Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 581
Filtrer
1.
BMC Cardiovasc Disord ; 24(1): 333, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961333

RÉSUMÉ

BACKGROUND: Oxidative stress may contribute to cardiac ryanodine receptor (RyR2) dysfunction in diabetic cardiomyopathy. Ginsenoside Rb1 (Rb1) is a major pharmacologically active component of ginseng to treat cardiovascular diseases. Whether Rb1 treat diabetes injured heart remains unknown. This study was to investigate the effect of Rb1 on diabetes injured cardiac muscle tissue and to further investigate its possible molecular pharmacology mechanisms. METHODS: Male Sprague-Dawley rats were injected streptozotocin solution for 2 weeks, followed 6 weeks Rb1 or insulin treatment. The activity of SOD, CAT, Gpx, and the levels of MDA was measured; histological and ultrastructure analyses, RyR2 activity and phosphorylated RyR2(Ser2808) protein expression analyses; and Tunel assay were performed. RESULTS: There was decreased activity of SOD, CAT, Gpx and increased levels of MDA in the diabetic group from control. Rb1 treatment increased activity of SOD, CAT, Gpx and decreased the levels of MDA as compared with diabetic rats. Neutralizing the RyR2 activity significantly decreased in diabetes from control, and increased in Rb1 treatment group from diabetic group. The expression of phosphorylation of RyR2 Ser2808 was increased in diabetic rats from control, and were attenuated with insulin and Rb1 treatment. Diabetes increased the apoptosis rate, and Rb1 treatment decreased the apoptosis rate. Rb1 and insulin ameliorated myocardial injury in diabetic rats. CONCLUSIONS: These data indicate that Rb1 could be useful for mitigating oxidative damage, reduced phosphorylation of RyR2 Ser2808 and decreased the apoptosis rate of cardiomyocytes in diabetic cardiomyopathy.


Sujet(s)
Antioxydants , Apoptose , Diabète expérimental , Cardiomyopathies diabétiques , Ginsénosides , Myocytes cardiaques , Stress oxydatif , Rat Sprague-Dawley , Canal de libération du calcium du récepteur à la ryanodine , Streptozocine , Animaux , Diabète expérimental/traitement médicamenteux , Mâle , Stress oxydatif/effets des médicaments et des substances chimiques , Canal de libération du calcium du récepteur à la ryanodine/métabolisme , Canal de libération du calcium du récepteur à la ryanodine/effets des médicaments et des substances chimiques , Ginsénosides/pharmacologie , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/traitement médicamenteux , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/physiopathologie , Cardiomyopathies diabétiques/étiologie , Apoptose/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Phosphorylation , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/métabolisme , Myocarde/anatomopathologie , Myocarde/métabolisme , Insuline , Malonaldéhyde/métabolisme
2.
Cardiovasc Diabetol ; 23(1): 217, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38915040

RÉSUMÉ

BACKGROUND: Diabetic peripheral neuropathy (DPN) is the most prevalent complication of diabetes, and has been demonstrated to be independently associated with cardiovascular events and mortality. This aim of this study was to investigate the subclinical left ventricular (LV) myocardial dysfunction in type 2 diabetes mellitus (T2DM) patients with and without DPN. METHODS: One hundred and thirty T2DM patients without DPN, 61 patients with DPN and 65 age and sex-matched controls who underwent cardiovascular magnetic resonance (CMR) imaging were included, all subjects had no symptoms of heart failure and LV ejection fraction ≥ 50%. LV myocardial non-infarct late gadolinium enhancement (LGE) was determined. LV global strains, including radial, circumferential and longitudinal peak strain (PS) and peak systolic and diastolic strain rates (PSSR and PDSR, respectively), were evaluated using CMR feature tracking and compared among the three groups. Multivariable linear regression analyses were performed to determine the independent factors of reduced LV global myocardial strains in T2DM patients. RESULTS: The prevalence of non-infarct LGE was higher in patients with DPN than those without DPN (37.7% vs. 19.2%, p = 0.008). The LV radial and longitudinal PS (radial: 36.60 ± 7.24% vs. 33.57 ± 7.30% vs. 30.72 ± 8.68%; longitudinal: - 15.03 ± 2.52% vs. - 13.39 ± 2.48% vs. - 11.89 ± 3.02%), as well as longitudinal PDSR [0.89 (0.76, 1.05) 1/s vs. 0.80 (0.71, 0.93) 1/s vs. 0.77 (0.63, 0.87) 1/s] were decreased significantly from controls through T2DM patients without DPN to patients with DPN (all p < 0.001). LV radial and circumferential PDSR, as well as circumferential PS were reduced in both patient groups (all p < 0.05), but were not different between the two groups (all p > 0.05). Radial and longitudinal PSSR were decreased in patients with DPN (p = 0.006 and 0.003, respectively) but preserved in those without DPN (all p > 0.05). Multivariable linear regression analyses adjusting for confounders demonstrated that DPN was independently associated with LV radial and longitudinal PS (ß = - 3.025 and 1.187, p = 0.014 and 0.003, respectively) and PDSR (ß = 0.283 and - 0.086, p = 0.016 and 0.001, respectively), as well as radial PSSR (ß = - 0.266, p = 0.007). CONCLUSIONS: There was more severe subclinical LV dysfunction in T2DM patients complicated with DPN than those without DPN, suggesting further prospective study with more active intervention in this cohort of patients.


Sujet(s)
Maladies asymptomatiques , Diabète de type 2 , Cardiomyopathies diabétiques , Neuropathies diabétiques , IRM dynamique , Valeur prédictive des tests , Dysfonction ventriculaire gauche , Fonction ventriculaire gauche , Humains , Mâle , Femelle , Adulte d'âge moyen , Dysfonction ventriculaire gauche/physiopathologie , Dysfonction ventriculaire gauche/imagerie diagnostique , Dysfonction ventriculaire gauche/étiologie , Diabète de type 2/complications , Diabète de type 2/diagnostic , Neuropathies diabétiques/physiopathologie , Neuropathies diabétiques/imagerie diagnostique , Neuropathies diabétiques/étiologie , Neuropathies diabétiques/diagnostic , Neuropathies diabétiques/épidémiologie , Sujet âgé , Études cas-témoins , Cardiomyopathies diabétiques/physiopathologie , Cardiomyopathies diabétiques/imagerie diagnostique , Cardiomyopathies diabétiques/étiologie , Facteurs de risque , Prévalence , Études transversales , Débit systolique , Contraction myocardique
3.
Cardiovasc Diabetol ; 23(1): 202, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867293

RÉSUMÉ

The specific pathophysiological pathways through which diabetes exacerbates myocardial ischemia/reperfusion (I/R) injury remain unclear; however, dysregulation of immune and inflammatory cells, potentially driven by abnormalities in their number and function due to diabetes, may play a significant role. In the present investigation, we simulated myocardial I/R injury by inducing ischemia through ligation of the left anterior descending coronary artery in mice for 40 min, followed by reperfusion for 24 h. Previous studies have indicated that protein kinase Cß (PKCß) is upregulated under hyperglycemic conditions and is implicated in the development of various diabetic complications. The Y4 RNA fragment is identified as the predominant small RNA component present in the extracellular vesicles of cardio sphere-derived cells (CDCs), exhibiting notable anti-inflammatory properties in the contexts of myocardial infarction and cardiac hypertrophy. Our investigation revealed that the administration of Y4 RNA into the ventricular cavity of db/db mice following myocardial I/R injury markedly enhanced cardiac function. Furthermore, Y4 RNA was observed to facilitate M2 macrophage polarization and interleukin-10 secretion through the suppression of PKCß activation. The mechanism by which Y4 RNA affects PKCß by regulating macrophage activation within the inflammatory environment involves the inhibition of ERK1/2 phosphorylation In our study, the role of PKCß in regulating macrophage polarization during myocardial I/R injury was investigated through the use of PKCß knockout mice. Our findings indicate that PKCß plays a crucial role in modulating the inflammatory response associated with macrophage activation in db/db mice experiencing myocardial I/R, with a notable exacerbation of this response observed upon significant upregulation of PKCß expression. In vitro studies further elucidated the protective mechanism by which Y4 RNA modulates the PKCß/ERK1/2 signaling pathway to induce M2 macrophage activation. Overall, our findings suggest that Y4 RNA plays an anti-inflammatory role in diabetic I/R injury, suggesting a novel therapeutic approach for managing myocardial I/R injury in diabetic individuals.


Sujet(s)
Modèles animaux de maladie humaine , Macrophages , Souris de lignée C57BL , Lésion de reperfusion myocardique , Protein kinase C beta , Transduction du signal , Animaux , Protein kinase C beta/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Lésion de reperfusion myocardique/enzymologie , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/prévention et contrôle , Lésion de reperfusion myocardique/génétique , Macrophages/métabolisme , Macrophages/enzymologie , Mâle , Interleukine-10/métabolisme , Interleukine-10/génétique , Souris , Cardiomyopathies diabétiques/enzymologie , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/étiologie , Cardiomyopathies diabétiques/génétique , Cardiomyopathies diabétiques/physiopathologie , Cellules cultivées , Phénotype , Myocytes cardiaques/enzymologie , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/métabolisme , Mitogen-Activated Protein Kinase 3/métabolisme , Activation des macrophages , Mitogen-Activated Protein Kinase 1/métabolisme , Fonction ventriculaire gauche , Phosphorylation
4.
Cardiovasc Diabetol ; 23(1): 197, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849829

RÉSUMÉ

OBJECTIVE: Sodium glucose cotransporter 2 (SGLT2) inhibitors significantly improve cardiovascular outcomes in diabetic patients; however, the mechanism is unclear. We hypothesized that dapagliflozin improves cardiac outcomes via beneficial effects on systemic and cardiac inflammation and cardiac fibrosis. RESEARCH AND DESIGN METHODS: This randomized placebo-controlled clinical trial enrolled 62 adult patients (mean age 62, 17% female) with type 2 diabetes (T2D) without known heart failure. Subjects were randomized to 12 months of daily 10 mg dapagliflozin or placebo. For all patients, blood/plasma samples and cardiac magnetic resonance imaging (CMRI) were obtained at time of randomization and at the end of 12 months. Systemic inflammation was assessed by plasma IL-1B, TNFα, IL-6 and ketone levels and PBMC mitochondrial respiration, an emerging marker of sterile inflammation. Global myocardial strain was assessed by feature tracking; cardiac fibrosis was assessed by T1 mapping to calculate extracellular volume fraction (ECV); and cardiac tissue inflammation was assessed by T2 mapping. RESULTS: Between the baseline and 12-month time point, plasma IL-1B was reduced (- 1.8 pg/mL, P = 0.003) while ketones were increased (0.26 mM, P = 0.0001) in patients randomized to dapagliflozin. PBMC maximal oxygen consumption rate (OCR) decreased over the 12-month period in the placebo group but did not change in patients receiving dapagliflozin (- 158.9 pmole/min/106 cells, P = 0.0497 vs. - 5.2 pmole/min/106 cells, P = 0.41), a finding consistent with an anti-inflammatory effect of SGLT2i. Global myocardial strain, ECV and T2 relaxation time did not change in both study groups. GOV REGISTRATION: NCT03782259.


Sujet(s)
Composés benzhydryliques , Marqueurs biologiques , Diabète de type 2 , Glucosides , Médiateurs de l'inflammation , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Humains , Composés benzhydryliques/usage thérapeutique , Composés benzhydryliques/effets indésirables , Glucosides/usage thérapeutique , Glucosides/effets indésirables , Femelle , Diabète de type 2/traitement médicamenteux , Diabète de type 2/diagnostic , Diabète de type 2/sang , Diabète de type 2/complications , Mâle , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Inhibiteurs du cotransporteur sodium-glucose de type 2/effets indésirables , Adulte d'âge moyen , Sujet âgé , Résultat thérapeutique , Médiateurs de l'inflammation/sang , Marqueurs biologiques/sang , Facteurs temps , Anti-inflammatoires/usage thérapeutique , Fibrose , Inflammation/traitement médicamenteux , Inflammation/sang , Inflammation/diagnostic , Méthode en double aveugle , Myocarde/anatomopathologie , Myocarde/métabolisme , Cardiomyopathies diabétiques/étiologie , Cardiomyopathies diabétiques/prévention et contrôle , Cardiomyopathies diabétiques/imagerie diagnostique , Cardiomyopathies diabétiques/traitement médicamenteux , Cardiomyopathies diabétiques/sang
5.
Sci Rep ; 14(1): 12978, 2024 06 05.
Article de Anglais | MEDLINE | ID: mdl-38839927

RÉSUMÉ

Diabetic cardiomyopathy is a specific type of cardiomyopathy. In DCM, glucose uptake and utilization are impaired due to insulin deficiency or resistance, and the heart relies more heavily on fatty acid oxidation for energy, resulting in myocardial lipid toxicity-related injury. MARK4 is a member of the AMPK-related kinase family, and improves ischaemic heart failure through microtubule detyrosination. However, the role of MARK4 in cardiac regulation of metabolism is unclear. In this study, after successful establishment of a diabetic cardiomyopathy model induced by streptozotocin and a high-fat diet, MARK4 expression was found to be significantly increased in STZ-induced DCM mice. After AAV9-shMARK4 was administered through the tail vein, decreased expression of MARK4 alleviated diabetic myocardial damage, reduced oxidative stress and apoptosis, and facilitated cardiomyocyte mitochondrial fusion, and promoted myocardial lipid oxidation metabolism. In addition, through the RNA-seq analysis of differentially expressed genes, we found that MARK4 deficiency promoted lipid decomposition and oxidative metabolism by downregulating the expression of ACSL4, thus reducing myocardial lipid accumulation in the STZ-induced DCM model.


Sujet(s)
Coenzyme A ligases , Cardiomyopathies diabétiques , Métabolisme lipidique , Myocarde , Animaux , Mâle , Souris , Apoptose , Coenzyme A ligases/métabolisme , Coenzyme A ligases/génétique , Diabète expérimental/métabolisme , Diabète expérimental/complications , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/génétique , Cardiomyopathies diabétiques/étiologie , Modèles animaux de maladie humaine , Souris de lignée C57BL , Myocarde/métabolisme , Myocarde/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Stress oxydatif , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Streptozocine
6.
Cardiovasc Diabetol ; 23(1): 169, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750502

RÉSUMÉ

Diabetic heart disease (DHD) is a serious complication in patients with diabetes. Despite numerous studies on the pathogenic mechanisms and therapeutic targets of DHD, effective means of prevention and treatment are still lacking. The pathogenic mechanisms of DHD include cardiac inflammation, insulin resistance, myocardial fibrosis, and oxidative stress. Macrophages, the primary cells of the human innate immune system, contribute significantly to these pathological processes, playing an important role in human disease and health. Therefore, drugs targeting macrophages hold great promise for the treatment of DHD. In this review, we examine how macrophages contribute to the development of DHD and which drugs could potentially be used to target macrophages in the treatment of DHD.


Sujet(s)
Cardiomyopathies diabétiques , Macrophages , Stress oxydatif , Transduction du signal , Humains , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Macrophages/métabolisme , Cardiomyopathies diabétiques/immunologie , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/traitement médicamenteux , Cardiomyopathies diabétiques/étiologie , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Fibrose , Anti-inflammatoires/usage thérapeutique , Myocarde/anatomopathologie , Myocarde/métabolisme , Myocarde/immunologie , Insulinorésistance , Médiateurs de l'inflammation/métabolisme , Thérapie moléculaire ciblée
7.
Front Immunol ; 15: 1393392, 2024.
Article de Anglais | MEDLINE | ID: mdl-38774880

RÉSUMÉ

This review provides a comprehensive analysis of the critical role played by macrophages and their underlying mechanisms in the progression of diabetic cardiomyopathy (DCM). It begins by discussing the origins and diverse subtypes of macrophages, elucidating their spatial distribution and modes of intercellular communication, thereby emphasizing their significance in the pathogenesis of DCM. The review then delves into the intricate relationship between macrophages and the onset of DCM, particularly focusing on the epigenetic regulatory mechanisms employed by macrophages in the context of DCM condition. Additionally, the review discusses various therapeutic strategies aimed at targeting macrophages to manage DCM. It specifically highlights the potential of natural food components in alleviating diabetic microvascular complications and examines the modulatory effects of existing hypoglycemic drugs on macrophage activity. These findings, summarized in this review, not only provide fresh insights into the role of macrophages in diabetic microvascular complications but also offer valuable guidance for future therapeutic research and interventions in this field.


Sujet(s)
Cardiomyopathies diabétiques , Macrophages , Animaux , Humains , Cardiomyopathies diabétiques/immunologie , Cardiomyopathies diabétiques/traitement médicamenteux , Cardiomyopathies diabétiques/étiologie , Cardiomyopathies diabétiques/anatomopathologie , Hypoglycémiants/usage thérapeutique , Macrophages/immunologie , Macrophages/métabolisme
8.
Cardiovasc Diabetol ; 23(1): 160, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38715043

RÉSUMÉ

BACKGROUND: Diabetic cardiomyopathy (DCM) is a crucial complication of long-term chronic diabetes that can lead to myocardial hypertrophy, myocardial fibrosis, and heart failure. There is increasing evidence that DCM is associated with pyroptosis, a form of inflammation-related programmed cell death. Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor ß superfamily, which regulates oxidative stress, inflammation, and cell survival to mitigate myocardial hypertrophy, myocardial infarction, and vascular injury. However, the role of GDF11 in regulating pyroptosis in DCM remains to be elucidated. This research aims to investigate the role of GDF11 in regulating pyroptosis in DCM and the related mechanism. METHODS AND RESULTS: Mice were injected with streptozotocin (STZ) to induce a diabetes model. H9c2 cardiomyocytes were cultured in high glucose (50 mM) to establish an in vitro model of diabetes. C57BL/6J mice were preinjected with adeno-associated virus 9 (AAV9) intravenously via the tail vein to specifically overexpress myocardial GDF11. GDF11 attenuated pyroptosis in H9c2 cardiomyocytes after high-glucose treatment. In diabetic mice, GDF11 alleviated cardiomyocyte pyroptosis, reduced myocardial fibrosis, and improved cardiac function. Mechanistically, GDF11 inhibited pyroptosis by preventing inflammasome activation. GDF11 achieved this by specifically binding to apoptosis-associated speck-like protein containing a CARD (ASC) and preventing the assembly and activation of the inflammasome. Additionally, the expression of GDF11 during pyroptosis was regulated by peroxisome proliferator-activated receptor α (PPARα). CONCLUSION: These findings demonstrate that GDF11 can treat diabetic cardiomyopathy by alleviating pyroptosis and reveal the role of the PPARα-GDF11-ASC pathway in DCM, providing ideas for new strategies for cardioprotection.


Sujet(s)
Diabète expérimental , Cardiomyopathies diabétiques , Fibrose , Facteurs de croissance et de différenciation , Inflammasomes , Souris de lignée C57BL , Myocytes cardiaques , Pyroptose , Transduction du signal , Animaux , Pyroptose/effets des médicaments et des substances chimiques , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/prévention et contrôle , Cardiomyopathies diabétiques/étiologie , Cardiomyopathies diabétiques/physiopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Diabète expérimental/métabolisme , Lignée cellulaire , Inflammasomes/métabolisme , Mâle , Facteurs de croissance et de différenciation/métabolisme , Rats , Glycémie/métabolisme , Souris , Glucose/métabolisme , Glucose/toxicité , Protéines morphogénétiques osseuses , Récepteur PPAR alpha
9.
Am J Chin Med ; 52(3): 841-864, 2024.
Article de Anglais | MEDLINE | ID: mdl-38716618

RÉSUMÉ

A high-glucose environment is involved in the progression of diabetes mellitus (DM). This study aims to explore the regulatory effects of quercetin (QUE) on autophagy and apoptosis after myocardial injury in rats with DM. The type 2 DM rat models were constructed using low-dose streptozotocin (STZ) treatment combined with a high-carbohydrate (HC) diet in vivo. Compared with the control group, the body weight was decreased, whereas blood pressure, blood glucose, and the LVW/BW ratio were increased in the diabetic group. The results showed that the myocardial fibers were disordered in the diabetic group. Moreover, we found that the myocardial collagen fibers, PAS-positive cells, and apoptosis were increased, whereas the mitochondrial structure was destroyed and autophagic vacuoles were significantly reduced in the diabetic group compared with the control group. The expression levels of autophagy-related proteins LC3 and Beclin1 were decreased, whereas the expression levels of P62, Caspae-3, and Bax/Bcl-2 were increased in the diabetic group in vitro and in vivo. Moreover, QUE treatment alleviated the cellular oxidative stress reaction under high-glucose environments. The results of immunoprecipitation (IP) showed that the autophagy protein Beclin1 was bound to Bcl-2, and the binding capacity increased in the HG group, whereas it decreased after QUE treatment, suggesting that QUE inhibited the binding capacity between Beclin1 and Bcl-2, thus leading to the preservation of Beclin1-induced autophagy. In addition, the blood pressure, blood glucose, and cardiac function of rats were improved following QUE treatment. In conclusion, QUE suppressed diabetic myocardial injury and ameliorated cardiac function by regulating myocardial autophagy and inhibition of apoptosis in diabetes through the AMPK/mTOR signaling pathway.


Sujet(s)
AMP-Activated Protein Kinases , Apoptose , Autophagie , Diabète expérimental , Quercétine , Transduction du signal , Sérine-thréonine kinases TOR , Animaux , Autophagie/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Quercétine/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Diabète expérimental/traitement médicamenteux , Diabète expérimental/métabolisme , Mâle , AMP-Activated Protein Kinases/métabolisme , Rat Sprague-Dawley , Rats , Modèles animaux de maladie humaine , Myocarde/métabolisme , Myocarde/anatomopathologie , Streptozocine , Cardiomyopathies diabétiques/étiologie , Cardiomyopathies diabétiques/traitement médicamenteux , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/prévention et contrôle , Phytothérapie , Bécline-1/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Diabète de type 2/complications
10.
Cardiovasc Diabetol ; 23(1): 164, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38724987

RÉSUMÉ

Dynamin-related protein 1 (Drp1) is a crucial regulator of mitochondrial dynamics, the overactivation of which can lead to cardiovascular disease. Multiple distinct posttranscriptional modifications of Drp1 have been reported, among which S-nitrosylation was recently introduced. However, the detailed regulatory mechanism of S-nitrosylation of Drp1 (SNO-Drp1) in cardiac microvascular dysfunction in diabetes remains elusive. The present study revealed that mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) was consistently upregulated in diabetic cardiomyopathy (DCM) and promoted SNO-Drp1 in cardiac microvascular endothelial cells (CMECs), which in turn led to mitochondrial dysfunction and cardiac microvascular disorder. Further studies confirmed that MAP4K4 promoted SNO-Drp1 at human C644 (mouse C650) by inhibiting glutathione peroxidase 4 (GPX4) expression, through which MAP4K4 stimulated endothelial ferroptosis in diabetes. In contrast, inhibition of MAP4K4 via DMX-5804 significantly reduced endothelial ferroptosis, alleviated cardiac microvascular dysfunction and improved cardiac dysfunction in db/db mice by reducing SNO-Drp1. In parallel, the C650A mutation in mice abolished SNO-Drp1 and the role of Drp1 in promoting cardiac microvascular disorder and cardiac dysfunction. In conclusion, our findings demonstrate that MAP4K4 plays an important role in endothelial dysfunction in DCM and reveal that SNO-Drp1 and ferroptosis activation may act as downstream targets, representing potential therapeutic targets for DCM.


