Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.123
Filtrer
1.
PLoS One ; 19(6): e0304227, 2024.
Article de Anglais | MEDLINE | ID: mdl-38870103

RÉSUMÉ

INTRODUCTION: Acute kidney injury (AKI) is a common complication of septic shock and together these conditions carry a high mortality risk. In septic patients who develop severe AKI, renal cortical perfusion is deficient despite normal macrovascular organ blood flow. This intra-renal perfusion abnormality may be amenable to pharmacological manipulation, which may offer mechanistic insight into the pathophysiology of septic AKI. The aim of the current study is to investigate the effects of vasopressin and angiotensin II on renal microcirculatory perfusion in a cohort of patients with septic shock. METHODS AND ANALYSIS: In this single centre, mechanistically focussed, randomised controlled study, 45 patients with septic shock will be randomly allocated to either of the study vasopressors (vasopressin or angiotensin II) or standard therapy (norepinephrine). Infusions will be titrated to maintain a mean arterial pressure (MAP) target set by the attending clinician. Renal microcirculatory assessment will be performed for the cortex and medulla using contrast-enhanced ultrasound (CEUS) and urinary oxygen tension (pO2), respectively. Renal macrovascular flow will be assessed via renal artery ultrasound. Measurement of systemic macrovascular flow will be performed through transthoracic echocardiography (TTE) and microvascular flow via sublingual incident dark field (IDF) video microscopy. Measures will be taken at baseline, +1 and +24hrs following infusion of the study drug commencing. Blood and urine samples will also be collected at the measurement time points. Longitudinal data will be compared between groups and over time. DISCUSSION: Vasopressors are integral to the management of patients with septic shock. This study aims to further understanding of the relationship between this therapy, renal perfusion and the development of AKI. In addition, using CEUS and urinary pO2, we hope to build a more complete picture of renal perfusion in septic shock by interrogation of the constituent parts of the kidney. Results will be published in peer-reviewed journals and presented at academic meetings. TRIAL REGISTRATION: The REPERFUSE study was registered on Clinical Trials.gov (NCT06234592) on the 30th Jan 24.


Sujet(s)
Atteinte rénale aigüe , Microcirculation , Choc septique , Vasoconstricteurs , Humains , Choc septique/traitement médicamenteux , Choc septique/physiopathologie , Vasoconstricteurs/usage thérapeutique , Vasoconstricteurs/administration et posologie , Microcirculation/effets des médicaments et des substances chimiques , Atteinte rénale aigüe/traitement médicamenteux , Atteinte rénale aigüe/étiologie , Rein/effets des médicaments et des substances chimiques , Rein/physiopathologie , Rein/vascularisation , Vasopressines/administration et posologie , Vasopressines/usage thérapeutique , Angiotensine-II/administration et posologie , Mâle , Femelle , Norépinéphrine/administration et posologie , Norépinéphrine/usage thérapeutique , Circulation rénale/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Adulte
2.
AAPS PharmSciTech ; 25(5): 115, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38755324

RÉSUMÉ

More than 1 billion people worldwide suffer from hypertension; therefore, hypertension management has been categorized as a global health priority. Losartan potassium (LP) is an antihypertensive drug with a limited oral bioavailability of about 33% since it undergoes the initial metabolic cycle. Thus, nasal administration is a unique route to overcome first-pass metabolism. The investigation focused on the potential effects of LP-loaded spanlastic vesicles (SNVs) on LP pharmacodynamics and pharmacokinetic parameters, utilizing a thin-film hydration methodology established on a 3122 full factorial design. Entrapment efficiency (EE%) ranged from 39.8 ± 3.87.8 to 83.8 ± 2.92% for LP-SNVs. Vesicle size (VS) varied from 205.5 ± 6.5.10 to 445.1 ± 13.52 nm, and the percentage of LP released after 8 h (Q8h) ranged from 30.8 ± 3.10 to 68.8 ± 1.45%. LP permeated through the nasal mucosa during 24 h and flocculated from 194.1 ± 4.90 to 435.3 ± 13.53 µg/cm2. After twenty-four hours, the optimal LP-SNVs in-situ gel showed 2.35 times more permeation through the nasal mucosa than the LP solution. It also lowered systolic blood pressure, so it is thought to be better than the reference formulation in terms of pharmacodynamics. The pharmacokinetics studies demonstrated that the intranasal LP-SNVs gel boosted its bioavailability approximately 6.36 times compared to the oral LP solution. Our research showed that intranasal LP-SNVs could be a good nanoplatform because they are well-tolerated and have possible pharmacokinetics and pharmacodynamics.


Sujet(s)
Antihypertenseurs , Gels , Hypertension artérielle , Losartan , Losartan/pharmacocinétique , Losartan/administration et posologie , Losartan/pharmacologie , Antihypertenseurs/pharmacocinétique , Antihypertenseurs/administration et posologie , Antihypertenseurs/pharmacologie , Animaux , Hypertension artérielle/traitement médicamenteux , Mâle , Rats , Biodisponibilité , Administration par voie nasale , Nanoparticules/composition chimique , Muqueuse nasale/métabolisme , Muqueuse nasale/effets des médicaments et des substances chimiques , Taille de particule , Angiotensine-II/pharmacocinétique , Angiotensine-II/administration et posologie , Angiotensine-II/pharmacologie , Pression sanguine/effets des médicaments et des substances chimiques , Rat Wistar , Chimie pharmaceutique/méthodes
3.
Anesth Analg ; 139(1): 165-173, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38289858

RÉSUMÉ

BACKGROUND: Vasoplegia is common after cardiac surgery, is associated with hyperreninemia, and can lead to acute kidney stress. We aimed to conduct a pilot study to test the hypothesis that, in vasoplegic cardiac surgery patients, angiotensin-II (AT-II) may not increase kidney stress (measured by [TIMP-2]*[IGFBP7]). METHODS: We randomly assigned patients with vasoplegia (cardiac index [CI] > 2.1l/min, postoperative hypotension requiring vasopressors) and Δ-renin (4-hour postoperative-preoperative value) ≥3.7 µU/mL, to AT-II or placebo targeting a mean arterial pressure ≥65 mm Hg for 12 hours. The primary end point was the incidence of kidney stress defined as the difference between baseline and 12 hours [TIMP-2]*[IGFBP7] levels. Secondary end points included serious adverse events (SAEs). RESULTS: We randomized 64 patients. With 1 being excluded, 31 patients received AT-II, and 32 received placebo. No significant difference was observed between AT-II and placebo groups for kidney stress (Δ-[TIMP-2]*[IGFBP7] 0.06 [ng/mL] 2 /1000 [Q1-Q3, -0.24 to 0.28] vs -0.08 [ng/mL] 2 /1000 [Q1-Q3, -0.35 to 0.14]; P = .19; Hodges-Lehmann estimation of the location shift of 0.12 [ng/mL] 2 /1000 [95% confidence interval, CI, -0.1 to 0.36]). AT-II patients received less fluid during treatment than placebo patients (2946 vs 3341 mL, P = .03), and required lower doses of norepinephrine equivalent (0.19 mg vs 4.18mg, P < .001). SAEs were reported in 38.7% of patients in the AT-II group and in 46.9% of patients in the placebo group. CONCLUSIONS: The infusion of AT-II for 12 hours appears feasible and did not lead to an increase in kidney stress in a high-risk cohort of cardiac surgery patients. These findings support the cautious continued investigation of AT-II as a vasopressor in hyperreninemic cardiac surgery patients.


Sujet(s)
Angiotensine-II , Procédures de chirurgie cardiaque , Rénine , Vasoplégie , Humains , Projets pilotes , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Procédures de chirurgie cardiaque/effets indésirables , Rénine/sang , Angiotensine-II/administration et posologie , Angiotensine-II/sang , Vasoplégie/traitement médicamenteux , Vasoplégie/étiologie , Méthode en double aveugle , Perfusions veineuses , Rein/effets des médicaments et des substances chimiques , Rein/physiopathologie , Inhibiteur tissulaire de métalloprotéinase-2 , Résultat thérapeutique , Marqueurs biologiques/sang , Protéines de liaison aux IGF
4.
Pediatrics ; 153(1)2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-38098437

RÉSUMÉ

Infants with congenital bilateral renal agenesis are at significant risk for morbidity and mortality, despite substantial and continuing advances in fetal and neonatal therapeutics. Infants with bilateral renal agenesis may episodically develop severe hypotension that can be refractory to traditional vasopressors. Synthetic angiotensin-II has been successfully used in adult and a few pediatric patients with refractory hypotension but has not been extensively studied in infants. We describe the use of angiotensin-II in treating refractory hypotension in a premature infant with congenital bilateral renal agenesis admitted to the NICU. Within 48 hours, he no longer required other vasopressors. Subsequently, angiotensin-II was gradually weaned and discontinued over 10 days and the patient was ultimately discharged from the hospital. This case demonstrates that angiotensin-II may be a helpful agent to treat refractory hypotension in infants with bilateral renal agenesis.


Sujet(s)
Angiotensine-II , Hypotension artérielle , Maladies du rein , Vasoconstricteurs , Hypotension artérielle/traitement médicamenteux , Malformations/traitement médicamenteux , Rein/malformations , Maladies du rein/congénital , Maladies du rein/traitement médicamenteux , Angiotensine-II/administration et posologie , Vasoconstricteurs/administration et posologie , Humains , Nouveau-né , Nourrisson
5.
Biomolecules ; 13(6)2023 05 28.
Article de Anglais | MEDLINE | ID: mdl-37371479

RÉSUMÉ

BACKGROUND: Gasdermin D, a molecule downstream of the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing inflammasome, forms the membrane pore for the secretion of interleukin (IL)-1ß and IL-18, and also mediates pyroptosis. This study was to explore the influence of treatment with disulfiram, a small molecule inhibitor to gasdermin D, on the formation and progression of experimental abdominal aortic aneurysms (AAA). METHODS: AAAs were induced in 10-week-old male apolipoprotein E deficient mice by subcutaneous infusion of angiotensin II (1000 ng/min/kg body weight) for 28 days via osmotic minipumps. Three days prior to angiotensin II infusion, disulfiram (50 mg/kg) or an equal volume of saline as the vehicle control was administered daily via oral gavage. The influence on experimental AAAs was analyzed by serial measurements of aortic diameters via ultrasonography, grading AAA severity and histopathology at sacrifice. Serum IL-1ß and IL-18 levels, systolic blood pressure, total cholesterol, and triglyceride were also measured. Additional experiments assayed the influences on the cell viability and IL-1ß secretion of in vitro activated macrophages. RESULTS: Disulfiram significantly reduced the enlargement, incidence, and severity of angiotensin II-induced experimental AAAs with attenuation of medial elastin breaks, mural macrophage accumulation, and systolic blood pressure. The AAA suppression was also associated with reduced systemic levels of IL-1ß but not IL-18. However, disulfiram treatment had no impact on body weight gain and lipid levels in aneurysmal mice. Additionally, disulfiram treatment also markedly reduced the secretion of IL-1ß from activated macrophages with a limited effect on cell viability in vitro. CONCLUSIONS: Gasdermin D inhibition by disulfiram attenuated angiotensin II-induced experimental AAAs with reduced systemic IL-1ß levels and in vitro activated macrophage IL-1ß secretion. Our study suggests that pharmacological gasdermin D inhibition may have translational potential for limiting clinical AAA progression.


Sujet(s)
Angiotensine-II , Anévrysme de l'aorte abdominale , Animaux , Mâle , Souris , Angiotensine-II/administration et posologie , Angiotensine-II/effets indésirables , Angiotensine-II/usage thérapeutique , Anévrysme de l'aorte abdominale/induit chimiquement , Anévrysme de l'aorte abdominale/traitement médicamenteux , Anévrysme de l'aorte abdominale/anatomopathologie , Poids , Modèles animaux de maladie humaine , Disulfirame/pharmacologie , Gasdermines/antagonistes et inhibiteurs , Souris de lignée C57BL
6.
Peptides ; 153: 170786, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35304156

RÉSUMÉ

Fibroblast growth factors (FGFs) mediate key cardiac functions from development to homeostasis and disease. The current research was to explore the effects of FGF12 in the fibrosis of cardiac function and heart failure, and whether FGF12 alleviated cardiac fibrosis via inhibition of oxidative stress. Ligation of left coronary artery in mice induced heart failure and myocardial infarction (MI). Angiotensin II (Ang II) was administered to cardiac fibroblasts (CFs). FGF12 upregulation or FGF12 transgenic (Tg) mice could improve cardiac dysfunction of MI mice, and attenuated cardiac fibrosis of heart failure induced by MI in mice. FGF12 overexpression suppressed the increases of collagen I, collagen III and fibronectin which was induced by Ang II in CFs. The oxidative stress was enhanced in the heart of MI mice and CFs treated with Ang II, and these enhances were attenuated via FGF12 overexpression. Superoxide dismutase (SOD) overexpression inhibited the enhancements of collagen I, collagen III and fibronectin in the heart of MI mice, and in the CFs treated with Ang II. Overexpression of nicotinamide adenine dinucleotide phosphate oxidases (Nox1) reversed the attenuating influences of FGF12 on the enhancements of collagen I, collagen III and fibronectin in the CFs induced by Ang II. These outcomes showed that FGF12 upregulation can improve cardiac dysfunction and heart fibrosis of heart failure. FGF12 attenuates oxidative stress to suppress the cardiac fibrosis.


Sujet(s)
Facteurs de croissance fibroblastique , Défaillance cardiaque , Infarctus du myocarde , Myocarde , Stress oxydatif , Remodelage ventriculaire , Angiotensine-II/administration et posologie , Animaux , Collagène/métabolisme , Facteurs de croissance fibroblastique/génétique , Facteurs de croissance fibroblastique/métabolisme , Fibronectines/génétique , Fibronectines/métabolisme , Fibrose , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/génétique , Défaillance cardiaque/métabolisme , Souris , Infarctus du myocarde/métabolisme , Infarctus du myocarde/anatomopathologie , Myocarde/métabolisme , Myocarde/anatomopathologie
7.
Oxid Med Cell Longev ; 2022: 5044046, 2022.
Article de Anglais | MEDLINE | ID: mdl-35222798

RÉSUMÉ

Cardiac lymphatic vessel growth (lymphangiogenesis) and integrity play an essential role in maintaining tissue fluid balance. Inhibition of lymphatic lymphangiogenesis is involved in cardiac edema and cardiac remodeling after ischemic injury or pressure overload. However, whether lymphatic vessel integrity is disrupted during angiotensin II- (Ang II-) induced cardiac remodeling remains to be investigated. In this study, cardiac remodeling models were established by Ang II (1000 ng/kg/min) in VEGFR-3 knockdown (Lyve-1Cre VEGFR-3f/-) and wild-type (VEGFR-3f/f) littermates. Our results indicated that Ang II infusion not only induced cardiac lymphangiogenesis and upregulation of VEGF-C and VEGFR-3 expression in the time-dependent manner but also enhanced proteasome activity, MKP5 and VE-cadherin degradation, p38 MAPK activation, and lymphatic vessel hyperpermeability. Moreover, VEGFR-3 knockdown significantly inhibited cardiac lymphangiogenesis in mice, resulting in exacerbation of tissue edema, hypertrophy, fibrosis superoxide production, inflammation, and heart failure (HF). Conversely, administration of epoxomicin (a selective proteasome inhibitor) markedly mitigated Ang II-induced cardiac edema, remodeling, and dysfunction; upregulated MKP5 and VE-cadherin expression; inactivated p38 MAPK; and reduced lymphatic vessel hyperpermeability in WT mice, indicating that inhibition of proteasome activity is required to maintain lymphatic endothelial cell (LEC) integrity. Our results show that both cardiac lymphangiogenesis and lymphatic barrier hyperpermeability are implicated in Ang II-induced adaptive hypertrophic remodeling and dysfunction. Proteasome-mediated hyperpermeability of LEC junctions plays a predominant role in the development of cardiac remodeling. Selective stimulation of lymphangiogenesis or inhibition of proteasome activity may be a potential therapeutic option for treating hypertension-induced cardiac remodeling.


Sujet(s)
Angiotensine-II/métabolisme , Cardiomégalie/métabolisme , Oedème cardiaque/métabolisme , Vaisseaux lymphatiques/métabolisme , Angiotensine-II/administration et posologie , Animaux , Cardiomégalie/traitement médicamenteux , Cardiomégalie/anatomopathologie , Cardiomégalie/physiopathologie , Oedème cardiaque/traitement médicamenteux , Oedème cardiaque/anatomopathologie , Oedème cardiaque/physiopathologie , Cellules endothéliales/métabolisme , Lymphangiogenèse/effets des médicaments et des substances chimiques , Souris , Souris knockout , Myocarde/métabolisme , Myocarde/anatomopathologie , Perméabilité/effets des médicaments et des substances chimiques , Proteasome endopeptidase complex/métabolisme , Inhibiteurs du protéasome/pharmacologie , Inhibiteurs du protéasome/usage thérapeutique , Récepteur-3 au facteur croissance endothéliale vasculaire/déficit , Récepteur-3 au facteur croissance endothéliale vasculaire/métabolisme
8.
Arterioscler Thromb Vasc Biol ; 42(3): 277-288, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35045728

RÉSUMÉ

AngII (angiotensin II) infusion in mice has been used to provide mechanistic insight into human abdominal aortic aneurysms for over 2 decades. This is a technically facile animal model that recapitulates multiple facets of the human disease. Although numerous publications have reported abdominal aortic aneurysms with AngII infusion in mice, there remain many fundamental unanswered questions such as uniformity of describing the pathological characteristics and which cell type is stimulated by AngII to promote abdominal aortic aneurysms. Extrapolation of the findings to provide insight into the human disease has been hindered by the preponderance of studies designed to determine the effects on initiation of abdominal aortic aneurysms, rather than a more clinically relevant scenario of determining efficacy on the established disease. The purpose of this review is to enhance understanding of AngII-induced abdominal aortic pathologies in mice, thereby providing greater insight into the human disease.


Sujet(s)
Angiotensine-II/toxicité , Anévrysme de l'aorte abdominale/étiologie , Facteurs âges , Angiotensine-II/administration et posologie , Animaux , Anévrysme de l'aorte abdominale/anatomopathologie , Anévrysme de l'aorte abdominale/thérapie , Pression sanguine/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Femelle , Facteurs de risque de maladie cardiaque , Humains , Hypercholestérolémie/complications , Mâle , Souris , Souris transgéniques , Modèles cardiovasculaires , Récepteurs aux angiotensines/effets des médicaments et des substances chimiques , Facteurs sexuels
9.
Biomolecules ; 11(12)2021 12 10.
Article de Anglais | MEDLINE | ID: mdl-34944506

RÉSUMÉ

Renin-angiotensin systems produce angiotensin II (Ang II) and angiotensin 1-7 (Ang 1-7), which are able to induce opposite effects on circulation. This study in vivo assessed the effects induced by Ang II or Ang 1-7 on rat pial microcirculation during hypoperfusion-reperfusion, clarifying the mechanisms causing the imbalance between Ang II and Ang 1-7. The fluorescence microscopy was used to quantify the microvascular parameters. Hypoperfusion and reperfusion caused vasoconstriction, disruption of blood-brain barrier, reduction of capillary perfusion and an increase in reactive oxygen species production. Rats treated with Ang II showed exacerbated microvascular damage with stronger vasoconstriction compared to hypoperfused rats, a further increase in leakage, higher decrease in capillary perfusion and marker oxidative stress. Candesartan cilexetil (specific Ang II type 1 receptor (AT1R) antagonist) administration prior to Ang II prevented the effects induced by Ang II, blunting the hypoperfusion-reperfusion injury. Ang 1-7 or ACE2 activator administration, preserved the pial microcirculation from hypoperfusion-reperfusion damage. These effects of Ang 1-7 were blunted by a Mas (Mas oncogene-encoded protein) receptor antagonist, while Ang II type 2 receptor antagonists did not affect Ang 1-7-induced changes. In conclusion, Ang II and Ang 1-7 triggered different mechanisms through AT1R or MAS receptors able to affect cerebral microvascular injury.


Sujet(s)
Angiotensine-II/administration et posologie , Angiotensine-I/administration et posologie , Benzimidazoles/administration et posologie , Dérivés du biphényle/administration et posologie , Fragments peptidiques/administration et posologie , Pie-mère/vascularisation , Lésion d'ischémie-reperfusion/métabolisme , Tétrazoles/administration et posologie , Angiotensine-I/effets indésirables , Angiotensine-II/effets indésirables , Animaux , Benzimidazoles/pharmacologie , Dérivés du biphényle/pharmacologie , Femelle , Mâle , Microcirculation/effets des médicaments et des substances chimiques , Microscopie de fluorescence , Fragments peptidiques/effets indésirables , Pie-mère/effets des médicaments et des substances chimiques , Pie-mère/métabolisme , Proto-oncogène Mas/métabolisme , Rats , Espèces réactives de l'oxygène/métabolisme , Lésion d'ischémie-reperfusion/traitement médicamenteux , Tétrazoles/pharmacologie
10.
Int J Mol Sci ; 22(24)2021 Dec 20.
Article de Anglais | MEDLINE | ID: mdl-34948475

RÉSUMÉ

Compound 21 (C21), an AT2 receptor agonist, and Angiotensin 1-7 (Ang 1-7), through Mas receptor, play an important role in the modulation of the protective arm of the renin-angiotensin system. The aim of this study was to investigate in an experimental model of angiotensin II-dependent hypertension whether the activation of the potentially protective arm of the renin-angiotensin system, through AT2 or Mas receptor stimulation, counteracts the onset of myocardial fibrosis and hypertrophy, and whether these effects are mediated by inflammatory mechanism and/or sympathetic activation. Sprague Dawley rats (n = 67) were treated for 1 (n = 25) and 4 (n = 42) weeks and divided in the following groups: (a) Angiotensin II (Ang II, 200 ng/kg/min, osmotic minipumps, sub cutis); (b) Ang II+Compound 21 (C21, 0.3 mg/kg/day, intraperitoneal); (c) Ang II+Ang 1-7 (576 µg/kg/day, intraperitoneal); (d) Ang II+Losartan (50 mg/kg/day, per os); (e) control group (physiological saline, sub cutis). Systolic blood pressure was measured by tail cuff method and, at the end of the experimental period, the rats were euthanized and the heart was excised to evaluate myocardial fibrosis, hypertrophy, inflammatory cell infiltration and tyrosine hydroxylase expression, used as marker of sympathetic activity. Ang II caused a significant increase of blood pressure, myocardial interstitial and perivascular fibrosis and myocardial hypertrophy, as compared to control groups. C21 or Ang 1-7 administration did not modify the increase in blood pressure in Ang II treated rats, but both prevented the development of myocardial fibrosis and hypertrophy. Treatment with losartan blocked the onset of hypertension and myocardial fibrosis and hypertrophy in Ang II treated rats. Activation of AT2 receptors or Mas receptors prevents the onset of myocardial fibrosis and hypertrophy in Ang II-dependent hypertension through the reduction of myocardial inflammatory cell infiltration and tyrosine hydroxylase expression. Unlike what happens in case of treatment with losartan, the antifibrotic and antihypertrophic effects that follow the activation of the AT2 or Mas receptors are independent on the modulation of blood pressure.


Sujet(s)
Angiotensine-II/administration et posologie , Angiotensine-I/administration et posologie , Cardiomégalie/prévention et contrôle , Hypertension artérielle/traitement médicamenteux , Imidazoles/administration et posologie , Losartan/administration et posologie , Fragments peptidiques/administration et posologie , Sulfonamides/administration et posologie , Thiophènes/administration et posologie , Angiotensine-I/pharmacologie , Angiotensine-II/pharmacologie , Animaux , Cardiomégalie/métabolisme , Modèles animaux de maladie humaine , Fibrose , Hypertension artérielle/induit chimiquement , Hypertension artérielle/métabolisme , Imidazoles/pharmacologie , Injections péritoneales , Losartan/pharmacologie , Mâle , Fragments peptidiques/pharmacologie , Proto-oncogène Mas/métabolisme , Rats , Rat Sprague-Dawley , Sulfonamides/pharmacologie , Thiophènes/pharmacologie , Tyrosine 3-monooxygenase/métabolisme
11.
Oxid Med Cell Longev ; 2021: 2860488, 2021.
Article de Anglais | MEDLINE | ID: mdl-34950416

RÉSUMÉ

Acute myocardial infarction (AMI) is a major cause of morbidity and mortality worldwide. Angiotensin (Ang) IV possesses many biological properties that are not yet completely understood. Therefore, we investigated the function and mechanism of Ang IV in AMI in in vivo and in vitro conditions. AMI was performed by ligation of the left anterior descending coronary artery (LAD) in male C57 mice. Ang IV was continuously infused by a minipump 3 d before AMI for 33 d. The neonatal rat ventricular myocytes (NRVCs) were stimulated with Ang IV and cultured under hypoxic conditions. In vivo, Ang IV infusion significantly reduced the mortality after AMI. By the 7th day after AMI, compared with the AMI group, Ang IV reduced the inflammatory cytokine expression. Moreover, terminal deoxyribonucleotidyl transferase- (TDT-) mediated dUTP nick-end labeling (TUNEL) assay showed that Ang IV infusion reduced AMI-induced cardiomyocyte apoptosis. Compared with AMI, Ang IV reduced autophagosomes in cardiomyocytes and improved mitochondrial swelling and disarrangement, as assessed by transmission electron microscopy. By 30th day after AMI, Ang IV significantly reduced the ratio of heart weight to body weight. Echocardiography showed that Ang IV improved impaired cardiac function. Hematoxylin and eosin (H&E) and Masson staining showed that Ang IV infusion reduced the infarction size and myocardial fibrosis. In vitro, dihydroethidium (DHE) staining and comet assay showed that, compared with the hypoxia group, Ang IV reduced oxidative stress and DNA damage. Enzyme-linked immunosorbent assay (ELISA) showed that Ang IV reduced hypoxia-induced secretion of the tumor necrosis factor- (TNF-) ɑ and interleukin- (IL-) 1ß. In addition, compared with the hypoxia group, Ang IV reduced the transformation of light chain 3- (LC3-) I to LC3-II but increased p62 expression and decreased cardiomyocyte apoptosis. Overall, the present study showed that Ang IV reduced the inflammatory response, autophagy, and fibrosis after AMI, leading to reduced infarction size and improved cardiac function. Therefore, administration of Ang IV may be a feasible strategy for the treatment of AMI.


Sujet(s)
Angiotensine-II/analogues et dérivés , Autophagie , Cardiomyopathies/prévention et contrôle , Inflammation/traitement médicamenteux , Infarctus du myocarde/prévention et contrôle , Myocytes cardiaques/effets des médicaments et des substances chimiques , Agents protecteurs/pharmacologie , Angiotensine-II/administration et posologie , Angiotensine-II/pharmacologie , Animaux , Apoptose , Cardiomyopathies/étiologie , Cardiomyopathies/métabolisme , Cardiomyopathies/anatomopathologie , Cellules cultivées , Inflammation/étiologie , Inflammation/métabolisme , Inflammation/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Infarctus du myocarde/étiologie , Infarctus du myocarde/métabolisme , Infarctus du myocarde/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Stress oxydatif , Rats
12.
Sci Rep ; 11(1): 21193, 2021 10 27.
Article de Anglais | MEDLINE | ID: mdl-34707201

RÉSUMÉ

Endothelial dysfunction (ED) is a key factor for the development of cardiovascular diseases. Due to its chronic, life-threatening nature, ED only can be studied experimentally in animal models. Therefore, this work was aimed to characterize a murine model of ED induced by a daily intraperitoneal administration of angiotensin II (AGII) for 10 weeks. Oxidative stress, inflammation, vascular remodeling, hypertension, and damage to various target organs were evaluated in treated animals. The results indicated that a chronic intraperitoneal administration of AGII increases the production of systemic soluble VCAM, ROS and ICAM-1 expression, and the production of TNFα, IL1ß, IL17A, IL4, TGFß, and IL10 in the kidney, as well as blood pressure levels; it also promotes vascular remodeling and induces non-alcoholic fatty liver disease, glomerulosclerosis, and proliferative retinopathy. Therefore, the model herein proposed can be a representative model for ED; additionally, it is easy to implement, safe, rapid, and inexpensive.


Sujet(s)
Angiotensine-II/administration et posologie , Modèles animaux de maladie humaine , Endothélium vasculaire/métabolisme , Maladies vasculaires/métabolisme , Angiotensine-II/toxicité , Animaux , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/anatomopathologie , Perfusions parentérales , Molécule-1 d'adhérence intercellulaire/métabolisme , Interleukines/métabolisme , Rein/métabolisme , Rein/anatomopathologie , Foie/métabolisme , Foie/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Stress oxydatif , Facteur de croissance transformant bêta/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Molécule-1 d'adhérence des cellules vasculaires/métabolisme , Maladies vasculaires/étiologie , Maladies vasculaires/anatomopathologie , Remodelage vasculaire
13.
Peptides ; 146: 170669, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34624429

RÉSUMÉ

Alarin could alleviate myocardial infarction-induced heart failure. The present study was to explore whether alarin could alleviate myocardial hypertrophy via inhibiting cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) signaling pathway to attenuate autophagy. Myocardial hypertrophy was induced by angiotensin (Ang) II infusion in vivo in mice and by Ang II treatment of neonatal rat cardiomyocytes (NRCMs) in vitro. The Ang II-induced hypertrophy and fibrosis of the heart were alleviated after alarin administration in mice. The increased atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP) and beta-myosin heavy chain (ß-MHC), and the decreased alpha-myosin heavy chain (α-MHC) induced by Ang II were reversed by alarin treatment in NRCMs. Alarin inhibited the increases of cAMP and PKA in NRCMs. Treatment with cAMP or overexpression of PKA blocked the attenuating effects of alarin on Ang II-induced hypertrophy in NRCMs. Alarin reduced the Ang II-induced increases of LC3, Beclin 1, autophagy-related gene (Atg)3 and Atg5 in NRCMs. The overexpression of cAMP and PKA reversed the alleviating effects of alarin on the increased autophagy induced by Ang II in NRCMs. These results indicated that alarin could moderate cardiac remodeling. Alarin improved myocardial hypertrophy via inhibiting the cAMP/PKA signaling pathway to attenuate autophagy.


Sujet(s)
Autophagie/effets des médicaments et des substances chimiques , Cardiomégalie/prévention et contrôle , Cyclic AMP-Dependent Protein Kinases/métabolisme , AMP cyclique/métabolisme , Peptide galanine-like/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Angiotensine-II/administration et posologie , Animaux , Lignée cellulaire , Mâle , Souris , Souris de lignée C57BL , Myocytes cardiaques/effets des médicaments et des substances chimiques , Rats , Rat Sprague-Dawley , Récepteurs à la galanine/antagonistes et inhibiteurs
14.
Physiol Rep ; 9(17): e15000, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34491001

RÉSUMÉ

Autophagy is an evolutionarily conserved cellular catabolic process essential for cell homeostasis, and thus its failure is associated with several diseases. While autophagy has been reported to play a role in vascular smooth muscle cells (SMCs) in vascular disorders, its precise role in the pathogenesis of abdominal aortic aneurysm (AAA) has not yet been elucidated. In this study, we investigated the role of SMC autophagy in AAA formation. As a mouse model of AAA, we used control apolipoprotein E-deficient (apoeKO) mice and Atg7cKO (SMC-specific Atg7-deficient mice):apoeKO mice administered angiotensin II for 4 weeks. Intriguingly, Kaplan-Meier curves showed that the survival rates of Atg7cKO:apoeKO mice were significantly higher than those of apoeKO mice. The hematoma area in AAA of Atg7cKO:apoeKO mice was smaller than in apoeKO mice despite the lack of a significant difference in AAA incidence between the two groups. Furthermore, the amount of granulomatous tissues was significantly larger and the collagen-positive area within AAA was significantly larger in Atg7cKO:apoeKO mice than in apoeKO mice. In accordance with these findings, SMCs cultured from Atg7cKO mice showed increased expression of collagens, independent of angiotensin II action. Taken together, our data suggest that defective autophagy in SMCs elicits AAA healing that may underlie the better survival rate under dyslipidemia and angiotensin II infusion.


Sujet(s)
Angiotensine-II/toxicité , Anévrysme de l'aorte abdominale/anatomopathologie , Autophagie/physiologie , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/anatomopathologie , Angiotensine-II/administration et posologie , Animaux , Anévrysme de l'aorte abdominale/induit chimiquement , Autophagie/effets des médicaments et des substances chimiques , Cellules cultivées , Pompes à perfusion implantables , Souris , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/effets des médicaments et des substances chimiques
15.
Exp Cell Res ; 406(1): 112761, 2021 09 01.
Article de Anglais | MEDLINE | ID: mdl-34339675

RÉSUMÉ

Stresses, such as neurohumoral activation, induced pathological cardiac hypertrophy is the main risk factor for heart failure. The ubiquitin-proteasome system (UPS) plays a key role in maintaining protein homeostasis and cardiac function. However, research on the role and mechanism of deubiquitinating enzymes (DUBs) in cardiac hypertrophy is limited. Here, we observe that the deubiquitinating enzyme ubiquitin-specific protease 12(USP12) is upregulated in Ang II-induced hypertrophic hearts and primary neonatal rat cardiomyocytes (NRCMs). Inhibition of USP12 ameliorate Ang II-induced myocardial hypertrophy, while overexpression of USP12 have the opposite effect. USP12 deficiency also significantly attenuate the phenotype of Ang II-induced cardiac hypertrophy in vivo. Moreover, we demonstrate that USP12 aggravate Ang II-induced cardiac hypertrophy by enhancing METTL3, a methyltransferase which catalyze N6-methyladenosine (m6A) modification on messenger RNA and acts as a harmful factor in pathological cardiac hypertrophy. Upregulation of METTL3 reverse the reduction of myocardial hypertrophy induced by USP12 silencing in NRCMs. In contrast, knockdown of METTL3 attenuate the aggravation of myocardial hypertrophy in USP12-overexpressing NRCMs. Furthermore, we discover that USP12 promote the expression of METTL3 via upregulating p300. Mechanistically, USP12 binds and stabilizes p300, thereby activating the transcription of its downstream gene METTL3. Finally, our data show that USP12 is partially dependent on the stabilization of p300 to activate METTL3 expression and promote myocardial hypertrophy. Taken together, our results demonstrate that USP12 acts as a pro-hypertrophic deubiquitinating enzyme via enhancing p300/METTL3 axis, indicating that targeting USP12 could be a potential treatment strategy for pathological cardiac hypertrophy.


Sujet(s)
Cardiomégalie/génétique , Protéine p300-E1A/génétique , Methyltransferases/génétique , Myocytes cardiaques/métabolisme , Ubiquitin thiolesterase/génétique , Adénosine/analogues et dérivés , Adénosine/métabolisme , Angiotensine-II/administration et posologie , Animaux , Animaux nouveau-nés , Cardiomégalie/induit chimiquement , Cardiomégalie/métabolisme , Cardiomégalie/anatomopathologie , Protéine p300-E1A/métabolisme , Régulation de l'expression des gènes , Mâle , Methyltransferases/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Myocytes cardiaques/cytologie , Culture de cellules primaires , Rats , Rat Sprague-Dawley , Transduction du signal , Ubiquitin thiolesterase/métabolisme , Ubiquitination
16.
J Cardiovasc Pharmacol ; 78(4): 551-559, 2021 10 01.
Article de Anglais | MEDLINE | ID: mdl-34269699

RÉSUMÉ

ABSTRACT: We have previously shown increased vascular reactivity to angiotensin (Ang) II in familial combined hyperlipidemia. However, this has not been well studied in familial hypercholesterolemia (FH), a condition with incipient endothelial dysfunction. This study aimed to examine microvascular and macrovascular responses to Ang II in FH. Therefore, we investigated the effects of a 3-hour infusion of Ang II on blood pressure and forearm skin microvascular function in 16 otherwise healthy patients with FH and matched healthy controls. Skin microvascular hyperemia was studied by laser Doppler fluxmetry during local heating. Microvascular resistance was determined by the ratio of mean arterial pressure to microvascular hyperemia. Macrovascular reactivity was assessed by changes in brachial blood pressure. Compared with the controls, the FH group had increased baseline systolic blood pressure (127 ± 14 vs. 115 ± 12 mm Hg; P = 0.02), while systolic blood pressure responses were similar (+24 ± 9 vs. +21 ± 7 mm Hg; P = 0.26) after 3 hours of Ang II infusion. At baseline, there were no group differences in microvascular hyperemia or resistance. However, after 3 hours of Ang II infusion, heat-induced microvascular hyperemia was less pronounced in FH (126 ± 95 vs. 184 ± 102 arbitrary units; P = 0.01), while microvascular resistance during heat-induced hyperemia was increased (1.9 ± 0.9 vs. 0.9 ± 0.8, P = 0.01), as compared to controls. Both these responses were further pronounced 1 hour after stopping Ang II. In conclusion, despite similar blood pressure responses to Ang II in the FH group and controls, microvascular dilatation capacity was impaired in the FH group, indicating endothelial dysfunction. These findings and increased microvascular resistance may lead to hypertension and microvascular complications in FH.


Sujet(s)
Angiotensine-II/administration et posologie , Pression artérielle/effets des médicaments et des substances chimiques , Artère brachiale/effets des médicaments et des substances chimiques , Hyperlipoprotéinémie de type II/physiopathologie , Microcirculation/effets des médicaments et des substances chimiques , Peau/vascularisation , Vasodilatation/effets des médicaments et des substances chimiques , Adulte , Angiotensine-II/sang , Artère brachiale/physiopathologie , Études cas-témoins , Femelle , Avant-bras , Humains , Hyperlipoprotéinémie de type II/sang , Hyperlipoprotéinémie de type II/diagnostic , Perfusions veineuses , Fluxmétrie laser Doppler , Mâle , Adulte d'âge moyen , Facteurs temps
17.
Nitric Oxide ; 113-114: 57-69, 2021 09 01.
Article de Anglais | MEDLINE | ID: mdl-34091009

RÉSUMÉ

Arterial hypertension is one of the major health risk factors leading to coronary artery disease, stroke or peripheral artery disease. Dietary uptake of inorganic nitrite (NO2-) and nitrate (NO3-) via vegetables leads to enhanced vascular NO bioavailability and provides antihypertensive effects. The present study aims to understand the underlying vasoprotective effects of nutritional NO2- and NO3- co-therapy in mice with angiotensin-II (AT-II)-induced arterial hypertension. High-dose AT-II (1 mg/kg/d, 1w, s. c.) was used to induce arterial hypertension in male C57BL/6 mice. Additional inorganic nitrite (7.5 mg/kg/d, p. o.) or nitrate (150 mg/kg/d, p. o.) were administered via the drinking water. Blood pressure (tail-cuff method) and endothelial function (isometric tension) were determined. Oxidative stress and inflammation markers were quantified in aorta, heart, kidney and blood. Co-treatment with inorganic nitrite, but not with nitrate, normalized vascular function, oxidative stress markers and inflammatory pathways in AT-II treated mice. Of note, the highly beneficial effects of nitrite on all parameters and the less pronounced protection by nitrate, as seen by improvement of some parameters, were observed despite no significant increase in plasma nitrite levels by both therapies. Methemoglobin levels tended to be higher upon nitrite/nitrate treatment. Nutritional nitric oxide precursors represent a non-pharmacological treatment option for hypertension that could be applied to the general population (e.g. by eating certain vegetables). The more beneficial effects of inorganic nitrite may rely on superior NO bioactivation and stronger blood pressure lowering effects. Future large-scale clinical studies should investigate whether hypertension and cardiovascular outcome in general can be influenced by dietary inorganic nitrite therapy.


Sujet(s)
Antihypertenseurs/pharmacologie , Hypertension artérielle/traitement médicamenteux , Nitrates/pharmacologie , Nitrites/pharmacologie , Administration par voie orale , Angiotensine-II/administration et posologie , Animaux , Antihypertenseurs/administration et posologie , Antihypertenseurs/sang , Pression sanguine/effets des médicaments et des substances chimiques , Hypertension artérielle/induit chimiquement , Inflammation/induit chimiquement , Inflammation/traitement médicamenteux , Mâle , Souris , Souris de lignée C57BL , Nitrates/administration et posologie , Nitrates/sang , Nitrites/administration et posologie , Nitrites/sang , Stress oxydatif/effets des médicaments et des substances chimiques
18.
Nephron ; 145(5): 518-527, 2021.
Article de Anglais | MEDLINE | ID: mdl-34062539

RÉSUMÉ

BACKGROUND: Increasing evidence suggests that angiotensin II (Ang II), the bioactive pro-oxidant in the renin-angiotensin system, aggravates fibrosis, and the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is involved in multiple diseases, such as renal fibrosis. However, the role and underlying mechanism of Ang II in renal fibrosis remain unclear. Here, we investigated whether the NLRP3 inflammasome mediated Ang II-induced renal fibrosis, as well as the downstream pathways involved in this process. METHODS: NLRP3-/- mice were used as a model to study Ang II-infused renal fibrosis. Mice were divided into 4 groups: sham wild type, Ang II-infused wild type, sham NLRP3-/-, and Ang II-infused NLRP3-/- groups. Ang II infusion-induced renal injury was confirmed by periodic acid-Schiff and Masson's staining, immunohistochemistry, and transmission electron microscopy (TEM). Mitochondrial morphology was presented on TEM micrographs, and mitochondrial function was reflected by the protein levels of peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), mitochondrial transcription factor A (TFAM), dynamin-related protein 1 (DRP1), and mitofusin 2 (MFN2), as assessed by Western blotting. Endoplasmic reticulum (ER) stress was characterized by changes in the levels of ER chaperones, such as GRP94, BiP, CHOP, and caspase 12. RESULTS: Ang II infusion increased cell proliferation, extracellular matrix overproduction, inflammatory cell infiltration, and glomerulosclerosis and induced obvious morphological abnormalities in podocytes. Ang II infusion promoted mitochondrial damage, as indicated by TEM, and induced mitochondrial dysfunction, as evidenced by downregulation of PGC-1α, TFAM, and increased mitochondrial ROS. In addition, DRP1 expression was upregulated, while MFN2 expression was markedly decreased. The levels of GRP94, BiP, CHOP, and caspase 12 were significantly increased. However, all these detrimental effects were attenuated by NLRP3 deletion. CONCLUSIONS: NLRP3 deletion may attenuate angiotensin II-induced renal fibrosis by improving mitochondrial dysfunction and ER stress.


Sujet(s)
Angiotensine-II/physiologie , Stress du réticulum endoplasmique , Maladies du rein/physiopathologie , Mitochondries/physiologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Angiotensine-II/administration et posologie , Animaux , Humains , Maladies du rein/génétique , Mâle , Souris , Souris de lignée C57BL , Mitochondries/métabolisme , Espèces réactives de l'oxygène/métabolisme
19.
J Am Heart Assoc ; 10(9): e018716, 2021 05 04.
Article de Anglais | MEDLINE | ID: mdl-33870711

RÉSUMÉ

Background Increased potassium intake lowers blood pressure in patients with hypertension, but increased potassium intake also elevates plasma concentrations of the blood pressure-raising hormone aldosterone. Besides its well-described renal effects, aldosterone is also believed to have vascular effects, acting through mineralocorticoid receptors present in endothelial and vascular smooth muscle cells, although mineralocorticoid receptors-independent actions are also thought to be involved. Methods and Results To gain further insight into the effect of increased potassium intake and potassium-stimulated hyperaldosteronism on the human cardiovascular system, we conducted a randomized placebo-controlled double-blind crossover study in 25 healthy normotensive men, where 4 weeks treatment with a potassium supplement (90 mmol/day) was compared with 4 weeks on placebo. At the end of each treatment period, we measured potassium and aldosterone in plasma and performed an angiotensin II (AngII) infusion experiment, during which we assessed the aldosterone response in plasma. Hemodynamics were also monitored during the AngII infusion using ECG, impedance cardiography, finger plethysmography (blood pressure-monitoring), and Doppler ultrasound. The study showed that higher potassium intake increased plasma potassium (mean±SD, 4.3±0.2 versus 4.0±0.2 mmol/L; P=0.0002) and aldosterone (median [interquartile range], 440 [336-521] versus 237 [173-386] pmol/L; P<0.0001), and based on a linear mixed model for repeated measurements, increased potassium intake potentiated AngII-stimulated aldosterone secretion (P=0.0020). In contrast, the hemodynamic responses (blood pressure, total peripheral resistance, cardiac output, and renal artery blood flow) to AngII were similar after potassium and placebo. Conclusions Increased potassium intake potentiates AngII-stimulated aldosterone secretion without affecting systemic cardiovascular hemodynamics in healthy normotensive men. Registration EudraCT Number: 2013-004460-66; URL: https://www.ClinicalTrials.gov; Unique identifier: NCT02380157.


Sujet(s)
Angiotensine-II/administration et posologie , Pression sanguine/physiologie , Hypertension artérielle/thérapie , Potassium alimentaire/pharmacocinétique , Potassium/sang , Adulte , Aldostérone/sang , Marqueurs biologiques/sang , Études croisées , Méthode en double aveugle , Femelle , Études de suivi , Volontaires sains , Humains , Hypertension artérielle/physiopathologie , Perfusions veineuses , Mâle , Adulte d'âge moyen , Résultat thérapeutique , Vasoconstricteurs/administration et posologie , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...