Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.372
Filtrer
1.
Expert Opin Pharmacother ; 25(8): 1095-1104, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38822807

RÉSUMÉ

BACKGROUND: This study aimed to investigate the association between clinical and laboratory parameters and response to therapy with sodium-glucose cotransporter 2 inhibitors (SGLT2i) in patients with type 2 diabetes mellitus (T2D). RESEARCH DESIGN AND METHODS: We retrospectively analyzed the medical records of people with T2D in whom SGLT2i was started. Clinical and laboratory parameters were recorded before, 3 and 6 months after starting treatment. Specific criteria were applied to classify participants into good and poor responders in terms of weight loss (primary outcome) and glycemic control (secondary outcome), separately. RESULTS: Fifty individuals (64% men) with a mean age of 65.8 ± 8.5 years were included in the analysis. 86% and 64% of the participants were classified into good response categories for glycemic control and weight loss, respectively. Good responders in terms of glycemic control had lower high-density lipoprotein cholesterol levels at baseline compared to poor responders (43.3 vs 57.4 mg/dl, p = 0.044). In the logistic regression analysis, a higher baseline weight was associated with a better response to therapy in terms of weight loss (p = 0.04). CONCLUSIONS: Our findings suggest that specific clinical and laboratory parameters are associated with response to SGLT2i treatment and can contribute to a more personalized approach to T2D care.


Sujet(s)
Glycémie , Diabète de type 2 , Hypoglycémiants , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Perte de poids , Humains , Diabète de type 2/traitement médicamenteux , Diabète de type 2/sang , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Études rétrospectives , Mâle , Femelle , Sujet âgé , Adulte d'âge moyen , Perte de poids/effets des médicaments et des substances chimiques , Hypoglycémiants/usage thérapeutique , Glycémie/effets des médicaments et des substances chimiques , Glycémie/métabolisme , Résultat thérapeutique , Cholestérol HDL/sang
2.
Drug Des Devel Ther ; 18: 1855-1864, 2024.
Article de Anglais | MEDLINE | ID: mdl-38828023

RÉSUMÉ

Purpose: Henagliflozin is an original, selective sodium-glucose cotransporter 2 (SGLT2) inhibitor. Hydrochlorothiazide (HCTZ) is a common anti-hypertensive drug. This study aimed to evaluate the potential interaction between henagliflozin and HCTZ. Methods: This was a single-arm, open-label, multi-dose, three-period study that was conducted in healthy Chinese volunteers. Twelve subjects were treated in three periods, period 1: 25 mg HCTZ for four days, period 2: 10 mg henagliflozin for four days and period 3: 25 mg HCTZ + 10 mg henagliflozin for four days. Blood samples and urine samples were collected before and up to 24 hours after drug administrations on day 4, day 10 and day 14. The plasma concentrations of henagliflozin and HCTZ were analyzed using LC-MS/MS. The urine samples were collected for pharmacodynamic glucose and electrolyte analyses. Tolerability was also evaluated. Results: The 90% CI of the ratio of geometric means (combination: monotherapy) for AUCτ,ss of henagliflozin and HCTZ was within the bioequivalence interval of 0.80-1.25. For henagliflozin, co-administration increased Css, max by 24.32% and the 90% CI of the GMR was (108.34%, 142.65%), and the 24-hour urine volume and glucose excretion decreased by 0.43% and 19.6%, respectively. For HCTZ, co-administration decreased Css, max by 19.41% and the 90% CI of the GMR was (71.60%, 90.72%), and the 24-hour urine volume and urinary calcium, potassium, phosphorus, chloride, and sodium excretion decreased by 11.7%, 20.8%, 11.8%, 11.9%, 22.0% and 15.5%, respectively. All subjects (12/12) reported adverse events (AEs), but the majority of theses AEs were mild and no serious AEs were reported. Conclusion: Although Css,max was affected by the combination of henagliflozin and HCTZ, there was no clinically meaningful safety interaction between them. Given these results, coadministration of HCTZ should not require any adaptation of henagliflozin dosing. Trial Registration: ClinicalTrials.gov NCT06083116.


Sujet(s)
Interactions médicamenteuses , Volontaires sains , Hydrochlorothiazide , Humains , Hydrochlorothiazide/administration et posologie , Hydrochlorothiazide/pharmacocinétique , Hydrochlorothiazide/pharmacologie , Adulte , Mâle , Jeune adulte , Femelle , Glucosides/administration et posologie , Glucosides/pharmacocinétique , Glucosides/pharmacologie , Asiatiques , Relation dose-effet des médicaments , Inhibiteurs du cotransporteur sodium-glucose de type 2/administration et posologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacocinétique , Peuples d'Asie de l'Est
3.
Clin Sci (Lond) ; 138(11): 687-697, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38835256

RÉSUMÉ

Endothelin A and B receptors, together with sodium-glucose cotransporter-2 (SGLT-2) channels are important targets in improving endothelial function and intervention with inhibitors has been the subject of multiple mechanistic and clinical outcome trials over recent years. Notable successes include the treatment of pulmonary hypertension with endothelin receptor antagonists, and the treatment of heart failure and chronic kidney disease with SGLT-2 inhibitors. With distinct and complementary mechanisms, in this review, we explore the logic of combination therapy for a number of diseases which have endothelial dysfunction at their heart.


Sujet(s)
Endothéline-1 , Endothélium vasculaire , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Humains , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Endothéline-1/métabolisme , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/métabolisme , Animaux , Association de médicaments , Antagonistes des récepteurs de l'endothéline/usage thérapeutique , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/physiopathologie
4.
J Affect Disord ; 360: 229-241, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38823591

RÉSUMÉ

A high-fat diet can modify the composition of gut microbiota, resulting in dysbiosis. Changes in gut microbiota composition can lead to increased permeability of the gut barrier, allowing bacterial products like lipopolysaccharides (LPS) to enter circulation. This process can initiate systemic inflammation and contribute to neuroinflammation. Empagliflozin (EF), an SGLT2 inhibitor-type hypoglycemic drug, has been reported to treat neuroinflammation. However, there is a lack of evidence showing that EF regulates the gut microbiota axis to control neuroinflammation in HFD models. In this study, we explored whether EF could improve neuroinflammation caused by an HFD via regulation of the gut microbiota and the mechanism underlying this phenomenon. Our data revealed that EF alleviates pathological brain injury, reduces the reactive proliferation of astrocytes, and increases the expression of synaptophysin. In addition, the levels of inflammatory factors in hippocampal tissue were significantly decreased after EF intervention. Subsequently, the results of 16S rRNA gene sequencing showed that EF could change the microbial community structure of mice, indicating that the abundance of Lactococcus, Ligilactobacillus and other microbial populations decreased dramatically. Therefore, EF alleviates neuroinflammation by inhibiting gut microbiota-mediated astrocyte activation in the brains of high-fat diet-fed mice. Our study focused on the gut-brain axis, and broader research on neuroinflammation can provide a more holistic understanding of the mechanisms driving neurodegenerative diseases and inform the development of effective strategies to mitigate their impact on brain health. The results provide strong evidence supporting the larger clinical application of EF.


Sujet(s)
Astrocytes , Composés benzhydryliques , Alimentation riche en graisse , Microbiome gastro-intestinal , Glucosides , Maladies neuro-inflammatoires , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Alimentation riche en graisse/effets indésirables , Astrocytes/effets des médicaments et des substances chimiques , Glucosides/pharmacologie , Souris , Composés benzhydryliques/pharmacologie , Maladies neuro-inflammatoires/traitement médicamenteux , Mâle , Souris de lignée C57BL , Encéphale/effets des médicaments et des substances chimiques , Axe cerveau-intestin/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Dysbiose
5.
JCI Insight ; 9(11)2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38855868