Sujet(s)
Cardiomyopathies diabétiques , Dynamines , Cellules endothéliales , Transduction du signal , Animaux , Humains , Mâle , Souris , Cellules cultivées , Circulation coronarienne , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/génétique , Cardiomyopathies diabétiques/physiopathologie , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/enzymologie , Cardiomyopathies diabétiques/étiologie , Modèles animaux de maladie humaine , Dynamines/métabolisme , Dynamines/génétique , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Cellules endothéliales/enzymologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Ferroptose/effets des médicaments et des substances chimiques , Protéines et peptides de signalisation intracellulaire , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/anatomopathologie , Mitochondries du myocarde/enzymologie , Maturation post-traductionnelle des protéines , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique
11.
Int J Mol Sci ; 25(9)2024 May 05.
Article de Anglais | MEDLINE | ID: mdl-38732253

RÉSUMÉ

Diabetes mellitus (DM) is known as the first non-communicable global epidemic. It is estimated that 537 million people have DM, but the condition has been properly diagnosed in less than half of these patients. Despite numerous preventive measures, the number of DM cases is steadily increasing. The state of chronic hyperglycaemia in the body leads to numerous complications, including diabetic cardiomyopathy (DCM). A number of pathophysiological mechanisms are behind the development and progression of cardiomyopathy, including increased oxidative stress, chronic inflammation, increased synthesis of advanced glycation products and overexpression of the biosynthetic pathway of certain compounds, such as hexosamine. There is extensive research on the treatment of DCM, and there are a number of therapies that can stop the development of this complication. Among the compounds used to treat DCM are antiglycaemic drugs, hypoglycaemic drugs and drugs used to treat myocardial failure. An important element in combating DCM that should be kept in mind is a healthy lifestyle-a well-balanced diet and physical activity. There is also a group of compounds-including coenzyme Q10, antioxidants and modulators of signalling pathways and inflammatory processes, among others-that are being researched continuously, and their introduction into routine therapies is likely to result in greater control and more effective treatment of DM in the future. This paper summarises the latest recommendations for lifestyle and pharmacological treatment of cardiomyopathy in patients with DM.


Sujet(s)
Cardiomyopathies diabétiques , Humains , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/thérapie , Cardiomyopathies diabétiques/traitement médicamenteux , Cardiomyopathies diabétiques/étiologie , Hypoglycémiants/usage thérapeutique , Stress oxydatif , Antioxydants/usage thérapeutique , Diabète/métabolisme , Diabète/traitement médicamenteux , Animaux
12.
Int J Med Sci ; 21(7): 1194-1203, 2024.
Article de Anglais | MEDLINE | ID: mdl-38818468

RÉSUMÉ

This study aims to elucidate the roles of Phosphoglycerate Mutase Family Member 5 (Pgam5) and Prohibitin 2 (Phb2) in the context of hyperglycemia-induced myocardial dysfunction, a critical aspect of diabetic cardiomyopathy. The research employed primary cardiomyocytes, which were then subjected to hyperglycemia treatment to mimic diabetic conditions. We used siRNA transfection to knock down Pgam5 and overexpressed Phb2 using adenovirus transfection to assess their individual and combined effects on cardiomyocyte health. Mitochondrial function was evaluated through measurements of mitochondrial membrane potential using the JC-1 probe, and levels of mitochondrial reactive oxygen species (ROS) were assessed. Additionally, the study involved qPCR analysis to quantify the transcriptional changes in genes related to mitochondrial fission and mitophagy. Our findings indicate that hyperglycemia significantly reduces cardiomyocyte viability and impairs mitochondrial function, as evidenced by decreased mitochondrial membrane potential and increased ROS levels. Pgam5 knockdown was observed to mitigate these adverse effects, preserving mitochondrial function and cardiomyocyte viability. On the molecular level, Pgam5 was found to regulate genes associated with mitochondrial fission (such as Drp1, Mff, and Fis1) and mitophagy (including Parkin, Bnip3, and Fundc1). Furthermore, overexpression of Phb2 countered the hyperglycemia-induced mitochondrial dysfunction and normalized the levels of key mitochondrial antioxidant enzymes. The combined data suggest a protective role for both Pgam5 knockdown and Phb2 overexpression against hyperglycemia-induced cellular and mitochondrial damage. The study elucidates the critical roles of Pgam5 and Phb2 in regulating mitochondrial dynamics in the setting of hyperglycemia-induced myocardial dysfunction. By modulating mitochondrial fission and mitophagy, Pgam5 and Phb2 emerge as key players in preserving mitochondrial integrity and cardiomyocyte health under diabetic conditions. These findings contribute significantly to our understanding of the molecular mechanisms underlying diabetic cardiomyopathy and suggest potential therapeutic targets for mitigating myocardial dysfunction in diabetes.


Sujet(s)
Cardiomyopathies diabétiques , Hyperglycémie , Potentiel de membrane mitochondriale , Dynamique mitochondriale , Myocytes cardiaques , Prohibitines , Espèces réactives de l'oxygène , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Dynamique mitochondriale/génétique , Hyperglycémie/métabolisme , Hyperglycémie/complications , Hyperglycémie/génétique , Humains , Cardiomyopathies diabétiques/génétique , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/étiologie , Espèces réactives de l'oxygène/métabolisme , Animaux , Mitophagie/génétique , Phosphoprotein Phosphatases/génétique , Phosphoprotein Phosphatases/métabolisme , Protéines de répression/génétique , Protéines de répression/métabolisme , Mitochondries du myocarde/métabolisme , Protéines mitochondriales/génétique , Protéines mitochondriales/métabolisme , Rats
13.
Cardiovasc Diabetol ; 23(1): 116, 2024 Apr 02.
Article de Anglais | MEDLINE | ID: mdl-38566123

RÉSUMÉ

BACKGROUND: Diabetic cardiomyopathy (DCM) is a serious complication in patients with type 1 diabetes mellitus (T1DM), which still lacks adequate therapy. Irisin, a cleavage peptide off fibronectin type III domain-containing 5, has been shown to preserve cardiac function in cardiac ischemia-reperfusion injury. Whether or not irisin plays a cardioprotective role in DCM is not known. METHODS AND RESULTS: T1DM was induced by multiple low-dose intraperitoneal injections of streptozotocin (STZ). Our current study showed that irisin expression/level was lower in the heart and serum of mice with STZ-induced TIDM. Irisin supplementation by intraperitoneal injection improved the impaired cardiac function in mice with DCM, which was ascribed to the inhibition of ferroptosis, because the increased ferroptosis, associated with increased cardiac malondialdehyde (MDA), decreased reduced glutathione (GSH) and protein expressions of solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4), was ameliorated by irisin. In the presence of erastin, a ferroptosis inducer, the irisin-mediated protective effects were blocked. Mechanistically, irisin treatment increased Sirtuin 1 (SIRT1) and decreased p53 K382 acetylation, which decreased p53 protein expression by increasing its degradation, consequently upregulated SLC7A11 and GPX4 expressions. Thus, irisin-mediated reduction in p53 decreases ferroptosis and protects cardiomyocytes against injury due to high glucose. CONCLUSION: This study demonstrated that irisin could improve cardiac function by suppressing ferroptosis in T1DM via the SIRT1-p53-SLC7A11/GPX4 pathway. Irisin may be a therapeutic approach in the management of T1DM-induced cardiomyopathy.


Sujet(s)
Diabète de type 1 , Cardiomyopathies diabétiques , Ferroptose , Humains , Animaux , Souris , Cardiomyopathies diabétiques/traitement médicamenteux , Cardiomyopathies diabétiques/étiologie , Cardiomyopathies diabétiques/prévention et contrôle , Sirtuine-1 , Fibronectines , Diabète de type 1/complications , Diabète de type 1/traitement médicamenteux , Protéine p53 suppresseur de tumeur , Myocytes cardiaques
14.
Biomed Pharmacother ; 174: 116589, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38636400

RÉSUMÉ

Diabetic cardiomyopathy (DCM) is a common severe complication of diabetes that occurs independently of hypertension, coronary artery disease, and valvular cardiomyopathy, eventually leading to heart failure. Previous studies have reported that Tectorigenin (TEC) possesses extensive anti-inflammatory and anti-oxidative stress properties. In this present study, the impact of TEC on diabetic cardiomyopathy was examined. The model of DCM in mice was established with the combination of a high-fat diet and STZ treatment. Remarkably, TEC treatment significantly attenuated cardiac fibrosis and improved cardiac dysfunction. Concurrently, TEC was also found to mitigate hyperglycemia and hyperlipidemia in the DCM mouse. At the molecular level, TEC is involved in the activation of AMPK, both in vitro and in vivo, by enhancing its phosphorylation. This is achieved through the regulation of endothelial-mesenchymal transition via the AMPK/TGFß/Smad3 pathway. Furthermore, it was demonstrated that the level of ubiquitination of the adiponectin receptor 1 (AdipoR1) protein is associated with TEC-mediated improvement of cardiac dysfunction in DCM mice. Notably the substantial reduction of myocardial fibrosis. In conclusion, TEC improves cardiac fibrosis in DCM mice by modulating the AdipoR1/AMPK signaling pathway. These findings suggest that TEC could be an effective therapeutic agent for the treatment of diabetic cardiomyopathy.


Sujet(s)
Diabète expérimental , Cardiomyopathies diabétiques , Isoflavones , Animaux , Souris , AMP-Activated Protein Kinases/effets des médicaments et des substances chimiques , AMP-Activated Protein Kinases/métabolisme , Diabète expérimental/traitement médicamenteux , Diabète expérimental/complications , Cardiomyopathies diabétiques/traitement médicamenteux , Cardiomyopathies diabétiques/prévention et contrôle , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/étiologie , Alimentation riche en graisse/effets indésirables , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Fibrose/traitement médicamenteux , Isoflavones/pharmacologie , Isoflavones/usage thérapeutique , Souris de lignée C57BL , Myocarde/anatomopathologie , Myocarde/métabolisme , Récepteurs à l'adiponectine/effets des médicaments et des substances chimiques , Récepteurs à l'adiponectine/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéine Smad-3/métabolisme , Streptozocine
15.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167158, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38588780

RÉSUMÉ

OBJECTIVES: Diabetic cardiomyopathy (DCM) is the leading cause of mortality in type 2 diabetes mellitus (T2DM) patients, with its underlying mechanisms still elusive. This study aims to investigate the role of cholesterol-25-monooxygenase (CH25H) in T2DM induced cardiomyopathy. METHODS: High fat diet combined with streptozotocin (HFD/STZ) were used to establish a T2DM model. CH25H and its product 25-hydroxycholesterol (25HC) were detected in the hearts of T2DM model. Gain- or loss-of-function of CH25H were performed by receiving AAV9-cTNT-CH25H or CH25H knockout (CH25H-/-) mice with HFD/STZ treatment. Cardiac function was evaluated using echocardiography, and cardiac tissues were collected for immunoblot analysis, histological assessment and quantitative polymerase chain reaction (qPCR). Mitochondrial morphology and function were evaluated using transmission electron microscopy (TEM) and Seahorse XF Cell Mito Stress Test Kit. RNA-sequence analysis was performed to determine the molecular changes associated with CH25H deletion. RESULTS: CH25H and 25HC were significantly decreased in the hearts of T2DM mice. CH25H-/- mice treated with HFD/STZ exhibited impaired mitochondrial function and structure, increased lipid accumulation, and aggregated cardiac dysfunction. Conversely, T2DM mice receiving AAV9-CH25H displayed cardioprotective effects. Mechanistically, RNA sequencing and qPCR analysis revealed that CH25H deficiency decreased peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) and its target gene expression. Additionally, administration of ZLN005, a potent PGC-1α activator, partially protected against high glucose and palmitic acid induced mitochondria dysfunction and lipid accumulation in vitro. CONCLUSION: Our study provides compelling evidence supporting the protective role of CH25H in T2DM-induced cardiomyopathy. Furthermore, the regulation of PGC-1α may be intricately involved in this cardioprotective process.


Sujet(s)
Diabète expérimental , Diabète de type 2 , Cardiomyopathies diabétiques , Souris knockout , Animaux , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/prévention et contrôle , Cardiomyopathies diabétiques/étiologie , Diabète de type 2/complications , Diabète de type 2/métabolisme , Souris , Mâle , Diabète expérimental/complications , Diabète expérimental/métabolisme , Diabète expérimental/anatomopathologie , Steroid hydroxylases/métabolisme , Steroid hydroxylases/génétique , Alimentation riche en graisse/effets indésirables , Souris de lignée C57BL , Hydroxycholestérols/métabolisme , Myocarde/métabolisme , Myocarde/anatomopathologie , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique
16.
Cardiovasc Diabetol ; 23(1): 96, 2024 Mar 14.
Article de Anglais | MEDLINE | ID: mdl-38486199

RÉSUMÉ

Diabetic cardiomyopathy (DCM) is a major contributor to mortality in diabetic patients, characterized by a multifaceted pathogenesis and limited therapeutic options. While lactate, a byproduct of glycolysis, is known to be significantly elevated in type 2 diabetes, its specific role in DCM remains uncertain. This study reveals an abnormal upregulation of monocarboxylate transporter 4 (MCT4) on the plasma membrane of cardiomyocytes in type 2 diabetes, leading to excessive lactate efflux from these cells. The disruption in lactate transport homeostasis perturbs the intracellular lactate-pyruvate balance in cardiomyocytes, resulting in oxidative stress and inflammatory responses that exacerbate myocardial damage. Additionally, our findings suggest increased lactate efflux augments histone H4K12 lactylation in macrophages, facilitating inflammatory infiltration within the microenvironment. In vivo experiments have demonstrated that inhibiting MCT4 effectively alleviates myocardial oxidative stress and pathological damage, reduces inflammatory macrophage infiltration, and enhances cardiac function in type 2 diabetic mice. Furthermore, a clinical prediction model has been established, demonstrating a notable association between peripheral blood lactate levels and diastolic dysfunction in individuals with type 2 diabetes. This underscores the potential of lactate as a prognostic biomarker for DCM. Ultimately, our findings highlight the pivotal involvement of MCT4 in the dysregulation of cardiac energy metabolism and macrophage-mediated inflammation in type 2 diabetes. These insights offer novel perspectives on the pathogenesis of DCM and pave the way for the development of targeted therapeutic strategies against this debilitating condition.


Sujet(s)
Diabète expérimental , Diabète de type 2 , Cardiomyopathies diabétiques , Animaux , Humains , Souris , Diabète de type 2/diagnostic , Diabète de type 2/traitement médicamenteux , Cardiomyopathies diabétiques/étiologie , Métabolisme énergétique , Inflammation , Acide lactique/métabolisme , Modèles statistiques , Pronostic
17.
Int J Mol Sci ; 25(4)2024 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-38396795

RÉSUMÉ

Diabetes mellitus is a metabolic disorder with global economic implications that can lead to complications such as diabetic cardiomyopathy. The aim of this study was to compare the effects of chitosan versus dapagliflozin in mouse diabetic cardiomyopathy. We used 32 C57Bl/6 male mice aged between 8 and 10 weeks, which were randomly divided into Control-without diabetes mellitus (DM), type 1 DM (T1DM), T1DM + Chitosan, and T1DM + Dapapgliflozin groups. We induced diabetes with streptozotocin and treated the animals for 12 weeks. The analysis showed a reduction in intramyocardial fibrosis in the T1DM + Dapapgliflozin compared to T1DM animals. In T1DM + CHIT, a reduction in intramyocardial fibrosis was observed although, accordingly, there was also no significant decrease in blood glucose. The level of oxidative stress was reduced in the groups of treated animals compared to T1DM. All these observed changes in the structure and function of hearts were highlighted in the echocardiographic examination. In the treated groups, there was delayed appearance of left ventricular (LV) hypertrophy, a slight decrease in the ejection fraction of the LV, and an improved diastolic profile. The results demonstrate that chitosan has promising effects on diabetic cardiomyopathy that are comparable to the beneficial effects of dapagliflozin.


Sujet(s)
Composés benzhydryliques , Chitosane , Diabète de type 1 , Cardiomyopathies diabétiques , Glucosides , Mâle , Souris , Animaux , Cardiomyopathies diabétiques/traitement médicamenteux , Cardiomyopathies diabétiques/étiologie , Diabète de type 1/métabolisme , Chitosane/pharmacologie , Chitosane/usage thérapeutique , Fonction ventriculaire gauche , Modèles animaux de maladie humaine , Fibrose
18.
Front Immunol ; 15: 1348043, 2024.
Article de Anglais | MEDLINE | ID: mdl-38390337

RÉSUMÉ

Introduction: Diabetes is a debilitating disease that leads to complications like cardiac dysfunction and heart failure. In this study, we investigated the pathophysiology of diabetes-induced cardiac dysfunction in mice with dyslipidemia. We hypothesize diabetes in ApoE knockout (ApoE-/-) mice induces cardiac dysfunction by increasing inflammation and necroptosis. Methods: ApoE-/- mice were divided into experimental groups: Control, Streptozotocin (STZ), STZ + MSC-Exo (mesenchymal stem cell-derived exosomes), and STZ+MEF-Exo (Mouse embryonic fibroblast derived exosomes). At Day 42, we assessed cardiac function, collected blood and heart tissues. Heart tissue samples were analyzed for inflammation, necroptosis, signaling mechanism, hypertrophy and adverse structural remodeling using histology, immunohistochemistry, western blotting, RT-PCR, cytokine array and TF array. Results and Discussion: STZ treated ApoE-/- mice developed diabetes, with significantly (p<0.05) increased blood glucose and body weight loss. These mice developed cardiac dysfunction with significantly (p<0.05) increased left ventricular internal diameter end diastole and end systole, and decreased ejection fraction, and fractional shortening. We found significant (p<0.05) increased expression of inflammatory cytokines TNF- a, IL-6, IL-1a, IL-33 and decreased IL-10 expression. Diabetic mice also exhibited significantly (p<0.05) increased necroptosis marker expression and infiltration of inflammatory monocytes and macrophages. MSC-Exos treated mice showed recovery of diabetes associated pathologies with significantly reduced blood glucose, recovered body weight, increased IL-10 secretion and M2 polarized macrophages in the heart. These mice showed reduced TAK1-pJNK-NFKB inflammation associated expression and improved cardiac function with significantly reduced cardiac hypertrophy and fibrosis compared to diabetic mice. Treatment with MEF-Exos did not play a significant role in attenuating diabetes-induced cardiomyopathy as these treatment mice presented with cardiac dysfunction and underlying pathologies observed in STZ mice. Conclusion: Thus, we conclude that cardiac dysfunction develops in diabetic ApoE-/- mice, arising from inflammation, necroptosis, and adverse tissue remodeling, which is ameliorated by MSC-Exos, a potential therapeutic for diabetes-induced cardiomyopathy.


Sujet(s)
Diabète expérimental , Cardiomyopathies diabétiques , Exosomes , Cardiopathies , Animaux , Souris , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Glycémie/métabolisme , Cytokines/métabolisme , Diabète expérimental/métabolisme , Cardiomyopathies diabétiques/étiologie , Cardiomyopathies diabétiques/anatomopathologie , Exosomes/métabolisme , Fibroblastes/anatomopathologie , Cardiopathies/métabolisme , Inflammation/métabolisme , Interleukine-10/métabolisme , Souris invalidées pour les gènes ApoE , Nécroptose
19.
Cardiovasc Diabetol ; 23(1): 49, 2024 02 01.
Article de Anglais | MEDLINE | ID: mdl-38302936

RÉSUMÉ

BACKGROUND: Diabetic cardiomyopathy (DbCM) is a form of Stage B heart failure (HF) at high risk for progression to overt disease. Using baseline characteristics of study participants from the Aldose Reductase Inhibition for Stabilization of Exercise Capacity in Heart Failure (ARISE-HF) Trial we sought to characterize clinical characteristics of individuals with findings consistent with DbCM. METHODS: Among study participants meeting inclusion criteria, clinical characteristics, laboratory testing, imaging, Kansas City Cardiomyopathy Questionnaire (KCCQ), Physical Activity Scale of the Elderly (PASE) and cardiopulmonary exercise testing (CPET) results were tabulated. Cluster phenogroups were identified. RESULTS: Among 691 study participants (mean age 67.4 years; 50% were female), mean duration of type 2 diabetes mellitus (T2DM) was 14.5 years. The median (Q1, Q3) N-terminal pro-B type natriuretic peptide and high sensitivity cardiac troponin T were 71 (35, 135) ng/L and 9 [6, 12] ng/L. The most common echocardiographic abnormalities were reduced global longitudinal strain in 25.3% and impaired diastolic relaxation in 17.7%. Despite rather well-preserved KCCQ scores the average PASE score was markedly impaired at 155 accompanied by an average maximal oxygen consumption of 15.7 mL/Kg/minute on CPET. In K-means clustering, 4 phenogroups were identified including a higher-risk group with more advanced age, greater elevation of cardiac biomarkers, and more prevalent evidence for diastolic dysfunction and left ventricular hypertrophy. CONCLUSIONS: Baseline data from the ARISE-HF Trial provide clinical characterization of individuals with T2DM and features of stage B HF, and may help clarify the diagnosis of DbCM. TRIAL REGISTRATION: ARISE-HF, NCT04083339.


Sujet(s)
Diabète de type 2 , Cardiomyopathies diabétiques , Défaillance cardiaque , Humains , Femelle , Sujet âgé , Mâle , Cardiomyopathies diabétiques/imagerie diagnostique , Cardiomyopathies diabétiques/étiologie , Diabète de type 2/complications , Diabète de type 2/diagnostic , Diabète de type 2/traitement médicamenteux , Débit systolique , Défaillance cardiaque/diagnostic , Hypertrophie ventriculaire gauche , Fonction ventriculaire gauche
20.
Cardiovasc Diabetol ; 23(1): 19, 2024 01 09.
Article de Anglais | MEDLINE | ID: mdl-38195474

RÉSUMÉ

AIMS: Diabetic cardiomyopathy (DCM) is a major cause of mortality in patients with diabetes, and the potential strategies for treating DCM are insufficient. Melatonin (Mel) has been shown to attenuate DCM, however, the underlying mechanism remains unclear. The role of vascular endothelial growth factor-B (VEGF-B) in DCM is little known. In present study, we aimed to investigate whether Mel alleviated DCM via regulation of VEGF-B and explored its underlying mechanisms. METHODS AND RESULTS: We found that Mel significantly alleviated cardiac dysfunction and improved autophagy of cardiomyocytes in type 1 diabetes mellitus (T1DM) induced cardiomyopathy mice. VEGF-B was highly expressed in DCM mice in comparison with normal mice, and its expression was markedly reduced after Mel treatment. Mel treatment diminished the interaction of VEGF-B and Glucose-regulated protein 78 (GRP78) and reduced the interaction of GRP78 and protein kinase RNA -like ER kinase (PERK). Furthermore, Mel increased phosphorylation of PERK and eIF2α, then up-regulated the expression of ATF4. VEGF-B-/- mice imitated the effect of Mel on wild type diabetic mice. Interestingly, injection with Recombinant adeno-associated virus serotype 9 (AAV9)-VEGF-B or administration of GSK2656157 (GSK), an inhibitor of phosphorylated PERK abolished the protective effect of Mel on DCM. Furthermore, rapamycin, an autophagy agonist displayed similar effect with Mel treatment; while 3-Methyladenine (3-MA), an autophagy inhibitor neutralized the effect of Mel on high glucose-treated neonatal rat ventricular myocytes. CONCLUSIONS: These results demonstrated that Mel attenuated DCM via increasing autophagy of cardiomyocytes, and this cardio-protective effect of Mel was dependent on VEGF-B/GRP78/PERK signaling pathway.


Sujet(s)
Diabète expérimental , Cardiomyopathies diabétiques , Mélatonine , Humains , Souris , Rats , Animaux , Cardiomyopathies diabétiques/traitement médicamenteux , Cardiomyopathies diabétiques/étiologie , Cardiomyopathies diabétiques/prévention et contrôle , Myocytes cardiaques , Facteur de croissance endothéliale vasculaire de type B , Mélatonine/pharmacologie , Chaperonne BiP du réticulum endoplasmique , Diabète expérimental/traitement médicamenteux , Transduction du signal , Autophagie , Glucose
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...