Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.362
Filtrer
1.
J Biochem Mol Toxicol ; 38(8): e23795, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39132761

RÉSUMÉ

Renal fibrosis (RF) is one of the underlying pathological conditions leading to progressive loss of renal function and end-stage renal disease (ESRD). Over the years, various therapeutic approaches have been explored to combat RF and prevent ESRD. Despite significant advances in understanding the underlying molecular mechanism(s), effective therapeutic interventions for RF are limited. Current therapeutic strategies primarily target these underlying mechanisms to halt or reverse fibrotic progression. Inhibition of transforming growth factor-ß (TGF-ß) signaling, a pivotal mediator of RF has emerged as a central strategy to manage RF. Small molecules, peptides, and monoclonal antibodies that target TGF-ß receptors or downstream effectors have demonstrated potential in preclinical models. Modulating the renin-angiotensin system and targeting the endothelin system also provide established approaches for controlling fibrosis-related hemodynamic changes. Complementary to pharmacological strategies, lifestyle modifications, and dietary interventions contribute to holistic management. This comprehensive review aims to summarize the underlying mechanisms of RF and provide an overview of the therapeutic strategies and novel antifibrotic agents that hold promise in its treatment.


Sujet(s)
Antifibrotiques , Fibrose , Humains , Fibrose/traitement médicamenteux , Antifibrotiques/usage thérapeutique , Antifibrotiques/pharmacologie , Maladies du rein/traitement médicamenteux , Maladies du rein/anatomopathologie , Animaux , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/antagonistes et inhibiteurs , Rein/anatomopathologie , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques
2.
CMAJ ; 196(27): E931-E939, 2024 Aug 11.
Article de Anglais | MEDLINE | ID: mdl-39134317

RÉSUMÉ

BACKGROUND: Hepatitis B virus (HBV) infection is a common cause of liver-related morbidity and mortality. Evidence suggests that angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin II receptor blockers (ARBs) decrease liver fibrosis, an intermediate step between liver injury and hepatocellular carcinoma (HCC). Our aim was to investigate the association between the use of ACEIs and ARBs on incident HCC and liver-related mortality among patients with HBV infection. METHODS: We conducted a population-based study on a new-user cohort of patients seen at 24 hospitals across China. We included adult patients with HBV infection who started ACEIs or ARBs (ACEIs/ARBs), or calcium channel blockers or thiazide diuretics (CCBs/THZs) from January 2012 to December 2022. The primary outcome was incident HCC; secondary outcomes were liver-related mortality and new-onset cirrhosis. We used propensity score matching and Cox proportional hazards regression to estimate the hazard ratio (HR) and 95% confidence intervals (CIs) of study outcomes. RESULTS: Among 32 692 eligible patients (median age 58 [interquartile range (IQR) 48-68] yr, and 18 804 male [57.5%]), we matched 9946 pairs of patients starting ACEIs/ARBs or CCBs/THZs. During a mean follow-up of 2.3 years, the incidence rate of HCC per 1000 person-years was 4.11 and 5.94 among patients who started ACEIs/ARBs and CCBs/THZs, respectively, in the matched cohort. Use of ACEIs/ARBs was associated with lower risks of incident HCC (HR 0.66, 95% CI 0.50-0.86), liver-related mortality (HR 0.77, 95% CI 0.64-0.93), and new-onset cirrhosis (HR 0.81, 95% CI 0.70-0.94). INTERPRETATION: In this cohort of patients with HBV infection, new users of ACEIs/ARBs had a lower risk of incident HCC, liver-related mortality, and new-onset cirrhosis than new users of CCBs/THZs.


Sujet(s)
Antagonistes des récepteurs aux angiotensines , Inhibiteurs de l'enzyme de conversion de l'angiotensine , Carcinome hépatocellulaire , Tumeurs du foie , Humains , Carcinome hépatocellulaire/épidémiologie , Mâle , Tumeurs du foie/épidémiologie , Femelle , Adulte d'âge moyen , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique , Inhibiteurs de l'enzyme de conversion de l'angiotensine/effets indésirables , Sujet âgé , Antagonistes des récepteurs aux angiotensines/usage thérapeutique , Antagonistes des récepteurs aux angiotensines/effets indésirables , Chine/épidémiologie , Hépatite B/complications , Cirrhose du foie , Incidence , Hépatite B chronique/complications , Hépatite B chronique/traitement médicamenteux , Score de propension , Modèles des risques proportionnels , Facteurs de risque , Système rénine-angiotensine/effets des médicaments et des substances chimiques
3.
PLoS One ; 19(7): e0304135, 2024.
Article de Anglais | MEDLINE | ID: mdl-39074114

RÉSUMÉ

Renin-angiotensin system (RAS) modulators, including Angiotensin receptor blockers (ARB) and angiotensin-converting enzyme inhibitors (ACEI), are effective medications for controlling blood pressure. Cognitive deficits, including lack of concentration, memory loss, and confusion, were reported after COVID-19 infection. ARBs or ACEI increase the expression of angiotensin-converting enzyme-2 (ACE-2), a functional receptor that allows binding of SARS-CoV-2 spike protein for cellular invasion. To date, the association between the use of RAS modulators and the severity of COVID-19 cognitive dysfunction is still controversial. PURPOSE: This study addressed the following questions: 1) Does prior treatment with RAS modulator worsen COVID-19-induced cerebrovascular and cognitive dysfunction? 2) Can post-treatment with RAS modulator improve cognitive performance and cerebrovascular function following COVID-19? We hypothesize that pre-treatment exacerbates COVID-19-induced detrimental effects while post-treatment displays protective effects. METHODS: Clinical study: Patients diagnosed with COVID-19 between May 2020 and December 2022 were identified through the electronic medical record system. Inclusion criteria comprised a documented medical history of hypertension treated with at least one antihypertensive medication. Subsequently, patients were categorized into two groups: those who had been prescribed ACEIs or ARBs before admission and those who had not received such treatment before admission. Each patient was evaluated on admission for signs of neurologic dysfunction. Pre-clinical study: Humanized ACE-2 transgenic knock-in mice received the SARS-CoV-2 spike protein via jugular vein injection for 2 weeks. One group had received Losartan (10 mg/kg), an ARB, in their drinking water for two weeks before the injection, while the other group began Losartan treatment after the spike protein injection. Cognitive functions, cerebral blood flow, and cerebrovascular density were determined in all experimental groups. Moreover, vascular inflammation and cell death were assessed. RESULTS: Signs of neurological dysfunction were observed in 97 out of 177 patients (51%) taking ACEIs/ARBs prior to admission, compared to 32 out of 118 patients (27%) not receiving ACEI or ARBs. In animal studies, spike protein injection increased vascular inflammation, increased endothelial cell apoptosis, and reduced cerebrovascular density. In parallel, spike protein decreased cerebral blood flow and cognitive function. Our results showed that pretreatment with Losartan exacerbated these effects. However, post-treatment with Losartan prevented spike protein-induced vascular and neurological dysfunctions. CONCLUSION: Our clinical data showed that the use of RAS modulators before encountering COVID-19 can initially exacerbate vascular and neurological dysfunctions. Similar findings were demonstrated in the in-vivo experiments; however, the protective effects of targeting the RAS become apparent in the animal model when the treatment is initiated after spike protein injection.


Sujet(s)
Antagonistes des récepteurs aux angiotensines , Angiotensin-converting enzyme 2 , Inhibiteurs de l'enzyme de conversion de l'angiotensine , COVID-19 , Dysfonctionnement cognitif , Système rénine-angiotensine , SARS-CoV-2 , Animaux , COVID-19/complications , Humains , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Souris , Mâle , Antagonistes des récepteurs aux angiotensines/usage thérapeutique , Antagonistes des récepteurs aux angiotensines/pharmacologie , Femelle , Dysfonctionnement cognitif/traitement médicamenteux , Dysfonctionnement cognitif/étiologie , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique , Inhibiteurs de l'enzyme de conversion de l'angiotensine/pharmacologie , Sujet âgé , Angiotensin-converting enzyme 2/métabolisme , Adulte d'âge moyen , Angiopathies intracrâniennes/traitement médicamenteux , Traitements médicamenteux de la COVID-19 , Cognition/effets des médicaments et des substances chimiques
4.
Eur J Pharmacol ; 978: 176794, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-38968980

RÉSUMÉ

Heart failure (HF) remains a huge medical burden worldwide, with aging representing a major risk factor. Here, we report the effects of sacubitril/valsartan, an approved drug for HF with reduced EF, in an experimental model of aging-related HF with preserved ejection fraction (HFpEF). Eighteen-month-old female Fisher 344 rats were treated for 12 weeks with sacubitril/valsartan (60 mg/kg/day) or with valsartan (30 mg/kg/day). Three-month-old rats were used as control. No differential action of sacubitril/valsartan versus valsartan alone, either positive or negative, was observed. The positive effects of both sacubitril/valsartan and valsartan on cardiac hypertrophy was evidenced by a significant reduction of wall thickness and myocyte cross-sectional area. Contrarily, myocardial fibrosis in aging heart was not reduced by any treatment. Doppler echocardiography and left ventricular catheterization evidenced diastolic dysfunction in untreated and treated old rats. In aging rats, both classical and non-classical renin-angiotensin-aldosterone system (RAAS) were modulated. In particular, with respect to untreated animals, both sacubitril/valsartan and valsartan showed a partial restoration of cardioprotective non-classical RAAS. In conclusion, this study evidenced the favorable effects, by both treatments, on age-related cardiac hypertrophy. The attenuation of cardiomyocyte size and hypertrophic response may be linked to a shift towards cardioprotective RAAS signaling. However, diastolic dysfunction and cardiac fibrosis persisted despite of treatment and were accompanied by myocardial inflammation, endothelial activation, and oxidative stress.


Sujet(s)
Vieillissement , Amino-butyrates , Dérivés du biphényle , Association médicamenteuse , Défaillance cardiaque , Rats de lignée F344 , Tétrazoles , Valsartan , Animaux , Amino-butyrates/pharmacologie , Amino-butyrates/usage thérapeutique , Dérivés du biphényle/pharmacologie , Valsartan/pharmacologie , Valsartan/usage thérapeutique , Vieillissement/effets des médicaments et des substances chimiques , Vieillissement/anatomopathologie , Femelle , Tétrazoles/pharmacologie , Tétrazoles/usage thérapeutique , Rats , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/physiopathologie , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Fibrose , Stress oxydatif/effets des médicaments et des substances chimiques , Antagonistes des récepteurs aux angiotensines/pharmacologie , Antagonistes des récepteurs aux angiotensines/usage thérapeutique , Débit systolique/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Néprilysine/antagonistes et inhibiteurs , Néprilysine/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie
5.
Am J Physiol Heart Circ Physiol ; 327(2): H454-H459, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38968163

RÉSUMÉ

Chronic lung disease, also known as bronchopulmonary dysplasia, affects thousands of infants worldwide each year. The impact on resources is second only to bronchial asthma, with lung function affected well into adolescence. Diagnostic and therapeutic constructs have almost exclusively focused on pulmonary architecture (alveoli/airways) and pulmonary hypertension. Information on systemic hemodynamics indicates major artery thickness/stiffness, elevated systemic afterload, and/or primary left ventricular dysfunction may play a part in a subset of infants with severe neonatal-pediatric lung disease. Understanding the underlying principles with attendant effectors would aid in identifying the pathophysiological course where systemic afterload reduction with angiotensin-converting enzyme inhibitors could become the preferred treatment strategy over conventional pulmonary artery vasodilatation.NEW & NOTEWORTHY Extremely preterm infants are at a higher risk of developing severe bronchopulmonary dysplasia. In a subset of infants, diuretic and pulmonary vasodilator therapy is ineffective. Recent information points toward systemic hemodynamic disease (systemic arterial stiffness and left ventricular dysfunction) as a contributor via back-pressure changes. Mechanistic links include heightened renin angiotensin aldosterone system activity, inflammation, and oxygen toxicity. Angiotensin-converting enzyme inhibition may be operationally more suited compared with induced pulmonary artery vasodilatation.


Sujet(s)
Dysplasie bronchopulmonaire , Hémodynamique , Humains , Dysplasie bronchopulmonaire/physiopathologie , Dysplasie bronchopulmonaire/traitement médicamenteux , Dysplasie bronchopulmonaire/métabolisme , Enfant , Nouveau-né , Nourrisson , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique , Poumon/physiopathologie , Poumon/vascularisation , Poumon/effets des médicaments et des substances chimiques , Animaux , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Rigidité vasculaire , Artère pulmonaire/physiopathologie , Artère pulmonaire/effets des médicaments et des substances chimiques , Enfant d'âge préscolaire
6.
Front Endocrinol (Lausanne) ; 15: 1329945, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994012

RÉSUMÉ

Introduction: There is a lack of real-world evidence regarding the impact of concomitant metformin and renin-angiotensin system inhibitors (RASis) on sodium-glucose cotransporter-2 inhibitor (SGLT2i)-associated kidney outcomes. This study was aimed to investigate whether SGLT2i-associated kidney outcomes were modified by the concomitant use of metformin or RASis in patients with type 2 diabetes. Methods: SGLT2i users were identified from three electronic health record databases during May 2016 and December 2017 and categorized into those with and without concomitant use of metformin or RASis. Propensity score matching was performed to minimize baseline differences between groups. Study outcomes were mean estimated glomerular filtration rate (eGFR) change and time to 30%, 40%, and 50% eGFR reductions. A meta-analysis was performed to combine the estimates across databases. Results: After matching, there were 6,625 and 3,260 SGLT2i users with and without metformin, and 6,654 and 2,746 SGLT2i users with and without RASis, respectively. The eGFR dip was similar in SGLT2i users with and without metformin therapy, but was greater in SGLT2i users with RASis compared to those without RASis. Neither metformin nor RASi use had a significant effect on SGLT2i-associated eGFR reductions, as evidenced by the hazard ratios (95% CIs) of 30% eGFR reductions for SGLT2is with versus without metformin/RASis, namely 1.02 (0.87-1.20)/1.09 (0.92-1.31). Such findings were also observed in the outcomes of 40% and 50% eGFR reductions. Conclusion: Using metformin or RASis did not modify SGLT2i-associated kidney outcomes in type 2 diabetes.


Sujet(s)
Diabète de type 2 , Débit de filtration glomérulaire , Hypoglycémiants , Metformine , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Humains , Diabète de type 2/traitement médicamenteux , Diabète de type 2/complications , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Metformine/usage thérapeutique , Mâle , Femelle , Débit de filtration glomérulaire/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Hypoglycémiants/usage thérapeutique , Sujet âgé , Rein/effets des médicaments et des substances chimiques , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Résultat thérapeutique , Études rétrospectives
7.
Biosci Rep ; 44(8)2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39046229

RÉSUMÉ

Human somatic angiotensin-converting enzyme (ACE) is a key zinc metallopeptidase that plays a pivotal role in the renin-angiotensin-aldosterone system (RAAS) by regulating blood pressure and electrolyte balance. Inhibition of ACE is a cornerstone in the management of hypertension, cardiovascular diseases, and renal disorders. Recent advances in structural biology techniques have provided invaluable insights into the molecular mechanisms underlying ACE inhibition, facilitating the design and development of more effective therapeutic agents. This review focuses on the latest advancements in elucidating the structural basis for ACE inhibition. High-resolution crystallographic studies of minimally glycosylated individual domains of ACE have revealed intricate molecular details of the ACE catalytic N- and C-domains, and their detailed interactions with clinically relevant and newly designed domain-specific inhibitors. In addition, the recently elucidated structure of the glycosylated form of full-length ACE by cryo-electron microscopy (cryo-EM) has shed light on the mechanism of ACE dimerization and revealed continuous conformational changes which occur prior to ligand binding. In addition to these experimental techniques, computational approaches have also played a pivotal role in elucidating the structural basis for ACE inhibition. Molecular dynamics simulations and computational docking studies have provided atomic details of inhibitor binding kinetics and energetics, facilitating the rational design of novel ACE inhibitors with improved potency and selectivity. Furthermore, computational analysis of the motions observed by cryo-EM allowed the identification of allosteric binding sites on ACE. This affords new opportunities for the development of next-generation allosteric inhibitors with enhanced pharmacological properties. Overall, the insights highlighted in this review could enable the rational design of novel ACE inhibitors with improved efficacy and safety profiles, ultimately leading to better therapeutic outcomes for patients with hypertension and cardiovascular diseases.


Sujet(s)
Inhibiteurs de l'enzyme de conversion de l'angiotensine , Peptidyl-Dipeptidase A , Humains , Inhibiteurs de l'enzyme de conversion de l'angiotensine/composition chimique , Inhibiteurs de l'enzyme de conversion de l'angiotensine/pharmacologie , Peptidyl-Dipeptidase A/composition chimique , Peptidyl-Dipeptidase A/métabolisme , Simulation de dynamique moléculaire , Relation structure-activité , Simulation de docking moléculaire , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Cryomicroscopie électronique , Liaison aux protéines , Animaux
8.
Int Immunopharmacol ; 139: 112654, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-38996777

RÉSUMÉ

Hypertension causes platelet activation and adhesion in the brain resulting in glial activation and neuroinflammation. Further, activation of Angiotensin-Converting Enzyme 2/Angiotensin (1-7)/Mas Receptor (ACE2/Ang (1-7)/MasR) axis of central Renin-Angiotensin System (RAS), is known to reduce glial activation and neuroinflammation, thereby exhibiting anti-hypertensive and anti-neuroinflammatory properties. Therefore, in the present study, the role of ACE2/Ang (1-7)/MasR axis was studied on platelet-induced glial activation and neuroinflammation using Diminazene Aceturate (DIZE), an ACE2 activator, in astrocytes and microglial cells as well as in rat model of hypertension. We found that the ACE2 activator DIZE, independently of its BP-lowering properties, efficiently prevented hypertension-induced glial activation, neuroinflammation, and platelet CD40-CD40L signaling via upregulation of ACE2/Ang (1-7)/MasR axis. Further, DIZE decreased platelet deposition in the brain by reducing the expression of adhesion molecules on the brain endothelium. Activation of ACE2 also reduced hypertension-induced endothelial dysfunction by increasing eNOS bioavailability. Interestingly, platelets isolated from hypertensive rats or activated with ADP had significantly increased sCD40L levels and induced significantly more glial activation than platelets from DIZE treated group. Therefore, injection of DIZE pre-treated ADP-activated platelets into normotensive rats strongly reduced glial activation compared to ADP-treated platelets. Moreover, CD40L-induced glial activation, CD40 expression, and NFкB-NLRP3 inflammatory signaling are reversed by DIZE. Furthermore, the beneficial effects of ACE2 activation, DIZE was found to be significantly blocked by MLN4760 (ACE2 inhibitor) as well as A779 (MasR antagonist) treatments. Hence, our study demonstrated that ACE2 activation reduced the platelet CD40-CD40L induced glial activation and neuroinflammation, hence imparted neuroprotection.


Sujet(s)
Angiotensin-converting enzyme 2 , Ligand de CD40 , Diminazène , Modèles animaux de maladie humaine , Hypertension artérielle , Peptidyl-Dipeptidase A , Transduction du signal , Animaux , Diminazène/analogues et dérivés , Diminazène/pharmacologie , Diminazène/usage thérapeutique , Angiotensin-converting enzyme 2/métabolisme , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Hypertension artérielle/traitement médicamenteux , Ligand de CD40/métabolisme , Peptidyl-Dipeptidase A/métabolisme , Rats , Plaquettes/effets des médicaments et des substances chimiques , Plaquettes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Proto-oncogène Mas , Névroglie/effets des médicaments et des substances chimiques , Névroglie/métabolisme , Fragments peptidiques , Angiotensine-I , Cellules cultivées , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Rat Wistar , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Récepteurs couplés aux protéines G/métabolisme , Antigènes CD40/métabolisme , Humains , Activation plaquettaire/effets des médicaments et des substances chimiques , Antihypertenseurs/pharmacologie , Antihypertenseurs/usage thérapeutique
10.
Curr Treat Options Oncol ; 25(8): 1038-1054, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39066853

RÉSUMÉ

OPINION STATEMENT: Anthracycline (ANT)-induced cardiotoxicity (AIC) is a particularly prominent form of cancer therapy-related cardiovascular toxicity leading to the limitations of ANTs in clinical practice. Even though AIC has drawn particular attention, the best way to treat it is remaining unclear. Updates to AIC therapy have been made possible by recent developments in research on the underlying processes of AIC. We review the current molecular pathways leading to AIC: 1) oxidative stress (OS) including enzymatic-induced and other mechanisms; 2) topoisomerase; 3) inflammatory response; 4) cardiac progenitor cell damage; 5) epigenetic changes; 6) renin-angiotensin-aldosterone system (RAAS) dysregulation. And we systematically discuss current prevention and treatment strategies and novel pathogenesis-based therapies for AIC: 1) dose reduction and change; 2) altering drug delivery methods; 3) antioxidants, dexrezosen, statina, RAAS inhibitors, and hypoglycemic drugs; 4) miRNA, natural phytochemicals, mesenchymal stem cells, and cardiac progenitor cells. We also offer a fresh perspective on the management of AIC by outlining the current dilemmas and challenges associated with its prevention and treatment.


Sujet(s)
Anthracyclines , Cardiotoxicité , Humains , Cardiotoxicité/étiologie , Cardiotoxicité/prévention et contrôle , Anthracyclines/effets indésirables , Anthracyclines/usage thérapeutique , Médecine de précision/méthodes , Animaux , Tumeurs/traitement médicamenteux , Stress oxydatif/effets des médicaments et des substances chimiques , Prise en charge de la maladie , Prédisposition aux maladies , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Marqueurs biologiques
11.
Rev Prat ; 74(6): s19-s22, 2024 Jun.
Article de Français | MEDLINE | ID: mdl-39011717

RÉSUMÉ

RENOPROTECTIVE TREATMENTS: RENIN-ANGIOTENSINALDOSTERONE SYSTEM BLOCKADE AND BEYOND. Patients with chronic kidney disease (CKD) require both etiological and symptomatic treatments to slow renal function decline. Reductions of protein and salt intakes are required. Pharmacological treatments combines blockade of the renin-angiotensin system, sodium-glucose co-transporter type 2 inhibitors and mineralocorticoid receptor antagonists in most diabetic patients. The albumin creatinine ration (ACR) in morning spot urine samples is now a therapeutic target both in diabetic and non-diabetic CKD patients.


NÉPHROPROTECTION MÉDICAMENTEUSE : AU-DELÀ DU BLOCAGE DU SYSTÈME RÉNINE-ANGIOTENSINE. Au-delà du traitement étiologique éventuel d'une maladie rénale chronique (MRC), et du contrôle de l'ensemble des facteurs de risque vasculaire, un traitement symptomatique est nécessaire pour ralentir le déclin de la fonction rénale. La prise en charge diététique permet de limiter les apports en protéines et en sel. Le traitement pharmacologique comporte nécessairement une association d'un bloqueur du système rénine-angiotensine-aldostérone, le plus souvent un inhibiteur du co-transport sodiumglucose de type 2 (SGLT2i) et chez certains diabétiques un antagoniste des récepteurs des minéralocorticoïdes (ARM). Le rapport albuminurie/créatininurie (RAC) sur un échantillon d'urine devient une cible thérapeutique aussi bien chez les non-diabétiques que chez les diabétiques.


Sujet(s)
Antagonistes des récepteurs des minéralocorticoïdes , Système rénine-angiotensine , Humains , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Système rénine-angiotensine/physiologie , Antagonistes des récepteurs des minéralocorticoïdes/usage thérapeutique , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique , Insuffisance rénale chronique/traitement médicamenteux , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Antagonistes des récepteurs aux angiotensines/usage thérapeutique
12.
Nutrients ; 16(12)2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38931311

RÉSUMÉ

Post-traumatic stress disorder (PTSD) is a persistent psychiatric condition that arises following exposure to traumatic events such as warfare, natural disasters, or other catastrophic incidents, typically characterized by heightened anxiety, depressive symptoms, and cognitive dysfunction. In this study, animals subjected to single prolonged stress (SPS) were administered evodiamine (EVO) and compared to a positive control group receiving sertraline. The animals were then assessed for alterations in anxiety, depression, and cognitive function. Histological analysis was conducted to examine neuronal changes in the hippocampus. In order to predict the core targets and related mechanisms of evodiamine intervention in PTSD, network pharmacology was used. The metabolic markers pre- and post-drug administration were identified using nontargeted serum metabolomics techniques, and the intersecting Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were screened. Finally, the core targets were validated through molecular docking, enzyme-linked immunosorbent assays, and immunofluorescence staining to confirm the anti-PTSD effects and mechanisms of these targets. As well as improving cognitive impairment, evodiamine reversed anxiety- and depression-like behaviors. It also inhibited the reduction in the number of hippocampal neuronal cells and Nissl bodies in SPS mice inhibited angiotensin converting enzyme (ACE) levels in the hippocampus of SPS mice, and modulated the renin angiotensin pathway and its associated serum metabolites in brain tissue. Evodiamine shows promise as a potential candidate for alleviating the symptoms of post-traumatic stress disorder.


Sujet(s)
Modèles animaux de maladie humaine , Hippocampe , Neurones , Quinazolines , Système rénine-angiotensine , Troubles de stress post-traumatique , Animaux , Troubles de stress post-traumatique/traitement médicamenteux , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Quinazolines/pharmacologie , Souris , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Mâle , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Comportement animal/effets des médicaments et des substances chimiques , Inhibiteurs de l'enzyme de conversion de l'angiotensine/pharmacologie , Dépression/traitement médicamenteux , Simulation de docking moléculaire , Anxiété/traitement médicamenteux , Souris de lignée C57BL , Pharmacologie des réseaux
13.
Physiol Rep ; 12(12): e16094, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38924381

RÉSUMÉ

The renin-angiotensin system (RAS)-a classical blood pressure regulator-largely contributes to healthy organ development and function. Besides, RAS activation promotes age-related changes and age-associated diseases, which are attenuated/abolished by RAS-blockade in several mammalian species. RAS-blockers also increase rodent lifespan. In previous work, we discussed how RAS-blockade downregulates mTOR and growth hormone/IGF-1 signaling, and stimulates AMPK activity (together with klotho, sirtuin, and vitamin D-receptor upregulation), and proposed that at least some of RAS-blockade's aging benefits are mediated through regulation of these intermediaries and their signaling to mitochondria. Here, we included RAS-blockade's impact on other aging regulatory pathways, that is, TGF-ß, NF-kB, PI3K, MAPK, PKC, Notch, and Wnt, all of which affect mitochondria. No direct evidence is available on RAS/RAS-blockade-aging regulatory pathway-mitochondria interactions. However, existing results allow to conjecture that RAS-blockers neutralize mitochondrial dysfunction by acting on the discussed pathways. The reviewed evidence led us to propose that the foundation is laid for conducting clinical trials aimed at testing whether angiotensin-converting enzyme inhibitors (ACEi) or angiotensin receptor blockers (ARB)-even at subclinical doses-offer the possibility to live longer and in better health. As ACEi and ARB are low cost and well-tolerated anti-hypertension therapies in use for over 35 years, investigating their administration to attenuate/prevent aging effects seems simple to implement.


Sujet(s)
Vieillissement , Inhibiteurs de l'enzyme de conversion de l'angiotensine , Système rénine-angiotensine , Humains , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Vieillissement/effets des médicaments et des substances chimiques , Vieillissement/métabolisme , Vieillissement/physiologie , Animaux , Inhibiteurs de l'enzyme de conversion de l'angiotensine/pharmacologie , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Antagonistes des récepteurs aux angiotensines/pharmacologie , Antagonistes des récepteurs aux angiotensines/usage thérapeutique
14.
Chin Med Sci J ; 39(2): 79-90, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38845179

RÉSUMÉ

Objective Variations are present in common clinical practices regarding best practice in managing hyperkalaemia (HK), there is therefore a need to establish a multi-specialty approach to optimal renin-angiotension-aldosterone system inhibitors (RAASi) usage and HK management in patients with chronic kidney disease (CKD) & heart failure (HF).This study aimed to establish a multi-speciality approach to the optimal use of RAASi and the management of HK in patients with CKD and HF. Methods A steering expert group of cardiology and nephrology experts across China were convened to discuss challenges to HK management through a nominal group technique. The group then created a list of 41 statements for a consensus questionnaire, which was distributed for a further survey in extended panel group of cardiologists and nephrologists across China. Consensus was assessed using a modified Delphi technique, with agreement defined as "strong" (≥75% and <90%) and "very strong" (≥90%). The steering group, data collection, and analysis were aided by an independent facilitator. Results A total of 150 responses from 21 provinces across China were recruited in the survey. Respondents were comprised of an even split (n=75, 50%) between cardiologists and nephrologists. All 41 statements achieved the 75% consensus agreement threshold, of which 27 statements attained very strong consensus (≥90% agreement) and 14 attained strong consensus (agreement between 75% and 90%). Conclusion Based on the agreement levels from respondents, the steering group agreed a set of recommendations intended to improve patient outcomes in the use of RAASi therapy and HK management in China.


Sujet(s)
Consensus , Méthode Delphi , Défaillance cardiaque , Hyperkaliémie , Insuffisance rénale chronique , Humains , Hyperkaliémie/traitement médicamenteux , Défaillance cardiaque/traitement médicamenteux , Insuffisance rénale chronique/thérapie , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/complications , Chine , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Enquêtes et questionnaires , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique , Peuples d'Asie de l'Est
15.
Food Res Int ; 189: 114570, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38876598

RÉSUMÉ

Edible insects are recognized as promising food sources due to their nutritional composition. Some species, such as Gryllus assimilis, contain proteins, lipids, and carbohydrates of high biological value, which regulate several metabolic functions, including the Renin-Angiotensin System (RAS). In this context, the present study aimed to assess the effects of dietary supplementation with whole Gryllus assimilis powder on the metabolism of malnourished mice. Thirty-two male Swiss mice were used and divided into four treatment groups. The groups were identified as (AIN93-M); AIN93-M + Gryllus assimilis diet (AIN93-M + GA); AIN93-M + Renutrition diet (AIN93-M + REN) and AIN93-M + Renutrition diet + Gryllus assimilis (AIN93-M + REN + GA). The results showed that whole Gryllus assimilis powder inclusion promotes recovery from protein-energy malnutrition, reduces adiposity, and improves glucose tolerance and insulin sensitivity. It also reduces total cholesterol, triglycerides, VLDL, and adipocyte area. We also observed a significant increase in the expression of RAS-related genes, such as ACE2 and MasR, followed by a reduction in Angiotensinogen and ACE. The main findings of the present study suggest the use of black cricket as a viable strategy for the prevention and treatment of protein-energy malnutrition, as well as the reduction of adiposity, and improvement of lipid and glycemic parameters, with antihypertensive potential.


Sujet(s)
Tissu adipeux , Compléments alimentaires , Gryllidae , Malnutrition protéinocalorique , Système rénine-angiotensine , Animaux , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Mâle , Souris , Malnutrition protéinocalorique/métabolisme , Malnutrition protéinocalorique/diétothérapie , Tissu adipeux/métabolisme , Adiposité , Insulinorésistance
16.
Cancer Chemother Pharmacol ; 94(1): 1-23, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38914751

RÉSUMÉ

Glioblastoma multiforme (GBM) is a highly aggressive and incurable disease accounting for about 10,000 deaths in the USA each year. Despite the current treatment approach which includes surgery with chemotherapy and radiation therapy, there remains a high prevalence of recurrence. Notable improvements have been observed in persons receiving concurrent antihypertensive drugs such as renin angiotensin inhibitors (RAS) or the antidiabetic drug metformin with standard therapy. Anti-tumoral effects of RAS inhibitors and metformin have been observed in in vitro and in vivo studies. Although clinical trials have shown mixed results, the potential for the use of RAS inhibitors and metformin as adjuvant GBM therapy remains promising. Nevertheless, evidence suggest that these drugs exert multimodal antitumor actions; by particularly targeting several cancer hallmarks. In this review, we highlight the results of clinical studies using multidrug cocktails containing RAS inhibitors and or metformin added to standard therapy for GBM. In addition, we highlight the possible molecular mechanisms by which these repurposed drugs with an excellent safety profile might elicit their anti-tumoral effects. RAS inhibition elicits anti-inflammatory, anti-angiogenic, and immune sensitivity effects in GBM. However, metformin promotes anti-migratory, anti-proliferative and pro-apoptotic effects mainly through the activation of AMP-activated protein kinase. Also, we discussed metformin's potential in targeting both GBM cells as well as GBM associated-stem cells. Finally, we summarize a few drug interactions that may cause an additive or antagonistic effect that may lead to adverse effects and influence treatment outcome.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Metformine , Système rénine-angiotensine , Humains , Metformine/usage thérapeutique , Metformine/pharmacologie , Glioblastome/traitement médicamenteux , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Tumeurs du cerveau/traitement médicamenteux , Animaux , Inhibiteurs de l'enzyme de conversion de l'angiotensine/pharmacologie , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique , Hypoglycémiants/usage thérapeutique , Hypoglycémiants/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie
17.
Clin Chim Acta ; 561: 119750, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38885756

RÉSUMÉ

BACKGROUND: Renin-angiotensin system inhibitors (RASi) treatment is the basic therapy for IgA nephropathy (IgAN) patients. However, there is few of biomarker that can predict the efficacy of RASi. This study aimed to find urinary exosomal mRNAs related to the therapeutic effect of RASi in the treatment of proteinuria in IgAN patients. METHODS: We divided IgAN patients in screening cohort into A1 (proteinuria increase at 3 months), B1 (proteinuria decrease less than 50 % at 3 months), C1 (proteinuria decrease more than 50 % at 3 months) groups according to changes of proteinuria after treatment. The urinary exosomes were collected before biopsy, RNAs were extracted and analyzed with the microarray assay. The candidate genes were screened by differentially expressed genes (DEGs) analysis and then validated by quantitative real-time polymerase chain reaction (qPCR) in a validation cohort. A receiver operating characteristic (ROC) curve was used to evaluate gene performance in predicting therapeutic effect on RASi reducing proteinuria in IgAN patients. RESULTS: ECE1 and PDE1A mRNAs were significantly different among the three groups, and were gradually decreased among A1, B1 and C1 groups. In the validation cohort, the level of urinary exosomal ECE1 and PDE1A mRNAs were also significantly lower in A2 group compared with C2 group(ECE1, P < 0.001;PDE1A, P < 0.01). Besides, the level of ECE1 mRNA was also lower in B2 group compared with C2 group (P < 0.01). The ROC curve verified that urinary exosomal ECE1 and PDE1A gene level predicted RASi efficacy in IgAN patients with area under curve (AUC) 0.68 and 0.63 respectively. CONCLUSION: Urinary exosomal ECE1 and PDE1A mRNAs expression can serve as potential biomarkers for predicting the RASi efficacy to reduce proteinuria in IgAN patients.


Sujet(s)
Marqueurs biologiques , Exosomes , Glomérulonéphrite à dépôts d'IgA , ARN messager , Système rénine-angiotensine , Humains , Glomérulonéphrite à dépôts d'IgA/traitement médicamenteux , Glomérulonéphrite à dépôts d'IgA/urine , Glomérulonéphrite à dépôts d'IgA/génétique , Mâle , ARN messager/génétique , ARN messager/urine , Adulte , Femelle , Exosomes/génétique , Exosomes/métabolisme , Marqueurs biologiques/urine , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Système rénine-angiotensine/génétique , Adulte d'âge moyen , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique
18.
Ren Fail ; 46(2): 2359033, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38836372

RÉSUMÉ

OBJECTIVE: To determine the efficacy and safety of Astragalus combined with renin-angiotensin-aldosterone system (RAAS) blockers in treating stage III diabetic nephropathy (DN) by meta-analysis. METHODS: PubMed, Embase, Cochrane Library, Wiley, and Web of Science databases were searched for articles published between August 2007 and August 2022. Clinical studies on Astragalus combined with RAAS blockers for the treatment of stage III DN were included. Meta-analysis was performed by RevMan 5.1 and Stata 14.3 software. RESULTS: A total of 32 papers were included in this meta-analysis, containing 2462 patients from randomized controlled trials, with 1244 receiving the combination treatment and 1218 solely receiving RAAS blockers. Astragalus combined with RAAS blockers yielded a significantly higher total effective rate (TER) (mean difference [MD] 3.63, 95% confidence interval [CI] 2.59-5.09) and significantly reduced urinary protein excretion rate (UPER), serum creatinine (Scr), blood urine nitrogen (BUN) and glycosylated hemoglobin (HbAlc) levels. In subgroup analysis, combining astragalus and angiotensin receptor blocker significantly lowered fasting plasma glucose (FPG) and 24 h urinary protein (24hUTP) levels, compared with the combined astragalus and angiotensin-converting enzyme inhibitor treatment. Meanwhile, the latter significantly decreased the urinary microprotein (ß2-MG). Importantly, the sensitivity analysis confirmed the study's stability, and publication bias was not detected for UPER, BUN, HbAlc, FPG, or ß2-MG. However, the TER, SCr, and 24hUTP results suggested possible publication bias. CONCLUSIONS: The astragalus-RAAS blocker combination treatment is safe and improves outcomes; however, rigorous randomized, large-scale, multi-center, double-blind trials are needed to evaluate its efficacy and safety in stage III DN.


Renin-angiotensin-aldosterone system (RAAS) inhibitors are commonly used to treat diabetic neuropathy (DN) and Astragalus membranaceus components are known to improve DN symptoms.We aimed to establish the efficacy and safety of using Astragalus combined with RAAS inhibitors.Astragalus combined with RAAS inhibitors enhances the total effective rate of diabetic neuropathy response to treatment and reduces urinary protein excretion rate, serum creatinine, blood urea nitrogen and HbAlc.Sensitivity analysis affirms study stability, while publication bias was detected for total effective rate, serum creatinine, and 24 h urinary protein levels.


Sujet(s)
Antagonistes des récepteurs aux angiotensines , Inhibiteurs de l'enzyme de conversion de l'angiotensine , Néphropathies diabétiques , Association de médicaments , Système rénine-angiotensine , Humains , Néphropathies diabétiques/traitement médicamenteux , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Antagonistes des récepteurs aux angiotensines/usage thérapeutique , Astragalus , Essais contrôlés randomisés comme sujet , Médicaments issus de plantes chinoises/usage thérapeutique , Médicaments issus de plantes chinoises/administration et posologie , Résultat thérapeutique , Créatinine/sang , Hémoglobine glyquée , Protéinurie/traitement médicamenteux
19.
Am J Respir Crit Care Med ; 210(4): 497-507, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38941161

RÉSUMÉ

Rationale: Sarcoidosis is a granulomatous disorder of unclear cause notable for abnormal elevation of blood and tissue ACE1 (angiotensin converting enzyme 1) levels and activity. ACE1 regulates the renin-angiotensin-aldosterone system (RAAS), the terminal product of which is aldosterone, which selectively engages mineralocorticoid receptors to promote inflammation. Objectives: We sought to determine whether the RAAS promotes sarcoidosis granuloma formation and related inflammatory responses. Methods: Using an established ex vivo model, we first determined whether aldosterone was produced by sarcoidosis granulomas and verified the presence of CYP11B2, the enzyme required for its production. We then evaluated the effects of selective inhibitors of ACE1 (captopril), angiotensin type 1 receptor (losartan), and mineralocorticoid receptors (spironolactone, eplerenone) on granuloma formation, reflected by computer image analysis-generated granuloma area, and selected cytokines incriminated in sarcoidosis pathogenesis. Measurements and Main Results: Aldosterone was spontaneously produced by sarcoidosis peripheral blood mononuclear cells, and both intra- and extracellular levels steadily increased during granuloma formation. In parallel, peripheral blood mononuclear cells were shown to express more CYP11B2 during granuloma formation. Significant inhibition of sarcoidosis granulomas and related cytokines (TNFα, IL-1ß, IFNγ, IL-10) was observed in response to pretreatments with captopril, losartan, spironolactone, or eplerenone, comparable to that of prednisone. Conclusions: The RAAS is intact in sarcoidosis granulomas and contributes significantly to early granuloma formation and to related inflammatory mediator responses, with important implications for clinical management.


Sujet(s)
Aldostérone , Cytochrome P-450 CYP11B2 , Granulome , Système rénine-angiotensine , Sarcoïdose , Humains , Système rénine-angiotensine/effets des médicaments et des substances chimiques , Système rénine-angiotensine/physiologie , Granulome/traitement médicamenteux , Aldostérone/métabolisme , Sarcoïdose/traitement médicamenteux , Sarcoïdose/physiopathologie , Mâle , Femelle , Losartan/pharmacologie , Losartan/usage thérapeutique , Éplérénone/pharmacologie , Éplérénone/usage thérapeutique , Inflammation , Spironolactone/usage thérapeutique , Spironolactone/pharmacologie , Adulte d'âge moyen , Captopril/pharmacologie , Captopril/usage thérapeutique , Cytokines/métabolisme , Inhibiteurs de l'enzyme de conversion de l'angiotensine/pharmacologie , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique , Peptidyl-Dipeptidase A/métabolisme , Agranulocytes/métabolisme , Agranulocytes/effets des médicaments et des substances chimiques , Antagonistes des récepteurs des minéralocorticoïdes/usage thérapeutique , Antagonistes des récepteurs des minéralocorticoïdes/pharmacologie
20.
Pharmacol Res ; 204: 107210, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38740146

RÉSUMÉ

Heart failure with reduced ejection fraction (HFrEF) is a clinical syndrome characterized by volume overload, impaired exercise capacity, and recurrent hospital admissions. A major contributor to the pathophysiology and clinical presentation of heart failure is the activation of the renin-angiotensin-aldosterone system (RAAS). Normally, RAAS is responsible for the homeostatic regulation of blood pressure, extracellular fluid volume, and serum sodium concentration. In HFrEF, RAAS gets chronically activated in response to decreased cardiac output, further aggravating the congestion and cardiotoxic effects. Hence, inhibition of RAAS is a major approach in the pharmacologic treatment of those patients. The most recently introduced RAAS antagonizing medication class is angiotensin receptor blocker/ neprilysin inhibitor (ARNI). In this paper, we discuss ARNIs' superiority over traditional RAAS antagonizing agents in reducing heart failure hospitalization and mortality. We also tease out the evidence that shows ARNIs' renoprotective functions in heart failure patients including those with chronic or end stage kidney disease. We also discuss the evidence showing the added benefit resulting from combining ARNIs with a sodium-glucose cotransporter-2 (SGLT-2) inhibitor. Moreover, how ARNIs decrease the risk of arrhythmias and reverse cardiac remodeling, ultimately lowering the risk of cardiovascular death, is also discussed. We then present the positive outcome of ARNIs' use in patients with diabetes mellitus and those recovering from acute decompensated heart failure. ARNIs' side effects are also appreciated and discussed. Taken together, the provided insight and critical appraisal of the evidence justifies and supports the implementation of ARNIs in the guidelines for the treatment of HFrEF.


Sujet(s)
Antagonistes des récepteurs aux angiotensines , Défaillance cardiaque , Néprilysine , Débit systolique , Humains , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/physiopathologie , Néprilysine/antagonistes et inhibiteurs , Débit systolique/effets des médicaments et des substances chimiques , Animaux , Antagonistes des récepteurs aux angiotensines/usage thérapeutique , Antagonistes des récepteurs aux angiotensines/effets indésirables , Système rénine-angiotensine/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE