Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Circ Res ; 135(7): 777-798, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39145385

RÉSUMÉ

BACKGROUND: Apelin is an endogenous prepropeptide that regulates cardiac homeostasis and various physiological processes. Intravenous injection has been shown to improve cardiac contractility in patients with heart failure. However, its short half-life prevents studying its impact on left ventricular remodeling in the long term. Here, we aim to study whether microparticle-mediated slow release of apelin improves heart function and left ventricular remodeling in mice with myocardial infarction (MI). METHODS: A cardiac patch was fabricated by embedding apelin-containing microparticles in a fibrin gel scaffold. MI was induced via permanent ligation of the left anterior descending coronary artery in adult C57BL/6J mice followed by epicardial patch placement immediately after (acute MI) or 28 days (chronic MI) post-MI. Four groups were included in this study, namely sham, MI, MI plus empty microparticle-embedded patch treatment, and MI plus apelin-containing microparticle-embedded patch treatment. Cardiac function was assessed by transthoracic echocardiography. Cardiomyocyte morphology, apoptosis, and cardiac fibrosis were evaluated by histology. Cardioprotective pathways were determined by RNA sequencing, quantitative polymerase chain reaction, and Western blot. RESULTS: The level of endogenous apelin was largely reduced in the first 7 days after MI induction and it was normalized by day 28. Apelin-13 encapsulated in poly(lactic-co-glycolic acid) microparticles displayed a sustained release pattern for up to 28 days. Treatment with apelin-containing microparticle-embedded patch inhibited cardiac hypertrophy and reduced scar size in both acute and chronic MI models, which is associated with improved cardiac function. Data from cellular and molecular analyses showed that apelin inhibits the activation and proliferation of cardiac fibroblasts by preventing transforming growth factor-ß-mediated activation of Smad2/3 (supporessor of mothers against decapentaplegic 2/3) and downstream profibrotic gene expression. CONCLUSIONS: Poly(lactic-co-glycolic acid) microparticles prolonged the apelin release time in the mouse hearts. Epicardial delivery of the apelin-containing microparticle-embedded patch protects mice from both acute and chronic MI-induced cardiac dysfunction, inhibits cardiac fibrosis, and improves left ventricular remodeling.


Sujet(s)
Apeline , Infarctus du myocarde , Animaux , Mâle , Souris , Apeline/administration et posologie , Apeline/métabolisme , Apeline/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Fibrose , Souris de lignée C57BL , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/métabolisme , Infarctus du myocarde/physiopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Remodelage ventriculaire/effets des médicaments et des substances chimiques
2.
J Control Release ; 372: 571-586, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38897292

RÉSUMÉ

Microvascular dysfunction following myocardial infarction exacerbates coronary flow obstruction and impairs the preservation of ventricular function. The apelinergic system, known for its pleiotropic effects on improving vascular function and repairing ischemic myocardium, has emerged as a promising therapeutic target for myocardial infarction. Despite its potential, the natural apelin peptide has an extremely short circulating half-life. Current apelin analogs have limited receptor binding efficacy and poor targeting, which restricts their clinical applications. In this study, we utilized an enzyme-responsive peptide self-assembly technique to develop an enzyme-responsive small molecule peptide that adapts to the expression levels of matrix metalloproteinases in myocardial infarction lesions. This peptide is engineered to respond to the high concentration of matrix metalloproteinases in the lesion area, allowing for precise and abundant presentation of the apelin motif. The changes in hydrophobicity allow the apelin motif to self-assemble into a supramolecular multivalent peptide ligand-SAMP. This self-assembly behavior not only prolongs the residence time of apelin in the myocardial infarction lesion but also enhances the receptor-ligand interaction through increased receptor binding affinity due to multivalency. Studies have demonstrated that SAMP significantly promotes angiogenesis after ischemia, reduces cardiomyocyte apoptosis, and improves cardiac function. This novel therapeutic strategy offers a new approach to restoring coronary microvascular function and improving damaged myocardium after myocardial infarction.


Sujet(s)
Apeline , Infarctus du myocarde , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/métabolisme , Animaux , Apeline/administration et posologie , Apeline/métabolisme , Ligands , Mâle , Souris de lignée C57BL , Humains , Néovascularisation physiologique/effets des médicaments et des substances chimiques
3.
J Endocrinol ; 249(1): 1-18, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33504680

RÉSUMÉ

Microcirculatory injuries had been reported to be involved in diabetic cardiomyopathy, which was mainly related to endothelial cell dysfunction. Apelin, an adipokine that is upregulated in diabetes mellitus, was reported to improve endothelial cell dysfunction and attenuate cardiac insufficiency induced by ischemia and reperfusion. Therefore, it is hypothesized that apelin might be involved in alleviating endothelial cell dysfunction and followed cardiomyopathy in diabetes mellitus. The results showed that apelin improved endothelial cell dysfunction via decreasing apoptosis and expression of adhesion molecules and increasing proliferation, angiogenesis, and expression of E-cadherin, VEGFR 2 and Tie-2 in endothelial cells, which resulted in the attenuation of the capillary permeability in cardiac tissues and following diabetic cardiomyopathy. Meanwhile, the results from endothelial cell-specific APJ knockout mice and cultured endothelial cells confirmed that the effects of apelin on endothelial cells were dependent on APJ and the downstream NFκB pathways. In conclusion, apelin might reduce microvascular dysfunction induced by diabetes mellitus via improving endothelial dysfunction dependent on APJ activated NFκB pathways.


