nl/min or injection of angiotensin II (Ang II) at 80 ng increased blood pressure (BP) by 15-22 mmHg, whereas mannitol with the same osmolarity as the Na(+)-rich aCSF had no effects. Intra SFO infusion of the AT1 receptor blocker candesartan abolished the pressor response induced by intra SFO administration of Na(+)-rich aCSF or Ang II. Intra cerebro-ventricular (icv) infusion of Na(+)-rich aCSF (0.3M Na(+)) at 3.8 µl/min for 10 min increased BP by 15-20 mmHg. Electrolytic lesion of the SFO attenuated these BP increases by 50-70%. Intra SFO infusion of candesartan also prevented 50% of these pressor responses. These data suggest that SFO neurons are indeed sensitive to Na(+), the SFO is a major - but not only - site in the brain to sense an increase in CSF [Na(+)], and activation of AT1 receptors in the SFO mediates the SFO component of the Na(+)-induced pressor response.


Subject(s)
Cerebrospinal Fluid/chemistry , Receptor, Angiotensin, Type 1/metabolism , Renin-Angiotensin System/physiology , Sodium/administration & dosage , Subfornical Organ/drug effects , Animals , Blood Pressure/physiology , Infusions, Intraventricular , Male , Rats , Rats, Wistar , Subfornical Organ/metabolism
6.
Hypertension ; 62(3): 564-71, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23856493

ABSTRACT

Circulating angiotensin II (Ang II) activates a central aldosterone-mineralocorticoid receptor neuromodulatory pathway, which mediates most of the Ang II-induced hypertension. This study examined whether specific central infusion of Ang II also activates this central aldosterone-mineralocorticoid receptor pathway. Intracerebroventricular infusion of Ang II at 1.0, 2.5, and 12.5 ng/min for 2 weeks caused dose-related increases in water intake, Ang II concentration in the cerebrospinal fluid, and blood pressure. Intracerebroventricular Ang II, at 2.5 and 12.5 ng/min, increased hypothalamic aldosterone and corticosterone, as well as plasma aldosterone and corticosterone without affecting plasma Ang II levels. Intracerebroventricular infusion of the aldosterone synthase inhibitor FAD286-but not the mineralocorticoid receptor blocker eplerenone-inhibited by ≈60% the Ang II-induced increase in hypothalamic aldosterone. Both blockers attenuated by ≈50% the increase in plasma aldosterone and corticosterone with only minimal effects on hypothalamic corticosterone. By telemetry, intracerebroventricular infusion of Ang II maximally increased blood pressure within the first day with no further increase over the next 2 weeks. Intracerebroventricular infusion of FAD286 or eplerenone did not affect the initial pressor responses but similarly prevented 60% to 70% of the chronic pressor responses to intracerebroventricular infusion of Ang II. These results indicate distinctly different patterns of blood pressure increase by circulating versus central Ang II and support the involvement of a brain aldosterone-mineralocorticoid receptor-activated neuromodulatory pathway in the chronic hypertension caused by both circulating and central Ang II.


Subject(s)
Aldosterone/metabolism , Brain/metabolism , Corticosterone/metabolism , Hypertension/metabolism , Angiotensin II , Animals , Blood Pressure/drug effects , Brain/drug effects , Cytochrome P-450 CYP11B2/antagonists & inhibitors , Dose-Response Relationship, Drug , Eplerenone , Fadrozole/pharmacology , Heart Rate/drug effects , Hypertension/chemically induced , Male , Mineralocorticoid Receptor Antagonists/pharmacology , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Receptors, Mineralocorticoid/metabolism , Spironolactone/analogs & derivatives , Spironolactone/pharmacology
7.
Cardiovasc Res ; 97(3): 424-31, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23257024

ABSTRACT

AIMS: In rats post-myocardial infarction (MI), activation of angiotensinergic pathways in the brain contributes to sympathetic hyperactivity and progressive left ventricle (LV) dysfunction. The present study examined whether angiotensin III (Ang III) is one of the main effector peptides of the brain renin-angiotensin system controlling these effects. METHODS AND RESULTS: After coronary artery ligation, Wistar rats were infused intracerebroventricularly for 4 weeks via minipumps with vehicle, the aminopeptidase A (APA) inhibitor RB150 (0.3 mg/day), which blocks the formation of brain Ang III, or losartan (0.25 mg/day). Blood pressure (BP), heart rate, and renal sympathetic nerve activity in response to air stress and acute changes in BP were measured, and LV function was evaluated by echocardiography and Millar catheter. At 4 weeks post-MI, brain APA activity was increased, sympatho-excitatory and pressor responses to air stress enhanced, and arterial baroreflex function impaired. LV end-diastolic pressure (LVEDP) was increased and ejection fraction (EF) and maximal first derivative of change in pressure over time (dP/dt(max)) were decreased. Central infusion of RB150 during 4 weeks post-MI normalized brain APA activity and responses to stress and baroreflex function, and improved LVEDP, EF, and dP/dt(max). Central infusion of losartan had similar effects but was somewhat less effective, and had no effect on brain APA activity. CONCLUSION: These results indicate that brain APA and Ang III appear to play a pivotal role in the sympathetic hyperactivity and LV dysfunction in rats post-MI. RB150 may be a potential candidate for central nervous system-targeted therapy post-MI.


Subject(s)
Angiotensin III/antagonists & inhibitors , Brain/metabolism , Disease Progression , Myocardial Infarction/physiopathology , Sympathetic Nervous System/physiopathology , Ventricular Dysfunction, Left/prevention & control , Ventricular Dysfunction, Left/physiopathology , Aldosterone/metabolism , Angiotensin III/drug effects , Animals , Baroreflex/drug effects , Baroreflex/physiology , Blood Pressure/drug effects , Blood Pressure/physiology , Brain/drug effects , Disease Models, Animal , Disulfides/administration & dosage , Disulfides/pharmacology , Glutamyl Aminopeptidase/antagonists & inhibitors , Heart Rate/drug effects , Heart Rate/physiology , Infusions, Intraventricular , Rats , Rats, Wistar , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Stroke Volume/drug effects , Stroke Volume/physiology , Sulfonic Acids/administration & dosage , Sulfonic Acids/pharmacology , Sympathetic Nervous System/drug effects
8.
Am J Physiol Regul Integr Comp Physiol ; 303(2): R236-45, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22647294

ABSTRACT

In Dahl salt-sensitive (S) rats, Na(+) entry into the cerebrospinal fluid (CSF) and sympathoexcitatory and pressor responses to CSF Na(+) are enhanced. Salt-inducible kinase 1 (SIK1) increases Na(+)/K(+)-ATPase activity in kidney cells. We tested the possible role of SIK1 in regulation of CSF [Na(+)] and responses to Na(+) in the brain. SIK1 protein and activity were lower in hypothalamic tissue of Dahl S (SS/Mcw) compared with salt-resistant SS.BN13 rats. Intracerebroventricular infusion of the protein kinase inhibitor staurosporine at 25 ng/day, to inhibit SIK1 further increased mean arterial pressure (MAP) and HR but did not affect the increase in CSF [Na(+)] or hypothalamic aldosterone in Dahl S on a high-salt diet. Intracerebroventricular infusion of Na(+)-rich artificial CSF caused significantly larger increases in renal sympathetic nerve activity, MAP, and HR in Dahl S vs. SS.BN13 or Wistar rats on a normal-salt diet. Intracerebroventricular injection of 5 ng staurosporine enhanced these responses, but the enhancement in Dahl S rats was only one-third that in SS.BN13 and Wistar rats. Staurosporine had no effect on MAP and HR responses to intracerebroventricular ANG II or carbachol, whereas the specific protein kinase C inhibitor GF109203X inhibited pressor responses to intracerebroventricular Na(+)-rich artificial CSF or ANG II. These results suggest that the SIK1-Na(+)/K(+)-ATPase network in neurons acts to attenuate sympathoexcitatory and pressor responses to increases in brain [Na(+)]. The lower hypothalamic SIK1 activity and smaller effect of staurosporine in Dahl S rats suggest that impaired activation of neuronal SIK1 by Na(+) may contribute to their enhanced central responses to sodium.


Subject(s)
Brain/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats, Inbred Dahl/metabolism , Sodium/cerebrospinal fluid , Angiotensin II/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Brain/drug effects , Carbachol/pharmacology , Enzyme Inhibitors/pharmacology , Heart Rate/drug effects , Heart Rate/physiology , Hypothalamus/drug effects , Hypothalamus/metabolism , Indoles/pharmacology , Male , Maleimides/pharmacology , Models, Animal , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/drug effects , Rats , Rats, Wistar , Sodium Chloride, Dietary/pharmacology , Staurosporine/pharmacology
9.
Am J Physiol Regul Integr Comp Physiol ; 302(7): R825-32, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22262304

ABSTRACT

Central infusion of an angiotensin type 1 (AT(1)) receptor blocker prevents sympathetic hyperactivity and hypertension in Dahl salt-sensitive (S) rats on high salt. In the present study, we examined whether central infusion of a direct renin inhibitor exerts similar effects. Intracerebroventricular infusion of aliskiren at the rate of 0.05 mg/day markedly inhibited the increase in ANG II levels in the cerebrospinal fluid and in blood pressure (BP) caused by intracerebroventricular infusion of rat renin. In Dahl S rats on high salt, intracerebroventricular infusion of aliskiren at 0.05 and 0.25 mg/day for 2 wk similarly decreased resting BP in Dahl S rats on high salt. In other groups of Dahl S rats, high salt intake for 2 wk increased resting BP by ∼25 mmHg, enhanced pressor and sympathoexcitatory responses to air-stress, and desensitized arterial baroreflex function. All of these effects were largely prevented by intracerebroventricular infusion of aliskiren at 0.05 mg/day. Aliskiren had no effects in rats on regular salt. Neither high salt nor aliskiren affected hypothalamic ANG II content. These results indicate that intracerebroventricular infusions of aliskiren and an AT(1) receptor blocker are similarly effective in preventing salt-induced sympathetic hyperactivity and hypertension in Dahl S rats, suggesting that renin in the brain plays an essential role in the salt-induced hypertension. The absence of an obvious increase in hypothalamic ANG II by high salt, or decrease in ANG II by aliskiren, suggests that tissue levels do not reflect renin-dependent ANG II production in sympathoexcitatory angiotensinergic neurons.


Subject(s)
Amides/administration & dosage , Fumarates/administration & dosage , Hyperkinesis/prevention & control , Hypertension/prevention & control , Renin/antagonists & inhibitors , Sympathetic Nervous System/drug effects , Angiotensin II/cerebrospinal fluid , Animals , Baroreflex/drug effects , Blood Pressure/drug effects , Heart Rate/drug effects , Hyperkinesis/physiopathology , Hypertension/physiopathology , Infusions, Intraventricular , Male , Rats , Rats, Inbred Dahl , Rats, Wistar , Sodium Chloride, Dietary/administration & dosage
10.
Exp Physiol ; 96(10): 1028-38, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21824999

ABSTRACT

In rats with salt-induced hypertension or postmyocardial infarction, angiotensin II type 1 receptor (AT(1)R) densities and oxidative stress increase and neuronal NO synthase (nNOS) levels decrease in the paraventricular nucleus (PVN). The present study was designed to determine whether these changes may depend on activation of the aldosterone -'ouabain' neuromodulatory pathway. After intracerebroventricular (i.c.v.) infusion of aldosterone (20 ng h(-1)) for 14 days, blood pressure (BP) and heart rate (HR) were recorded in conscious Wistar rats, and mRNA and protein for nNOS, endothelial NO synthase (eNOS), AT(1)R and NADPH oxidase subunits were assessed in brain tissue. Blood pressure and HR were significantly increased by aldosterone. Aldosterone significantly increased mRNA and protein of AT(1)R, P22phox, P47phox, P67phox and Nox2, and decreased nNOS but not eNOS mRNA and protein in the PVN, as well as increased the angiotensin-converting enzyme and AT(1)R binding densities in the PVN and supraoptic nucleus. The increases in BP and HR, as well as the changes in mRNA, proteins and angiotensin-converting enzyme and AT(1)R binding densities were all largely prevented by concomitant i.c.v. infusion of Digibind (to bind 'ouabain') or benzamil (to block presumed epithelial sodium channels). These data indicate that aldosterone, via 'ouabain', increases in the PVN angiotensin-converting enzyme, AT(1)R and oxidative stress, but decreases nNOS, and suggest that endogenous aldosterone may cause the similar pattern of changes observed in salt-sensitive hypertension and heart failure postmyocardial infarction.


Subject(s)
Aldosterone/pharmacology , Oxidative Stress/drug effects , Receptor, Angiotensin, Type 1/physiology , Renin-Angiotensin System/drug effects , Aldosterone/administration & dosage , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Blood Pressure/drug effects , Heart Rate/drug effects , Immunoglobulin Fab Fragments/pharmacology , Infusions, Intraventricular , Male , NADPH Oxidases/biosynthesis , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Peptidyl-Dipeptidase A/metabolism , Phosphoproteins/biosynthesis , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/drug effects , Renin-Angiotensin System/physiology , Supraoptic Nucleus/drug effects , Supraoptic Nucleus/metabolism , Up-Regulation
11.
Curr Hypertens Rep ; 13(3): 214-20, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21298576

ABSTRACT

Mineralocorticoid receptors (MR) and epithelial sodium channels (ENaC) in the brain mediate central aldosterone-induced sympathetic hyperactivity and hypertension. Enzymes for biosynthesis of aldosterone are present in the brain, and aldosterone can be produced locally in the brain. Hypothalamic aldosterone levels increase in Dahl salt-sensitive rats on high-salt diet, and in Wistar rats with chronic central infusion of sodium-rich artificial cerebrospinal fluid (CSF) or with subcutaneous infusion of angiotensin II. Functional studies using antagonists of MR, ENaC, and ouabain-like compounds ("ouabain"), as well as specific aldosterone synthase inhibitors, suggest that an increase in local synthesis of aldosterone via MR and ENaC in the brain increases "ouabain" and thereby causes enhanced AT(1) receptor stimulation, leading to sympathoexcitation and hypertension. An increase in CSF sodium or an increase in angiotensinergic output from circumventricular organs such as the subfornical organ projecting to hypothalamic nuclei may increase local production of aldosterone and "ouabain" in magnocellular neurons in the supraoptic nucleus and paraventricular nucleus. This aldosterone-"ouabain" neuromodulatory mechanism appears to play a major role in salt-induced or angiotensin II-induced hypertension.


Subject(s)
Aldosterone/biosynthesis , Brain/physiopathology , Epithelial Sodium Channels , Hypertension/physiopathology , Mineralocorticoids , Adrenal Cortex Hormones/biosynthesis , Angiotensin II , Cerebrospinal Fluid , Cytochrome P-450 CYP11B2 , Humans , Ouabain , Pressoreceptors , Renin-Angiotensin System
12.
Am J Physiol Heart Circ Physiol ; 299(2): H422-30, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20511409

ABSTRACT

An increase in plasma ANG II causes neuronal activation in hypothalamic nuclei and a slow pressor response, presumably by increasing sympathetic drive. We evaluated whether the activation of a neuromodulatory pathway, involving aldosterone and "ouabain," is involved in these responses. In Wistar rats, the subcutaneous infusion of ANG II at 150 and 500 ng x kg(-1) x min(-1) gradually increased blood pressure up to 60 mmHg at the highest dose. ANG II at 500 ng x kg(-1) x min(-1) increased plasma ANG II by 4-fold, plasma aldosterone by 25-fold, and hypothalamic aldosterone by 3-fold. The intracerebroventricular infusion of an aldosterone synthase (AS) inhibitor prevented the ANG II-induced increase in hypothalamic aldosterone without affecting the increase in plasma aldosterone. Neuronal activity, as assessed by Fra-like immunoreactivity, increased transiently in the subfornical organ (SFO) but progressively in the paraventricular nucleus (PVN) and supraoptic nucleus (SON). The central infusion of the AS inhibitor or a mineralocorticoid receptor blocker markedly attenuated the ANG II-induced neuronal activation in the PVN but not in the SON. Pressor responses to ANG II at 150 ng x kg(-1) x min(-1) were abolished by an intracerebroventricular infusion of the AS inhibitor. Pressor responses to ANG II at 500 ng x kg(-1) x min(-1) were attenuated by the central infusion of the AS inhibitor or the mineralocorticoid receptor blocker by 70-80% and by Digibind (to bind "ouabain") by 50%. These results suggest a novel central nervous system mechanism for the ANG II-induced slow pressor response, i.e., circulating ANG II activates the SFO, leading to the direct activation of the PVN and SON, and, in addition, via aldosterone-dependent amplifying mechanisms, causes sustained activation of the PVN and thereby hypertension.


Subject(s)
Aldosterone/metabolism , Angiotensin II/blood , Blood Pressure , Brain/metabolism , Cardenolides/metabolism , Neurons/metabolism , Renin-Angiotensin System , Saponins/metabolism , Angiotensin II/administration & dosage , Animals , Blood Pressure/drug effects , Blood Pressure Monitoring, Ambulatory , Brain/cytology , Brain/drug effects , Cytochrome P-450 CYP11B2/antagonists & inhibitors , Cytochrome P-450 CYP11B2/metabolism , Enzyme Inhibitors/administration & dosage , Fadrozole , Heart Rate , Hypothalamus/metabolism , Imidazoles/administration & dosage , Immunoglobulin Fab Fragments/administration & dosage , Immunohistochemistry , Infusion Pumps, Implantable , Infusions, Parenteral , Infusions, Subcutaneous , Male , Mineralocorticoid Receptor Antagonists/administration & dosage , Neural Pathways/metabolism , Neurons/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Pyridines/administration & dosage , Rats , Rats, Wistar , Renin-Angiotensin System/drug effects , Spironolactone/administration & dosage , Subfornical Organ/metabolism , Telemetry , Time Factors
13.
Am J Physiol Regul Integr Comp Physiol ; 299(1): R222-33, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20427723

ABSTRACT

We evaluated the effects of intracerebroventricular (icv) infusion of Na(+)-rich artificial cerebrospinal fluid (aCSF), with or without the mineralocorticoid receptor (MR) blocker spironolactone, on epithelial Na(+) channel (ENaC) subunits and regulators, such as MR, serum/glucocorticoid-inducible kinase 1, neural precursor cells expressed developmentally downregulated 4-like gene, 11beta-hydroxylase, and aldosterone synthase, in brain regions of Wistar rats. The effects of icv infusion of the amiloride analog benzamil on brain tissue and CSF Na(+) concentration ([Na(+)]) were also assessed. In the choroid plexus and ependyma of the anteroventral third ventricle, ENaC subunits are present in apical and basal membranes. Na(+)-rich aCSF increased beta-ENaC mRNA and immunoreactivity in the choroid plexus and increased alpha- and beta-ENaC immunoreactivities in the ependyma. Na(+)-rich aCSF increased alpha- and beta-ENaC-gold-labeled particles in the microvilli of the choroid plexus and in basolateral membranes of the ependyma. Spironolactone only prevented the increase in beta-ENaC immunoreactivity in the choroid plexus and ependyma. In the supraoptic nucleus, paraventricular nucleus, and subfornical organ, Na(+)-rich aCSF did not affect mRNA expression levels of the studied genes. Benzamil significantly increased CSF [Na(+)] in the control, but not Na(+)-rich, aCSF group. In contrast, benzamil prevented the increase in hypothalamic tissue [Na(+)] by Na(+)-rich aCSF. These results suggest that CSF Na(+) upregulates ENaC expression in the brain epithelia, but not in the neurons of hypothalamic nuclei. ENaC in the choroid plexus and ependyma appear to contribute to regulation of Na(+) homeostasis in the brain.


Subject(s)
Brain/metabolism , Epithelial Sodium Channels/metabolism , Sodium , Amiloride/analogs & derivatives , Animals , Biological Transport/drug effects , Choroid Plexus/metabolism , Cytochrome P-450 CYP11B2/metabolism , Epithelial Sodium Channels/genetics , Epithelium/metabolism , Glucocorticoids/metabolism , Glucocorticoids/pharmacology , Hypothalamus/metabolism , Male , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Wistar , Receptors, Mineralocorticoid , Sodium/cerebrospinal fluid , Sodium/metabolism , Sodium/pharmacology , Sodium, Dietary/pharmacology , Spironolactone/metabolism , Spironolactone/pharmacology
14.
Can J Physiol Pharmacol ; 87(12): 979-88, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20029534

ABSTRACT

After a myocardial infarct (MI), a variety of mechanisms contribute to progressive cardiac remodeling and dysfunction. Progressive activation of central sympathoexcitatory pathways appears to depend on a neuromodulatory pathway, involving local production of aldosterone and release of endogenous ouabain-like compounds ('ouabain') possibly from magnocellular neurons in the supraoptic and paraventricular nuclei. 'Ouabain' may lower the membrane potential of neurons and thereby enhance activity of angiotensinergic pathways. These central pathways appear to coordinate progressive activation of several peripheral mechanisms such as sympathetic tone and circulating and cardiac renin-angiotensin-aldosterone system (RAAS). Central blockade of aldosterone production, mineralocorticoid receptors, 'ouabain' activity, or AT1 receptors similarly prevents activation of these peripheral mechanisms. Cardiac remodeling after MI involves progressive left ventricular dilation, fibrosis, and decrease in contractile performance. Central blockade of this neuromodulatory pathway causes a marked attenuation of the remodeling and dysfunction, presumably by inhibiting increases in (cardiac) sympathetic activity and RAAS. At the cellular level, these systems may contribute to the cardiac remodeling by activating proinflammatory cytokines and cardiac myocyte apoptosis. New therapeutic approaches, specifically preventing activation of this brain neuromodulatory pathway, may lead to more optimal and specific approaches to the prevention of heart failure after MI.


Subject(s)
Myocardial Infarction/physiopathology , Renin-Angiotensin System/physiology , Ventricular Remodeling/physiology , Aldosterone/physiology , Animals , Apoptosis/physiology , Brain/physiopathology , Humans , Myocarditis/physiopathology , Myocytes, Cardiac/physiology
15.
Am J Physiol Heart Circ Physiol ; 297(3): H968-75, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19617416

ABSTRACT

In rats, both central and systemic ANG II type 1 (AT(1)) receptor blockade attenuate sympathetic hyperactivity, but central blockade more effectively attenuates left ventricular (LV) dysfunction post-myocardial infarction (MI). In protocol I, we examined whether functional effects on cardiac load may play a role and different cardiac effects disappear after withdrawal of the blockade. Wistar rats were infused for 4 wk post-MI intracerebroventricularly (1 mg.kg(-1).day(-1)) or injected subcutaneously daily (100 mg x kg(-1) x day(-1)) with losartan. LV dimensions and function were assessed at 4 wk and at 6 wk post-MI, i.e., 2 wk after discontinuing treatments. At 4 and 6 wk post-MI, LV dimensions were increased and ejection fraction was decreased. Intracerebroventricular but not subcutaneous losartan significantly improved these parameters. At 6 wk, LV peak systolic pressure (LVPSP) and maximal or minimal first derivative of change in pressure over time (dP/dt(max/min)) were decreased and LV end-diastolic pressure (LVEDP) was increased. All four indexes were improved by previous intracerebroventricular losartan, whereas subcutaneous losartan improved LVEDP only. In protocol II, we evaluated effects of oral instead of subcutaneous administration of losartan for 4 wk post-MI. Losartan ( approximately 200 mg x kg(-1) x day(-1)) either via drinking water or by gavage similarly decreased AT(1) receptor binding densities in brain nuclei and improved LVEDP but further decreased LVPSP and dP/dt(max). These results indicate that effects on cardiac load by peripheral AT(1) receptor blockade or the pharmacokinetic profile of subcutaneous versus oral dosing do not contribute to the different cardiac effects of central versus systemic AT(1) receptor blockade post-MI.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Losartan/pharmacology , Myocardial Infarction/metabolism , Receptor, Angiotensin, Type 1/metabolism , Sympathetic Nervous System/physiology , Administration, Oral , Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Animals , Echocardiography , Injections, Intraventricular , Injections, Subcutaneous , Losartan/pharmacokinetics , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Rats , Rats, Wistar , Renin-Angiotensin System/physiology , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology , Ventricular Remodeling/physiology
16.
Curr Heart Fail Rep ; 6(2): 81-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19486591

ABSTRACT

Following a myocardial infarction (MI), increases in plasma angiotensin II may activate central nervous system (CNS) pathways and thereby peripheral mechanisms (eg, sympathetic activity and the circulating/cardiac renin-angiotensin-aldosterone system ). Plasma angiotensin II may directly activate CNS pathways through the subfornical organ and chronically enhance activity by way of a neuromodulatory system. The latter involves an increase in CNS aldosterone-causing "ouabain" release (eg, from magnocellular neurons of the supraoptic and paraventricular nuclei). "Ouabain" may lower membrane potential, thereby enhancing activity of angiotensinergic pathways. The resulting increases in sympathetic activity, and circulating/cardiac RAAS contributes to progressive left ventricular remodeling and dysfunction after MI and can be largely prevented by central administration of a blocker for any of the components of this neuromodulatory system. These new insights into the crucial role of the CNS may lead to new therapeutic approaches for the prevention of heart failure after MI with minimal peripheral adverse effects.


Subject(s)
Angiotensin II/blood , Myocardial Infarction/physiopathology , Renin-Angiotensin System/physiology , Sympathetic Nervous System/physiology , Ventricular Dysfunction, Left/physiopathology , Brain/physiology , Heart Failure/prevention & control , Humans , Myocardial Infarction/complications , Ventricular Remodeling/physiology
17.
Am J Physiol Regul Integr Comp Physiol ; 296(4): R994-R1000, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19118098

ABSTRACT

In Dahl salt-sensitive (S) rats, high salt intake increases cerebrospinal fluid (CSF) Na(+) concentration ([Na(+)]) and blood pressure (BP). Intracerebroventricular (ICV) infusion of a mineralocorticoid receptor (MR) blocker prevents the hypertension. To assess the role of aldosterone locally produced in the brain, we evaluated the effects of chronic central blockade with the aldosterone synthase inhibitor FAD286 and the MR blocker spironolactone on changes in aldosterone and corticosterone content in the hypothalamus and the increase in CSF [Na(+)] and hypertension induced by high salt intake in Dahl S rats. After 4 wk of high salt intake, plasma aldosterone and corticosterone were not changed, but hypothalamic aldosterone increased by approximately 35% and corticosterone tended to increase in Dahl S rats, whereas both steroids decreased by approximately 65% in Dahl salt-resistant rats. In Dahl S rats fed the high-salt diet, ICV infusion of FAD286 or spironolactone did not affect the increase in CSF [Na(+)]. ICV infusion of FAD286 prevented the increase in hypothalamic aldosterone and 30 mmHg of the 50-mmHg BP increase induced by high salt intake. ICV infusion of spironolactone fully prevented the salt-induced hypertension. These results suggest that, in Dahl S rats, high salt intake increases aldosterone synthesis in the hypothalamus and aldosterone acts as the main MR agonist activating central pathways contributing to salt-induced hypertension.


Subject(s)
Aldosterone/metabolism , Blood Pressure , Corticosterone/metabolism , Hypertension/enzymology , Hypothalamus/enzymology , Receptors, Mineralocorticoid/metabolism , Sodium/cerebrospinal fluid , Steroid Hydroxylases/metabolism , Aldosterone/blood , Animals , Blood Pressure/drug effects , Corticosterone/blood , Cytochrome P-450 CYP11B2 , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Heart Rate , Hippocampus/enzymology , Hypertension/etiology , Hypertension/physiopathology , Hypertension/prevention & control , Hypothalamus/drug effects , Male , Mineralocorticoid Receptor Antagonists/administration & dosage , Rats , Rats, Inbred Dahl , Sodium Chloride, Dietary/metabolism , Spironolactone/administration & dosage , Steroid Hydroxylases/antagonists & inhibitors , Time Factors
18.
Cardiovasc Res ; 81(3): 574-81, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-18689429

ABSTRACT

AIMS: Blockade of mineralocorticoid receptors in the central nervous system (CNS) prevents sympathetic hyperactivity and improves left ventricle (LV) function in rats post-myocardial infarction (MI). We examined whether aldosterone produced locally in the brain may contribute to the activation of mineralocorticoid receptors in the CNS. METHODS AND RESULTS: Two days after coronary artery ligation, Wistar rats received an intra-cerebroventricular (icv) infusion via osmotic mini-pumps of the aldosterone synthase inhibitor FAD286 at 100 microg/kg/day or vehicle for 4 weeks. LV function was assessed by echocardiography at 2 and 4 weeks, and by Millar catheter at 4 weeks. At 4 weeks post-MI, aldosterone in the hippocampus was increased by 70% and tended to increase in the hypothalamus by 20%. These increases were prevented by FAD286. Across groups, aldosterone in the hippocampus and hypothalamus showed a high correlation. There were no differences in brain corticosterone levels. Compared to sham rats, at both 2 and 4 weeks post-MI rats treated with vehicle showed increased LV dimensions and decreased LV ejection fraction. Icv infusion of FAD286 attenuated these changes in LV dimensions and ejection fraction by approximately 30%. At 4 weeks post-MI, LV peak systolic pressure (LVPSP) and dP/dt(max/min) were decreased and LV end-diastolic pressure (LVEDP) was increased. In rats treated with icv FAD286, LVPSP and dP/dt(min) remained normal and LVEDP and dP/dt(max) were markedly improved. Post-MI increases in cardiac fibrosis and cardiomyocyte diameter were substantially attenuated by icv FAD286. CONCLUSION: These data suggest that aldosterone produced locally in the brain acts as the main agonist of mineralocorticoid receptors in the CNS and contributes substantially to the progressive heart failure post MI.


Subject(s)
Aldosterone/metabolism , Enzyme Inhibitors/administration & dosage , Hippocampus/drug effects , Hypothalamus/drug effects , Myocardial Infarction/drug therapy , Steroid Hydroxylases/antagonists & inhibitors , Ventricular Dysfunction, Left/prevention & control , Ventricular Remodeling/drug effects , Aldosterone/blood , Animals , Cardiac Catheterization , Corticosterone/metabolism , Cytochrome P-450 CYP11B2 , Disease Models, Animal , Echocardiography , Fibrosis , Hippocampus/enzymology , Hypothalamus/enzymology , Infusions, Parenteral , Myocardial Infarction/complications , Myocardial Infarction/enzymology , Myocardial Infarction/physiopathology , Myocardium/pathology , Rats , Rats, Wistar , Steroid Hydroxylases/metabolism , Stroke Volume/drug effects , Time Factors , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/physiopathology , Ventricular Pressure/drug effects
19.
Am J Physiol Regul Integr Comp Physiol ; 295(1): R166-72, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18495825

ABSTRACT

In Wistar rats, increasing cerebrospinal fluid (CSF) Na+ concentration ([Na+]) by intracerebroventricular (ICV) infusion of hypertonic saline causes sympathetic hyperactivity and hypertension that can be prevented by blockade of brain mineralocorticoid receptors (MR). To assess the role of aldosterone produced locally in the brain in the activation of MR in the central nervous system (CNS), Wistar rats were infused ICV with artificial CSF (aCSF), Na+ -rich (800 mmol/l) aCSF, aCSF plus the aldosterone synthase inhibitor FAD286 (100 microg x kg(-1) x day(-1)), or Na+ -rich aCSF plus FAD286. After 2 wk of infusion, rats treated with Na+ -rich aCSF exhibited significant increases in aldosterone and corticosterone content in the hypothalamus but not in the hippocampus, as well as increases in resting blood pressure (BP) and sympathoexcitatory responses to air stress, and impairment of arterial baroreflex function. Concomitant ICV infusion of FAD286 prevented the Na+ -induced increase in hypothalamic aldosterone but not corticosterone and prevented most of the increases in resting BP and sympathoexcitatory and pressor responses to air stress and the baroreflex impairment. FAD286 had no effects in rats infused with ICV aCSF. In another set of rats, 24-h BP and heart rate were recorded via telemetry before and during a 14-day ICV infusion of Na+ -rich aCSF with or without FAD286. Na+ -rich aCSF without FAD286 caused sustained increases ( approximately 10 mmHg) in resting mean arterial pressure that were absent in the rats treated with FAD286. These data suggest that in Wistar rats, an increase in CSF [Na+] may increase the biosynthesis of corticosterone and aldosterone in the hypothalamus, and mainly aldosterone activates MR in the CNS leading to sympathetic hyperactivity and hypertension.


Subject(s)
Cytochrome P-450 CYP11B2/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hypertension/chemically induced , Sodium/pharmacology , Sympathetic Nervous System/drug effects , Aldosterone/blood , Aldosterone/metabolism , Angiotensin II/blood , Angiotensin II/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Brain/drug effects , Brain/metabolism , Corticosterone/blood , Corticosterone/metabolism , Enzyme Inhibitors/administration & dosage , Male , Rats , Rats, Wistar , Sodium/administration & dosage , Sympathetic Nervous System/physiopathology
20.
J Cardiovasc Pharmacol ; 51(6): 565-72, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18496146

ABSTRACT

BACKGROUND: The brain renin-angiotensin system contributes significantly to progressive left ventricular (LV) dysfunction in rats after myocardial infarction (MI). The present study evaluated the effects of central versus peripheral plus central angiotensin-converting enzyme (ACE) blockade on sympathetic activity, and LV anatomy and function after MI. METHODS: Wistar rats were treated for 4 weeks after MI with the lipophilic ACE inhibitor trandolapril at 5 mg/kg/day or the hydrophilic blocker lisinopril at 50 mg/kg/day by once daily subcutaneous injection, or with a central infusion of lisinopril at 0.1 mg/kg/day. RESULTS: At 24 hours after the last dose, subcutaneous trandolapril caused 70% to 80% ACE inhibition in both brain and kidneys; lisinopril caused 10% to 20% less. Central infusion of lisinopril caused 70% inhibition of brain ACE and minimal (6%) inhibition in the kidneys. All three treatments similarly improved sympathetic reactivity and arterial baroreflex function. All three treatments lowered cardiac Ang I and II, and similarly attenuated the increases in LV end diastolic pressure, circumference, and fibrosis. Both subcutaneous treatments further decreased LV peak systolic pressure and dP/dt max, whereas icv lisinopril caused no change. CONCLUSION: Despite marked differences in the extent of peripheral blockade, all three treatments similarly affected sympathetic activity and decreased cardiac Ang II, preload and remodeling after MI. One may speculate that central and peripheral ACE-mediated mechanisms are sequential and therefore only minor additional effects of peripheral ACE blockade are noted.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Lisinopril/pharmacology , Myocardial Infarction/complications , Ventricular Dysfunction, Left/prevention & control , Angiotensin I/metabolism , Angiotensin II/metabolism , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Animals , Baroreflex/drug effects , Brain/drug effects , Brain/enzymology , Fibrosis , Hemodynamics , Indoles/administration & dosage , Infusions, Intravenous , Injections, Subcutaneous , Kidney/drug effects , Kidney/enzymology , Lisinopril/administration & dosage , Male , Myocardial Infarction/pathology , Myocardium/pathology , Rats , Rats, Wistar , Renin-Angiotensin System/drug effects , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiopathology , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...