RÉSUMÉ

Lactate elevation is a well-characterized biomarker of mitochondrial dysfunction, but its role in diabetic kidney disease (DKD) is not well defined. Urine lactate was measured in patients with type 2 diabetes (T2D) in 3 cohorts (HUNT3, SMART2D, CRIC). Urine and plasma lactate were measured during euglycemic and hyperglycemic clamps in participants with type 1 diabetes (T1D). Patients in the HUNT3 cohort with DKD had elevated urine lactate levels compared with age- and sex-matched controls. In patients in the SMART2D and CRIC cohorts, the third tertile of urine lactate/creatinine was associated with more rapid estimated glomerular filtration rate decline, relative to first tertile. Patients with T1D demonstrated a strong association between glucose and lactate in both plasma and urine. Glucose-stimulated lactate likely derives in part from proximal tubular cells, since lactate production was attenuated with sodium-glucose cotransporter-2 (SGLT2) inhibition in kidney sections and in SGLT2-deficient mice. Several glycolytic genes were elevated in human diabetic proximal tubules. Lactate levels above 2.5 mM potently inhibited mitochondrial oxidative phosphorylation in human proximal tubule (HK2) cells. We conclude that increased lactate production under diabetic conditions can contribute to mitochondrial dysfunction and become a feed-forward component to DKD pathogenesis.


Sujet(s)
Diabète de type 1 , Diabète de type 2 , Néphropathies diabétiques , Glycolyse , Acide lactique , Humains , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Animaux , Souris , Acide lactique/métabolisme , Acide lactique/sang , Femelle , Mâle , Adulte d'âge moyen , Diabète de type 2/métabolisme , Diabète de type 2/complications , Diabète de type 1/métabolisme , Diabète de type 1/complications , Mitochondries/métabolisme , Adulte , Débit de filtration glomérulaire , Sujet âgé , Tubules contournés proximaux/métabolisme , Glucose/métabolisme , Phosphorylation oxydative , Marqueurs biologiques/métabolisme , Transporteur-2 sodium-glucose/métabolisme , Transporteur-2 sodium-glucose/génétique , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie
8.
Circ Heart Fail ; 17(6): e011107, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38847102

RÉSUMÉ

BACKGROUND: Clinical studies demonstrated beneficial effects of sodium-glucose-transporter 2 inhibitors on the risk of cardiovascular death in patients with heart failure with preserved ejection fraction (HFpEF). However, underlying processes for cardioprotection remain unclear. The present study focused on the impact of empagliflozin (Empa) on myocardial function in a rat model with established HFpEF and analyzed underlying molecular mechanisms. METHODS: Obese ZSF1 (Zucker fatty and spontaneously hypertensive) rats were randomized to standard care (HFpEF, n=18) or Empa (HFpEF/Empa, n=18). ZSF1 lean rats (con, n=18) served as healthy controls. Echocardiography was performed at baseline and after 4 and 8 weeks, respectively. After 8 weeks of treatment, hemodynamics were measured invasively, mitochondrial function was assessed and myocardial tissue was collected for either molecular and histological analyses or transmission electron microscopy. RESULTS: In HFpEF Empa significantly improved diastolic function (E/é: con: 17.5±2.8; HFpEF: 24.4±4.6; P<0.001 versus con; HFpEF/Empa: 19.4±3.2; P<0.001 versus HFpEF). This was accompanied by improved hemodynamics and calcium handling and by reduced inflammation, hypertrophy, and fibrosis. Proteomic analysis demonstrated major changes in proteins involved in mitochondrial oxidative phosphorylation. Cardiac mitochondrial respiration was significantly impaired in HFpEF but restored by Empa (Vmax complex IV: con: 0.18±0.07 mmol O2/s/mg; HFpEF: 0.13±0.05 mmol O2/s/mg; P<0.041 versus con; HFpEF/Empa: 0.21±0.05 mmol O2/s/mg; P=0.012 versus HFpEF) without alterations of mitochondrial content. The expression of cardiolipin, an essential stability/functionality-mediating phospholipid of the respiratory chain, was significantly decreased in HFpEF but reverted by Empa (con: 15.9±1.7 nmol/mg protein; HFpEF: 12.5±1.8 nmol/mg protein; P=0.002 versus con; HFpEF/Empa: 14.5±1.8 nmol/mg protein; P=0.03 versus HFpEF). Transmission electron microscopy revealed a reduced size of mitochondria in HFpEF, which was restored by Empa. CONCLUSIONS: The study demonstrates beneficial effects of Empa on diastolic function, hemodynamics, inflammation, and cardiac remodeling in a rat model of HFpEF. These effects were mediated by improved mitochondrial respiratory capacity due to modulated cardiolipin and improved calcium handling.


Sujet(s)
Composés benzhydryliques , Diastole , Modèles animaux de maladie humaine , Glucosides , Défaillance cardiaque , Mitochondries du myocarde , Rat Zucker , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Débit systolique , Animaux , Glucosides/pharmacologie , Composés benzhydryliques/pharmacologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/physiopathologie , Défaillance cardiaque/métabolisme , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/ultrastructure , Diastole/effets des médicaments et des substances chimiques , Débit systolique/effets des médicaments et des substances chimiques , Mâle , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Rats de lignée SHR , Transport d'électrons/effets des médicaments et des substances chimiques , Rats
9.
Turk Kardiyol Dern Ars ; 52(4): 237-243, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38829634

RÉSUMÉ

OBJECTIVE: This study aims to explore the impact of sodium-glucose cotransporter-2 (SGLT-2) inhibitors, a newer class of oral antidiabetic drugs, on atrial electromechanical delay (EMD) in patients with type 2 diabetes mellitus (DM). This is particularly relevant given the significantly higher incidence of atrial fibrillation (AF) in diabetic patients compared to the general population. Atrial electromechanical delay is recognized as an important factor influencing the development of atrial fibrillation. METHODS: This study included 30 type 2 DM patients (53.3% female, mean age 60.07 ± 10.03 years), initiating treatment with SGLT-2 inhibitors. The patients were assessed using echocardiography at baseline and again at 6 months, focusing on basic echocardiographic parameters and atrial electromechanical delay times (EMD) measured via tissue Doppler imaging. RESULTS: No significant changes were observed in intra-atrial EMD times. However, significant reductions were noted in interatrial EMD times, decreasing from 15.13 ± 5.87 ms to 13.20 ± 6.12 ms (P = 0.029). Statistically significant shortening occurred in lateral pulmonary acceleration (PA) times (from 58.73 ± 6.41 ms to 54.37 ± 6.97 ms, P < 0.001), septal PA times (from 50.90 ± 6.02 ms to 48.23 ± 5), and tricuspid PA times (from 43.60 ± 6.28 ms to 41.30 ± 5.60 ms, P = 0.003). Additionally, there was a significant reduction in the E/e' ratio from 8.13 ± 4.0 to 6.50 ± 2.37 (P = 0.003). CONCLUSION: SGLT-2 inhibitors might positively influence atrial electromechanical conduction, reducing DM-related functional impairments and the risk of arrhythmias, particularly AF.


Sujet(s)
Fibrillation auriculaire , Diabète de type 2 , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Humains , Femelle , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Diabète de type 2/traitement médicamenteux , Diabète de type 2/complications , Adulte d'âge moyen , Mâle , Fibrillation auriculaire/traitement médicamenteux , Fibrillation auriculaire/physiopathologie , Atrium du coeur/imagerie diagnostique , Atrium du coeur/effets des médicaments et des substances chimiques , Atrium du coeur/physiopathologie , Sujet âgé , Système de conduction du coeur/effets des médicaments et des substances chimiques , Système de conduction du coeur/physiopathologie , Échocardiographie
10.
Physiol Rep ; 12(11): e16093, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38845596

RÉSUMÉ

Regular exercise and antihyperglycemic drugs are front-line treatments for type-2 diabetes and related metabolic disorders. Leading drugs are metformin, sodium-glucose cotransporter-2 inhibitors, and glucagon-like peptide 1 receptor agonists. Each class has strong individual efficacy to treat hyperglycemia, yet the combination with exercise can yield varied results, some of which include blunting of expected metabolic benefits. Skeletal muscle insulin resistance contributes to the development of type-2 diabetes while improvements in skeletal muscle insulin signaling are among key adaptations to exercise training. The current review identifies recent advances into the mechanisms, with an emphasis on skeletal muscle, of the interaction between exercise and these common antihyperglycemic drugs. The review is written toward researchers and thus highlights specific gaps in knowledge and considerations for future study directions.


Sujet(s)
Exercice physique , Hypoglycémiants , Muscles squelettiques , Humains , Muscles squelettiques/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Hypoglycémiants/pharmacologie , Hypoglycémiants/usage thérapeutique , Exercice physique/physiologie , Animaux , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Insulinorésistance/physiologie , Metformine/pharmacologie , Metformine/usage thérapeutique , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique
11.
J Cell Mol Med ; 28(11): e18364, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38837668

RÉSUMÉ

Diabetic kidney disease (DKD) is a leading cause of end stage renal disease with unmet clinical demands for treatment. Lipids are essential for cell survival; however, renal cells have limited capability to metabolize overloaded lipids. Dyslipidaemia is common in DKD patients and renal ectopic lipid accumulation is associated with disease progression. Unveiling the molecular mechanism involved in renal lipid regulation is crucial for exploring potential therapeutic targets. In this review, we focused on the mechanism underlying cholesterol, oxysterol and fatty acid metabolism disorder in the context of DKD. Specific regulators of lipid accumulation in different kidney compartment and TREM2 macrophages, a lipid-related macrophages in DKD, were discussed. The role of sodium-glucose transporter 2 inhibitors in improving renal lipid accumulation was summarized.


Sujet(s)
Néphropathies diabétiques , Rein , Métabolisme lipidique , Humains , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Animaux , Rein/métabolisme , Rein/anatomopathologie , Macrophages/métabolisme , Cholestérol/métabolisme , Acides gras/métabolisme , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Oxystérols/métabolisme , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique
12.
Sci Rep ; 14(1): 13802, 2024 06 14.
Article de Anglais | MEDLINE | ID: mdl-38877312

RÉSUMÉ

Sodium-glucose cotransporter (SGLT) 2 inhibition is a well-known target for the treatment of type 2 diabetes, renal disease and chronic heart failure. The protein SGLT2 is encoded by SLC5A2 (Solute Carrier Family 5 Member 2), which is highly expressed in renal cortex, but also in the testes where glucose uptake may be essential for spermatogenesis and androgen synthesis. We postulated that in healthy males, SGLT2 inhibitor therapy may affect gonadal function. We examined the impact on gonadal and steroid hormones in a post-hoc analysis of a double-blind, randomized, placebo-controlled research including 26 healthy males who were given either placebo or empagliflozin 10 mg once daily for four weeks. After one month of empagliflozin, there were no discernible changes in androgen, pituitary gonadotropin hormones, or inhibin B. Regardless of BMI category, the administration of empagliflozin, a highly selective SGLT2 inhibitor, did not alter serum androgen levels in men without diabetes. While SGLT2 is present in the testes, its inhibition does not seem to affect testosterone production in Leydig cells nor inhibin B secretion by the Sertoli cells.


Sujet(s)
Composés benzhydryliques , Glucosides , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Mâle , Humains , Composés benzhydryliques/pharmacologie , Glucosides/pharmacologie , Adulte , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Méthode en double aveugle , Testicule/métabolisme , Testicule/effets des médicaments et des substances chimiques , Testostérone/sang , Inhibines/sang , Inhibines/métabolisme , Adulte d'âge moyen , Transporteur-2 sodium-glucose/métabolisme , Androgènes/métabolisme , Cellules de Leydig/métabolisme , Cellules de Leydig/effets des médicaments et des substances chimiques , Cellules de Sertoli/métabolisme , Cellules de Sertoli/effets des médicaments et des substances chimiques
13.
Int J Mol Sci ; 25(11)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38891889

RÉSUMÉ

The sodium-glucose co-transporter-2 (SGLT2) inhibitor dapagliflozin is increasingly used in the treatment of diabetes and heart failure. Dapagliflozin has been associated with reduced incidence of atrial fibrillation (AF) in clinical trials. We hypothesized that the favorable antiarrhythmic outcome of dapagliflozin use may be caused in part by previously unrecognized effects on atrial repolarizing potassium (K+) channels. This study was designed to assess direct pharmacological effects of dapagliflozin on cloned ion channels Kv11.1, Kv1.5, Kv4.3, Kir2.1, K2P2.1, K2P3.1, and K2P17.1, contributing to IKur, Ito, IKr, IK1, and IK2P K+ currents. Human channels coded by KCNH2, KCNA5, KCND3, KCNJ2, KCNK2, KCNK3, and KCNK17 were heterologously expressed in Xenopus laevis oocytes, and currents were recorded using the voltage clamp technique. Dapagliflozin (100 µM) reduced Kv11.1 and Kv1.5 currents, whereas Kir2.1, K2P2.1, and K2P17.1 currents were enhanced. The drug did not significantly affect peak current amplitudes of Kv4.3 or K2P3.1 K+ channels. Biophysical characterization did not reveal significant effects of dapagliflozin on current-voltage relationships of study channels. In conclusion, dapagliflozin exhibits direct functional interactions with human atrial K+ channels underlying IKur, IKr, IK1, and IK2P currents. Substantial activation of K2P2.1 and K2P17.1 currents could contribute to the beneficial antiarrhythmic outcome associated with the drug. Indirect or chronic effects remain to be investigated in vivo.


Sujet(s)
Composés benzhydryliques , Glucosides , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Xenopus laevis , Humains , Glucosides/pharmacologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Composés benzhydryliques/pharmacologie , Animaux , Canaux potassiques/métabolisme , Ovocytes/métabolisme , Ovocytes/effets des médicaments et des substances chimiques , Transporteur-2 sodium-glucose/métabolisme , Transporteur-2 sodium-glucose/génétique
14.
Cardiovasc Diabetol ; 23(1): 199, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867314

RÉSUMÉ

BACKGROUND: Metformin and sodium-glucose-cotransporter-2 inhibitors (SGLT2i) are cornerstone therapies for managing hyperglycemia in diabetes. However, their detailed impacts on metabolic processes, particularly within the citric acid (TCA) cycle and its anaplerotic pathways, remain unclear. This study investigates the tissue-specific metabolic effects of metformin, both as a monotherapy and in combination with SGLT2i, on the TCA cycle and associated anaplerotic reactions in both mice and humans. METHODS: Metformin-specific metabolic changes were initially identified by comparing metformin-treated diabetic mice (MET) with vehicle-treated db/db mice (VG). These findings were then assessed in two human cohorts (KORA and QBB) and a longitudinal KORA study of metformin-naïve patients with Type 2 Diabetes (T2D). We also compared MET with db/db mice on combination therapy (SGLT2i + MET). Metabolic profiling analyzed 716 metabolites from plasma, liver, and kidney tissues post-treatment, using linear regression and Bonferroni correction for statistical analysis, complemented by pathway analyses to explore the pathophysiological implications. RESULTS: Metformin monotherapy significantly upregulated TCA cycle intermediates such as malate, fumarate, and α-ketoglutarate (α-KG) in plasma, and anaplerotic substrates including hepatic glutamate and renal 2-hydroxyglutarate (2-HG) in diabetic mice. Downregulated hepatic taurine was also observed. The addition of SGLT2i, however, reversed these effects, such as downregulating circulating malate and α-KG, and hepatic glutamate and renal 2-HG, but upregulated hepatic taurine. In human T2D patients on metformin therapy, significant systemic alterations in metabolites were observed, including increased malate but decreased citrulline. The bidirectional modulation of TCA cycle intermediates in mice influenced key anaplerotic pathways linked to glutaminolysis, tumorigenesis, immune regulation, and antioxidative responses. CONCLUSION: This study elucidates the specific metabolic consequences of metformin and SGLT2i on the TCA cycle, reflecting potential impacts on the immune system. Metformin shows promise for its anti-inflammatory properties, while the addition of SGLT2i may provide liver protection in conditions like metabolic dysfunction-associated steatotic liver disease (MASLD). These observations underscore the importance of personalized treatment strategies.


Sujet(s)
Cycle citrique , Diabète de type 2 , Hypoglycémiants , Rein , Foie , Metformine , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Metformine/pharmacologie , Animaux , Cycle citrique/effets des médicaments et des substances chimiques , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Humains , Hypoglycémiants/pharmacologie , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Diabète de type 2/sang , Mâle , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Rein/métabolisme , Rein/effets des médicaments et des substances chimiques , Femelle , Association de médicaments , Souris de lignée C57BL , Métabolomique , Marqueurs biologiques/sang , Adulte d'âge moyen , Glycémie/métabolisme , Glycémie/effets des médicaments et des substances chimiques , Études longitudinales , Souris , Sujet âgé , Résultat thérapeutique
17.
Pak J Pharm Sci ; 37(2): 337-347, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38767101

RÉSUMÉ

Heart failure is a condition in which the heart's one or both ventricles are unable to either receive an adequate amount of blood or eject an adequate amount of blood. Diabetes is considered one of the major risk factors for cardiovascular diseases. The current research is designed to evaluate the cardioprotective effects of dapagliflozin in streptozotocin and isoproterenol-induced comorbid rats. The COX-2, TNF-α, NF-КB, NLRP3, PPAR-γ, CKMB, TROP-I, AR, GP and SGLT were docked against dapagliflozin, propranolol and metformin. Dapagliflozin restored adequate blood flow and halted myofibril damage. Moreover, it's evident from this study that dapagliflozin significantly decreased serum concentration of various blood markers, decreased relative growth rate and QT interval prolongation, as compared to the negative control group. However, it improved the ventricular ejection fraction in rats of the treatment group. The GST, GSH and CAT levels were increased, as compared to normal. On the contrary, a decrease in LPO concentrations was observed. Evaluation of the coronal section of heart tissues showed the anti-inflammatory expressions evaluated through H & E staining and immunohistochemical techniques and with ELISA and PCR. In a nutshell, dapagliflozin reverses myocardial necrosis and apoptosis.


Sujet(s)
Composés benzhydryliques , Glucosides , Défaillance cardiaque , Isoprénaline , Protéine-3 de la famille des NLR contenant un domaine pyrine , Récepteur PPAR gamma , Transduction du signal , Streptozocine , Animaux , Glucosides/pharmacologie , Isoprénaline/toxicité , Défaillance cardiaque/induit chimiquement , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/métabolisme , Composés benzhydryliques/pharmacologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Récepteur PPAR gamma/métabolisme , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Rat Wistar , Diabète expérimental/traitement médicamenteux , Cardiotoniques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Myocarde/métabolisme , Myocarde/anatomopathologie
18.
Int J Mol Sci ; 25(9)2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38732178

RÉSUMÉ

Some of the most common conditions affecting people are kidney diseases. Among them, we distinguish chronic kidney disease and acute kidney injury. Both entities pose serious health risks, so new drugs are still being sought to treat and prevent them. In recent years, such a role has begun to be assigned to sodium-glucose cotransporter-2 (SGLT2) inhibitors. They increase the amount of glucose excreted in the urine. For this reason, they are currently used as a first-line drug in type 2 diabetes mellitus. Due to their demonstrated cardioprotective effect, they are also used in heart failure treatment. As for the renal effects of SGLT2 inhibitors, they reduce intraglomerular pressure and decrease albuminuria. This results in a slower decline in glomelular filtration rate (GFR) in patients with kidney disease. In addition, these drugs have anti-inflammatory and antifibrotic effects. In the following article, we review the evidence for the effectiveness of this group of drugs in kidney disease and their nephroprotective effect. Further research is still needed, but meta-analyses indicate SGLT2 inhibitors' efficacy in kidney disease, especially the one caused by diabetes. Development of new drugs and clinical trials on specific patient subgroups will further refine their nephroprotective effects.


Sujet(s)
Diabète de type 2 , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Humains , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/métabolisme , Débit de filtration glomérulaire/effets des médicaments et des substances chimiques , Maladies du rein/traitement médicamenteux , Animaux
19.
Cell Mol Biol Lett ; 29(1): 80, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38811901

RÉSUMÉ

BACKGROUND: Sodium-glucose transporter 2 (SGLT2) inhibitors (iSGLT2) are approved medications for type 2 diabetes. Recent studies indicate that iSGLT2 inhibit the growth of some cancer cells. However, the mechanism(s) remains to be fully elucidated. METHODS: The SGLT2 levels were determined in normal colon CCD 841 CoN and, HCT 116, HT-29, SW480 and LoVo colorectal cancer (CRC) cell lines by quantitative real-time PCR and western blot. The effect of iSGLT2 canagliflozin on cell proliferation was examined using CCK-8, as its role on CRC cells metabolism and tumorigenesis has been evaluated by XF HS Seahorse Bioanalyzer and flow cytometric analyses. Transient gene silencing experiments and analysis of protein-protein interaction network were conducted to evaluate the SGLT2 molecular targets in CRC cells. RESULTS: Data showed that the treatment with iSGLT2 (50 µM) for 72 h induced cell cycle arrest (p < 0.001), impaired glucose and energetic metabolism (p < 0.001), promoted apoptotic cell death and ER stress flowing into autophagy (p < 0.001) in HCT 116 and HT-29 cells. These cellular events were accompanied by sirtuin 3 (SIRT3) upregulation (p < 0.01), as also supported by SIRT3 transient silencing experiments resulting in the attenuation of the effects of iSGLT2 on the cellular metabolic/energetic alterations and the induction of programmed cell death. The identification and validation of dipeptidyl peptidase 4 (DPP4) as potential common target of SGLT2 and SIRT3 were also assessed. CONCLUSIONS: These results deepened knowledge on the iSGLT2 contribution in limiting CRC tumorigenesis unveiling the SGLT2/SIRT3 axis in the cytotoxic mechanisms.


Sujet(s)
Apoptose , Prolifération cellulaire , Tumeurs colorectales , Stress du réticulum endoplasmique , Mitochondries , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Transporteur-2 sodium-glucose , Humains , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transporteur-2 sodium-glucose/métabolisme , Transporteur-2 sodium-glucose/génétique , Apoptose/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Canagliflozine/pharmacologie , Cellules HT29 , Cellules HCT116 , Sirtuine-3/métabolisme , Sirtuine-3/génétique , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Glucose/métabolisme
20.
Free Radic Biol Med ; 220: 288-300, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38734268

RÉSUMÉ

A tumour suppressor miRNA, miR-128-3p, is widely involved in various biological processes and has been found to get downregulated in breast cancer patients. We previously published that ectopically expressed miR-128-3p suppressed migration, invasion, cell cycle arrest, and breast cancer stem cells. In the present study, we explored the role of Empagliflozin (EMPA) as a miR-128-3p functionality-mimicking drug in inducing ferroptosis by inhibiting CD98hc. Given that CD98hc is one of the proteins critical in triggering ferroptosis, we confirmed that miR-128-3p and EMPA inhibited SP1, leading to inhibition of CD98hc expression. Further, transfection with siCD98hc, miR-128-3p mimics, and inhibitors was performed to assess their involvement in the ferroptosis of anoikis-resistant cells. We proved that anoikis-resistant cells possess high ROS and iron levels. Further, miR-128-3p and EMPA treatments induced ferroptosis by inhibiting GSH and enzymatic activity of GPX4 and also induced lipid peroxidation. Moreover, EMPA suppressed bioluminescence of 4T1-Red-FLuc induced thoracic cavity, peritoneal tumour burden and lung nodules in an in-vivo metastatic model of breast cancer. Collectively, we revealed that EMPA sensitized the ECM detached cells to ferroptosis by synergically activating miR-128-3p and lowering the levels of SP1 and CD98hc, making it a potential adjunct drug for breast cancer chemotherapy.


Sujet(s)
Anoïkis , Composés benzhydryliques , Tumeurs du sein , Ferroptose , Régulation de l'expression des gènes tumoraux , Glucosides , microARN , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/génétique , microARN/génétique , microARN/métabolisme , Humains , Femelle , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Glucosides/pharmacologie , Animaux , Anoïkis/effets des médicaments et des substances chimiques , Anoïkis/génétique , Souris , Composés benzhydryliques/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Espèces réactives de l'oxygène/métabolisme , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Peroxydation lipidique/effets des médicaments et des substances chimiques , Cotransporteurs sodium-phosphate de type IIb
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...