Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Peptides ; 146: 170660, 2021 12.
Article in English | MEDLINE | ID: mdl-34571055

ABSTRACT

Spinal cord neurons contribute to elevated sympathetic vasomotor activity in renovascular hypertension (2K1C), particularly, increased actions of angiotensin II. However, the origin of these spinal angiotensinergic inputs remains unclear. The present study aimed to investigate the role of spinal angiotensin II type 1 receptor (AT1) receptors in the sympathoexcitatory responses evoked by the activation of the rostral ventrolateral medulla (RVLM) in control and 2K1C Goldblatt rats. Hypertension was induced by clipping of the left renal artery. After 6 weeks, a catheter (PE-10) filled with losartan was inserted into the subarachnoid space and advanced to the T10-11 vertebral level in urethane-anesthetized rats. The effects of glutamate microinjection into the RVLM on blood pressure (BP), heart rate (HR), and renal and splanchnic sympathetic nerve activity (rSNA and sSNA, respectively) were evaluated in the presence or absence of spinal AT1 blockade. Tachycardic, pressor, and renal sympathoexcitatory effects caused by RVLM activation were significantly blunted by losartan in 2K1C rats, but not in control rats. However, no differences were found in the gene expression of angiotensin-converting enzyme, angiotensinogen, and renin in the spinal cord segments between the groups. In conclusion, acute sympathoexcitation induced by RVLM activation is dependent on the spinal AT1 receptor in Goldblatt, but not in control, rats. The involvement of other central cardiovascular nuclei in spinal angiotensinergic actions, as well as the source of angiotensin II, remains to be determined in the Goldblatt model.


Subject(s)
Hypertension/physiopathology , Kidney/innervation , Medulla Oblongata/physiology , Receptor, Angiotensin, Type 1/physiology , Spinal Cord/metabolism , Sympathetic Nervous System/physiology , Animals , Hypertension/metabolism , Male , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism
3.
J Neurochem ; 152(6): 675-696, 2020 03.
Article in English | MEDLINE | ID: mdl-31386177

ABSTRACT

Neuropathic and inflammatory pain results from cellular and molecular changes in dorsal root ganglion (DRG) neurons. The type-2 receptor for Angiotensin-II (AT2R) has been involved in this type of pain. However, the underlying mechanisms are poorly understood, including the role of the type-1 receptor for Angiotensin-II (AT1R). Here, we used a combination of immunohistochemistry and immunocytochemistry, RT-PCR and in vitro and in vivo pharmacological manipulation to examine how cutaneous inflammation affected the expression of AT1R and AT2R in subpopulations of rat DRG neurons and studied their impact on inflammation-induced neuritogenesis. We demonstrated that AT2R-neurons express C- or A-neuron markers, primarily IB4, trkA, and substance-P. AT1R expression was highest in small neurons and co-localized significantly with AT2R. In vitro, an inflammatory soup caused significant elevation of AT2R mRNA, whereas AT1R mRNA levels remained unchanged. In vivo, we found a unique pattern of change in the expression of AT1R and AT2R after cutaneous inflammation. AT2R increased in small neurons at 1 day and in medium size neurons at 4 days. Interestingly, cutaneous inflammation increased AT1R levels only in large neurons at 4 days. We found that in vitro and in vivo AT1R and AT2R acted co-operatively to regulate DRG neurite outgrowth. In vivo, AT2R inhibition impacted more on non-peptidergic C-neurons neuritogenesis, whereas AT1R blockade affected primarily peptidergic nerve terminals. Thus, cutaneous-induced inflammation regulated AT1R and AT2R expression and function in different DRG neuronal subpopulations at different times. These findings must be considered when targeting AT1R and AT2R to treat chronic inflammatory pain. Cover Image for this issue: doi: 10.1111/jnc.14737.


Subject(s)
Dermatitis/physiopathology , Receptor, Angiotensin, Type 1/physiology , Receptor, Angiotensin, Type 2/physiology , Sensory Receptor Cells/physiology , Animals , Cells, Cultured , Dermatitis/etiology , Female , Freund's Adjuvant/administration & dosage , Ganglia, Spinal/cytology , Neurites/physiology , Pain/physiopathology , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/analysis , Receptor, Angiotensin, Type 2/analysis , Sensory Receptor Cells/chemistry , Skin/innervation
4.
J Neuroendocrinol ; 31(4): e12703, 2019 04.
Article in English | MEDLINE | ID: mdl-30803087

ABSTRACT

Angiotensin II (Ang II) acts on Ang II type 1 (AT1) receptors located in the organum vasculosum and subfornical organ (SFO) of the lamina terminalis as a main facilitatory mechanism of sodium appetite. The brain serotonin (5-HT) system with soma located in the dorsal raphe nucleus (DRN) provides a main inhibitory mechanism. In the present study, we first investigated the existence of Ang II AT1 receptors in serotonergic DRN neurones. Then, we examined whether whole body sodium depletion affects the gene expression of the AT1a receptor subtype and the presumed functional significance of AT1 receptors. Using confocal microscopy, we found that tryptophan hydroxylase-2 and serotonin neurones express AT1 receptors in the DRN. Immunofluorescence quantification showed a significant reduction in 5-HT content but no change in AT1 receptor expression or AT1/5-HT colocalisation in the DRN after sodium depletion. Whole body sodium depletion also significantly increased Agtr1a mRNA expression in the SFO and DRN. Oral treatment with the AT1 receptor antagonist losartan reversed the changes in Agtr1a expression in the SFO but not the DRN. Losartan injection into either the DRN or the mesencephalic aqueduct had no influence on sodium depletion-induced 0.3 mol L-1 NaCl intake. The results indicate the expression of Agtr1a mRNA in the DRN and SFO as a marker of sodium depletion. They also suggest that serotonergic DRN neurones are targets for Ang II. However, the function of their AT1 receptors remains elusive.


Subject(s)
Dorsal Raphe Nucleus/metabolism , Gene Expression , Receptor, Angiotensin, Type 1/genetics , Serotonin/analysis , Sodium/deficiency , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Appetite/physiology , Dorsal Raphe Nucleus/chemistry , Fluorescent Antibody Technique , Gene Expression/physiology , Losartan/pharmacology , Male , Neurons/chemistry , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/analysis , Receptor, Angiotensin, Type 1/physiology , Sodium/blood , Subfornical Organ/chemistry , Subfornical Organ/metabolism , Tryptophan Hydroxylase/analysis
5.
Braz J Med Biol Res ; 51(12): e7526, 2018 Nov 14.
Article in English | MEDLINE | ID: mdl-30462770

ABSTRACT

It has been previously demonstrated that the hemodynamic effect induced by angiotensin II (AII) in the liver was completely abolished by losartan while glucose release was partially affected by losartan. Angiotensin II type 1 (AT1) and adrenergic (∝1- and ß-) receptors (AR) belong to the G-proteins superfamily, which signaling promote glycogen breakdown and glucose release. Interactive relationship between AR and AT1-R was shown after blockade of these receptors with specific antagonists. The isolated perfused rat liver was used to study hemodynamic and metabolic responses induced by AII and adrenaline (Adr) in the presence of AT1 (losartan) and ∝1-AR and ß-AR antagonists (prazosin and propranolol). All antagonists diminished the hemodynamic response induced by Adr. Losartan abolished hemodynamic response induced by AII, and AR antagonists had no effect when used alone. When combined, the antagonists caused a decrease in the hemodynamic response. The metabolic response induced by Adr was mainly mediated by ∝1-AR. A significant decrease in the hemodynamic response induced by Adr caused by losartan confirmed the participation of AT1-R. The metabolic response induced by AII was impaired by propranolol, indicating the participation of ß-AR. When both ARs were blocked, the hemodynamic and metabolic responses were impaired in a cumulative effect. These results suggested that both ARs might be responsible for AII effects. This possible cross-talk between ß-AR and AT1-R signaling in the hepatocytes has yet to be investigated and should be considered in the design of specific drugs.


Subject(s)
Glucose/metabolism , Hypertension, Portal/metabolism , Liver/metabolism , Receptor, Angiotensin, Type 1/physiology , Receptors, Adrenergic, alpha/physiology , Receptors, Adrenergic, beta/physiology , Adrenergic beta-Antagonists/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin Receptor Antagonists/pharmacology , Animals , Hemodynamics/drug effects , Hemodynamics/physiology , Liver/drug effects , Losartan/pharmacology , Male , Prazosin/pharmacology , Propranolol/pharmacology , Rats, Wistar , Receptor, Angiotensin, Type 1/drug effects , Receptors, Adrenergic, alpha/drug effects , Receptors, Adrenergic, beta/drug effects , Time Factors
6.
Cell Mol Neurobiol ; 38(1): 305-316, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28695320

ABSTRACT

The prelimbic cortex (PL) is an important structure in the neural pathway integrating stress responses. Brain angiotensin is involved in cardiovascular control and modulation of stress responses. Blockade of angiotensin receptors has been reported to reduce stress responses. Acute restraint stress (ARS) is a stress model, which evokes sustained blood pressure increase, tachycardia, and reduction in tail temperature. We therefore hypothesized that PL locally generated angiotensin and angiotensin receptors modulate stress autonomic responses. To test this hypothesis, we microinjected an angiotensin-converting enzyme (ACE) inhibitor or angiotensin antagonists into the PL, prior to ARS. Male Wistar rats were used; guide cannulas were bilaterally implanted in the PL for microinjection of vehicle or drugs. A polyethylene catheter was introduced into the femoral artery to record cardiovascular parameters. Tail temperature was measured using a thermal camera. ARS was started 10 min after PL treatment with drugs. Pretreatment with ACE inhibitor lisinopril (0.5 nmol/100 nL) reduced the pressor response, but did not affect ARS-evoked tachycardia. At a dose of 1 nmol/100 nL, it reduced both ARS pressor and tachycardic responses. Pretreatment with candesartan, AT1 receptor antagonist reduced ARS-evoked pressor response, but not tachycardia. Pretreatment with PD123177, AT2 receptor antagonist, reduced tachycardia, but did not affect ARS pressor response. No treatment affected ARS fall in tail temperature. Results suggest involvement of PL angiotensin in the mediation of ARS cardiovascular responses, with participation of both AT1 and AT2 receptors. In conclusion, results indicate that PL AT1-receptors modulate the ARS-evoked pressor response, while AT2-receptors modulate the tachycardic component of the autonomic response.


Subject(s)
Blood Pressure/physiology , Cerebral Cortex/metabolism , Heart Rate/physiology , Receptor, Angiotensin, Type 1/physiology , Receptor, Angiotensin, Type 2/physiology , Stress, Psychological/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Blood Pressure/drug effects , Cerebral Cortex/drug effects , Dose-Response Relationship, Drug , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Heart Rate/drug effects , Limbic Lobe/drug effects , Limbic Lobe/metabolism , Male , Rats , Rats, Wistar , Restraint, Physical/physiology , Restraint, Physical/psychology , Stress, Psychological/psychology
7.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;51(12): e7526, 2018. graf
Article in English | LILACS | ID: biblio-974255

ABSTRACT

It has been previously demonstrated that the hemodynamic effect induced by angiotensin II (AII) in the liver was completely abolished by losartan while glucose release was partially affected by losartan. Angiotensin II type 1 (AT1) and adrenergic (∝1- and β-) receptors (AR) belong to the G-proteins superfamily, which signaling promote glycogen breakdown and glucose release. Interactive relationship between AR and AT1-R was shown after blockade of these receptors with specific antagonists. The isolated perfused rat liver was used to study hemodynamic and metabolic responses induced by AII and adrenaline (Adr) in the presence of AT1 (losartan) and ∝1-AR and β-AR antagonists (prazosin and propranolol). All antagonists diminished the hemodynamic response induced by Adr. Losartan abolished hemodynamic response induced by AII, and AR antagonists had no effect when used alone. When combined, the antagonists caused a decrease in the hemodynamic response. The metabolic response induced by Adr was mainly mediated by ∝1-AR. A significant decrease in the hemodynamic response induced by Adr caused by losartan confirmed the participation of AT1-R. The metabolic response induced by AII was impaired by propranolol, indicating the participation of β-AR. When both ARs were blocked, the hemodynamic and metabolic responses were impaired in a cumulative effect. These results suggested that both ARs might be responsible for AII effects. This possible cross-talk between β-AR and AT1-R signaling in the hepatocytes has yet to be investigated and should be considered in the design of specific drugs.


Subject(s)
Animals , Male , Receptors, Adrenergic, alpha/physiology , Receptors, Adrenergic, beta/physiology , Receptor, Angiotensin, Type 1/physiology , Glucose/metabolism , Hypertension, Portal/metabolism , Liver/metabolism , Propranolol/pharmacology , Time Factors , Prazosin/pharmacology , Receptors, Adrenergic, alpha/drug effects , Receptors, Adrenergic, beta/drug effects , Rats, Wistar , Adrenergic beta-Antagonists/pharmacology , Losartan/pharmacology , Receptor, Angiotensin, Type 1/drug effects , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin Receptor Antagonists/pharmacology , Hemodynamics/drug effects , Hemodynamics/physiology , Liver/drug effects
8.
Neurobiol Learn Mem ; 127: 27-33, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26642920

ABSTRACT

Inappropriate defense-alerting reaction to fear is a common feature of neuropsychiatric diseases. Therefore, impairments in brain circuits, as well as in molecular pathways underlying the neurovegetative adjustments to fear may play an essential role on developing neuropsychiatric disorders. Here we tested the hypothesis that interfering with angiotensin-(1-7) [Ang-(1-7)]/Mas receptor axis homeostasis, which appears to be essential to arterial pressure control, would affect fear memory and extinction. Mas knockout (MasKO) mice, in FVB/N background, showed normal cued fear memory and extinction, but increased freezing in response to context. Next, as FVB/N has poor performance in contextual fear memory, we tested MasKO in mixed 129xC57BL/6 background. MasKO mice behaved similarly to wild-type (WT), but memory extinction was slower in contextual fear conditioning to a weak protocol (1CS/US). In addition, delayed extinction in MasKO mice was even more pronounced after a stronger protocol (3CS/US). We showed previously that Angiotensin II receptor AT1 antagonist, losantan, rescued object recognition memory deficit in MasKO mice. Here, losartan was also effective. Memory extinction was accelerated in MasKO mice after treatment with losartan. In conclusion, we showed for the first time that Ang-(1-7)/Mas axis may modulate fear memory extinction. Furthermore, we suggest MasKO mice as an animal model to study post-traumatic stress disorder (PTSD).


Subject(s)
Angiotensin I/physiology , Conditioning, Classical/physiology , Extinction, Psychological/physiology , Fear/physiology , Peptide Fragments/physiology , Proto-Oncogene Proteins/physiology , Receptor, Angiotensin, Type 1/physiology , Receptors, G-Protein-Coupled/physiology , Acoustic Stimulation , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Electroshock , Losartan/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics
9.
Bauru; s.n; 2016. 127 p. tab, ilus, graf.
Thesis in Portuguese | LILACS, BBO - Dentistry | ID: biblio-882111

ABSTRACT

O Sistema renina-angiotensina (SRA) tem sido relatado como um importante modulador de processos inflamatórios e imunológicos, incluindo a doença periodontal (DP). Estudos sugerem neste sistema um eixo alternativo (ECA-2 /ANG(1-7) /MAS) que atuaria como um contra-regulador de efeitos mediados pelo clássico eixo (ECA /ANGII /AT1). Sabe-se que bactérias periodontopatogênicas, como a Porphyromonas gingivalis (Pg), possuem componentes bioativos de membrana (ex. lipopolissacarídeos-LPS) capazes de induzir uma forte resposta imune no hospedeiro devido à liberação de citocinas nas células, entre elas Interleucina (IL)- 1ß. Neste contexto, fibroblastos são as células mais abundantes nos tecidos periodontais e possuem em sua superfície celular receptores necessários para o reconhecimento da invasão bacteriana, ativando cascatas intracelulares, que levam à produção de citocinas. O objetivo deste estudo foi verificar se os eixos ECA/ ANGII/ AT1 e ECA-2/ ANG(1-7)/ MAS contribuem para a produção e/ ou regulação de citocinas inflamatórias (CI) por fibroblastos de gengiva humana (HGF) e ligamento periodontal humano (HPLF) estimulados por IL-1ß. Após o pré-tratamento com Losartan e Ang (1-7) ou silenciamento mediado por RNA de interferência (RNAi) de AT1, HGF e HPLF foram estimulados por IL-1ß por 3 horas (RNAm) ou 24 horas (proteína). Expressão de RNAm para AT1, MAS, ECA, ECA-2, IL-1ß, TNF-α, IL-6, IL-8, IL-10, TGF-ß, CXCL12, RANK-L e OPG foram avaliados por RT-qPCR e das proteínas IL-6, IL-8, ECA e ECA-2 por ELISA. Foi realizado também Western Blot para detecção de AT1 e ECA nos extratos celulares e dosagem de nitrito no sobrenadante das culturas. Ambos os subtipos de fibroblastos mostraram aumento da expressão de RNAm para AT1, IL-1ß, IL-6, IL-8, TNF-α e OPG, quando estimulados por IL-1ß. No entanto, apenas em HPLF foi observado aumento para MAS, ECA e TGF-ß. Losartan e Ang (1-7) não modularam o transcrito, a secreção de CI e nem a produção de nitrito no sobrenadante das culturas, tanto em HGF como em HPLF. O silenciamento do receptor AT1 reduziu a secreção de IL-6 e IL-8 induzida por IL-1ß em cultura de HGF e HPLF e aumentou a expressão gênica de OPG somente em HGF. Estes resultados sugerem que o silenciamento de AT1, mas não o bloqueio farmacológico deste receptor pelo antagonista Losartan, em HGF e HPLF, pode controlar a produção de IL-6 e IL-8, que por sua vez contribuem para a patogênese periodontal.(AU)


The renin-angiotensin system (RAS) has been reported as an important modulator of inflammatory and immune responses, including periodontal disease (PD). Studies suggest an alternative axis as part of this system (ACE-2 / ANG (1-7) / MAS) that would act as counter-regulatory to the classical axis (ECA / ANGII / AT1). It is known that periodontal bacteria such as Porphyromonas gingivalis (Pg) have bioactive components in their membrane (such as lipopolysaccharide-LPS) capable of inducing a strong immune response in the host due to the release of cytokines in cells, including interleukin (IL) - 1ß. In this regard, fibroblasts are the most abundant cells in periodontal tissues and receptors needed for the recognition of bacterial invasion by activating intracellular cascades that lead to cytokine production. The aim of this study was to determine whether the axes ACE / ANGII / AT1 and ACE-2 / ANG (1-7) / MAS contribute to the production and / or regulation of inflammatory cytokines (IC) by fibroblasts of human gingiva (HGF) and human periodontal ligament (HPLF) stimulated IL-1ß. After pre-treatment with Losartan, Ang (1-7) or silencing mediated by RNA interference (RNAi) of AT1, HGF and HPLF were stimulated by IL-1ß for 3 hours (RNAm) or 24 hours (protein). Expression mRNA for AT1, MAS, ACE, ACE-2, IL-1ß, TNF-α, IL-6, IL-8, IL-10, TGF-ß, CXCL12, RANK-L and OPG was assessed by RT- qPCR and proteins IL-6, IL-8, ACE and ACE-2 by ELISA. Western Blot for the detection of AT1 and ECA and dosage of nitrite was also performed. Experiments stimulated by IL-1ß showed a positive control for gene expression AT1, IL-1ß, IL-6, IL-8, TNF-α and OPG in HGF and HPLF and MAS, ACE and TGF-ß only HPLF. Losartan and Ang (1-7) did not modulate the transcription and secretion of IC and no nitrite production in the culture supernatant of HGF and HPLF. The silencing AT1 reduced IL-6 secretion and IL-8 induced by IL- ß in cultured HGF and HPLF and increased OPG gene expression only HGF. These results suggest that silencing AT1, but not pharmacological blockade of this receptor by Losartan in HPLF and HGF, can control the production of IL-6 and IL-8, which in turn contribute to the pathogenesis of periodontal disease.(AU)


Subject(s)
Humans , Chemokines/metabolism , Cytokines/metabolism , Fibroblasts/physiology , Interleukin-1beta/physiology , Renin-Angiotensin System/physiology , Analysis of Variance , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II/analysis , Angiotensin II/physiology , Angiotensin I/analysis , Angiotensin I/physiology , Blotting, Western , Cells, Cultured , Chemokines/analysis , Cytokines/analysis , Gingiva/cytology , Losartan/pharmacology , Peptide Fragments/analysis , Peptide Fragments/physiology , Peptidyl-Dipeptidase A/analysis , Peptidyl-Dipeptidase A/physiology , Periodontal Ligament/cytology , Polymerase Chain Reaction , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/physiology , Receptor, Angiotensin, Type 1/analysis , Receptor, Angiotensin, Type 1/physiology
10.
Clin Sci (Lond) ; 127(5): 295-306, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24827941

ABSTRACT

The RAS (renin-angiotensin system) is composed of two arms: the pressor arm containing AngII (angiotensin II)/ACE (angiotensin-converting enzyme)/AT1Rs (AngII type 1 receptors), and the depressor arm represented by Ang-(1-7) [angiotensin-(1-7)]/ACE2/Mas receptors. All of the components of the RAS are present in the brain. Within the brain, Ang-(1-7) contributes to the regulation of BP (blood pressure) by acting at regions that control cardiovascular function such that, when Ang-(1-7) is injected into the nucleus of the solitary tract, caudal ventrolateral medulla, paraventricular nucleus or anterior hypothalamic area, a reduction in BP occurs; however, when injected into the rostral ventrolateral medulla, Ang-(1-7) stimulates an increase in BP. In contrast with AngII, Ang-(1-7) improves baroreflex sensitivity and has an inhibitory neuromodulatory role in hypothalamic noradrenergic neurotransmission. Ang-(1-7) not only exerts effects related to BP regulation, but also acts as a cerebroprotective component of the RAS by reducing cerebral infarct size and neuronal apoptosis. In the present review, we provide an overview of effects elicited by Ang-(1-7) in the brain, which suggest a potential role for Ang-(1-7) in controlling the central development of hypertension.


Subject(s)
Brain/physiology , Renin-Angiotensin System/physiology , Angiotensin I/metabolism , Angiotensin II/physiology , Angiotensin-Converting Enzyme 2 , Animals , Baroreflex/drug effects , Blood Pressure/drug effects , Brain/metabolism , Neurotransmitter Agents/metabolism , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/physiology , Proto-Oncogene Mas , Proto-Oncogene Proteins/physiology , Rats , Receptor, Angiotensin, Type 1/physiology , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology
12.
Learn Mem ; 20(7): 379-87, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23782508

ABSTRACT

Some memories enter into a labile state after retrieval, requiring reconsolidation in order to persist. One functional role of memory reconsolidation is the updating of existing memories. There are reports suggesting that reconsolidation can be modulated by a particular endogenous process taking place concomitantly to its natural course, such as water or sleep deprivation. Here, we investigated whether an endogenous process activated during a natural/physiological experience, or a pharmacological intervention, can also contribute to memory content updating. Using the contextual fear conditioning paradigm in rats, we found that the endogenous content of an aversive memory can be updated during its reconsolidation incorporating consequences of natural events such as water deprivation, transforming a previously stored memory into a state-dependent one. This updating seems to be mediated by the activation of angiotensin AT1 receptors in the dorsal hippocampus and local infusion of human angiotensin II (ANGII) was shown to mimic the water deprivation effects on memory reconsolidation. Systemic morphine injection was also able to turn a previously acquired experience into a state-dependent memory, reproducing the very same effects obtained by water deprivation or local angiotensin II infusion, and suggesting that other state-dependent-inducing protocols would also be able to contribute to memory updating. These findings trigger new insights about the influence of ordinary daily life events upon memory in its continuing reconstruction, adding the realm of reconsolidation to the classical view of endogenous modulation of consolidation.


Subject(s)
Memory/physiology , Water Deprivation/physiology , Angiotensin II/pharmacology , Animals , Conditioning, Psychological , Fear , Hippocampus/drug effects , Hippocampus/physiology , Male , Memory/drug effects , Morphine/pharmacology , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/physiology
13.
Auton Neurosci ; 177(2): 163-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23623788

ABSTRACT

The objective of this study was to explore the influence of the renin-angiotensin system on cardiac prooxidants and antioxidants levels and its association to autonomic imbalance induced by hyperthyroidism. Male Wistar rats were divided into four groups: control, losartan (10mg/kg/day by gavage, 28 day), thyroxine (T4) (12 mg/L in drinking water for 28 days), and T4+losartan. Spectral analysis (autonomic balance), angiotensin II receptor (AT1R), NADPH oxidase, Nrf2 and heme-oxygenase-1 (HO-1) myocardial protein expression, and hydrogen peroxide (H2O2) concentration were quantified. Autonomic imbalance induced by hyperthyroidism (~770%) was attenuated in the T4+losartan group (~32%) (P<0.05). AT1R, NADPH oxidase, H2O2, as well as concentration, Nrf2 and HO-1 protein expression were elevated (~172%, 43%, 40%, 133%, and 154%, respectively) in T4 group (P<0.05). H2O2 and HO-1 levels were returned to control values in the T4+losartan group (P<0.05). The overall results demonstrate a positive impact of RAS blockade in the autonomic control of heart rate, which was associated with an attenuation of H2O2 levels, as well as with a reduced counter-regulatory response of HO-1 in experimental hyperthyroidism.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Hyperthyroidism/metabolism , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/physiology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Heart Rate/drug effects , Heart Rate/physiology , Hyperthyroidism/drug therapy , Male , Rats , Rats, Wistar
14.
J Am Soc Hypertens ; 6(6): 369-74, 2012.
Article in English | MEDLINE | ID: mdl-23063532

ABSTRACT

The influence of mechanical stretch on the effect of angiotensin II (Ang II) on potassium current was investigated on cardiomyocytes isolated from the left ventricle of adult Sprague Dawley rats. Measurements of total potassium current were performed using the voltage clamp whole cell configuration. The results indicated: that mechanical stretch increased the potassium current appreciably, an effect inhibited by valsartan (10(-8) M), which is a strong inverse agonist of AT1 receptors; Ang II (10(-8) M) administered to the bath increased the potassium current by 60 ± 5.2% (n = 22) in cardiomyocytes isolated from Sprague Dawley rats; mechanical stretch (3 µm) applied in the longitudinal direction for a duration of 10 minutes, reduced the effect of Ang II (10(-8) M) on potassium current to 25 ± 4.3% (n = 24); 4) Bis-1 (300 nM), which is a specific inhibitor of protein kinase C, inhibited the effect of mechanical stretch on the increment of potassium current elicited by Ang II. In conclusion, the mechanical stretch of cardiomyocytes increases the potassium currents, an effect greatly dependent on the mechanical activation of AT1 receptors independently of Ang II. In addition, the increment of potassium currents caused by Ang II was greatly reduced by mechanical stretch, an effect abolished by protein kinase C inhibition.


Subject(s)
Angiotensin II/pharmacology , Myocytes, Cardiac/drug effects , Potassium Channels/drug effects , Receptor, Angiotensin, Type 1/physiology , Stress, Mechanical , Vasoconstrictor Agents/pharmacology , Action Potentials/drug effects , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Heart Ventricles/drug effects , Heart Ventricles/pathology , Indoles/pharmacology , Male , Maleimides/pharmacology , Mechanotransduction, Cellular/drug effects , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Tetrazoles/pharmacology , Valine/analogs & derivatives , Valine/pharmacology , Valsartan
15.
Horm Behav ; 62(1): 43-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22565127

ABSTRACT

There is an association between hypertension and reproductive dysfunction. Angiotensin II (Ang II) is involved in the pathogenesis of hypertension and the regulation of reproduction. The present study aimed to determine whether the angiotensinergic system mediates the effects of hypertension on reproductive function in male rats subjected to a two-kidney, one-clip (2K1C) model. Sexual behavior parameters, gametogenesis and plasma concentrations of Ang II, testosterone, prolactin and corticosterone were evaluated in male rats 28days after 2K1C or sham surgery and losartan (Los) treatment (a type 1 angiotensin II (AT1) receptor antagonist) or vehicle (V) treatment. The animals were divided into Sham+V, 2K1C+V, Sham+Los and 2K1C+Los groups. The 2K1C+V group showed a hypertensive response, inhibition of sexual behavior, spermatogenesis dysfunction, and increases in plasma Ang II and prolactin. Conversely, plasma testosterone decreased, and plasma corticosterone remained constant. Losartan treatment normalized blood pressure and prevented the changes in plasma testosterone and prolactin, sexual behavior and spermatogenesis in the 2K1C+Los group. In addition, losartan treatment caused an additional increase in circulating Ang ll in both groups (Sham+Los and 2K1C+Los). Together, these results suggest that Ang ll, acting through the AT1 receptor, modulates behavioral and endocrine parameters of reproductive function during renovascular hypertension. In addition, the effects of circulating Ang II on plasma testosterone and prolactin seem to contribute to the spermatogenic and sexual dysfunctions in hypertensive rats.


Subject(s)
Hypertension, Renovascular/physiopathology , Receptor, Angiotensin, Type 1/physiology , Spermatogenesis/physiology , Angiotensin II/blood , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Corticosterone/blood , Hypertension, Renovascular/drug therapy , Losartan/therapeutic use , Male , Prolactin/blood , Rats , Rats, Wistar , Sexual Behavior, Animal/drug effects , Sexual Behavior, Animal/physiology , Spermatogenesis/drug effects , Testosterone/blood
16.
Synapse ; 65(6): 505-12, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20936684

ABSTRACT

It has been shown that a single exposure to amphetamine is sufficient to induce long-term behavioral, neurochemical, and neuroendocrine sensitization in rats. Dopaminergic neurotransmission in the nucleus accumbens and the caudate-putamen plays a critical role in the addictive properties of drugs of abuse. Angiotensin (Ang) II receptors are found on the soma and terminals of mesolimbic dopaminergic neurons and it has been shown that Ang II acting through its AT1 receptors facilitates dopamine release. The hypothesis was tested that Ang II AT1 receptors are involved in the neuroadaptative changes induced by a single exposure to amphetamine and that such changes are related to the development of behavioral and neurochemical sensitization. For this purpose, the study examined the expression of amphetamine-enhanced (0.5 mg kg⁻¹ i.p.) locomotor activity in animals pretreated with candesartan, an AT1 blocker, (3 mg kg⁻¹ p.o. x 5 days), 3 weeks after an amphetamine injection (5 mg kg⁻¹ i.p.). Dopaminergic hyperreactivity was tested by measuring the 3H-DA release in vitro from caudate-putamen and nucleus accumbens slices, induced by K+ stimulus. It was confirmed the behavioral sensitization in the two-injection protocol and candesartan pretreatment attenuate this response. It was also found that AT1 blockade pretreatment did not affect the locomotor response to dopamine agonists. In respect to the neurochemical sensitization tested using ex vivo 3H-DA release experiments it was found that AT1 receptor pretreatment blunted the enhanced response induced by K+ stimulus. The results support the idea that the development of neuroadaptive changes induced by amphetamine involves brain AT1 Ang II receptor activation.


Subject(s)
Amphetamine-Related Disorders/metabolism , Amphetamine-Related Disorders/psychology , Amphetamine/toxicity , Angiotensin II Type 1 Receptor Blockers/pharmacology , Behavior, Animal/drug effects , Receptor, Angiotensin, Type 1/metabolism , Amphetamine/antagonists & inhibitors , Amphetamine-Related Disorders/physiopathology , Animals , Behavior, Animal/physiology , Disease Models, Animal , Injections, Intraperitoneal , Male , Organ Culture Techniques , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/physiology
17.
J Nutr ; 140(10): 1742-51, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20724490

ABSTRACT

High salt intake is a known cardiovascular risk factor and is associated with cardiac alterations. To better understand this effect, male Wistar rats were fed a normal (NSD: 1.3% NaCl), high 4 (HSD4: 4%), or high 8 (HSD8: 8%) salt diet from weaning until 18 wk of age. The HSD8 group was subdivided into HSD8, HSD8+HZ (15 mg . kg(-1) . d(-1) hydralazine in the drinking water), and HSD8+LOS (20 mg . kg(-1) . d(-1) losartan in the drinking water) groups. The cardiomyocyte diameter was greater in the HSD4 and HSD8 groups than in the HSD8+LOS and NSD groups. Interstitial fibrosis was greater in the HSD4 and HSD8 groups than in the HSD8+HZ and NSD groups. Hydralazine prevented high blood pressure (BP) and fibrosis, but not cardiomyocyte hypertrophy. Losartan prevented high BP and cardiomyocyte hypertrophy, but not fibrosis. Angiotensin II type 1 receptor (AT(1)) protein expression in both ventricles was greater in the HSD8 group than in the NSD group. Losartan, but not hydralazine, prevented this effect. Compared with the NSD group, the binding of an AT(1) conformation-specific antibody that recognizes the activated form of the receptor was lower in both ventricles in all other groups. Losartan further lowered the binding of the anti-AT(1) antibody in both ventricles compared with all other experimental groups. Angiotensin II was greater in both ventricles in all groups compared with the NSD group. Myocardial structural alterations in response to HSD are independent of the effect on BP. Salt-induced cardiomyocyte hypertrophy and interstitial fibrosis possibly are due to different mechanisms. Evidence from the present study suggests that salt-induced AT(1) receptor internalization is probably due to angiotensin II binding.


Subject(s)
Blood Pressure/physiology , Cardiomegaly/chemically induced , Cardiomegaly/physiopathology , Myocardium/pathology , Sodium Chloride, Dietary/administration & dosage , Aldosterone/blood , Angiotensin II/analysis , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Animals , Antihypertensive Agents/administration & dosage , Cardiomegaly/pathology , Collagen Type I/analysis , Collagen Type III/analysis , Disease Models, Animal , Drinking , Eating , Echocardiography , Fibrosis , Gene Expression , Heart Ventricles/chemistry , Heart Ventricles/pathology , Hydralazine/administration & dosage , Hypertension/physiopathology , Hypertension/prevention & control , Losartan/administration & dosage , Male , Potassium/blood , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/analysis , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/physiology , Receptor, Angiotensin, Type 2/analysis , Renin/blood , Renin-Angiotensin System/genetics , Renin-Angiotensin System/physiology , Sodium/blood , Sodium/urine , Transforming Growth Factor beta/analysis , Urine
18.
Basic Res Cardiol ; 105(3): 325-35, 2010 May.
Article in English | MEDLINE | ID: mdl-20155476

ABSTRACT

Although most of effects of Angiotensin II (Ang II) related to cardiac remodelling can be attributed to type 1 Ang II receptor (AT(1)R), the type 2 receptor (AT(2)R) has been shown to be involved in the development of some cardiac hypertrophy models. In the present study, we investigated whether the thyroid hormone (TH) action leading to cardiac hypertrophy is also mediated by increased Ang II levels or by change on AT(1)R and AT(2)R expression, which could contribute to this effect. In addition, we also evaluated the possible contribution of AT(2)R in the activation of Akt and in the development of TH-induced cardiac hypertrophy. To address these questions, Wistar rats were treated with thyroxine (T(4), 0.1 mg/kg BW/day, i.p.), with or without AT(2)R blocker (PD123319), for 14 days. Cardiac hypertrophy was identified based on heart/body weight ratio and confirmed by analysis of atrial natriuretic factor mRNA expression. Cardiomyocyte cultures were used to exclude the influence of TH-related hemodynamic effects. Our results demonstrate that the cardiac Ang II levels were significantly increased (80%, P < 0.001) as well as the AT(2)R expression (50%, P < 0.05) in TH-induced cardiac hypertrophy. The critical involvement of AT(2)R to the development of this cardiac hypertrophy in vivo was evidenced after administration of AT(2) blocker, which was able to prevent in 40% (P < 0.01) the cardiac mass gain and the Akt activation induced by TH. The role of AT(2)R to the TH-induced cardiomyocyte hypertrophy was also confirmed after using PD123319 in the in vitro studies. These findings improve understanding of the cardiac hypertrophy observed in hyperthyroidism and provide new insights into the generation of future therapeutic strategies.


Subject(s)
Angiotensin II Type 2 Receptor Blockers , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Myocardium/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Thyroxine/adverse effects , Angiotensin II/physiology , Animals , Cells, Cultured , Disease Models, Animal , Heart Diseases/physiopathology , Hyperthyroidism/physiopathology , Hypertrophy/chemically induced , Hypertrophy/physiopathology , Hypertrophy/prevention & control , Imidazoles/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/physiology , Pyridines/pharmacology , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/physiology , Receptor, Angiotensin, Type 2/physiology , Signal Transduction/physiology
20.
Nephron Exp Nephrol ; 112(1): e10-9, 2009.
Article in English | MEDLINE | ID: mdl-19342869

ABSTRACT

BACKGROUND/AIMS: Contributions to the understanding of acute renal failure (ARF) pathogenesis have not been translated into an effective clinical therapy. We studied the effects of pretreatment with the angiotensin II type 1 (AT1) receptor blocker, losartan, on renal function, tissue injury, inflammatory response and serum aldosterone levels in a model of ischemic ARF. METHODS: Rats underwent unilateral renal ischemia followed by 24 h of reperfusion (IR), and were pretreated or not with 8 (IRL8) or 80 (IRL80) mg/kg/day of losartan for 3 days. RESULTS: IR kidneys showed marked renal dysfunction, epithelial damage, capillary congestion, increased myeloperoxidase (MPO) activity and increased TNF-alpha, IL1-beta and IL-6 mRNA levels. IRL80 kidneys showed protection against dysfunction and tissue injury, associated with normal MPO activity and cytokine mRNA levels. The lower dose was not able to achieve the same degree of functional renoprotection and could not prevent an increase of MPO or proinflammatory cytokine mRNA levels. The high losartan dose completely prevented an increase of serum aldosterone levels induced by IR. CONCLUSION: Renoprotection of the high losartan dose would be mainly mediated by its anti-inflammatory actions. Our results show a potential pathophysiological role of AT1 activation in promoting renal dysfunction, structural injury, inflammation and aldosterone elevation after IR injury.


Subject(s)
Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Kidney/blood supply , Losartan/therapeutic use , Receptor, Angiotensin, Type 1/physiology , Reperfusion Injury/complications , Reperfusion Injury/prevention & control , Acute Disease , Animals , Kidney/drug effects , Kidney/pathology , Male , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL