Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
Plus de filtres











Gamme d'année
1.
Biology (Basel) ; 13(6)2024 May 26.
Article de Anglais | MEDLINE | ID: mdl-38927262

RÉSUMÉ

Currently, it is known that angiotensin II (AngII) induces inflammation, and an AT1R blockade has anti-inflammatory effects. The use of an AT1 receptor antagonist promotes the inhibition of the secretion of multiple proinflammatory cytokines in macrophages, as well as a decrease in the concentration of reactive oxygen species. The aim of this study was to determine the effect of AT1 receptor gene silencing on the modulation of cytokines (e.g., IL-1ß, TNF-α, and IL-10) in THP-1 macrophages and the relation to the gene expression of NF-κB. MATERIALS AND METHODS: We evaluated the gene expression of PPAR-γ in THP-1 macrophages using PMA (60 ng/mL). For the silencing, cells were incubated with the siRNA for 72 h and telmisartan (10 µM) was added to the medium for 24 h. After that, cells were incubated during 1 and 24 h, respectively, with Ang II (1 µM). The gene expression levels of AT1R, NF-κB, and cytokines (IL-1ß, TNF-α, and IL-10) were measured by RT-qPCR. RESULTS: We observed that silencing of the AT1 receptor causes a decrease in the expression of mRNA of proinflammatory cytokines (IL-1ß and TNF-α), NF-κB, and PPAR-γ. CONCLUSIONS: We conclude that AT1R gene silencing is an alternative to modulating the production of proinflammatory cytokines such as TNF-α and IL-1ß via NF-κB in macrophages and having high blood pressure decrease.

2.
Eur J Neurosci ; 59(10): 2450-2464, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38480476

RÉSUMÉ

Amphetamine (AMPH) exposure induces behavioural and neurochemical sensitization observed in rodents as hyperlocomotion and increased dopamine release in response to a subsequent dose. Brain Angiotensin II modulates dopaminergic neurotransmission through its AT1 receptors (AT1-R), positively regulating striatal dopamine synthesis and release. This work aims to evaluate the AT1-R role in the development and maintenance of AMPH-induced sensitization. Also, the AT1-R involvement in striatal dopamine reuptake was analysed. The sensitization protocol consisted of daily AMPH administration for 5 days and tested 21 days after withdrawal. An AT1-R antagonist, candesartan, was administered before or after AMPH exposure to evaluate the participation of AT1-R in the development and maintenance of sensitization, respectively. Sensitization was evaluated by locomotor activity and c-Fos immunostaining. Changes in dopamine reuptake kinetics were evaluated 1 day after AT1-R blockade withdrawal treatment, with or without the addition of AMPH in vitro. The social interaction test was performed as another behavioural output. Repeated AMPH exposure induced behavioural and neurochemical sensitization, which was prevented and reversed by candesartan. The AT1-R blockade increased the dopamine reuptake kinetics. Neither the AMPH administration nor the AT1-R blockade altered the performance of social interaction. Our results highlight the AT1-R's crucial role in AMPH sensitization. The enhancement of dopamine reuptake kinetics induced by the AT1-R blockade might attenuate the neuroadaptive changes that lead to AMPH sensitization and its self-perpetuation. Therefore, AT1-R is a prominent candidate as a target for pharmacological treatment of pathologies related to dopamine imbalance, including drug addiction and schizophrenia.


Sujet(s)
Amfétamine , Antagonistes du récepteur de type 1 de l'angiotensine-II , Angiotensine-II , Benzimidazoles , Dérivés du biphényle , Corps strié , Dopamine , Animaux , Amfétamine/pharmacologie , Mâle , Dopamine/métabolisme , Corps strié/effets des médicaments et des substances chimiques , Corps strié/métabolisme , Angiotensine-II/pharmacologie , Dérivés du biphényle/pharmacologie , Benzimidazoles/pharmacologie , Antagonistes du récepteur de type 1 de l'angiotensine-II/pharmacologie , Rat Wistar , Rats , Récepteur de type 1 à l'angiotensine-II/métabolisme , Tétrazoles/pharmacologie , Stimulants du système nerveux central/pharmacologie , Interaction sociale/effets des médicaments et des substances chimiques , Activité motrice/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-fos/métabolisme
3.
Am J Med Sci ; 367(2): 128-134, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37984736

RÉSUMÉ

Cardiovascular diseases (CVD) are the main causes of death in hemodialysis patients, representing a public health challenge. We investigated the effect of different antihypertensive treatments on circulating levels of renin-angiotensin system (RAS) components in end-stage renal disease (ESRD) patients on hemodialysis. ESRD patients were grouped following the prescribed antihypertensive drugs: ß-blocker, ß-blocker+ACEi and ß-blocker+AT1R blocker. ESDR patients under no antihypertensive drug treatment were used as controls. Blood samples were collected before hemodialysis sessions. Enzymatic activities of the angiotensin-converting enzymes ACE and ACE2 were measured through fluorescence assays and plasma concentrations of the peptides Angiotensin II (Ang II) and Angiotensin-(1-7) [Ang-(1-7)] were quantified using mass spectrometry (LC-MS/MS). ACE activity was decreased only in the ß-blocker+ACEi group compared to the ß-blocker+AT1R, while ACE2 activity did not change according to the antihypertensive treatment. Both Ang II and Ang-(1-7) levels also did not change according to the antihypertensive treatment. We concluded that the treatment of ESRD patients on hemodialysis with different antihypertensive drugs do not alter the circulating levels of RAS components.


Sujet(s)
Antihypertenseurs , Défaillance rénale chronique , Humains , Antihypertenseurs/pharmacologie , Antihypertenseurs/usage thérapeutique , Angiotensin-converting enzyme 2/pharmacologie , Chromatographie en phase liquide , Spectrométrie de masse en tandem , Système rénine-angiotensine , Peptidyl-Dipeptidase A/métabolisme , Peptides/pharmacologie , Défaillance rénale chronique/traitement médicamenteux , Angiotensine-II/pharmacologie , Fragments peptidiques/métabolisme , Dialyse rénale
4.
Cell Signal ; 92: 110253, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35077849

RÉSUMÉ

Cardiovascular diseases are the leading cause of death worldwide. The renin-angiotensin-aldosterone system is one of the major regulators of cardiovascular homeostasis and the angiotensin II type 1 receptor (AT1R) mediates the main deleterious effects resulting from the hyperactivation of this hormonal system. Beta-arrestins are multifunctional proteins that regulate the desensitization and internalization of G protein-coupled receptors. After the discovery of beta-arrestins, many efforts have been made towards characterizing and distinguishing this new signaling pathway for drug discovery. Here, we summarize recent advances that address the beta-arrestin signaling in the cardiovascular system, focusing on the activation of the AT1R.


Sujet(s)
Antagonistes du récepteur de type 1 de l'angiotensine-II/usage thérapeutique , Maladies cardiovasculaires/anatomopathologie , Récepteur de type 1 à l'angiotensine-II/métabolisme , Système rénine-angiotensine/physiologie , bêta-Arrestines/métabolisme , Angiotensin-converting enzyme 2/métabolisme , COVID-19/anatomopathologie , Lignée cellulaire , Cellules HEK293 , Humains , Oligopeptides/usage thérapeutique , Transduction du signal/physiologie
5.
Biomedicines ; 11(1)2022 Dec 22.
Article de Anglais | MEDLINE | ID: mdl-36672525

RÉSUMÉ

Ischemia/reperfusion injury is a process associated with cardiologic interventions, such as percutaneous coronary angioplasty after an acute myocardial infarction. Blood flow restoration causes a quick burst of reactive oxygen species (ROS), which generates multiple organelle damage, leading to the activation of cell death pathways. Therefore, the intervention contributes to a greater necrotic zone, thus increasing the risk of cardiovascular complications. A major cardiovascular ROS source in this setting is the activation of multiple NADPH oxidases, which could result via the occupancy of type 1 angiotensin II receptors (AT1R); hence, the renin angiotensin system (RAS) is associated with the generation of ROS during reperfusion. In addition, ROS can promote the expression of NF-κΒ, a proinflammatory transcription factor. Recent studies have described an intracellular RAS pathway that is associated with increased intramitochondrial ROS through the action of isoform NOX4 of NADPH oxidase, thereby contributing to mitochondrial dysfunction. On the other hand, the angiotensin II/ angiotensin type 2 receptor (Ang II/AT2R) axis exerts its effects by counter-modulating the action of AT1R, by activating endothelial nitric oxide synthase (eNOS) and stimulating cardioprotective pathways such as akt. The aim of this review is to discuss the possible use of AT1R blockers to hamper both the Ang II/AT1R axis and the associated ROS burst. Moreover; we suggest that AT1R antagonist drugs should act synergistically with other cardioprotective agents, such as ascorbic acid, N-acetylcysteine and deferoxamine, leading to an enhanced reduction in the reperfusion injury. This therapy is currently being tested in our laboratory and has shown promising outcomes in experimental studies.

6.
Pharmaceuticals (Basel) ; 14(5)2021 May 16.
Article de Anglais | MEDLINE | ID: mdl-34065702

RÉSUMÉ

Activation of renin-angiotensin system (RAS) plays a role in bone deterioration associated with bone metabolic disorders, via increased Angiotensin II (AngII) targeting Angiotensin II type 1 receptor/Angiotensin II type 2 receptor (AT1R/AT2R). Despite the wide data availability, the RAS role remains controversial. This study analyzes the feasibility of using the embryonic chick femur organotypic model to address AngII/AT1R/AT2R axis in bone, which is an application not yet considered. Embryonic day-11 femurs were cultured ex vivo for 11 days in three settings: basal conditions, exposure to AngII, and modulation of AngII effects by prior receptor blockade, i.e., AT1R, AT2R, and AT1R + AT2R. Tissue response was evaluated by combining µCT and histological analysis. Basal-cultured femurs expressed components of RAS, namely ACE, AT1R, AT2R, and MasR (qPCR analysis). Bone formation occurred in the diaphyseal region in all conditions. In basal-cultured femurs, AT1R blocking increased Bone Surface/Bone Volume (BS/BV), whereas Bone Volume/Tissue Volume (BV/TV) decreased with AT2R or AT1R + AT2R blockade. Exposure to AngII greatly decreased BV/TV compared to basal conditions. Receptor blockade prior to AngII addition prevented this effect, i.e., AT1R blockade induced BV/TV, whereas blocking AT2R caused lower BV/TV increase but greater BS/BV; AT1R + AT2R blockade also improved BV/TV. Concluding, the embryonic chick femur model was sensitive to three relevant RAS research setups, proving its usefulness to address AngII/AT1R/AT2R axis in bone both in basal and activated conditions.

7.
J Cell Physiol ; 236(6): 4640-4654, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33345322

RÉSUMÉ

We have previously reported that angiotensin II receptor type 1 (AT1R) contributes to the hypertrophic effects of thyroid hormones (TH) in cardiac cells. Even though evidence indicates crosstalks between TH and AT1R, the underlying mechanisms are poorly understood. Beta-arrestin (ARRB) signaling has been described as noncanonical signal transduction pathway that exerts important effects in the cardiovascular system through G-protein-coupled receptors, as AT1R. Herein, we investigated the contribution of ARRB signaling in TH-induced cardiomyocyte hypertrophy. Primary cardiomyocyte cultures were treated with Triiodothyronine (T3) to induce cell hypertrophy. T3 rapidly activates extracellular signal-regulated kinase 1/2 (ERK1/2) signaling, which was partially inhibited by AT1R blockade. Also, ERK1/2 inhibition attenuated the hypertrophic effects of T3. ARRB2 was upregulated by T3, and small interfering RNA assays revealed the role of ARRB2-but not ARRB1-on ERK1/2 activation and cardiomyocyte hypertrophy. Corroborating these findings, the ARRB2-overexpressed cells showed increased expression of hypertrophic markers, which were attenuated by ERK1/2 inhibition. Immunocytochemistry and immunoprecipitation assays revealed the increased expression of nuclear AT1R after T3 stimulation and the increased interaction of AT1R/ARRB2. The inhibition of endocytosis also attenuated the T3 effects on cardiac cells. Our results evidence the contribution of ARRB2 on ERK1/2 activation and cardiomyocyte hypertrophy induced by T3 via AT1R.


Sujet(s)
Cardiomégalie/induit chimiquement , Myocytes cardiaques/effets des médicaments et des substances chimiques , Récepteur de type 1 à l'angiotensine-II/métabolisme , Tri-iodothyronine/toxicité , bêta-Arrestine 2/métabolisme , Animaux , Animaux nouveau-nés , Cardiomégalie/métabolisme , Cardiomégalie/anatomopathologie , Cellules cultivées , Endocytose/effets des médicaments et des substances chimiques , Activation enzymatique , Extracellular Signal-Regulated MAP Kinases/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Phosphorylation , Rat Wistar , Transduction du signal , bêta-Arrestine 2/génétique
8.
Biomed Pharmacother ; 109: 639-645, 2019 Jan.
Article de Anglais | MEDLINE | ID: mdl-30404071

RÉSUMÉ

Local renin-angiotensin system (RAS) in the pancreas is linked to the modulation of glucose-stimulated insulin secretion (GSIS) in beta cells and insulin sensitivity in target tissues, emerging as a promising tool in the prevention and/or treatment of obesity, diabetes, and systemic arterial hypertension. Insulin resistance alters pancreatic islet cell distribution and morphology and hypertrophied islets exhibit upregulated angiotensin II type 1 receptor, which drives oxidative stress, apoptosis, and fibrosis, configuring beta cell dysfunction and diminishing islet lifespan. Pharmacological modulation of RAS has shown beneficial effects in diet-induced obesity model, mainly related to the translational potential that angiotensin receptor blockers and ECA2/ANG (1-7)/MAS receptor axis modulation have when it comes to islet preservation and type 2 diabetes prevention and/or treatment. This review describes the existing evidence for different approaches to blocking RAS elements in the management of insulin resistance and diabetes and focuses on islet remodeling and GSIS in rodents and humans.


Sujet(s)
Systèmes de délivrance de médicaments/tendances , Homéostasie/physiologie , Ilots pancréatiques/métabolisme , Système rénine-angiotensine/physiologie , Angiotensine-I/antagonistes et inhibiteurs , Angiotensine-I/métabolisme , Angiotensine-II/métabolisme , Inhibiteurs de l'enzyme de conversion de l'angiotensine/pharmacologie , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique , Animaux , Systèmes de délivrance de médicaments/méthodes , Homéostasie/effets des médicaments et des substances chimiques , Humains , Insulinorésistance/physiologie , Ilots pancréatiques/effets des médicaments et des substances chimiques , Fragments peptidiques/antagonistes et inhibiteurs , Fragments peptidiques/métabolisme , Système rénine-angiotensine/effets des médicaments et des substances chimiques
9.
Pflugers Arch ; 470(3): 549-558, 2018 03.
Article de Anglais | MEDLINE | ID: mdl-29178049

RÉSUMÉ

We have previously demonstrated that calcium-binding protein S100A8 and myeloid differentiation factor-88 (MyD88) are important mediators of nuclear transcription factor kappa-B (NF-κB) activation in cardiomyocytes and that signalling molecules are involved in the hypertrophic response that is stimulated by thyroid hormones (TH). Angiotensin II (Ang II), the main active peptide of the renin-angiotensin system (RAS), binds to type 1 Ang II receptor (AT1R) and subsequently promotes cardiac hypertrophy and the inflammatory response with NF-κB activation underlying the cardiovascular effects. Considering the amount of evidence that RAS is an important mediator of TH actions on the cardiovascular system, we aimed to investigate whether cardiac expression of NF-κB and upstream associated molecules could be altered in hyperthyroidism, as well as whether AT1R could mediate the effects of TH on cardiac tissue and in cardiomyocytes in culture. Wistar rats were subjected to hyperthyroidism with or without the AT1R blocker losartan. The TH serum levels, haemodynamic parameters and cardiac mass were assessed to confirm the hyperthyroid status. The S100A8, MyD88 and nuclear NF-κB expression levels were increased in the hearts of the hyperthyroid rats, and the losartan treatment attenuated these TH effects. In addition, the cultured cardiomyocytes that had been stimulated with losartan exhibited blunted S100A8 upregulation and NF-κB activation compared with the TH-treated cells. Together, our results suggest that AT1R participates in TH-induced cardiac hypertrophy partly by mediating S100A8, MyD88 and NF-κB activation via TH. These findings indicate the important crosstalk between TH and RAS, highlighting the participation of AT1R in the triggered mechanisms of TH that contribute to the cardiac hypertrophy response.


Sujet(s)
Cardiomégalie/métabolisme , Hyperthyroïdie/métabolisme , Myocytes cardiaques/métabolisme , Récepteur de type 1 à l'angiotensine-II/métabolisme , Hormones thyroïdiennes/pharmacologie , Antagonistes du récepteur de type 1 de l'angiotensine-II/pharmacologie , Animaux , Calgranuline A/génétique , Calgranuline A/métabolisme , Cardiomégalie/étiologie , Cellules cultivées , Hyperthyroïdie/complications , Losartan/pharmacologie , Mâle , Facteur de différenciation myéloïde-88/génétique , Facteur de différenciation myéloïde-88/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/métabolisme , Rats , Rat Wistar , Hormones thyroïdiennes/sang
10.
Front Med (Lausanne) ; 5: 365, 2018.
Article de Anglais | MEDLINE | ID: mdl-30729109

RÉSUMÉ

Background: Tonin, a serine-protease that forms Angiotensin II (AngII) from angiotensinogen, is increased in failing human heart samples. Increased blood pressure (BP) and decreased heart rate (HR) variabilities are associated with higher risk of cardiovascular morbidity. Losartan has been used to reduce hypertension and, therefore, lowers the risk of fatal and non-fatal cardiovascular events. Determination of tonin's impact on BP and HR variabilities as well as the impact of losartan remain questions to be elucidated. Aim: Evaluation of cardiovascular autonomic profile in transgenic mice overexpressing the rat tonin enzyme TGM'(rton) and the impact of AT1 receptor blocker, losartan. Methods: Male C57BL/6 (WT) and TGM'(rTon) mice were cannulated for recording BP (Windaq, 4 MHz) for 30 min at baseline and 30 min after losartan injection (20 mg/kg). BP and HR variabilities were analyzed in time and frequency domain method. Low-frequency (LF) and high-frequency (HF) components were identified for sympathetic and parasympathetic modulations analysis. Ang I, AngII, and Ang1-7 were quantified by high performance liquid chromatography method. The total enzymatic activity for AngI, AngII, and Ang1-7 formation was evaluated in the heart and plasma by Liquid chromatography mass spectrometry (LC-MS/MS). Results: At the baseline TGM'(rTon) exhibited higher BP, lower cardiac LF, higher cardiac HF, lower LF/HF, and lower alpha index than wild type (WT). After losartan injection, TGM'(rTon) mice presented an additional decrease in cardiac LF and increase in HF in relation to baseline and WT. In the vasculature, losartan caused decreased in BP and LF of systolic BP in WT mice in relation to its baseline. A similar effect was observed in the BP of TGM'(rTon) mice; however, LF of systolic BP increased compared to baseline. Our data also indicates that AT1R receptor signaling has been altered in TGM'(rTon)mice. Interestingly, the dynamics of the renin-angiotensin system kinetics change, favoring production of Ang1-7. Conclusion: Autonomic evaluation of TGM'(rTon) mice indicates an unclear prognosis for diseases that affect the heart. HR variability in TGM'(rTon) mice indicates high risk of morbidity, and sympathetic and parasympathetic modulation indicate low risk of morbidity. The low risk of morbidity could be the biased production of Ang1-7 in the heart and circulation; however, the altered response of AT1R in the TGM'(rTon) remains to be elucidated, as well aswhether that signaling is pro-protection or pro-pathology.

11.
Front Pharmacol ; 9: 1553, 2018.
Article de Anglais | MEDLINE | ID: mdl-30804791

RÉSUMÉ

Hypertension is a disease associated to increased plasma levels of angiotensin II (Ang II). Ang II can regulate proliferation, migration, ROS production and hypertrophy of vascular smooth muscle cells (VSMCs). However, the mechanisms by which Ang II can affect VSMCs remain to be fully elucidated. In this context, autophagy, a process involved in self-digestion of proteins and organelles, has been described to regulate vascular remodeling. Therefore, we sought to investigate if Ang II regulates VSMC hypertrophy through an autophagy-dependent mechanism. To test this, we stimulated A7r5 cell line and primary rat aortic smooth muscle cells with Ang II 100 nM and measured autophagic markers at 24 h by Western blot. Autophagosomes were quantified by visualizing fluorescently labeled LC3 using confocal microscopy. The results showed that treatment with Ang II increases Beclin-1, Vps34, Atg-12-Atg5, Atg4 and Atg7 protein levels, Beclin-1 phosphorylation, as well as the number of autophagic vesicles, suggesting that this peptide induces autophagy by activating phagophore initiation and elongation. These findings were confirmed by the assessment of autophagic flux by co-administering Ang II together with chloroquine (30 µM). Pharmacological antagonism of the angiotensin type 1 receptor (AT1R) with losartan and RhoA/Rho Kinase inhibition prevented Ang II-induced autophagy. Moreover, Ang II-induced A7r5 hypertrophy, evaluated by α-SMA expression and cell size, was prevented upon autophagy inhibition. Taking together, our results suggest that the induction of autophagy by an AT1R/RhoA/Rho Kinase-dependent mechanism contributes to Ang II-induced hypertrophy in VSMC.

12.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;51(12): e7526, 2018. graf
Article de Anglais | LILACS | ID: biblio-974255

RÉSUMÉ

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.


Sujet(s)
Animaux , Mâle , Récepteurs alpha-adrénergiques/physiologie , Récepteurs bêta-adrénergiques/physiologie , Récepteur de type 1 à l'angiotensine-II/physiologie , Glucose/métabolisme , Hypertension portale/métabolisme , Foie/métabolisme , Propranolol/pharmacologie , Facteurs temps , Prazosine/pharmacologie , Récepteurs alpha-adrénergiques/effets des médicaments et des substances chimiques , Récepteurs bêta-adrénergiques/effets des médicaments et des substances chimiques , Rat Wistar , Antagonistes bêta-adrénergiques/pharmacologie , Losartan/pharmacologie , Récepteur de type 1 à l'angiotensine-II/effets des médicaments et des substances chimiques , Antagonistes du récepteur de type 1 de l'angiotensine-II/pharmacologie , Antagonistes des récepteurs aux angiotensines/pharmacologie , Hémodynamique/effets des médicaments et des substances chimiques , Hémodynamique/physiologie , Foie/effets des médicaments et des substances chimiques
13.
Am J Physiol Renal Physiol ; 313(2): F440-F449, 2017 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-28468964

RÉSUMÉ

ANG II has many biological effects in renal physiology, particularly in Ca2+ handling in the regulation of fluid and solute reabsorption. It involves the systemic endocrine renin-angiotensin system (RAS), but tissue and intracrine ANG II are also known. We have shown that ANG II induces heterodimerization of its AT1 and AT2 receptors (AT1R and AT2R) to stimulate sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) activity. Thus, we investigated whether ANG II-AT1R/AT2R complex is formed and internalized, and also examined the intracellular localization of this complex to determine how its effect might be exerted on renal intracrine RAS. Living cell imaging of LLC-PK1 cells, quantification of extracellular ANG II, and use of the receptor antagonists, losartan and PD123319, showed that ANG II is internalized with AT1R/AT2R heterodimers as a complex in a microtubule-dependent and clathrin-independent manner, since colchicine-but not Pitstop2-blocked this process. This result was confirmed by an increase of ß-arrestin phosphorylation after ANG II treatment, clathrin-mediated endocytosis being dependent on dephosphorylation of ß-arrestin. Internalized ANG II colocalized with an endoplasmic reticulum (ER) marker and increased levels of AT1R, AT2R, and PKCα in ER-enriched membrane fractions. This novel evidence suggests the internalization of an ANG II-AT1/AT2 complex to target ER, where it might trigger intracellular Ca2+ responses.


Sujet(s)
Angiotensine-II/métabolisme , Membrane cellulaire/métabolisme , Endocytose , Réticulum endoplasmique/métabolisme , Rein/métabolisme , Récepteur de type 1 à l'angiotensine-II/métabolisme , Récepteur de type 2 à l'angiotensine-II/métabolisme , Antagonistes du récepteur de type 1 de l'angiotensine-II/pharmacologie , Antagonistes du récepteur de type 2 de l'angiotensine-II/pharmacologie , Animaux , Calcium/métabolisme , Membrane cellulaire/effets des médicaments et des substances chimiques , Endocytose/effets des médicaments et des substances chimiques , Réticulum endoplasmique/effets des médicaments et des substances chimiques , Rein/effets des médicaments et des substances chimiques , Cellules LLC-PK1 , Microtubules/métabolisme , Complexes multiprotéiques , Phosphorylation , Protein kinase C-alpha/métabolisme , Transport des protéines , Récepteur de type 1 à l'angiotensine-II/effets des médicaments et des substances chimiques , Récepteur de type 2 à l'angiotensine-II/effets des médicaments et des substances chimiques , Sarcoplasmic Reticulum Calcium-Transporting ATPases/métabolisme , Suidae , bêta-Arrestines/métabolisme
14.
J Renin Angiotensin Aldosterone Syst ; 18(1): 1470320316689338, 2017 Jan.
Article de Anglais | MEDLINE | ID: mdl-28097883

RÉSUMÉ

Angiotensin-(3-4) (Ang-(3-4) or Val-Tyr) is the shorter angiotensin (Ang) II-derived peptide, formed through successive hydrolysis that culminates with the release of Val-Tyr as a dipeptide. It is formed both in plasma and in kidney from Ang II and Ang III, and can be considered a component of the systemic and organ-based renin-angiotensin system. It is potently antihypertensive in humans and rats, and its concerted actions on proximal tubule cells culminate in the inhibition of fluid reabsorption, hyperosmotic urinary excretion of Na+. At the renal cell signaling level, Ang-(3-4) counteracts Ang II-type 1 receptor-mediated responses by acting as an allosteric enhancer in Ang II-type 2 receptor populations that target adenosine triphosphate-dependent Ca2+ and Na+ transporters through a cyclic adenosine monophosphate-activated protein kinase pathway.


Sujet(s)
Angiotensine-II/métabolisme , Système rénine-angiotensine , Animaux , Humains , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Peptides/métabolisme , Récepteur de type 1 à l'angiotensine-II/métabolisme , Récepteur de type 2 à l'angiotensine-II/métabolisme
15.
Endocrine ; 50(2): 355-67, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-25854303

RÉSUMÉ

High activation of the angiotensin-converting enzyme (ACE)/(angiotensin-II type 1 receptor) AT1r axis is closely linked to pro-inflammatory effects and liver damage. The aim of this study was to evaluate the effects of the short-term administration of GW501516 on pro-inflammatory markers in white adipose tissue (WAT) and hepatic stellate cells (HSCs), lipogenesis and insulin resistance in the liver upon high-fructose diet (HFru)-induced ACE/AT1r axis activation. Three-month-old male C57Bl/6 mice were fed a standard chow diet or a HFru for 8 weeks. Then, the animals were separated randomly into four groups and treated with GW501516 for 3 weeks. Morphological variables, systolic blood pressure, and plasma determinations were analyzed. In the WAT, the ACE/AT1r axis and pro-inflammatory cytokines were assessed, and in the liver, the ACE/AT1r axis, HSCs, fatty acid oxidation, insulin resistance, and AMPK activation were evaluated. The HFru group displayed a high activation of the ACE/AT1r axis in both the WAT and liver; consequently, we detected inflammation and liver damage. Although GW501516 abolished the increased activation of the ACE/AT1r axis in the WAT, no differences were found in the liver. GW501516 blunted the inflammatory state in the WAT and reduced HSC activation in the liver. In addition, GW501516 alleviates damage in the liver by increasing the expression of the genes that regulate beta-oxidation and decreasing the expression of the genes and proteins that are involved in lipogenesis and gluconeogenesis. We conclude that GW501516 may serve as a therapeutic option for the treatment of a highly activated ACE/AT1r axis in WAT and liver.


Sujet(s)
Tissu adipeux blanc/effets des médicaments et des substances chimiques , Fructose/métabolisme , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Insulinorésistance , Lipogenèse/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Récepteur PPAR delta/agonistes , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Thiazoles/pharmacologie , Animaux , Mâle , Souris , Souris de lignée C57BL , Thiazoles/administration et posologie
16.
Life Sci ; 124: 81-90, 2015 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-25623850

RÉSUMÉ

AIMS: Activation of the renin-angiotensin system (RAS), renal oxidative stress and inflammation are constantly present in experimental hypertension. Nitric oxide (NO) inhibition with N(w)-nitro-L-arginine methyl ester (L-NAME) has previously been reported to produce hypertension, increased expression of Angiotensin II (Ang II) and renal dysfunction. The use of Losartan, an Ang II type 1 receptor (AT1R) antagonist has proven to be effective reducing hypertension and renal damage; however, the mechanism by which AT1R blockade reduced kidney injury and normalizes blood pressure in this experimental model is still complete unknown. The current study was designed to test the hypothesis that AT1R activation promotes renal NAD(P)H oxidase up-regulation, oxidative stress and cytokine production during L-NAME induced-hypertension. MAIN METHODS: Male Sprague-Dawley rats were distributed in three groups: L-NAME, receiving 70 mg/100ml of L-NAME, L-NAME+Los, receiving 70 mg/100ml of L-NAME and 40 mg/kg/day of Losartan; and Controls, receiving water instead of L-NAME or L-NAME and Losartan. KEY FINDINGS: After two weeks, L-NAME induced high blood pressure, renal overexpression of AT1R, NAD(P)H oxidase sub-units gp91, p22 and p47, increased levels of oxidative stress, interleukin-6 (IL-6) and interleukin-17 (IL-17). Also, we found increased renal accumulation of lymphocytes and macrophages. Losartan treatment abolished the renal expression of gp91, p22, p47, oxidative stress and reduced NF-κB activation and IL-6 expression. SIGNIFICANCE: These findings indicate that NO induced-hypertension is associated with up-regulation of NADPH oxidase, oxidative stress production and overexpression of key inflammatory mediators. These events are associated with up-regulation of AT1R, as evidenced by their reversal with AT1R blocker treatment.


Sujet(s)
Hypertension artérielle/physiopathologie , Inflammation/physiopathologie , Monoxyde d'azote/métabolisme , Stress oxydatif/physiologie , Récepteur de type 1 à l'angiotensine-II/métabolisme , Antagonistes du récepteur de type 1 de l'angiotensine-II/pharmacologie , Animaux , Pression sanguine/effets des médicaments et des substances chimiques , Hypertension artérielle/traitement médicamenteux , Inflammation/traitement médicamenteux , Interleukine-17/métabolisme , Interleukine-6/métabolisme , Losartan/pharmacologie , Mâle , NADH, NADPH oxidoreductases/métabolisme , NADPH Oxidase 4 , Facteur de transcription NF-kappa B/métabolisme , L-NAME/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Rats , Rat Sprague-Dawley , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Système rénine-angiotensine/physiologie , Régulation positive/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE