Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
BMC Neurosci ; 25(1): 29, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926677

ABSTRACT

BACKGROUND: Astrocytes are the most abundant cell type of the central nervous system and are fundamentally involved in homeostasis, neuroprotection, and synaptic plasticity. This regulatory function of astrocytes on their neighboring cells in the healthy brain is subject of current research. In the ischemic brain we assume disease specific differences in astrocytic acting. The renin-angiotensin-aldosterone system regulates arterial blood pressure through endothelial cells and perivascular musculature. Moreover, astrocytes express angiotensin II type 1 and 2 receptors. However, their role in astrocytic function has not yet been fully elucidated. We hypothesized that the angiotensin II receptors impact astrocyte function as revealed in an in vitro system mimicking cerebral ischemia. Astrocytes derived from neonatal wistar rats were exposed to telmisartan (angiotensin II type 1 receptor-blocker) or PD123319 (angiotensin II type 2 receptor-blocker) under normal conditions (control) or deprivation from oxygen and glucose. Conditioned medium (CM) of astrocytes was harvested to elucidate astrocyte-mediated indirect effects on microglia and cortical neurons. RESULT: The blockade of angiotensin II type 1 receptor by telmisartan increased the survival of astrocytes during ischemic conditions in vitro without affecting their proliferation rate or disturbing their expression of S100A10, a marker of activation. The inhibition of the angiotensin II type 2 receptor pathway by PD123319 resulted in both increased expression of S100A10 and proliferation rate. The CM of telmisartan-treated astrocytes reduced the expression of pro-inflammatory mediators with simultaneous increase of anti-inflammatory markers in microglia. Increased neuronal activity was observed after treatment of neurons with CM of telmisartan- as well as PD123319-stimulated astrocytes. CONCLUSION: Data show that angiotensin II receptors have functional relevance for astrocytes that differs in healthy and ischemic conditions and effects surrounding microglia and neuronal activity via secretory signals. Above that, this work emphasizes the strong interference of the different cells in the CNS and that targeting astrocytes might serve as a therapeutic strategy to influence the acting of glia-neuronal network in de- and regenerative context.


Subject(s)
Angiotensin II Type 1 Receptor Blockers , Angiotensin II Type 2 Receptor Blockers , Astrocytes , Ischemic Stroke , Microglia , Neurons , Rats, Wistar , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Telmisartan , Animals , Rats , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals, Newborn , Astrocytes/metabolism , Astrocytes/drug effects , Benzimidazoles/pharmacology , Cell Communication/physiology , Cell Communication/drug effects , Cells, Cultured , Imidazoles/pharmacology , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Microglia/metabolism , Microglia/drug effects , Neurons/metabolism , Neurons/drug effects , Pyridines/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Telmisartan/pharmacology
2.
J Am Heart Assoc ; 13(10): e033998, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38726925

ABSTRACT

BACKGROUND: The vasoconstrictor effects of angiotensin II via type 1 angiotensin II receptors in vascular smooth muscle cells are well established, but the direct effects of angiotensin II on vascular endothelial cells (VECs) in vivo and the mechanisms how VECs may mitigate angiotensin II-mediated vasoconstriction are not fully understood. The present study aimed to explore the molecular mechanisms and pathophysiological relevance of the direct actions of angiotensin II on VECs in kidney and brain microvessels in vivo. METHODS AND RESULTS: Changes in VEC intracellular calcium ([Ca2+]i) and nitric oxide (NO) production were visualized by intravital multiphoton microscopy of cadherin 5-Salsa6f mice or the endothelial uptake of NO-sensitive dye 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate, respectively. Kidney fibrosis by unilateral ureteral obstruction and Ready-to-use adeno-associated virus expressing Mouse Renin 1 gene (Ren1-AAV) hypertension were used as disease models. Acute systemic angiotensin II injections triggered >4-fold increases in VEC [Ca2+]i in brain and kidney resistance arterioles and capillaries that were blocked by pretreatment with the type 1 angiotensin II receptor inhibitor losartan, but not by the type 2 angiotensin II receptor inhibitor PD123319. VEC responded to acute angiotensin II by increased NO production as indicated by >1.5-fold increase in 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate fluorescence intensity. In mice with kidney fibrosis or hypertension, the angiotensin II-induced VEC [Ca2+]i and NO responses were significantly reduced, which was associated with more robust vasoconstrictions, VEC shedding, and microthrombi formation. CONCLUSIONS: The present study directly visualized angiotensin II-induced increases in VEC [Ca2+]i and NO production that serve to counterbalance agonist-induced vasoconstriction and maintain residual organ blood flow. These direct and endothelium-specific angiotensin II effects were blunted in disease conditions and linked to endothelial dysfunction and the development of vascular pathologies.


Subject(s)
Angiotensin II , Brain , Calcium , Hypertension , Kidney , Microvessels , Nitric Oxide , Vasoconstriction , Animals , Nitric Oxide/metabolism , Angiotensin II/pharmacology , Hypertension/metabolism , Hypertension/physiopathology , Hypertension/drug therapy , Kidney/blood supply , Kidney/metabolism , Calcium/metabolism , Vasoconstriction/drug effects , Microvessels/metabolism , Microvessels/drug effects , Microvessels/pathology , Brain/metabolism , Brain/blood supply , Mice , Disease Models, Animal , Male , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Mice, Inbred C57BL , Calcium Signaling/drug effects
3.
Biochem Pharmacol ; 216: 115793, 2023 10.
Article in English | MEDLINE | ID: mdl-37689272

ABSTRACT

With the discovery of the protective arm of the renin-angiotensin system (RAS), interest has grown in protective RAS-related receptors such as the angiotensin AT2-receptor [AT2R] as potential new drug targets. While it is known that AT2R couple to Gi, it is also apparent that they do not signal via inhibition of adenylyl cyclase/decrease in cAMP, as do many Gi-coupled receptors. Thus, standard commercially-available assays cannot be applied to test for agonistic or antagonistic properties of AT2R ligands. This lack of standard assays has hampered the development of new drugs targeting the AT2R. Therefore, we aimed at developing a reliable, technically easy assay for the determination of intrinsic activity of AT2R ligands, primarily for distinguishing between AT2R agonists and antagonists. We found that measurement of NO release by DAF-FM fluorescence in primary human aortic endothelial cells (HAEC) or in AT2R-transfected CHO cells is a reliable assay for the characterization of AT2R ligands. While testing the assay, we made several novel findings, including: a) C21 is a full agonist at the AT2R (with the same efficacy as angiotensin II); b) C21 has no intrinsic activity at the receptor Mas; c) AT2R-transfected HEK-293 cells are unresponsive to AT2R stimulation; d) EMA401 and PD123319, which are commonly regarded as AT2R antagonists, are partial agonists at the AT2R. Collectively, we have developed and tested an assay based on the measurement and quantification of NO release in HAEC or in AT2R-CHO cells that is suitable for the characterisation of novel and established AT2R ligands.


Subject(s)
Endothelial Cells , Receptor, Angiotensin, Type 2 , Animals , Cricetinae , Humans , Cricetulus , HEK293 Cells , Angiotensin II/pharmacology , Receptor, Angiotensin, Type 1
4.
J Renin Angiotensin Aldosterone Syst ; 21(3): 1470320320949850, 2020.
Article in English | MEDLINE | ID: mdl-32962526

ABSTRACT

INTRODUCTION: Angiotensin II (Ang II) is multifunctional peptide that plays an important role in blood pressure regulation and maintenance electrolyte homeostasis. It shows biological effects by activating two main receptors: AT1 and AT2. The aim of the present work was to investigate the effect of Ang II on NRK-52E cells in in vitro studies. Furthermore, an attempt was made to determine the effectiveness of the AT1 and AT2 receptor blocker activity (respectively, losartan and PD123319). METHODS: The study was carried out using adherent NRK-52E cell line. Immunofluorescence and Western Blot method were used to confirm the presence of AT1 and AT2 receptors in the cells. The SRB and MTT tests showed decrease in the viability of NRK-52E cells incubated with Ang II in comparison to the control (without Ang II). RESULTS: The blockade of the AT1 receptor caused an increase in cell viability in comparison to cells incubated with Ang II only. The blockade of AT2 receptor also triggered statistically significant increase in cell viability in comparison with cells only exposed to Ang II. Combined administration of blockers for both receptors (losartan and PD123319) decreased Ang II cytotoxicity against NRK-52E cell line. The apoptosis was only observed in cells incubated with Ang II in comparison with control cells. However, simultaneous use of both blockers caused statistically significant decrease in apoptosis. CONCLUSIONS: The result of our study indicates that Ang II causes damaging effect on NRK-52E cells by directing them to programmed cell death. It seems that not only does the AT2 receptor itself play an important role in the induction of apoptosis, but also its interaction with AT1 receptor does as well.


Subject(s)
Angiotensin II/pharmacology , Apoptosis/drug effects , Epithelial Cells/cytology , Kidney Tubules, Proximal/cytology , Animals , Caspase 3/metabolism , Cell Line , Cell Shape/drug effects , Cell Survival/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Imidazoles/pharmacology , Losartan/pharmacology , Pyridines/pharmacology , Rats , Receptor, Angiotensin, Type 1/metabolism
5.
Pflugers Arch ; 471(9): 1173-1182, 2019 09.
Article in English | MEDLINE | ID: mdl-31396758

ABSTRACT

The medial amygdaloid nucleus (MeA) is a limbic structure that has been demonstrated to be part of the central circuitry regulating baroreflex function. However, the local neurochemical mechanisms involved in baroreflex control by this forebrain structure is poorly understood. Thus, in the present study, we investigated the specific role of AT1, AT2, and MAS angiotensinergic receptors within the MeA in baroreflex responses in unanesthetized rats. For this, the baroreflex function was assessed using both the pharmacological approach via intravenous infusion of vasoactive agents and the sequence analysis technique. Using the pharmacological approach, we observed that bilateral microinjection of the selective AT2 receptor antagonist PD123319 into the MeA increased the tachycardia evoked by blood pressure decrease, but without affecting the reflex bradycardia caused by blood pressure increase. Besides, bilateral microinjection of the selective MAS receptor antagonist A-779 decreased both tachycardic and bradycardic responses of the baroreflex. The sequence analysis technique indicated that PD123319 into the MeA increased baroreflex effectiveness index while A-779 had an opposite effect. Treatment of the MeA with the selective AT1 receptor antagonist losartan did not affect baroreflex function assessed by either the pharmacological approach or sequence analysis technique. Overall, these findings provide evidence that MAS receptor within the MeA plays a facilitatory role in baroreflex function, whereas local AT2 receptor inhibits cardiac baroreflex responses. Results also indicate that AT1 receptor within the MeA is not involved in the control of baroreflex function.


Subject(s)
Baroreflex/physiology , Corticomedial Nuclear Complex/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Angiotensin, Type 2/metabolism , Receptors, G-Protein-Coupled/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Baroreflex/drug effects , Blood Pressure/drug effects , Blood Pressure/physiology , Corticomedial Nuclear Complex/drug effects , Heart/drug effects , Heart Rate/drug effects , Heart Rate/physiology , Imidazoles/pharmacology , Losartan/pharmacology , Male , Proto-Oncogene Mas , Pyridines/pharmacology , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism
6.
Can J Physiol Pharmacol ; 97(12): 1124-1131, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31361968

ABSTRACT

Our study aimed to determine the effects of losartan and PD123319 in ischemia-reperfusion (IR) injury in isolated perfused rat heart. The study used 40 male Wistar albino rats that were grouped as Control, IR, and IR treatment groups that received losartan (20 mg/kg), PD123319 (20 mg/kg), and losartan+PD123319. The hearts were attached to Langendorff isolated heart system by employing in situ cannulation method, and cardiodynamic parameters were recorded during the experiment. At the end of experiment, hearts were retained for biochemical analysis and all data were statistically evaluated. A partial recovery of cardiodynamic parameters was observed in all treatment groups. A significant increase in oxidative stress parameters were seen in the IR group, whereas all treatment groups exhibited lower increase. Furthermore, levels of all antioxidant parameters were significantly lower in the IR group, but higher in the treatment groups. Effects on all parameters were much more remarkable in the PD123319 group. Levels of angiotensin II and renin were increased (P < 0.001) with IR application and decreased (P < 0.001) with the treatment of both antagonists. In conclusion, treatment of losartan and PD123319 played a cardioprotective role against IR injury, PD123319 being more effective in this protection.


Subject(s)
Angiotensin II Type 2 Receptor Blockers/pharmacology , Imidazoles/pharmacology , Losartan/pharmacology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Oxidative Stress/drug effects , Pyridines/pharmacology , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II Type 2 Receptor Blockers/therapeutic use , Animals , Dose-Response Relationship, Drug , Imidazoles/therapeutic use , Male , Pyridines/therapeutic use , Rats , Rats, Wistar
7.
Pharmacol Res ; 135: 112-121, 2018 09.
Article in English | MEDLINE | ID: mdl-30048754

ABSTRACT

Angiotensin-(19), a peptide of the non-classical renin angiotensin system, has been shown to prevent and revert hypertension and cardiac hypertrophy. We hypothetized that systemic delivery of angiotensin-(1-9) following myocardial infarction will also be protective and extend to provide protection during reperfusion of the ischemic heart. Adult Sprague Dawley rats were subjected to left anterior descending artery ligation and treated with angiotensin-(1-9) via osmotic mini-pump for 2 weeks in the presence or absence of Mas receptor or AT2R antagonists (A779 and PD123319, respectively). Myocardial death and left ventricular function were evaluated after infarction. Infarct size and functional parameters were determined in isolated rat hearts after global ischemia/reperfusion in the presence of angiotensin-(1-9) plus receptor antagonists or Akt inhibitor at reperfusion. in vitro, neonatal rat ventricular cardiomyocytes underwent simulated ischemia/reperfusion and angiotensin-(1-9) was co-incubated with A779, PD123319 or Akt inhibitor. Systemic delivery of angiotensin-(1-9) significantly decreased cell death and improved left ventricular recovery after in vivo myocardial infarction. Perfusion with the peptide reduced the infarct size and improved functional recovery after ex vivo ischemia/reperfusion. In vitro, angiotensin-(1-9) decreased cell death in isolated neonatal rat ventricular cardiomyocytes subjected to simulated ischemia/reperfusion. The cardioprotective effects of angiotensin-(1-9) were blocked by PD123319 and Akti VIII but not by A779. Angiotensin-(1-9) limits reperfusion-induced cell death by an AT2R- and Aktdependent mechanism. Angiotensin-(1-9) is a novel strategy to protect against cardiac ischemia/reperfusion injury.


Subject(s)
Angiotensin I/therapeutic use , Cardiotonic Agents/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Peptide Fragments/therapeutic use , Angiotensin I/pharmacology , Animals , Animals, Newborn , Cardiotonic Agents/pharmacology , Cells, Cultured , Heart/drug effects , Heart/physiology , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 2/metabolism
8.
Biomed Pharmacother ; 102: 947-958, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29710550

ABSTRACT

In the present study, we investigated the effects of angiotensin (Ang II) receptor blockers in cerebral ischemia by administration of telmisartan (AT1 blocker) and/or PD123319 (AT2 blocker) in global ischemic mice model. The neuroprotective effect of AT antagonists was evaluated through monitoring muscle co-ordination and cerebral blood perfusion in ischemic mice. Gene expression studies (NF-κB, GSK-3ß, EAAT-2, AT1 & AT2 receptors) and staining of brain regions with cresyl violet, GFAP, synaptophysin and NSE methods were carried out in to understand the molecular mechanisms. Further, the brain glutamate, cytokines, and Ang II peptide levels were evaluated and their correlation with EAAT-2 mRNA expression was performed. Our results indicate that the induction of ischemia elevates brain Ang II, cytokines, and glutamate levels and reduced muscle co-ordination and cerebral blood perfusion. The expressions of NF-κB, GSK-3ß and AT1 were significantly increased, whereas, EAAT-2 expression was decreased. Blocking of AT1 receptors by telmisartan (TM) reversed the detrimental responses of cerebral ischemia and restored the cerebral blood flow denoting blockade of Ang II/AT1 pathway is beneficial in ischemia, whereas, blockade of AT2 receptors by PD123319 (PD) increased the ischemic injury in mice. This vulnerable effect of PD may be attributed through augmenting the Ang II/AT1 dependent cytokines mediated glutamate transporter (EAAT-2) dysfunction. Interestingly, the beneficial effects of AT1 blocker was remarkably antagonized by AT2 blocker in most of the parameters studied in ischemic conditions. Also, the expression of AT2 receptors was significantly increased compared to that of AT1 receptors upon ischemic induction. It denotes that the endogenous Ang II predominantly acts on AT2 receptor, thereby promoting its own mRNA transcription. Hence, the increased expression of AT2 receptors in ischemic condition could be used as target protein for therapeutic benefit.


Subject(s)
Brain Ischemia/pathology , Brain/metabolism , Brain/pathology , Glutamine/metabolism , Inflammation/pathology , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Brain Ischemia/drug therapy , Brain Ischemia/physiopathology , Cerebrovascular Circulation , Cytokines/metabolism , Glutamic Acid/metabolism , Inflammation/drug therapy , Inflammation/physiopathology , Laser-Doppler Flowmetry , Male , Mice , Muscles/metabolism , Neurons/metabolism , Neurons/pathology , Perfusion , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 2/genetics , Signal Transduction/drug effects
9.
J Am Soc Hypertens ; 12(1): 58-65, 2018 01.
Article in English | MEDLINE | ID: mdl-29100861

ABSTRACT

The objective was to investigate the role of angiotensin II type 2 receptor during electrophysiological remodeling of left ventricular hypertrophic myocardium in spontaneously hypertensive rats (SHRs). A total of 36, aged 10 weeks, male SHRs were divided into three groups: control, valsartan, and valsartan + PD123319 groups (n = 12 in each). The systolic blood pressure, left ventricular mass index, ventricular effective refractory period, and ventricular fibrillation threshold (VFT) were also measured after 8 weeks. At the same time, INa, ICaL, Ito, and membrane capacitance were measured in left ventricular myocytes by whole-cell patch-clamp. The VFT of valsartan was higher than that of control (valsartan vs. CONTROL: 17.4 ± 0.6 mA vs. 15.8 ± 0.5 mA, P < .05). The VFT of valsartan was higher than that of valsartan + PD123319 (valsartan vs. valsartan + PD123319: 17.4 ± 0.6 mA vs. 16.6 ± 0.9 mA, P < .05). The density of Ito of valsartan was higher than that of control (valsartan vs. CONTROL: 14.7 ± 0.42 pA/pF vs. 11.2 ± 0.15 pA/pF, P < .05). The density of Ito of valsartan was higher than that of valsartan + PD123319 (valsartan vs. valsartan + PD123319: 14.7 ± 0.42 pA/pF vs. 13.6 ± 0.30 pA/pF, P < .05). The density of ICaL of valsartan was lower than that of control (valsartan vs. CONTROL: -4.6 ± 0.2 pA/pF vs. -6.9 ± 0.1 pA/pF, P < .05). The density of ICaL of valsartan was lower than that of valsartan + PD123319 (valsartan vs. valsartan + PD123319: -4.6 ± 0.2 pA/pF vs. -5.4 ± 0.1 pA/pF, P < .05). These results demonstrated that the stimulation of angiotensin II type 2 receptor improved electrophysiological remodeling of left ventricular hypertrophic myocardium in SHR.


Subject(s)
Hypertension , Hypertrophy, Left Ventricular , Imidazoles/pharmacology , Pyridines/pharmacology , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Disease Models, Animal , Electrophysiological Phenomena , Hypertension/complications , Hypertension/drug therapy , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Rats , Rats, Inbred SHR , Treatment Outcome , Valsartan/pharmacology , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology
10.
Cells Tissues Organs ; 204(1): 25-37, 2017.
Article in English | MEDLINE | ID: mdl-28478436

ABSTRACT

Animal studies have reported on the benefits of ARB on bone mass. However, the underlying mechanism for angiotensin II (AngII)/AngII receptor blockade (ARB) in regulating bone mass remains elusive. Since high levels of plasma and urine cAMP are observed in osteoporotic and hypertensive patients, we hypothesized that cAMP may be an important molecule for the downstream events of the activation of AT receptors, members of the G-protein-coupled receptor family, in regulating bone turnover. In this study, micro-CT and X-ray analyses indicated that AngII decreased bone mass via biasing bone resorption over bone formation in osteoporotic mice. However, these adverse effects were blocked by olmesartan and PD123319. In vitro, AngII was shown to downregulate osteogenic differentiation and matrix mineralization, but to upregulate osteoclastic activity by mainly affecting osteoblasts producing osteoclastogenesis-associated key soluble factors, including M-CSF and RANKL. Similarly, ARB treatment exhibited antagonistic effects on AngII. In conclusion, osteoblasts are the directly targeted cells. ARB1 exhibits a greater capacity to increase bone mass than ARB2. The cAMP-dependent PKA pathway plays an important role in AngII/ARB on changing bone mass.


Subject(s)
Angiotensin II/metabolism , Angiotensin Receptor Antagonists/metabolism , Hypertension/drug therapy , Osteoporosis/drug therapy , Receptors, Angiotensin/drug effects , Animals , Female , Hypertension/pathology , Mice , Mice, Inbred C57BL , Osteoporosis/pathology
11.
Neural Regen Res ; 11(7): 1102-7, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27630693

ABSTRACT

Angiotensin II type 2 receptor (AT2R) activation has been shown to protect against stroke, but its precise mechanism remains poorly understood. We investigated whether the protective effect of AT2R against ischemia/reperfusion injury is mediated by the suppression of immune and inflammatory responses. Rat models of middle cerebral artery occlusion were intraperitoneally injected with physiological saline, the AT2R agonist CGP42112 (1 mg/kg per day) or antagonist PD123319 (1 mg/kg per day). In the CGP42112 group, AT2R expression increased, the infarct area decreased, interleukin-1ß and tumor necrosis factor-α expression decreased, and interleukin-10 expression increased compared with the saline group. Antagonisin AT2R using PD123319 produced the opposite effects. These results indicate that AT2R activation suppresses immune and inflammatory responses, and protects against cerebral ischemia/reperfusion injury.

12.
Naunyn Schmiedebergs Arch Pharmacol ; 389(12): 1333-1340, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27629578

ABSTRACT

To investigate the effects of PD123319, an antagonist of angiotensin II subtype-2 receptor (AT2R), on the electrophysiological characteristics of the left ventricular hypertrophic myocardium in spontaneously hypertensive rats (SHR). A total of twenty-four 10-week-old male SHR were divided into two groups: PD123319 and non-PD123319 groups (n = 12 in each). Twelve 10-week-old Wistar-Kyoto rats served as the control group. Systolic blood pressure, left ventricular mass index (LVMI), ventricular effective refractory period, and ventricular fibrillation threshold were also measured after 8 weeks. I Na, I CaL, I to, and membrane capacitance were measured in the left ventricular myocytes after 8 weeks by whole-cell patch clamp. PD123319 increased LVMI compared with the non-PD123319 group (PD123319 vs. non-PD123319, 3.83 ± 0.11 vs. 3.60 ± 0.19 mg/g; P < 0.01). PD123319 also decreased the ventricular fibrillation threshold compared with the non-PD123319 group (PD123319 vs. non-PD123319, 14.75 ± 0.65 vs. 16.0 ± 0.86 mA; P < 0.01). PD123319 enhanced membrane capacitance compared with the non-PD123319 group (PD123319 vs. non-PD123319, 283.63 ± 5.80 vs. 276.50 ± 4.28 pF; P < 0.05). PD123319 increased the density of I CaL compared with the non-PD123319 group (PD123319 vs. non-PD123319, -6.76 ± 0.48 vs. -6.13 ± 0.30 pA/pF; P < 0.05). PD123319 decreased the density of I to compared with the non-PD123319 group (PD123319 vs. non-PD123319, 11.49 ± 0.50 vs. 12.23 ± 0.36 pA/pF; P < 0.05). Long-term treatment with PD123319 worsened the development of myocyte hypertrophy and associated electrophysiological alterations in spontaneously hypertensive rat.


Subject(s)
Angiotensin II Type 2 Receptor Blockers/toxicity , Antihypertensive Agents/toxicity , Heart Ventricles/drug effects , Hypertension/drug therapy , Hypertrophy, Left Ventricular/chemically induced , Imidazoles/toxicity , Pyridines/toxicity , Ventricular Fibrillation/chemically induced , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Action Potentials/drug effects , Animals , Blood Pressure/drug effects , Disease Models, Animal , Electric Capacitance , Heart Ventricles/physiopathology , Hypertension/complications , Hypertension/physiopathology , Hypertrophy, Left Ventricular/physiopathology , Male , Rats, Inbred SHR , Rats, Inbred WKY , Refractory Period, Electrophysiological/drug effects , Time Factors , Ventricular Fibrillation/physiopathology
13.
Clin Exp Hypertens ; 38(2): 209-17, 2016.
Article in English | MEDLINE | ID: mdl-26818039

ABSTRACT

We hypothesized that blockade of angiotensin II type 2 receptors (AT2Rs) in the rostral ventrolateral medullary pressor area (RVLM) may elicit sympathoexcitatory responses which are smaller in hypertensive rats compared to normotensive rats. This hypothesis was tested in urethane-anesthetized, artificially ventilated male 14-week-old spontaneously hypertensive rats (SHR). Age-matched male Wistar-Kyoto rats (WKY) and Wistar rats were used as controls. PD123319 (AT2R antagonist) was microinjected into the RVLM and mean arterial pressure (MAP), heart rate (HR) and greater splanchnic nerve activity (GSNA) were recorded. Increases in MAP, HR and GSNA elicited by unilateral microinjections of PD123319 into the RVLM were significantly smaller in SHR when compared with those in WKY and Wistar rats. Unilateral microinjections of l-glutamate (l-Glu) into the RVLM elicited greater increases in MAP and GSNA in SHR compared to those in WKY. AT2R immunoreactivity was demonstrated in the RVLM neurons which were retrogradely labeled from the intermediolateral cell column (IML) of the spinal cord. These results indicate that AT2Rs are present on the RVLM neurons projecting to the IML and their blockade results in sympathoexcitatory responses. Activation of AT2Rs has an inhibitory influence in the RVLM and these receptors are tonically active. Attenuation of the function of AT2Rs in the RVLM may play a role in genesis and/or maintenance of hypertension in SHR.


Subject(s)
Angiotensin II Type 2 Receptor Blockers/pharmacology , Arterial Pressure/drug effects , Glutamic Acid/pharmacology , Heart Rate/drug effects , Imidazoles/pharmacology , Medulla Oblongata/drug effects , Pyridines/pharmacology , Splanchnic Nerves/drug effects , Animals , Blood Pressure/drug effects , Hypertension , Immunohistochemistry , Male , Medulla Oblongata/metabolism , Microinjections , Neurons/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Wistar , Receptor, Angiotensin, Type 2/metabolism , Spinal Cord Lateral Horn/metabolism
14.
Vascul Pharmacol ; 72: 93-100, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26141930

ABSTRACT

Norepinephrine (NE) responses are larger in renal and femoral veins compared to phenylephrine (PE). These differences may be due to the subtypes of adrenoceptor involved in these responses or to the involvement of local modulatory mechanisms. Therefore, the present study investigated in organ bath the adrenoceptor subtypes involved in the NE and PE responses in both renal and femoral veins as well as the influence of local mechanisms related to NO and to prostanoids upon these responses. The obtained data showed that the NE responses in these veins were not significantly modified by the selective inhibition of ß1 or ß2-adrenoceptors as well as AT1 or AT2 receptors. However, yohimbine reduced the NE Rmax in renal veins and, in parallel, right shifted the NE concentration-response curves in femoral veins. In both veins, prazosin reduced the NE Rmax and the clonidine induced a measurable contraction. The endothelium removal attenuated the NE responses in femoral veins, thereby abolishing the differences of NE and PE responses. Furthermore, the NE responses in renal and femoral veins were attenuated by indomethacin, which suppressed the statistical difference in relation to the PE response. In conclusion, a synergism between α1- and α2-adrenoceptors is essential to assure full NE contractile responses in both renal and femoral veins. Thus, by acting simultaneously in these adrenoceptors, NE induces more pronounced contractile responses, in comparison to PE, not only in renal but also in femoral veins. Moreover, this pronounced NE response in both renal and femoral veins appears to involve endothelium-derived vasoconstrictor prostanoids.


Subject(s)
Femoral Vein/drug effects , Kidney/drug effects , Norepinephrine/pharmacology , Prostaglandins/pharmacology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Clonidine/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Kidney/metabolism , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Phenylephrine/pharmacology , Prazosin/pharmacology , Rats , Rats, Wistar , Receptors, Adrenergic/metabolism , Receptors, Angiotensin/metabolism , Yohimbine/pharmacology
15.
Biochem Pharmacol ; 93(3): 343-51, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25482567

ABSTRACT

Growing body of evidence points out the crucial role of ACE2 in preventing atherosclerosis. However, data on how atherosclerosis affects ACE2 expression in heart and kidney remains unknown. Atherosclerosis was induced by feeding New Zealand White rabbits with high cholesterol diet (HCD - 2%) for 12 weeks and atorvastatin was administered (5mg/kg/day p.o) in last 3 weeks. ACE2 mRNA and protein expression was assessed by Western blotting and real time PCR. HCD fed rabbits developed atherosclerosis as confirmed by increase in plasma total cholesterol, LDL and triglycerides as well as formation atherosclerotic plaques in arch of aorta. The ACE2 protein but not mRNA expression was reduced in heart and kidney of HCD rabbits. Interestingly, atorvastatin increased the ACE2 protein expression in heart and kidney of HCD rabbits. However, atorvastatin increased ACE2 mRNA in heart but not in kidney of HCD rabbits. Atorvastatin increased the occupancy of histone H3 acetylation (H3-Ac) mark on ACE2 promoter region in heart of HCD rabbits indicating direct or indirect epigenetic up-regulation of ACE2 by atorvastatin. Further, atorvastatin suppressed Ang II-induced contractile responses and enhanced AT2 receptor mediated relaxant responses in atherosclerotic aorta. We propose that atherosclerosis is associated with reduced ACE2 expression in heart and kidney. We also show an unexplored potential of atorvastatin to up-regulate ACE2 via epigenetic histone modifications. Our data suggest a novel way of replenishing ACE2 expression for preventing not only atherosclerosis but also other cardiovascular disorders.


Subject(s)
Atherosclerosis/metabolism , Disease Models, Animal , Epigenesis, Genetic/drug effects , Heptanoic Acids/therapeutic use , Histones/biosynthesis , Peptidyl-Dipeptidase A/biosynthesis , Pyrroles/therapeutic use , Angiotensin-Converting Enzyme 2 , Animals , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atorvastatin , Epigenesis, Genetic/physiology , Heptanoic Acids/pharmacology , Histones/genetics , Male , Peptidyl-Dipeptidase A/genetics , Pyrroles/pharmacology , Rabbits , Tissue Distribution/drug effects , Tissue Distribution/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
16.
Biochem Pharmacol ; 86(5): 656-65, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23831951

ABSTRACT

Tobacco smoking is the leading preventable cause of death in the United States. A major negative health consequence of chronic smoking is hypertension. Untoward addictive and cardiovascular sequelae associated with chronic smoking are mediated by nicotine-induced activation of nicotinic receptors (nAChRs) within striatal dopaminergic and hypothalamic noradrenergic systems. Hypertension involves both brain and peripheral angiotensin systems. Activation of angiotensin type-1 receptors (AT1) release dopamine and norepinephrine. The current study determined the role of AT1 and angiotensin type-2 (AT2) receptors in mediating nicotine-evoked dopamine and norepinephrine release from striatal and hypothalamic slices, respectively. The potential involvement of nAChRs in mediating effects of AT1 antagonist losartan and AT2 antagonist, 1-[[4-(dimethylamino)-3-methylphenyl]methyl]-5-(diphenylacetyl)-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridine-6-carboxylic acid (PD123319) was evaluated by determining their affinities for α4ß2* and α7* nAChRs using [³H]nicotine and [³H]methyllycaconitine binding assays, respectively. Results show that losartan concentration-dependently inhibited nicotine-evoked [³H]dopamine and [³H]norepinephrine release (IC50: 3.9 ± 1.2 and 2.2 ± 0.7 µM; Imax: 82 ± 3 and 89 ± 6%, respectively). In contrast, PD123319 did not alter nicotine-evoked norepinephrine release, and potentiated nicotine-evoked dopamine release. These results indicate that AT1 receptors modulate nicotine-evoked striatal dopamine and hypothalamic norepinephrine release. Furthermore, AT1 receptor activation appears to be counteracted by AT2 receptor activation in striatum. Losartan and PD123319 did not inhibit [³H]nicotine or [³H]methyllycaconitine binding, indicating that these AT1 and AT2 antagonists do not interact with the agonist recognition sites on α4ß2* and α7* nAChRs to mediate these effects of nicotine. Thus, angiotensin receptors contribute to the effects of nicotine on dopamine and norepinephrine release in brain regions involved in nicotine reward and hypertension.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Dopamine/metabolism , Nicotine/pharmacology , Norepinephrine/metabolism , Animals , Hypothalamus/drug effects , Hypothalamus/metabolism , Imidazoles/pharmacology , In Vitro Techniques , Losartan/pharmacology , Male , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/metabolism , Tritium/metabolism
17.
Article in English | WPRIM (Western Pacific) | ID: wpr-90655

ABSTRACT

BACKGROUND: The aim of this study was to examine whether PD 123319 (an angiotensin II type 2 [AT2] receptor antagonist) can influence the release of catecholamines (CA) from the perfused model of the rat adrenal medulla. METHODS: The adrenal gland was isolated by the modification of Wakade method, and perfused with normal Krebs-bicarbonate solution. The content of CA was measured using the fluorospectrophotometer. RESULTS: During perfusion of PD 123319 (range, 5 to 50 nM) into an adrenal vein for 90 minutes the CA secretory responses evoked by acetylcholine (ACh), high K+, 1,1-dimethyl-4-phenylpiperazinium iodide (DMPP), and McN-A-343 was dose- and time-dependently inhibited. Furthermore, loading with PD 123319 for 90 minutes also markedly inhibited the CA secretory responses evoked by 4-dihydro-2,6-dimethyl-3-nitro-4-(2-trifluoro-methyl-phenyl)-pyridine-5-carboxylate (Bay-K-8644), cyclopiazonic acid, veratridine, and angiotensin II (Ang II). PD 123319 did not affect basal CA output. Simultaneous perfusion of PD 123319 and CGP 42112 perfused into an adrenal vein for 90 minutes rather more potently inhibited the CA seretory responses evoked by Ach, high K+, DMPP, Bay-K-8644, veratridine, and Ang II compared to the inhibitory effect by PD123319-treated alone. CONCLUSIONS: Taken together, these results show that PD 123319 inhibits the CA secretion evoked by both cholinergic and Ang II receptor stimulation from the perfused rat adrenal medulla. This inhibitory effect of PD 123319 seems to be exerted by blocking the influx of both Na+ and Ca2+ through their voltage-dependent channels into the rat adrenomedullary chromaffin cells as well as by reducing the Ca2+ release from its cytoplasmic calcium store, which may be relevant to AT2 receptor blockade. Based on these present data, it is thought that PD 123319 has different activity from previously known AT2 antagonist activity in the perfused adrenal medulla, and that AT2 receptors may be involved in the rat adrenomedullary CA secretion.


Subject(s)
Animals , Rats , (4-(m-Chlorophenylcarbamoyloxy)-2-butynyl)trimethylammonium Chloride , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester , Acetylcholine , Adrenal Glands , Adrenal Medulla , Angiotensin II , Angiotensin II Type 2 Receptor Blockers , Calcium , Catecholamines , Chromaffin Cells , Cytoplasm , Dimethylphenylpiperazinium Iodide , Imidazoles , Indoles , Oligopeptides , Perfusion , Pyridines , Veins , Veratridine
18.
J Renin Angiotensin Aldosterone Syst ; 2(1_suppl): S37-S41, 2001 Mar.
Article in English | MEDLINE | ID: mdl-28095233

ABSTRACT

Subcutaneous arteries have been used as a model for resistance arteries, which are potentially involved in enhanced blood pressure (BP) regulation in man. Angiotensin II (Ang II) is an important regulator of tone, acting via type 1 (AT1-) and type 2 (AT2-) receptor subtypes. The aim of this study was to characterise the Ang II receptors in isolated human subcutaneous arteries, using pharmacological and molecular methods. Subcutaneous arteries were obtained from patients undergoing elective gut surgery and were carefully dissected from the abdominal wall. Cylindrical segments were mounted on two L-shaped metal prongs, one of which was connected to a force-displacement transducer for continuous recording of isometric tension. Concentration-response curves to Ang II were constructed in the presence and absence of various selective AT1-receptor antagonists, candesartan, EXP3174, irbesartan and losartan, and the AT2-receptor antagonist, PD 123319. Responses to Ang II were measured as increases in force (mN) and expressed as a percentage of the response to 60 mM of KCl. Ang II caused a concentration-dependent contraction (pEC50=9.45±0.48, Emax=120±13%). Candesartan and EXP3174 caused concentration-dependent depression of the Emax of Ang II without any major shift of pEC50. Losartan and irbesartan caused a significant, dose-dependent rightward shift of the Ang II contraction-response curve in human subcutaneous arteries. The results show that contractile responses of human subcutaneous arteries are mediated via the AT1-receptor. The AT1-receptor antagonists, candesartan and EXP3174, acted in an insurmountable manner, while losartan and irbesartan were surmountable AT1-receptor antagonists. The AT2-receptor antagonist, PD 123319, (10, 100 nM) had no effect on Ang II-induced contraction. This is supported by the positive identification of mRNA for the human AT 1-receptor by RT-PCR.

19.
J Renin Angiotensin Aldosterone Syst ; 2(1_suppl): S42-S47, 2001 Mar.
Article in English | MEDLINE | ID: mdl-28095239

ABSTRACT

Enhanced responses to vasoconstriction induced by neuropeptide Y and α2-adrenoceptor agonists have been seen following pharmacological activation of the adenylyl cyclase (AC) system. Since preliminary studies revealed only minor responses to angiotensin II (Ang II) in human omental arteries, we have investigated whether enhanced activity of AC may unravel further functional Ang II receptors. Human omental arteries were obtained in conjunction with elective gut surgery. After dissection of the vessel, the endothelium was removed by 10 sec of Triton X-100 treatment. Ring segments (1-2 mm long) were mounted on a myograph and studied. Ang II produced small contractions, 27±5% relative to the response elicited by 60 mM K+. However, enhanced Ang II (105±10%, p<0.001) responses were seen during AC activation by forskolin (0.1-1 µM). This enhanced contractile response to Ang II was not inhibited by the angiotensin II type 2 (AT2-receptor antagonist PD 123319 (0.1 µM), but was blocked in an insurmountable way by the angiotensin II type 1 (AT1)-receptor antagonist candesartan (1 nM) and in a surmountable manner by losartan (0.1 µM) and irbesartan (0.1 µM). Pertussis toxin (a Gi-protein blocker) and the protein kinase C inhibitor, RO31-8220 (0.01, 0.1 and 1 µM), markedly reduced this response, while the protein kinase A inhibitor, H89 (1, 10 µM), had no effect. RT-PCR provided evidence for the presence of mRNA for both AT1- and AT2-receptors. The results suggest that both a cAMP-dependent and a cAMP-independent mechanism are involved in the contractile responses to Ang II in human omental arteries and that both responses are mediated via the AT1-receptor.

SELECTION OF CITATIONS
SEARCH DETAIL
...