Sujet(s)
Récepteur de l'apeline/physiologie , Apeline/physiologie , Cardiomyopathies diabétiques/traitement médicamenteux , Cellules endothéliales/effets des médicaments et des substances chimiques , Microvaisseaux/physiopathologie , Animaux , Apeline/administration et posologie , Récepteur de l'apeline/déficit , Glycémie/analyse , Molécules d'adhérence cellulaire/analyse , Cellules cultivées , Diabète expérimental/sang , Diabète de type 2/sang , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/physiopathologie , Cellules endothéliales/anatomopathologie , Cellules endothéliales/physiologie , Inflammation/traitement médicamenteux , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Microvaisseaux/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme
4.
Nat Commun ; 12(1): 305, 2021 01 12.
Article de Anglais | MEDLINE | ID: mdl-33436646

RÉSUMÉ

Apelin and arginine-vasopressin (AVP) are conversely regulated by osmotic stimuli. We therefore hypothesized that activating the apelin receptor (apelin-R) with LIT01-196, a metabolically stable apelin-17 analog, may be beneficial for treating the Syndrome of Inappropriate Antidiuresis, in which AVP hypersecretion leads to hyponatremia. We show that LIT01-196, which behaves as a potent full agonist for the apelin-R, has an in vivo half-life of 156 minutes in the bloodstream after subcutaneous administration in control rats. In collecting ducts, LIT01-196 decreases dDAVP-induced cAMP production and apical cell surface expression of phosphorylated aquaporin 2 via AVP type 2 receptors, leading to an increase in aqueous diuresis. In a rat experimental model of AVP-induced hyponatremia, LIT01-196 subcutaneously administered blocks the antidiuretic effect of AVP and the AVP-induced increase in urinary osmolality and induces a progressive improvement of hyponatremia. Our data suggest that apelin-R activation constitutes an original approach for hyponatremia treatment.


Sujet(s)
Apeline/analogues et dérivés , Apeline/métabolisme , Arginine vasopressine/effets indésirables , Diurèse , Hyponatrémie/anatomopathologie , Hyponatrémie/physiopathologie , Séquence d'acides aminés , Animaux , Apeline/administration et posologie , Apeline/sang , Récepteur de l'apeline/métabolisme , Arginine vasopressine/sang , Glycémie/métabolisme , Pression sanguine/effets des médicaments et des substances chimiques , Lignée cellulaire , Colforsine/pharmacologie , AMP cyclique/biosynthèse , Desmopressine/pharmacologie , Modèles animaux de maladie humaine , Diurèse/effets des médicaments et des substances chimiques , Électrolytes/sang , Période , Hyponatrémie/sang , Hyponatrémie/urine , Tubules collecteurs rénaux/effets des médicaments et des substances chimiques , Tubules collecteurs rénaux/métabolisme , Tubules collecteurs rénaux/physiopathologie , Mâle , Souris , Modèles biologiques , Contraction myocardique/effets des médicaments et des substances chimiques , Peptides/composition chimique , Peptides/pharmacologie , Phosphorylation/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Tolvaptan/pharmacologie
5.
Mol Med Rep ; 23(1)2021 01.
Article de Anglais | MEDLINE | ID: mdl-33179090

RÉSUMÉ

Apelin­36 is able to mediate a range of effects on various diseases, and is upregulated in lipopolysaccharide (LPS)­induced acute lung injury (ALI). However, to the best of our knowledge, whether apelin­36 is able to regulate LPS­induced ALI has yet to be investigated. The present study aimed to investigate the role of apelin­36 in LPS­induced ALI, and the putative underlying mechanisms. Rats were assigned to one of four treatment groups: The Control group, apelin­36 group, LPS group and LPS + apelin­36 group. At 4 h after intratracheal instillation of LPS (5 mg/kg), rats were intraperitoneally treated with 10 nmol/kg apelin­36. Subsequently, pathological manifestations and the extent of inflammation and apoptosis of the lung tissues were assessed. Untransfected and apoptosis signal­regulating kinase 1 (ASK1)­overexpressing Beas­2B cells were treated with LPS in the absence or presence of apelin­36, and subsequently the levels of inflammation and apoptosis were assessed. The results obtained showed that the level of apelin­36 was increased in the bronchoalveolar lavage fluid (BALF) of LPS­treated rats. Co­treatment with apelin­36 alleviated LPS­induced lung injury and pulmonary edema, reduced the levels of pro­inflammatory cytokines, including interleukin­6, monocyte chemoattractant protein­1 and tumor necrosis factor­α, in BALF, and inhibited apoptosis in the lung tissues. The presence of apelin­36 also blocked the activation of LPS­induced ASK1, p38, c­Jun N­terminal kinase and extracellular signal­regulated kinase in lung tissues. In vitro studies performed with Beas­2B cells showed that the addition of apelin­36 led to an increase in the cell viability of LPS­induced Beas­2B cells in a concentration­dependent manner. Additionally, co­treatment with 1 µM apelin­36 prevented LPS­induced inflammation and apoptosis. However, overexpression of ASK1 significantly reversed the inhibitory effects of apelin­36 on LPS­induced inflammation and apoptosis. Taken together, the results of the present study demonstrated that apelin­36 was able to protect against LPS­induced lung injury both in vivo and in vitro, and these actions may be dependent on inhibition of the ASK1/mitogen­activated protein kinase signaling pathway.


Sujet(s)
Lésion pulmonaire aigüe/traitement médicamenteux , Apeline/administration et posologie , Lipopolysaccharides/effets indésirables , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/génétique , Lésion pulmonaire aigüe/métabolisme , Animaux , Apeline/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Injections péritoneales , MAP Kinase Kinase Kinase 5/métabolisme , Mâle , Répartition aléatoire , Rats
6.
Mol Cell Endocrinol ; 504: 110695, 2020 03 15.
Article de Anglais | MEDLINE | ID: mdl-31904406

RÉSUMÉ

Stable apelin-13 peptide analogues have shown promising acute antidiabetic effects in mice with diet-induced obesity diabetes. Here the efficacy of (pGlu)apelin-13 amide (apelin amide) and the acylated analogue (pGlu)(Lys8GluPAL)apelin-13 amide (apelin FA), were examined following chronic administration in db/db mice, a genetic model of degenerative diabetes. Groups of 9-week old male db/db mice (n = 8) received twice daily injections (09:00 and 17:00 h; i.p.) or saline vehicle, apelin amide, apelin FA, or the established incretin therapies, exendin-4(1-39) or liraglutide, all at 25 nmol/kg body weight for 21 days. Control C57BL/6J mice were given saline twice daily. No changes in body weight or food intake were observed with either apelin or liraglutide treatments, but exendin-4 showed a reduction in cumulative food intake (p < 0.01) compared with saline-treated db/db mice. Apelin analogues and incretin mimetics induced sustained improvements of glycaemia (p < 0.05 to p < 0.001, from day 9-21), lowered HbA1c at 21 days (p < 0.05) and raised plasma insulin concentrations. The treatments also improved OGTT and ipGTT with enhanced insulin responses compared with saline-treated control db/db mice (p < 0.05 to p < 0.001). Apelin amide was superior to incretin mimetics in lowering plasma triglycerides by 34% (p < 0.05). Apelin analogues unlike both incretin mimetics reduced pancreatic α-cell area (p < 0.05 to p < 0.01) and all peptide treatments enhanced pancreatic insulin content (p < 0.05 to p < 0.01). In conclusion, longer-term administration of apelin-13 analogues, induced similar and in some respects more effective metabolic improvements than incretin mimetics in db/db mice, providing a viable alternative approach for counteracting metabolic dysfunction for mild and more degenerative forms of the disease.


Sujet(s)
Apeline/analogues et dérivés , Diabète expérimental/traitement médicamenteux , Hypoglycémiants/administration et posologie , Maladies métaboliques/traitement médicamenteux , Animaux , Apeline/administration et posologie , Glycémie/métabolisme , Diabète expérimental/complications , Diabète expérimental/génétique , Diabète expérimental/métabolisme , Relation dose-effet des médicaments , Incrétines/administration et posologie , Protéines et peptides de signalisation intercellulaire/administration et posologie , Protéines et peptides de signalisation intercellulaire/composition chimique , Maladies métaboliques/étiologie , Maladies métaboliques/métabolisme , Souris , Souris de lignée C57BL , Souris transgéniques , Récepteurs à la leptine/génétique , Résultat thérapeutique
7.
Brain Res ; 1726: 146493, 2020 01 01.
Article de Anglais | MEDLINE | ID: mdl-31586624

RÉSUMÉ

Parkinson's disease (PD), a common human neurodegenerative disorder, is characterized by the presence of intraneuronal Lewy bodies composed principally of abnormal aggregated and post-translationally modified α-synuclein. In our previous research, we have demonstrated the neuroprotective effect of Apelin-36, a neuroendocrine peptide in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin (MPTP)-lesioned PD model mice. Therefore, this study was designed to evaluate the neuroprotective mechanism of Apelin-36 against MPTP-induced neurotoxicity in mice. The results showed that MPTP-induced the depletion of dopamine in the striatum (STR) was partially reversed by Apelin-36. Apelin-36 also improved the activity of antioxidant system including superoxide dismutase (SOD) and glutathione (GSH), and decreased the overproduction of malondialdehyde (MDA) in the substantia nigra pars compacta (SNpc) and STR of MPTP-treated mice. Moreover, Apelin-36 downregulated inducible nitric oxide synthase (iNOS) and nitrated α-synuclein expression. Furthermore, Apelin-36 significantly promoted autophagy indicated by the up-regulation of LC3-II and Beclin1 and inhibition of p62 expression in the SNpc and STR of MPTP-treated mice. The protective effect of Apelin-36 was also associated with the inhibition of the apoptosis signal-regulating kinase 1 (ASK1)/c-Jun N-terminal kinase (JNK) signaling pathway and inactivation of caspase-3. Taken together, our findings demonstrated that the neuroprotective mechanism of Apelin-36 against MPTP-induced neurotoxicity in mice might be related to decreasing the aggregation of nitrated α-synuclein and alleviating oxidative stress as well as promoting autophagy and inhibiting ASK1/JNK/caspase-3 apoptotic pathway, which provides a novel strategy for PD treatment.


Sujet(s)
Apeline/administration et posologie , Apeline/métabolisme , Autophagie , Intoxication au MPTP/métabolisme , Neuroprotecteurs/administration et posologie , Stress oxydatif , Animaux , Autophagie/effets des médicaments et des substances chimiques , Corps strié/effets des médicaments et des substances chimiques , Corps strié/métabolisme , Modèles animaux de maladie humaine , Dopamine/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Mâle , Souris de lignée C57BL , Stress oxydatif/effets des médicaments et des substances chimiques , Pars compacta/effets des médicaments et des substances chimiques , Pars compacta/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
8.
Clin Exp Pharmacol Physiol ; 47(3): 393-402, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-31630435

RÉSUMÉ

The renin-angiotensin system (RAS) has a deleterious and apelin/APJ system has protective effect on the ischaemic heart. The collaboration between these systems in the pathophysiology of myocardial infarction is not clear. We determined the effect of chronic pretreatment with apelin, losartan and their combination on ischaemia-reperfusion (IR) injury in the isolated perfused rat heart and on the expression of apelin-13 receptor (APJ) and angiotensin type 1 receptor (AT1R) in the myocardium. During 5 days before the induction of IR, saline (vehicle), apelin-13 (Apl), F13A (apelin antagonist), losartan (Los, AT1R antagonist) and the combination of Apl and Los were administered intraperitoneally in rats. Ischaemia was induced by left anterior descending (LAD) artery occlusion for 30 minutes followed by reperfusion for 55 minutes in the Langendorff isolated heart perfusion system. Pretreatment with Apl, Los and the combination of Apl + Los significantly reduced infarct size by about 30, 33 and 48 percent respectively; and significantly improved the left ventricular function indices such as left ventricular developed pressure (LVDP), left ventricular end-diastolic pressure (LVEDP) and rate pressure product (RPP). IR increased AT1R protein level but it did not change APJ significantly. AT1R expression was reduced in groups treated with Apl, Los and Apl + Los. Findings showed that chronic pretreatment with apelin along with AT1R antagonist had more protective effects against IR injury. Combination therapy may diminish the risk of IR-induced heart damage, by reducing AT1R expression, in the heart of patients with coronary artery disease that are at the risk of MI and reperfusion injury.


Sujet(s)
Antagonistes du récepteur de type 1 de l'angiotensine-II/administration et posologie , Antiarythmiques/administration et posologie , Apeline/administration et posologie , Rythme cardiaque/effets des médicaments et des substances chimiques , Losartan/administration et posologie , Infarctus du myocarde/traitement médicamenteux , Animaux , Association de médicaments , Rythme cardiaque/physiologie , Mâle , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/physiopathologie , Rats , Rat Wistar
9.
United European Gastroenterol J ; 7(5): 689-698, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-31210947

RÉSUMÉ

Introduction: Hepatic ischemic reperfusion injury occurs in multiple clinical settings. Novel potential protective agents are still needed to attenuate this injury. Apelin preconditioning protects against ischemic reperfusion injury in different organs. However, the protective mechanism of apelin on hepatic ischemic reperfusion injury is not yet clear. Aim: Evaluate the effect of apelin-13 preconditioning on hepatic ischemic reperfusion injury and clarify possible interactions between apelinergic, renin-angiotensin systems and endothelial nitric oxide synthase. Methods: In total, 60 rats were assigned to four groups: control sham-operated, ischemic reperfusion, apelin-treated ischemic reperfusion and apelin + N-nitro-L-arginine methyl ester-treated ischemic reperfusion. Apelin 2 µg/kg/day and N-nitro-L-arginine methyl ester 10 mg/kg/day were injected intraperitoneally daily for 3 days and 2 weeks respectively before hepatic ischemic reperfusion. Serum aminotransferase, aspartate aminotransferase, hepatic malondialdehyde, apelin, gene expression of caspase-3, endothelial nitric oxide synthase and angiotensin type 1 receptor and liver histopathology were compared between groups. Results: Apelin significantly reduced serum aminotransferase, aspartate aminotransferase, hepatic malondialdehyde, caspase-3 and angiotensin type 1 receptor expression, whereas hepatic apelin and endothelial nitric oxide synthase expression were significantly increased with improved hepatic histopathology. N-nitro-L-arginine methyl ester co-administration partially reversed this hepatoprotective effect. Conclusion: Apelin-13 reduced hepatic ischemic reperfusion injury. This protection could be related to the suppression of hepatic angiotensin type 1 receptor expression and elevation of hepatic apelin level and endothelial nitric oxide synthase expression, which counteracts the pathologic effects of Ang II/angiotensin type 1 receptor. An interaction exists between apelinergic, renin-angiotensin systems and endothelial nitric oxide synthase in hepatic ischemic reperfusion pathophysiology.


Sujet(s)
Angiotensine-II/métabolisme , Apeline/administration et posologie , Apeline/métabolisme , Préconditionnement ischémique/méthodes , Nitric oxide synthase type III/métabolisme , Récepteur de type 1 à l'angiotensine-II/métabolisme , Lésion d'ischémie-reperfusion/prévention et contrôle , Alanine transaminase/sang , Animaux , Aspartate aminotransferases/sang , Caspase-3/génétique , Modèles animaux de maladie humaine , Expression des gènes , Foie/métabolisme , Foie/anatomopathologie , Mâle , Malonaldéhyde/métabolisme , L-NAME/administration et posologie , L-NAME/métabolisme , Rats , Récepteur de type 1 à l'angiotensine-II/génétique , Système rénine-angiotensine/physiologie , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie
10.
J Physiol Pharmacol ; 70(6)2019 12.
Article de Anglais | MEDLINE | ID: mdl-32084650

RÉSUMÉ

Apelin was thought to be an adipocyte-specific hormone, but recent studies have indicated a link between apelin and placenta function e.g. cell proliferation. The aim of the study was investigating dose- and time-dependent effect of apelin on hormone secretion including steroids: progesterone (P4) and estradiol (E2) and proteins: chorionic gonadotropin (hCG), human placental lactogen (hPL), placental growth factor (PLGF), as well as protein expression of steroid enzymes (3ßHSD, CYP19) and protein hormones (hCG, hPL and PLGF) in placental cells. Syncytiotrophoblast BeWo cells, as human trophoblast models, were treated for 24, 48, and 72 hours with the human recombinant apelin at doses 0.02, 0.2, 2.0, 20 and 200 ng/ml followed by culture medium. Concentrations of the above hormones were studied by ELISA kits. Furthermore, protein expression of steroid enzymes and protein hormones were measured using Western blot. Our results showed that apelin significantly decreased both steroid and protein hormones by inhibiting steroid enzymes or protein hormone expression. Moreover, we demonstrated that apelin at dose 2.0 ng/ml increased phosphorylation of protein kinase A (PKA) from 1 to 60 min of BeWo cell incubation. Inhibitory effect of apelin on P4, E2 and PLGF secretion were abolished when BeWo cells were cultured in the presence of ML221, an apelin receptor antagonist, PD98059, an extracellular signal-regulated kinases (ERK1/2) antagonist and KT5720, a PKA antagonist. In turn, secretion of hCG and hPL occurs only in the presence of ML221 and PD98059. In conclusion, our results indicate that apelin can be considered as a gestational hormone implied in the endocrine function of the human placenta, with an important role in controlling the production of steroid and protein hormones in placental BeWo cells.


Sujet(s)
Récepteur de l'apeline/métabolisme , Apeline/métabolisme , Placenta/métabolisme , Trophoblastes/métabolisme , Apeline/administration et posologie , Lignée cellulaire tumorale , Choriocarcinome/métabolisme , Gonadotrophine chorionique/métabolisme , Cyclic AMP-Dependent Protein Kinases/métabolisme , Relation dose-effet des médicaments , Femelle , Humains , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3/métabolisme , Facteur de croissance placentaire/métabolisme , Hormone lactogène placentaire/métabolisme , Grossesse , Facteurs temps
11.
Clin Exp Pharmacol Physiol ; 46(1): 29-39, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30225902

RÉSUMÉ

Exposure to stress induces gastrointestinal (GI) dysmotility. In rodents, acute restraint stress (ARS) inhibits gastric emptying (GE) and intestinal transit (IT) via central and peripheral corticotropin-releasing factor (CRF)-mediated pathways. Peripherally administered apelin-13 was shown to inhibit GI motor functions; moreover, stress-induced upregulation of gastric apelin content was demonstrated in rats suggesting that peripheral apelin may mediate stress-induced alterations in GI motility. We investigated the role of endogenous peripheral apelin in stress-induced GI dysfunction. GE, IT and gastro-duodenal fasting motility were measured in non-stressed (NS), CRF-injected and ARS-loaded rats. CRF and apelin receptor antagonists astressin or F13A was administered before ARS or peripheral CRF injection. Apelin and APJ receptor expressions were determined using immunohistochemistry and quantified by qRT-PCR. Double immunofluorescence was performed for enteric neuronal apelin. GE and IT were delayed by CRF and ARS. ARS-induced changes were attenuated by F13A, whereas astressin was ineffective. CRF-induced alterations in GE and IT were restored completely by astressin, while they were diminished by F13A. Antral phase III-like contractions were disturbed following ARS which were preserved by preadministration of astressin, but not F13A. CRF impaired gastric and duodenal fasting contractions, while these changes were not altered by F13A. ARS increased apelin expression in stomach and duodenum. Apelin immunoreactivity was detected in mucosa, smooth muscles and myenteric plexi, whereas dense APJ receptor expression was observed within tunica muscularis. APJ receptor was downregulated in rats fasted overnight. These results suggest that enteric apelin acts as an inhibitor stress mediator in the postprandial state.


Sujet(s)
Apeline/administration et posologie , Apeline/pharmacologie , Vidange gastrique/effets des médicaments et des substances chimiques , Transit gastrointestinal/effets des médicaments et des substances chimiques , État nutritionnel , Contention physique/psychologie , Stress physiologique/physiologie , Animaux , Apeline/génétique , Apeline/métabolisme , Récepteur de l'apeline/génétique , Récepteur de l'apeline/métabolisme , Corticolibérine/pharmacologie , Duodénum/cytologie , Duodénum/effets des médicaments et des substances chimiques , Duodénum/physiologie , Jeûne/physiologie , Vidange gastrique/physiologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Mâle , Fragments peptidiques/pharmacologie , Transport des protéines/effets des médicaments et des substances chimiques , ARN messager/génétique , Rats , Rat Wistar , Transduction du signal/effets des médicaments et des substances chimiques , Estomac/cytologie , Estomac/effets des médicaments et des substances chimiques , Estomac/physiologie , Stress physiologique/effets des médicaments et des substances chimiques
12.
Arch Physiol Biochem ; 125(3): 244-254, 2019 Jul.
Article de Anglais | MEDLINE | ID: mdl-29564917

RÉSUMÉ

Apelin and its receptor (APJ) are involved in the regulation of a variety of pathophysiological processes. We studied the effect of apelin treatment on obesity-induced type 2 diabetes mellitus (T2DM) and possible interaction between apelin/APJ system and renin-angiotensin system (RAS). Forty eight male albino rats were divided into two groups: control group and diabetic group. Diabetic group was subdivided into: control diabetic, apelin-treated, apelin + losartan-treated, apelin + l-NAME-treated and losartan-treated diabetic subgroup. Administration of apelin-13 yielded an improvement of IR, dyslipidaemia, inflammation, oxidative stress with significant decrease in AT1R gene expression and significant increase in ACE2 gene expression in adipose tissues. Losartan + apelin yielded a further significant decrease in ATR1 gene expression, glycaemic indices, serum TGs and TPA versus Apelin only. Adding l-NAME in subgroup (2D) reversed the effect of apelin. We suggested that the beneficial effect of Apelin is mainly mediated by NO-activated pathway and/or ACE2/Ang (1-7) dependent pathway.


Sujet(s)
Apeline/administration et posologie , Marqueurs biologiques/analyse , Diabète expérimental/prévention et contrôle , Diabète de type 2/prévention et contrôle , Monoxyde d'azote/métabolisme , Obésité/complications , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Antagonistes du récepteur de type 1 de l'angiotensine-II/administration et posologie , Animaux , Glycémie/analyse , Diabète expérimental/étiologie , Diabète expérimental/métabolisme , Diabète expérimental/anatomopathologie , Diabète de type 2/étiologie , Diabète de type 2/métabolisme , Diabète de type 2/anatomopathologie , Alimentation riche en graisse/effets indésirables , Association de médicaments , Antienzymes/administration et posologie , Insuline/métabolisme , Insulinorésistance , Losartan/administration et posologie , Mâle , L-NAME/administration et posologie , Obésité/étiologie , Obésité/anatomopathologie , Rats
13.
J Neuroendocrinol ; 30(9): e12635, 2018 09.
Article de Anglais | MEDLINE | ID: mdl-30044523

RÉSUMÉ

Exposure to an acute stressor induces up-regulation of apelin and cholecystokinin (CCK) in the hypothalamic paraventricular nucleus (PVN), which is the key brain centre integrating the stress-induced alterations in neuroendocrine, autonomic and behavioural functions. We tested the hypothesis that the release of CCK from the PVN is increased by centrally administered or stress-induced up-regulated endogenous apelin via the APJ receptor. Additionally, the effect of hypothalamic CCK on autonomic outflow was investigated under basal and stressed conditions. In vivo brain microdialysis was performed in rats that received (i) intra-PVN administration of apelin-13 or (ii) acute restraint stress (ARS). For chemical stimulation of the neurones in the PVN, a high concentration of KCl was applied by reverse microdialysis. CCK-8 levels in microdialysates were quantified by an enzyme immunoassay. The immunoreactivity of the APJ receptor and CCK was detected by immunofluorescence in hypothalamic sections. Heart rate variability was assessed in rats that received PVN stimulation or ARS following pre-administration of vehicle or CCK1 receptor antagonist lorglumide. Both intra-PVN exogenous apelin-13 and ARS increased the CCK-8 levels in dialysates significantly. The ARS-induced elevations in CCK levels were reversed by intra-PVN pre-administration of the APJ receptor antagonist F13A. Within the PVN, robust APJ receptor expression was detected on the CCK-producing mediocellular cells, in addition to the parvocellular neurones in the periventricular region. Dual immunoreactivity of APJ/CCK was observed in magnocellular cells to a lesser degree. Both exogenous apelin and ARS increased the CCK immunoreactivity markedly within the PVN, which was diminished significantly by F13A. Sympathetic tonus was increased markedly both by PVN stimulation and ARS, which was attenuated by lorglumide. These results revealed the interaction between apelin and CCK in the brain, suggesting that hypothalamic CCK may contribute to the apelin-induced alterations in autonomic outflow under stressed conditions.


Sujet(s)
Récepteur de l'apeline/métabolisme , Apeline/administration et posologie , Cholécystokinine/métabolisme , Hypothalamus/effets des médicaments et des substances chimiques , Neurones/effets des médicaments et des substances chimiques , Stress physiologique/physiologie , Stress psychologique/métabolisme , Animaux , Rythme cardiaque/effets des médicaments et des substances chimiques , Hypothalamus/métabolisme , Mâle , Neurones/métabolisme , Rats , Rat Wistar , Contention physique
14.
Auton Neurosci ; 212: 17-22, 2018 07.
Article de Anglais | MEDLINE | ID: mdl-29778241

RÉSUMÉ

Centrally administered apelin has been shown to inhibit gastric emptying (GE) in rodents, however, the relevant mechanism has been investigated incompletely. Using male Wistar rats, we investigated the efferent pathways involved in gastroinhibitory action of central apelin. Stereotaxic intracerebroventricular (icv) cannulation, subdiaphragmatic vagotomy (VGX) and/or celiac ganglionectomy (CGX) were performed 7 days prior to the experiments. Apelin-13 was administered (30 nmol, icv) 90 min prior to GE measurement. Nitric oxide synthase inhibitor L-NAME (100 mg/kg), sympatholytic agent guanethidine (5 mg/kg) and/or muscarinic receptor agonist bethanechol (1 mg/kg) were administered intraperitoneally 30 min prior to the central apelin-13 injection. Two strain gages were implanted serosally onto antrum and pylorus to monitor gastric postprandial motility. Heart rate variability (HRV) analysis was performed before and after central vehicle or apelin-13 administration. Apelin-13 delayed solid GE significantly by disturbing coordinated antral and pyloric postprandial contractions. The apelin-induced delayed GE was attenuated partially by CGX or VGX, whereas it was restored completely in rats underwent both CGX and VGX. L-NAME did not change the apelin-induced alterations. Guanethidine or bethanechol restored the apelin-induced gastroinhibition partially, while it was abolished completely in rats received both agents. Apelin-13 decreased the HRV spectral activity in high-frequency range by increasing low-frequency component and the ratio of LF:HF. The present data suggest that (1) both vagal parasympathetic and sympathetic pathways play a role in apelin-induced gastroinhibition, (2) central apelin attenuates vagal cholinergic pathway rather than activating nonadrenergic-noncholinergic pathway. Apelin/APJ receptor system might be candidate for the treatment of autonomic dysfunction and gastrointestinal motor disorders.


Sujet(s)
Apeline/pharmacologie , Système nerveux autonome/effets des médicaments et des substances chimiques , Vidange gastrique/effets des médicaments et des substances chimiques , Motilité gastrointestinale/effets des médicaments et des substances chimiques , Tube digestif/effets des médicaments et des substances chimiques , Animaux , Apeline/administration et posologie , Système nerveux autonome/physiologie , Vidange gastrique/physiologie , Motilité gastrointestinale/physiologie , Tube digestif/physiologie , Mâle , Période post-prandiale/effets des médicaments et des substances chimiques , Période post-prandiale/physiologie , Rat Wistar , Nerf vague/effets des médicaments et des substances chimiques , Nerf vague/physiologie
15.
Hypertens Res ; 40(8): 732-737, 2017 Aug.
Article de Anglais | MEDLINE | ID: mdl-28275232

RÉSUMÉ

The apelin/APJ system has an important role in the regulation of vascular tone and blood pressure. Opioid receptors (OPRs) are also important cardiovascular regulators and exert many of their effects by modulating the function of other G-protein-coupled receptors. The aim of this study was to analyze the interaction of apelin and the opioid system with respect to vascular responses to apelin in rats with renovascular hypertension (two-kidney, one clip (2K1C)). Homodynamic studies were carried out in 2K1C rats. Naloxone (a nonselective OPR inhibitor) or nor-binaltorphimine dihydrochloride (norBNI, a kappa OPR inhibitor) and signaling pathway inhibitors PTX (a Gi path inhibitor) and chelerythrine (a protein kinase C (PKC) inhibitor) were administered before apelin at 20 and 40 µg kg-1. Apelin at 20 and 40 µg kg-1 decreased the systolic blood pressure by 15% and 20%, respectively (P<0.05). The pressure drop caused by apelin 20 was inhibited by naloxone, norBNI and PTX, but it was not affected by chelerythrine. The pressure drop caused by apelin 40 was augmented by naloxone and chelerythrine, and it was not affected by norBNI or PTX. The lowering effect of apelin 20 on blood pressure is exerted through OPRs and stimulation of Gi and PKC pathways. However, apelin 40 functions independently of OPRs, Gi and PKC. This dose-dependent differential effect of apelin may have potential clinical applications as opioids are currently used, and apelin has been introduced as a potential therapeutic agent in cardiovascular complications.


Sujet(s)
Antihypertenseurs/administration et posologie , Antihypertenseurs/usage thérapeutique , Apeline/administration et posologie , Apeline/usage thérapeutique , Hypertension rénovasculaire/traitement médicamenteux , Récepteurs aux opioïdes/effets des médicaments et des substances chimiques , Animaux , Benzophénanthridines/pharmacologie , Relation dose-effet des médicaments , Antienzymes/pharmacologie , Hémodynamique/effets des médicaments et des substances chimiques , Hypertension rénovasculaire/physiopathologie , Mâle , Naloxone/pharmacologie , Naltrexone/analogues et dérivés , Naltrexone/pharmacologie , Antagonistes narcotiques/pharmacologie , Protéine kinase C/antagonistes et inhibiteurs , Rats , Rat Wistar
16.
Brain Res Bull ; 130: 67-74, 2017 04.
Article de Anglais | MEDLINE | ID: mdl-28065733

RÉSUMÉ

Apelin, a small bioactive peptide, plays an important role in the pathogenesis of mood disorders through the endogenous ligand APJ. Although the anxiolytic effect of apelin is well established, the mechanisms are poorly understood. In this study, we hypothesized that apelin played an anxiolytic role in chronic normobaric hypoxia (CNH)-induced anxiety like behavior in mice, which might be associated with an inhibition of nuclear factor-κB (NF-κB) activation in the hippocampus. To this end, mice were exposed in a normobaric hypoxic chamber with a fraction of inspired oxygen (FIO2, ∼10%, 23h/d) with or without apelin-13 application (20 nmolkg-1d-1, i.p.), for 4 weeks. The anxiety-like behavior was tested by elevated plus maze and open field. Activities of NF-κB, microglial, and related signaling pathways in the hippocampus during this pathological process were examined. We found that CNH treatment decreased APJ but increased Iba-1 proteins expression, as well as nucleus translocation of p50 and p65 in the hippocampus, which were reversed by apelin-13 treatment. In addition, apelin-13 treatment ameliorated CNH-induced anxiety-like behavior in mice, suggesting anxiogenic effect of apelin-13 might be mediated by an inhibition of NF-κB activation in microglial of the hippocampus. Furthermore, apelin-13 treatment reversed p-CAMKII decrease in the hippocampus under CNH treatment. Apelin-13 treatment did not affect anxiety-like behavior and relative proteins expression in normoxia control mice. Finally, we found that rats with CNH treatment decreased APJ expression while enhanced NF-κB activation in the hippocampus, providing additional evidences that NF-κB activation in hippocampus in CNH-induced anxiety-like behavior in rats we reported previously might be associated with an inhibition of APJ activity. In conclusion, the present results illustrated that inhibition of APJ and promotion of NF-κB activation in the microglial of hippocampus might be involved in anxiogenic effect in CNH-exposed mice, and apelin-13 ameliorates CNH-induced anxiety-like behavior might be associated with an inhibition of NF-κB activation.


Sujet(s)
Anxiété/métabolisme , Apeline/métabolisme , Hippocampe/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Animaux , Apeline/administration et posologie , Récepteur de l'apeline/métabolisme , Comportement animal , Hippocampe/effets des médicaments et des substances chimiques , Hypoxie , Mâle , Souris de lignée C57BL , Rat Sprague-Dawley
17.
Sci Rep ; 6: 31849, 2016 08 23.
Article de Anglais | MEDLINE | ID: mdl-27549402

RÉSUMÉ

Apelin is a bioactive peptide involved in the control of energy metabolism. In the hypothalamus, chronic exposure to high levels of apelin is associated with an increase in hepatic glucose production, and then contributes to the onset of type 2 diabetes. However, the molecular mechanisms behind deleterious effects of chronic apelin in the brain and consequences on energy expenditure and thermogenesis are currently unknown. We aimed to evaluate the effects of chronic intracerebroventricular (icv) infusion of apelin in normal mice on hypothalamic inflammatory gene expression, energy expenditure, thermogenesis and brown adipose tissue functions. We have shown that chronic icv infusion of apelin increases the expression of pro-inflammatory factors in the hypothalamus associated with an increase in plasma interleukin-1 beta. In parallel, mice infused with icv apelin exhibit a significant lower energy expenditure coupled to a decrease in PGC1alpha, PRDM16 and UCP1 expression in brown adipose tissue which could explain the alteration of thermogenesis in these mice. These data provide compelling evidence that central apelin contributes to the development of type 2 diabetes by altering energy expenditure, thermogenesis and fat browning.


Sujet(s)
Tissu adipeux brun/effets des médicaments et des substances chimiques , Apeline/pharmacologie , Métabolisme énergétique/effets des médicaments et des substances chimiques , Thermogenèse/effets des médicaments et des substances chimiques , Tissu adipeux brun/métabolisme , Animaux , Apeline/administration et posologie , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Diabète de type 2/sang , Diabète de type 2/génétique , Diabète de type 2/métabolisme , Expression des gènes/effets des médicaments et des substances chimiques , Hypothalamus/effets des médicaments et des substances chimiques , Hypothalamus/métabolisme , Perfusions intraventriculaires , Interleukine-1 bêta/génétique , Interleukine-1 bêta/métabolisme , Souris , Nitric oxide synthase type II/génétique , Nitric oxide synthase type II/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Protéine-1 de découplage/génétique , Protéine-1 de découplage/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE