Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Am J Physiol Heart Circ Physiol ; 314(4): H796-H804, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351470

ABSTRACT

The central nervous system plays an important role in essential hypertension in humans and in animal models of hypertension through modulation of sympathetic activity and Na+ and body fluid homeostasis. Data from animal models of hypertension suggest that the renin-angiotensin system in the subfornical organ (SFO) of the brain is critical for hypertension development. We recently reported that the brain (pro)renin receptor (PRR) is a novel component of the brain renin-angiotensin system and could be a key initiator of the pathogenesis of hypertension. Here, we examined the expression level and cellular distribution of PRR in the SFO of postmortem human brains to assess its association with the pathogenesis of human hypertension. Postmortem SFO tissues were collected from hypertensive and normotensive human subjects. Immunolabeling for the PRR and a retrospective analysis of clinical data were performed. We found that human PRR was prominently expressed in most neurons and microglia, but not in astrocytes, in the SFO. Importantly, PRR levels in the SFO were elevated in hypertensive subjects. Moreover, PRR immunoreactivity was significantly correlated with systolic blood pressure but not body weight, age, or diastolic blood pressure. Interestingly, this correlation was independent of antihypertensive drug therapy. Our data indicate that PRR in the SFO may be a key molecular player in the pathogenesis of human hypertension and, as such, could be an important focus of efforts to understand the neurogenic origin of hypertension. NEW & NOTEWORTHY This study provides evidence that, in the subfornical organ of the human brain, the (pro)renin receptor is expressed in neurons and microglia cells but not in astrocytes. More importantly, (pro)renin receptor immunoreactivity in the subfornical organ is increased in hypertensive humans and is significantly correlated with systolic blood pressure.


Subject(s)
Hypertension/enzymology , Receptors, Cell Surface/analysis , Subfornical Organ/enzymology , Vacuolar Proton-Translocating ATPases/analysis , Aged , Autopsy , Blood Pressure , Female , Humans , Hypertension/diagnosis , Hypertension/physiopathology , Immunohistochemistry , Male , Microglia/enzymology , Middle Aged , Neurons/enzymology , Retrospective Studies , Subfornical Organ/physiopathology , Up-Regulation
2.
Am J Physiol Regul Integr Comp Physiol ; 314(3): R447-R458, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29167166

ABSTRACT

We previously reported that microinjection of the proinflammatory cytokine interleukin-1ß (IL-1ß) into the subfornical organ (SFO) elicits a pressor response accompanied by increases in inflammation and renin-angiotensin system (RAS) activity in the SFO and hypothalamic paraventricular nucleus (PVN). The present study sought to determine whether blood-borne IL-1ß induces similar neurochemical changes in the SFO and PVN and, if so, whether increased inflammation and RAS activity at the SFO level orchestrate the sympathoexcitatory response to circulating IL-1ß. In urethane-anesthetized male Sprague-Dawley rats, intravenous injection of IL-1ß (500 ng) increased blood pressure, heart rate, renal sympathetic nerve activity, and mRNA for angiotensin-converting enzyme, angiotensin II type 1a receptor, cyclooxygenase-2, tumor necrosis factor-α, and IL-1ß, as well as the tumor necrosis factor-α p55 receptor and the IL-1 receptor, in the SFO and PVN. Pretreatment with SFO microinjections of the angiotensin II type 1a receptor blocker losartan (1 µg), the angiotensin-converting enzyme inhibitor captopril (1 µg), or the cyclooxygenase-2 inhibitor NS-398 (2 µg) attenuated expression of these excitatory mediators in the SFO and downstream in the PVN and the IL-1ß-induced pressor responses. An SFO lesion minimized the IL-1ß-induced expression of inflammatory and RAS components as well as c-Fos, an indicator of neuronal excitation, in the PVN. These studies demonstrate that circulating IL-1ß, which increases in cardiovascular disorders such as hypertension and heart failure, acts on the SFO to increase inflammation and RAS activity in the SFO and PVN and that intervening in these neurochemical processes in the SFO can significantly reduce the sympathetic response.


Subject(s)
Arterial Pressure/drug effects , Heart Rate/drug effects , Heart/innervation , Interleukin-1beta/administration & dosage , Kidney/innervation , Paraventricular Hypothalamic Nucleus/drug effects , Subfornical Organ/drug effects , Sympathetic Nervous System/drug effects , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Animals , Cyclooxygenase 2 Inhibitors/administration & dosage , Injections, Intravenous , Injections, Intraventricular , Interleukin-1beta/blood , Male , Microinjections , Paraventricular Hypothalamic Nucleus/physiopathology , Rats, Sprague-Dawley , Renin-Angiotensin System/drug effects , Subfornical Organ/physiopathology , Subfornical Organ/surgery , Sympathetic Nervous System/physiopathology
3.
Am J Physiol Heart Circ Physiol ; 313(4): H744-H756, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28710070

ABSTRACT

In systolic heart failure (HF), circulating proinflammatory cytokines upregulate inflammation and renin-angiotensin system (RAS) activity in cardiovascular regions of the brain, contributing to sympathetic excitation and cardiac dysfunction. Important among these is the subfornical organ (SFO), a forebrain circumventricular organ that lacks an effective blood-brain barrier and senses circulating humors. We hypothesized that the tumor necrosis factor-α (TNF-α) receptor 1 (TNFR1) in the SFO contributes to sympathetic excitation and cardiac dysfunction in HF rats. Rats received SFO microinjections of a TNFR1 shRNA or a scrambled shRNA lentiviral vector carrying green fluorescent protein, or vehicle. One week later, some rats were euthanized to confirm the accuracy of the SFO microinjections and the transfection potential of the lentiviral vector. Other rats underwent coronary artery ligation (CL) to induce HF or a sham operation. Four weeks after CL, vehicle- and scrambled shRNA-treated HF rats had significant increases in TNFR1 mRNA and protein, NF-κB activity, and mRNA for inflammatory mediators, RAS components and c-Fos protein in the SFO and downstream in the hypothalamic paraventricular nucleus, along with increased plasma norepinephrine levels and impaired cardiac function, compared with vehicle-treated sham-operated rats. In HF rats treated with TNFR1 shRNA, TNFR1 was reduced in the SFO but not paraventricular nucleus, and the central and peripheral manifestations of HF were ameliorated. In sham-operated rats treated with TNFR1 shRNA, TNFR1 expression was also reduced in the SFO but there were no other effects. These results suggest a key role for TNFR1 in the SFO in the pathophysiology of systolic HF.NEW & NOTEWORTHY Activation of TNF-α receptor 1 in the subfornical organ (SFO) contributes to sympathetic excitation in heart failure rats by increasing inflammation and renin-angiotensin system activity in the SFO and downstream in the hypothalamic paraventricular nucleus. Cytokine receptors in the SFO may be a target for central intervention in cardiovascular conditions characterized by peripheral inflammation.


Subject(s)
Coronary Circulation/genetics , Heart Failure/genetics , Heart Failure/physiopathology , Receptors, Tumor Necrosis Factor, Type I/genetics , Subfornical Organ/physiopathology , Sympathetic Nervous System/physiopathology , Animals , Electrocardiography , Gene Knockdown Techniques , Hemodynamics/drug effects , Male , NF-kappa B/metabolism , Norepinephrine/blood , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Renin-Angiotensin System , Tumor Necrosis Factor-alpha/pharmacology
4.
Acta Physiol (Oxf) ; 217(1): 33-44, 2016 May.
Article in English | MEDLINE | ID: mdl-26639993

ABSTRACT

AIM: Endoplasmic reticulum (ER) stress in the brain subfornical organ (SFO), a key cardiovascular regulatory centre, has been implicated in angiotensin (ANG) II-induced hypertension in males; however, the contribution of ER stress to ANG II-induced hypertension in females is unknown. Female hormones have been shown to prevent ER stress in the periphery. We tested the hypothesis that females are less susceptible to ANG II-induced SFO ER stress than males, leading to sex differences in hypertension. METHODS: Male, intact and ovariectomized (OVX) female rats received a continuous 2-week subcutaneous infusion of ANG II or saline. Additional male, intact and OVX female rats received intracerebroventricular (ICV) injection of ER stress inducer tunicamycin. RESULTS: ANG II, but not saline, increased blood pressure (BP) in both males and females, but intact females exhibited smaller increase in BP and less depressor response to ganglionic blockade compared with males or OVX females. Molecular studies revealed that ANG II elevated expression of ER stress biomarkers and Fra-like activity in the SFO in both males and females; however, elevations in these parameters were less in intact females than in males or OVX females. Moreover, ICV tunicamycin induced smaller elevation in BP and less increase in expression of ER stress biomarkers in the SFO in intact females compared with males or OVX females. CONCLUSION: The results suggest that differences in ANG II-induced brain ER stress between males and females contribute to sex differences in ANG II-mediated hypertension and that oestrogen protects females against ANG II-induced brain ER stress.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Hypertension/physiopathology , Subfornical Organ/physiopathology , Angiotensin II/toxicity , Animals , Blotting, Western , Endoplasmic Reticulum Stress/drug effects , Female , Hypertension/chemically induced , Immunohistochemistry , Male , Ovariectomy , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Sex Characteristics , Vasoconstrictor Agents/toxicity
5.
Hypertension ; 65(6): 1341-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25895586

ABSTRACT

Endoplasmic reticulum stress has become an important mechanism in hypertension. We examined the role of endoplasmic reticulum stress in mediating the increased saline-intake and hypertensive effects in response to deoxycorticosterone acetate (DOCA)-salt. Intracerebroventricular delivery of the endoplasmic reticulum stress-reducing chemical chaperone tauroursodeoxycholic acid did not affect the magnitude of hypertension, but markedly decreased saline-intake in response to DOCA-salt. Increased saline-intake returned after tauroursodeoxycholic acid was terminated. Decreased saline-intake was also observed after intracerebroventricular infusion of 4-phenylbutyrate, another chemical chaperone. Immunoreactivity to CCAAT homologous binding protein, a marker of irremediable endoplasmic reticulum stress, was increased in the subfornical organ and supraoptic nucleus of DOCA-salt mice, but the signal was absent in control and CCAAT homologous binding protein-deficient mice. Electron microscopy revealed abnormalities in endoplasmic reticulum structure (decrease in membrane length, swollen membranes, and decreased ribosome numbers) in the subfornical organ consistent with endoplasmic reticulum stress. Subfornical organ-targeted adenoviral delivery of GRP78, a resident endoplasmic reticulum chaperone, decreased DOCA-salt-induced saline-intake. The increase in saline-intake in response to DOCA-salt was blunted in CCAAT homologous binding protein-deficient mice, but these mice exhibited a normal hypertensive response. We conclude that (1) brain endoplasmic reticulum stress mediates the saline-intake, but not blood pressure response to DOCA-salt, (2) DOCA-salt causes endoplasmic reticulum stress in the subfornical organ, which when attenuated by GRP78 blunts saline-intake, and (3) CCAAT homologous binding protein may play a functional role in DOCA-salt-induced saline-intake. The results suggest a mechanistic distinction between the importance of endoplasmic reticulum stress in mediating effects of DOCA-salt on saline-intake and blood pressure.


Subject(s)
Brain/metabolism , Desoxycorticosterone Acetate/pharmacology , Endoplasmic Reticulum Stress/drug effects , Hypertension/physiopathology , Sodium Chloride/pharmacology , Analysis of Variance , Animals , Blood Pressure/drug effects , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/physiology , Infusions, Intraventricular , Mice , Mice, Inbred C57BL , Random Allocation , Reference Values , Sensitivity and Specificity , Sodium Chloride/metabolism , Statistics, Nonparametric , Subfornical Organ/drug effects , Subfornical Organ/physiopathology
6.
Am J Physiol Regul Integr Comp Physiol ; 308(6): R507-16, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25552661

ABSTRACT

The present study tested the hypotheses that 1) ERα in the brain plays a key role in the estrogen-protective effects against ANG II-induced hypertension, and 2) that the subfornical organ (SFO) is a key site where ERα mediates these protective actions. In this study, a "floxed" ERα transgenic mouse line (ERα(flox)) was used to create models in which ERα was knocked down in the brain or just in the SFO. Female mice with ERα ablated in the nervous system (Nestin-ERα(-) mice) showed greater increases in blood pressure (BP) in response to ANG II. Furthermore, females with ERα knockdown specifically in the SFO [SFO adenovirus-Cre (Ad-Cre) injected ERα(flox) mice] also showed an enhanced pressor response to ANG II. Immunohistochemical (IHC), RT-PCR, and Western blot analyses revealed a marked reduction in the expression of ERα in nervous tissues and, in particular, in the SFO. These changes were not present in peripheral tissues in Nestin-ERα(-) mice or Ad-Cre-injected ERα(flox) mice. mRNA expression of components of the renin-angiotensin system in the lamina terminalis were upregulated in Nestin-ERα(-) mice. Moreover, ganglionic blockade on day 7 after ANG II infusions resulted in a greater reduction of BP in Nestin-ERα(-) mice or SFO Ad-Cre-injected mice, suggesting that knockdown of ERα in the nervous system or the SFO alone augments central ANG II-induced increase in sympathetic tone. The results indicate that interfering with the action of estrogen on SFO ERα is sufficient to abolish the protective effects of estrogen against ANG II-induced hypertension.


Subject(s)
Angiotensin II , Blood Pressure , Estrogen Receptor alpha/deficiency , Gene Knockdown Techniques , Hypertension/metabolism , Subfornical Organ/metabolism , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Disease Models, Animal , Estrogen Receptor alpha/genetics , Female , Ganglionic Blockers/pharmacology , Genotype , Heart Rate , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Hypertension/prevention & control , Male , Mice, Knockout , Nestin/genetics , Nestin/metabolism , Phenotype , Subfornical Organ/physiopathology , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology
7.
Am J Physiol Regul Integr Comp Physiol ; 308(5): R370-8, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25519733

ABSTRACT

Endoplasmic reticulum (ER) stress was previously reported to contribute to neurogenic hypertension while neuronal angiotensin-converting enzyme type 2 (ACE2) overexpression blunts the disease. To assess which brain regions are important for ACE2 beneficial effects and the contribution of ER stress to neurogenic hypertension, we first used transgenic mice harboring a floxed neuronal hACE2 transgene (SL) and tested the impact of hACE2 knockdown in the subfornical organ (SFO) and paraventricular nucleus (PVN) on deoxycorticosterone acetate (DOCA)-salt hypertension. SL and nontransgenic (NT) mice underwent DOCA-salt or sham treatment while infected with an adenoassociated virus (AAV) encoding Cre recombinase (AAV-Cre) or a control virus (AAV-green fluorescent protein) to the SFO or PVN. DOCA-salt-induced hypertension was reduced in SL mice, with hACE2 overexpression in the brain. This reduction was only partially blunted by knockdown of hACE2 in the SFO or PVN, suggesting that both regions are involved but not essential for ACE2 regulation of blood pressure (BP). DOCA-salt treatment did not increase the protein levels of ER stress and autophagy markers in NT mice, despite a significant increase in BP. In addition, these markers were not affected by hACE2 overexpression in the brain, despite a significant reduction of hypertension in SL mice. To further assess the role of ER stress in neurogenic hypertension, NT mice were infused intracerebroventricularlly with tauroursodeoxycholic acid (TUDCA), an ER stress inhibitor, during DOCA-salt treatment. However, TUDCA infusion failed to blunt the development of hypertension in NT mice. Our data suggest that brain ER stress does not contribute to DOCA-salt hypertension and that ACE2 blunts neurogenic hypertension independently of ER stress.


Subject(s)
Brain/enzymology , Desoxycorticosterone Acetate , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/enzymology , Hypertension/prevention & control , Peptidyl-Dipeptidase A/metabolism , Sodium Chloride, Dietary , Angiotensin-Converting Enzyme 2 , Animals , Biomarkers/metabolism , Blood Pressure , Brain/drug effects , Brain/physiopathology , Disease Models, Animal , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Stress/drug effects , Gene Knockdown Techniques , Humans , Hypertension/enzymology , Hypertension/genetics , Hypertension/physiopathology , Infusions, Intraventricular , Mice, Inbred C57BL , Mice, Transgenic , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/physiopathology , Peptidyl-Dipeptidase A/genetics , Subfornical Organ/enzymology , Subfornical Organ/physiopathology , Taurochenodeoxycholic Acid/administration & dosage , Time Factors , Up-Regulation
8.
Am J Physiol Regul Integr Comp Physiol ; 308(4): R238-49, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25519738

ABSTRACT

It is critical for cells to maintain a homeostatic balance of water and electrolytes because disturbances can disrupt cellular function, which can lead to profound effects on the physiology of an organism. Dehydration can be classified as either intra- or extracellular, and different mechanisms have developed to restore homeostasis in response to each. Whereas the renin-angiotensin system (RAS) is important for restoring homeostasis after dehydration, the pathways mediating the responses to intra- and extracellular dehydration may differ. Thirst responses mediated through the angiotensin type 1 receptor (AT1R) and angiotensin type 2 receptors (AT2R) respond to extracellular dehydration and intracellular dehydration, respectively. Intracellular signaling factors, such as protein kinase C (PKC), reactive oxygen species (ROS), and the mitogen-activated protein (MAP) kinase pathway, mediate the effects of central angiotensin II (ANG II). Experimental evidence also demonstrates the importance of the subfornical organ (SFO) in mediating some of the fluid intake effects of central ANG II. The purpose of this review is to highlight the importance of the SFO in mediating fluid intake responses to dehydration and ANG II.


Subject(s)
Angiotensin II/metabolism , Blood Pressure , Dehydration/metabolism , Drinking , Renin-Angiotensin System , Subfornical Organ/metabolism , Animals , Dehydration/physiopathology , Humans , Receptors, Angiotensin/metabolism , Signal Transduction , Subfornical Organ/physiopathology , Water-Electrolyte Balance
9.
Am J Physiol Heart Circ Physiol ; 308(5): H435-46, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25539713

ABSTRACT

Sleep apnea is associated with hypertension. The mechanisms contributing to a sustained increase in mean arterial pressure (MAP) even during normoxic awake-state remain unknown. Rats exposed to chronic intermittent hypoxia for 7 days, a model of the hypoxemia associated with sleep apnea, exhibit sustained increases in MAP even during the normoxic dark phase. Activation of the renin-angiotensin system (RAS) has been implicated in chronic intermittent hypoxia (CIH) hypertension. Since the subfornical organ (SFO) serves as a primary target for the central actions of circulating ANG II, we tested the effects of ANG II type 1a receptor (AT1aR) knockdown in the SFO on the sustained increase in MAP in this CIH model. Adeno-associated virus carrying green fluorescent protein (GFP) and small-hairpin RNA against either AT1aR or a scrambled control sequence (SCM) was stereotaxically injected in the SFO of rats. After recovery, MAP, heart rate, respiratory rate, and activity were continuously recorded using radiotelemetry. In the normoxic groups, the recorded variables did not deviate from the baseline values. Both CIH groups exhibited significant increases in MAP during CIH exposures (P < 0.05). During the normoxic dark phase in the CIH groups, only the SCM-injected group exhibited a sustained increase in MAP (P < 0.05). The AT1aR-CIH group showed significant decreases in FosB/ΔFosB staining in the median preoptic nucleus and the paraventricular nuclei of the hypothalamus compared with the SCM-CIH group. Our data indicate that AT1aRs in the SFO are critical for the sustained elevation in MAP and increased FosB/ΔFosB expression in forebrain autonomic nuclei associated with CIH.


Subject(s)
Blood Pressure , Hypertension/metabolism , Hypoxia/metabolism , Receptor, Angiotensin, Type 1/metabolism , Sleep Apnea Syndromes/metabolism , Subfornical Organ/metabolism , Animals , Hypertension/etiology , Hypertension/physiopathology , Hypoxia/complications , Hypoxia/physiopathology , Male , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Sleep Apnea Syndromes/complications , Sleep Apnea Syndromes/physiopathology , Subfornical Organ/physiopathology
10.
Am J Physiol Heart Circ Physiol ; 306(1): H121-31, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24163080

ABSTRACT

Exercise training (ExT) has been shown to reduce sympathetic drive during heart failure (HF). The subfornical organ (SFO) is involved in the neural control of sympathetic drive. We hypothesized that an activated SFO contributes to enhanced sympathetic activity in HF. We also postulated that ExT would reduce the activation of the SFO and its contribution to the sympathetic drive during HF. Sprague-Dawley rats were subjected to coronary artery ligation to induce HF. Rats were assigned to ExT for 3-4 wk. Rats with HF had a 2.5-fold increase in FosB-positive cells in the SFO compared with sham-operated rats, and this was normalized by ExT. Microinjection of ANG II (100 pmol) into the SFO resulted in a greater increase in renal sympathetic nerve activity (RSNA), blood pressure, and heart rate in the HF group than in the sham-operated group. These responses were normalized after ExT (change in RSNA: 23 ± 3% vs. 8 ± 2%). ExT also abolished the decrease in RSNA in HF rats after the microinjection of losartan (200 pmol) into the SFO (-21 ± 4% vs. -2 ± 3%). Finally, there was elevated mRNA (5-fold) and protein expression (43%) of ANG II type 1 receptors in the SFO of rats with HF, which were reversed after ExT. These data suggest that the enhanced activity of the SFO by elevated tonic ANG II contributes to the enhanced sympathoexcitation exhibited in HF. The decrease in ANG II type 1 receptor expression in the SFO by ExT may be responsible for reversing the neuronal activation in the SFO and SFO-mediated sympathoexcitation in rats with HF.


Subject(s)
Heart Failure/physiopathology , Heart/innervation , Physical Exertion , Subfornical Organ/physiopathology , Sympathetic Nervous System/physiopathology , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Blood Pressure , Heart Failure/metabolism , Heart Rate , Kidney/innervation , Losartan/pharmacology , Male , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Subfornical Organ/drug effects , Subfornical Organ/metabolism
11.
Am J Physiol Heart Circ Physiol ; 305(10): H1451-61, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24014678

ABSTRACT

Regulation of blood pressure by angiotensin II (ANG II) is a process that involves the reactive oxygen species (ROS) and calcium. We have shown that ANG-II type 1 receptor (AT1R) and prostaglandin E2 (PGE2) type 1 receptors (EP1R) are required in the subfornical organ (SFO) for ROS-mediated hypertension induced by slow-pressor ANG-II infusion. However, the signaling pathway associated with this process remains unclear. We sought to determine mechanisms underlying the ANG II-induced ROS and calcium influx in mouse SFO cells. Ultrastructural studies showed that cyclooxygenase 1 (COX-1) codistributes with AT1R in the SFO, indicating spatial proximity. Functional studies using SFO cells revealed that ANG II potentiated PGE2 release, an effect dependent on AT1R, phospholipase A2 (PLA2) and COX-1. Furthermore, both ANG II and PGE2 increased ROS formation. While the increase in ROS initiated by ANG II, but not PGE2, required the activation of the AT1R/PLA2/COX-1 pathway, both ANG II and PGE2 were dependent on EP1R and Nox2 as downstream effectors. Finally, ANG II potentiated voltage-gated L-type Ca(2+) currents in SFO neurons via the same signaling pathway required for PGE2 production. Blockade of EP1R and Nox2-derived ROS inhibited ANG II and PGE2-mediated Ca(2+) currents. We propose a mechanism whereby ANG II increases COX-1-derived PGE2 through the AT1R/PLA2 pathway, which promotes ROS production by EP1R/Nox2 signaling in the SFO. ANG II-induced ROS are coupled with Ca(2+) influx in SFO neurons, which may influence SFO-mediated sympathoexcitation. Our findings provide the first evidence of a spatial and functional framework that underlies ANG-II signaling in the SFO and reveal novel targets for antihypertensive therapies.


Subject(s)
Angiotensin II/metabolism , Calcium Signaling , Cyclooxygenase 1/metabolism , Dinoprostone/metabolism , Hypertension/enzymology , Membrane Proteins/metabolism , Neurons/enzymology , Reactive Oxygen Species/metabolism , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Subfornical Organ/enzymology , Action Potentials , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Blood Pressure , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Hypertension/pathology , Hypertension/physiopathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neurons/drug effects , Neurons/ultrastructure , Phospholipases A2/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, Prostaglandin E, EP1 Subtype/deficiency , Receptors, Prostaglandin E, EP1 Subtype/genetics , Subfornical Organ/drug effects , Subfornical Organ/physiopathology , Subfornical Organ/ultrastructure
12.
Hypertension ; 62(5): 866-71, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24001896

ABSTRACT

Hypertension contributes to multiple forms of cardiovascular disease and thus morbidity and mortality. The mechanisms inducing hypertension remain unclear although the involvement of homeostatic systems, such as the renin-angiotensin and sympathetic nervous systems, is established. A pivotal role of the angiotensin type 1 receptor in the proximal tubule of the kidney for the development of experimental hypertension is established. Yet, other systems are involved. This study tests whether the expression of angiotensin type 1A receptors in catecholaminergic cells contributes to hypertension development. Using a Cre-lox approach, we deleted the angiotensin type 1A receptor from all catecholaminergic cells. This deletion did not alter basal metabolism or blood pressure but delayed the onset of angiotensin-dependent hypertension and reduced the maximal response. Cardiac hypertrophy was also reduced. The knockout mice showed attenuated activation of the sympathetic nervous system during angiotensin II infusion as measured by spectral analysis of the blood pressure. Increased reactive oxygen species production was observed in forebrain regions, including the subfornical organ, of the knockout mouse but was markedly reduced in the rostral ventrolateral medulla. These studies demonstrate that stimulation of the angiotensin type 1A receptor on catecholaminergic cells is required for the full development of angiotensin-dependent hypertension and support an important role for the sympathetic nervous system in this model.


Subject(s)
Blood Pressure/physiology , Cardiomegaly/metabolism , Catecholamines/metabolism , Hypertension/metabolism , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II , Animals , Blood Pressure/drug effects , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Mice , Mice, Knockout , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/genetics , Subfornical Organ/drug effects , Subfornical Organ/metabolism , Subfornical Organ/physiopathology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology
13.
Hypertension ; 61(3): 716-22, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23266541

ABSTRACT

Although elevated renin-angiotensin system activity and angiotensinergic signaling within the brain are required for hypertension, polydipsia, and increased metabolic rate induced by deoxycorticosterone acetate (DOCA)-salt, the contribution of specific receptor subtypes and brain nuclei mediating these responses remains poorly defined. We hypothesized that angiotensin type 1a receptors (AT(1a)R) within the subfornical organ (SFO) mediate these responses. Transgenic mice carrying a conditional allele of the endogenous AT(1a)R (AT(1a)R(flox)) were administered an adenovirus encoding Cre-recombinase and enhanced green fluorescent protein (eGFP) or adenovirus encoding eGFP alone into the lateral cerebral ventricle. Adenovirus encoding Cre-recombinase reduced AT(1a)R mRNA and induced recombination in AT(1a)R(flox) genomic DNA specifically in the SFO, without significant effect in the paraventricular or arcuate nuclei, and also induced SFO-specific recombination in ROSA(TdTomato) reporter mice. The effect of SFO-targeted ablation of endogenous AT(1a)R was evaluated in AT(1a)R(flox) mice at 3 time points: (1) baseline, (2) 1 week after virus injection but before DOCA-salt, and (3) after 3 weeks of DOCA-salt. DOCA-salt-treated mice with deletion of AT(1a)R in SFO exhibited a blunted increase in arterial pressure. Increased sympathetic cardiac modulation and urine copeptin, a marker of vasopressin release, were both significantly reduced in DOCA-salt mice when AT(1a)R was deleted in the SFO. Additionally, deletion of AT(1a)R in the SFO significantly attenuated the polydipsia, polyuria, and sodium intake in response to DOCA-salt. Together, these data highlight the contribution of AT(1a)R in the SFO to arterial pressure regulation potentially through changes on sympathetic cardiac modulation, vasopressin release, and hydromineral balance in the DOCA-salt model of hypertension.


Subject(s)
Desoxycorticosterone/adverse effects , Hypertension/chemically induced , Mineralocorticoids/adverse effects , Receptor, Angiotensin, Type 1/physiology , Subfornical Organ/drug effects , Subfornical Organ/physiopathology , Animals , Arterial Pressure/drug effects , Biomarkers/urine , Glycopeptides/urine , Heart/drug effects , Heart/innervation , Male , Mice , Mice, Transgenic , Polydipsia/chemically induced , Polyuria/chemically induced , Receptor, Angiotensin, Type 1/genetics , Recombination, Genetic , Sodium/metabolism , Sympathetic Nervous System/drug effects
14.
Hypertension ; 61(2): 382-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23248154

ABSTRACT

Reactive oxygen species and the NADPH oxidases contribute to hypertension via mechanisms that remain undefined. Reactive oxygen species produced in the central nervous system have been proposed to promote sympathetic outflow, inflammation, and hypertension, but the contribution of the NADPH oxidases to these processes in chronic hypertension is uncertain. We therefore sought to identify how NADPH oxidases in the subfornical organ (SFO) of the brain regulate blood pressure and vascular inflammation during sustained hypertension. We produced mice with loxP sites flanking the coding region of the NADPH oxidase docking subunit p22(phox). SFO-targeted injections of an adenovirus encoding cre-recombinase markedly diminished p22(phox), Nox2, and Nox4 mRNA in the SFO, as compared with a control adenovirus encoding red-fluorescent protein injection. Increased superoxide production in the SFO by chronic angiotensin II infusion (490 ng/kg min(-1) × 2 weeks) was blunted in adenovirus encoding cre-recombinase-treated mice, as detected by dihydroethidium fluorescence. Deletion of p22(phox) in the SFO eliminated the hypertensive response observed at 2 weeks of angiotensin II infusion compared with control adenovirus encoding red-fluorescent protein-treated mice (mean arterial pressures=97 ± 15 versus 154 ± 6 mm Hg, respectively; P=0.0001). Angiotensin II infusion also promoted marked vascular inflammation, as characterized by accumulation of activated T-cells and other leukocytes, and this was prevented by deletion of the SFO p22(phox). These experiments definitively identify the NADPH oxidases in the SFO as a critical determinant of the blood pressure and vascular inflammatory responses to chronic angiotensin II, and further support a role of reactive oxygen species in central nervous system signaling in hypertension.


Subject(s)
Hypertension/metabolism , NADPH Oxidases/metabolism , Subfornical Organ/metabolism , Angiotensin II , Animals , Blood Pressure/physiology , Hypertension/chemically induced , Hypertension/physiopathology , Inflammation/metabolism , Inflammation/physiopathology , Integrases/genetics , Mice , Mice, Transgenic , NADPH Oxidases/genetics , Oxidation-Reduction , Protein Subunits/genetics , Reactive Oxygen Species/metabolism , Subfornical Organ/physiopathology
15.
J Neurosci ; 32(14): 4878-86, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22492044

ABSTRACT

Hypertension, a powerful risk factor for stroke and dementia, has damaging effects on the brain and its vessels. In particular, hypertension alters vital cerebrovascular control mechanisms linking neural activity to cerebral perfusion. In experimental models of slow-developing hypertension, free radical signaling in the subfornical organ (SFO), one of the forebrain circumventricular organs, is critical for the hormonal release and sympathetic activation driving the elevation in arterial pressure. However, the contribution of this central mechanism to the cerebrovascular alterations induced by hypertension remains uncertain. We tested the hypothesis that free radical production in the SFO is involved in the alterations in cerebrovascular regulation produced by hypertension. In a mouse model of gradual hypertension induced by chronic administration of subpressor doses of angiotensin II (AngII), suppression of free radicals in the SFO by overexpression of CuZn-superoxide dismutase (CuZnSOD) prevented the alteration in neurovascular coupling and endothelium-dependent responses in somatosensory cortex induced by hypertension. The SFO mediates the dysfunction via two signaling pathways. One involves SFO-dependent activation of the paraventricular hypothalamic nucleus, elevations in plasma vasopressin, upregulation of endothelin-1 in cerebral resistance arterioles and activation of endothelin type A receptors. The other pathway depends on activation of cerebrovascular AngII type 1 (AT1) receptors by AngII. Both pathways mediate vasomotor dysfunction by inducing vascular oxidative stress. The findings implicate for the first time the SFO and its efferent hypothalamic pathways in the cerebrovascular alterations induced by AngII, and identify vasopressin and endothelin-1 as potential therapeutic targets to counteract the devastating effects of hypertension on the brain.


Subject(s)
Angiotensin II/physiology , Angiotensin II/toxicity , Cerebrovascular Circulation/physiology , Hypertension/physiopathology , Subfornical Organ/physiopathology , Animals , Cerebrovascular Circulation/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Gene Transfer Techniques , Hypertension/chemically induced , Hypertension/pathology , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/agonists , Receptor, Angiotensin, Type 1/physiology , Subfornical Organ/drug effects
16.
Exp Physiol ; 97(1): 80-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21967900

ABSTRACT

Hypertension caused by chronic infusion of angiotensin II (Ang II) in experimental animals is dependent, in part, on increased activity of the sympathetic nervous system. This chronic sympathoexcitatory response is amplified by a high-salt diet, suggesting an interaction of circulating Ang II and dietary salt on sympathetic regulatory pathways in the brain. The present study tested the hypothesis that the subfornical organ (SFO), a forebrain circumventricular organ known to be activated by circulating Ang II, is crucial to the pathogenesis of hypertension induced by chronic Ang II administration in rats on a high-salt diet (Ang II-salt model). Rats were randomly selected to undergo either subfornical organ lesion (SFOx) or sham surgery (Sham) and then placed on a high-salt (2% NaCl) diet. One week later, rats were instrumented for radiotelemetric measurement of mean arterial pressure (MAP) and heart rate (HR) and placed in metabolic cages to measure sodium and water balance. Baseline MAP was slightly (but not statistically) lower in SFOx compared with Sham rats during the 5 day control period. During the subsequent 10 days of Ang II administration, MAP was statistically lower in SFOx rats. However, when MAP responses to Ang II were analysed by comparing the change from the 5 day baseline period, only on the fifth day of Ang II was MAP significantly different between groups. There were no differences between groups for water or sodium balance throughout the protocol. We conclude that, although the SFO is required for the complete expression of Ang II-salt hypertension in the rat, other brain sites are also involved.


Subject(s)
Angiotensin II/pharmacology , Hypertension/chemically induced , Prosencephalon/drug effects , Sodium Chloride, Dietary/pharmacology , Subfornical Organ/drug effects , Sympathetic Nervous System/drug effects , Angiotensin II/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Drug Synergism , Heart Rate/drug effects , Heart Rate/physiology , Hypertension/metabolism , Hypertension/physiopathology , Male , Prosencephalon/physiology , Rats , Rats, Sprague-Dawley , Sodium Chloride, Dietary/metabolism , Subfornical Organ/metabolism , Subfornical Organ/physiopathology , Sympathetic Nervous System/physiology
17.
J Neuroendocrinol ; 24(3): 504-10, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22103447

ABSTRACT

The subfornical organ (SFO), a sensory circumventricular organ lacking the normal blood-brain barrier with well documented roles in cardiovascular regulation, has recently been identified as a potential site at which the adipokine, leptin, may act to influence central autonomic pathways. Systemic and central leptin administration has been shown to increase blood pressure and it has been suggested that selective leptin resistance contributes to obesity-related hypertension. Given the relationship between obesity and hypertension, the present study aimed to investigate the cardiovascular consequences of the direct administration of leptin into the SFO of young lean rats and in the diet-induced obesity (DIO) rat model, which has been shown to be leptin-resistant. Leptin administration (500 fmol) directly into the SFO of young rats resulted in rapid decreases in blood pressure (BP) [mean area under the curve (AUC) = -677.8 ± 167.1 mmHg*s; n = 9], without an effect on heart rate (mean AUC = -21.2 ± 13.4 beats; n = 9), and these effects were found to be dose-related as microinjection of 5 pmol of leptin into the SFO had a larger effect on BP (mean AUC = -972.3 ± 280.1 mmHg*s; n = 4). These BP effects were also shown to be site-specific as microinjection of leptin into non-SFO regions or into the ventricle was without effect on BP (non-SFO: mean AUC = -22.4 ± 55.3 mmHg*s; n = 4; ventricle: mean AUC = 194.0 ± 173.0 mmHg*s; n = 6). By contrast, microinjection of leptin into leptin-resistant DIO rats was without effect on BP (mean AUC = 205.2 ± 75.1 mmHg*s; n = 4). These observations suggest that the SFO may be an important relay centre through which leptin, in normal weight, leptin responsive rats, acts to maintain BP within normal physiological limits through descending autonomic pathways involved in cardiovascular control and that, in obese, leptin-resistant, rats leptin no longer influences SFO neurones, resulting in an elevated BP, thus contributing to obesity-related hypertension.


Subject(s)
Cardiovascular System/physiopathology , Diet , Leptin/physiology , Obesity/physiopathology , Subfornical Organ/physiopathology , Animals , Blood Pressure , Heart Rate , Male , Microinjections , Rats , Rats, Sprague-Dawley
18.
Am J Physiol Regul Integr Comp Physiol ; 298(4): R1098-110, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20106992

ABSTRACT

The mechanism and route whereby glucagon-like peptide 1 (GLP-1) receptor agonists, such as GLP-1 and exendin-4 (Ex-4), access the central nervous system (CNS) to exert their metabolic effects have yet to be clarified. The primary objective of the present study was to investigate the potential role of two circumventricular organs (CVOs), the area postrema (AP) and the subfornical organ (SFO), in mediating the metabolic and CNS-stimulating effects of Ex-4. We demonstrated that electrolytic ablation of the AP, SFO, or AP + SFO does not acutely prevent the anorectic effects of Ex-4. AP + SFO lesion chronically decreased food intake and body weight and also modulated the effect of Ex-4 on the neuronal activation of brain structures involved in the hypothalamic-pituitary-adrenal axis and glucose metabolism. The results of the study also showed that CVO lesions blunted Ex-4-induced expression of c-fos mRNA (a widely used neuronal activity marker) in 1) limbic structures (bed nucleus of the stria terminalis and central amygdala), 2) hypothalamus (paraventricular hypothalamic nucleus, supraoptic nucleus, and arcuate nucleus), and 3) hindbrain (lateral and lateral-external parabrachial nucleus, medial nucleus of the solitary tract, and ventrolateral medulla). In conclusion, although the present results do not support a role for the CVOs in the anorectic effect induced by a single injection of Ex-4, they suggest that the CVOs play important roles in mediating the actions of Ex-4 in the activation of CNS structures involved in homeostatic control.


Subject(s)
Area Postrema/physiopathology , Hypoglycemic Agents/pharmacology , Peptides/pharmacology , Subfornical Organ/physiopathology , Venoms/pharmacology , Animals , Body Composition/drug effects , Body Weight/drug effects , Brain/enzymology , Brain/physiopathology , Deglutition Disorders/chemically induced , Deglutition Disorders/physiopathology , Energy Intake/drug effects , Energy Metabolism/drug effects , Exenatide , Genes, fos , Glucokinase/genetics , Male , Organ Size , RNA, Heterogeneous Nuclear/genetics , RNA, Messenger/drug effects , RNA, Messenger/genetics , Rats , Rats, Wistar
19.
Am J Physiol Heart Circ Physiol ; 288(2): H680-5, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15458953

ABSTRACT

Previous studies clearly demonstrated acute actions of angiotensin II (ANG II) at one of the central circumventricular organs, the subfornical organ (SFO), but studies demonstrating a role for the SFO in the chronic actions of ANG II remain uncertain. The purpose of this study was to examine the role of the SFO in the chronic hypertensive phase of ANG II-induced hypertension. We hypothesized that the SFO is necessary for the full hypertensive response observed during the chronic phase of ANG II-induced hypertension. To test this hypothesis, male Sprague-Dawley rats were subjected to sham operation (sham rats) or electrolytic lesion of the SFO (SFOx rats). After 1 wk, the rats were instrumented with venous catheters and radiotelemetric transducers for intravenous administration of ANG II and measurement of blood pressure and heart rate, respectively. Rats were then allowed 1 wk for recovery. After 3 days of saline control infusion (7 ml of 0.9% NaCl/day), sham and SFOx rats were infused with ANG II at 10 ng.kg(-1).min(-1) i.v. for 10 consecutive days and then allowed to recover for 3 days. A 0.4% NaCl diet and distilled water were provided ad libitum. At day 5 of ANG II infusion, mean arterial pressure increased 11.7 +/- 3.0 mmHg in sham rats (n = 9) but increased only 3.7 +/- 1.4 mmHg in SFOx rats (n = 9). This trend continued through day 10 of ANG II treatment. These results support the hypothesis that the SFO is necessary for the full hypertensive response to chronic ANG II administration.


Subject(s)
Angiotensin II/pharmacology , Hypertension/physiopathology , Subfornical Organ/physiopathology , Vasoconstrictor Agents/pharmacology , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Drinking/physiology , Heart Rate/drug effects , Heart Rate/physiology , Hexamethonium/pharmacology , Hypertension/chemically induced , Male , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/physiology , Sodium/metabolism , Sympathetic Nervous System/physiology , Telemetry , Water/metabolism
20.
J Renin Angiotensin Aldosterone Syst ; 4(4): 207-12, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14689367

ABSTRACT

UNLABELLED: HYPOTHESIS/INTRODUCTION: Circumventricular organs are central nervous system brain sites thought to participate in neuroendocrine regulation of neural output. We have previously demonstrated a profound chronic hypotensive response to the angiotensin II (Ang II) AT(1) antagonist, losartan (10 mg/kg/day), in normal rats. In addition, we have demonstrated that the area postrema, one of the circumventricular organs, partially mediates this response. The subfornical organ (SFO) is another circumventricular organ which has been shown to mediate actions of Ang II. The present study was designed to test the hypothesis that the SFO mediates the chronic hypotensive effects of losartan in normal rats. MATERIALS AND METHODS: Rats were randomly chosen for lesion of the SFO or sham operation and instrumented with intravenous catheters and radiotelemetric blood pressure transducers. After a control period, rats were infused with losartan (10 mg/kg/day) for nine days. Mean arterial pressure and heart rate responses were measured continuously throughout the protocol and examined as 12-hour day/night averages. RESULTS: By day 7 of losartan treatment, night-time mean arterial pressure had dropped to 75+2 mmHg in sham rats (n=8) but only to 83+2 mmHg in SFO-lesioned rats (n=10). This trend continued throughout the treatment protocol. CONCLUSIONS: These results suggest that the SFO partially mediates the chronic hypotensive effects of chronic losartan treatment in normal rats.


Subject(s)
Antihypertensive Agents , Hypotension/chemically induced , Hypotension/physiopathology , Losartan , Sodium Chloride/metabolism , Subfornical Organ/physiopathology , Angiotensin II Type 1 Receptor Blockers , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Body Water/metabolism , Chronic Disease , Heart Rate/drug effects , Hypotension/metabolism , Losartan/pharmacology , Male , Rats , Rats, Sprague-Dawley , Renin/blood , Sodium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...