Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 471
Filtrer
1.
Cardiovasc Diabetol ; 23(1): 227, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951895

RÉSUMÉ

In recent years, the incidence of diabetes has been increasing rapidly, posing a serious threat to human health. Diabetic cardiomyopathy (DCM) is characterized by cardiomyocyte hypertrophy, myocardial fibrosis, apoptosis, ventricular remodeling, and cardiac dysfunction in individuals with diabetes, ultimately leading to heart failure and mortality. However, the underlying mechanisms contributing to DCM remain incompletely understood. With advancements in molecular biology technology, accumulating evidence has shown that numerous non-coding RNAs (ncRNAs) crucial roles in the development and progression of DCM. This review aims to summarize recent studies on the involvement of three types of ncRNAs (micro RNA, long ncRNA and circular RNA) in the pathophysiology of DCM, with the goal of providing innovative strategies for the prevention and treatment of DCM.


Sujet(s)
Cardiomyopathies diabétiques , ARN circulaire , ARN long non codant , Humains , Cardiomyopathies diabétiques/génétique , Cardiomyopathies diabétiques/physiopathologie , Cardiomyopathies diabétiques/métabolisme , Animaux , ARN circulaire/génétique , ARN circulaire/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme , microARN/génétique , microARN/métabolisme , Régulation de l'expression des gènes , ARN non traduit/génétique , ARN non traduit/métabolisme , Transduction du signal , Myocarde/anatomopathologie , Myocarde/métabolisme
2.
Sci Rep ; 14(1): 15324, 2024 07 03.
Article de Anglais | MEDLINE | ID: mdl-38961143

RÉSUMÉ

Diabetic cardiomyopathy (DCM) is a common cardiovascular complication of diabetes, which may threaten the quality of life and shorten life expectancy in the diabetic population. However, the molecular mechanisms underlying the diabetes cardiomyopathy are not fully elucidated. We analyzed two datasets from Gene Expression Omnibus (GEO). Differentially expressed and weighted gene correlation network analysis (WGCNA) was used to screen key genes and molecules. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and protein-protein interaction (PPI) network analysis were constructed to identify hub genes. The diagnostic value of the hub gene was evaluated using the receiver operating characteristic (ROC). Quantitative real-time PCR (RT-qPCR) was used to validate the hub genes. A total of 13 differentially co-expressed modules were selected by WGCNA and differential expression analysis. KEGG and GO analysis showed these DEGs were mainly enriched in lipid metabolism and myocardial hypertrophy pathway, cytomembrane, and mitochondrion. As a result, six genes were identified as hub genes. Finally, five genes (Pdk4, Lipe, Serpine1, Igf1r, and Bcl2l1) were found significantly changed in both the validation dataset and experimental mice with DCM. In conclusion, the present study identified five genes that may help provide novel targets for diagnosing and treating DCM.


Sujet(s)
Biologie informatique , Cardiomyopathies diabétiques , Réseaux de régulation génique , Cartes d'interactions protéiques , Cardiomyopathies diabétiques/génétique , Biologie informatique/méthodes , Animaux , Souris , Cartes d'interactions protéiques/génétique , Humains , Inhibiteur-1 d'activateur du plasminogène/génétique , Analyse de profil d'expression de gènes , Récepteur IGF de type 1/génétique , Gene Ontology , Régulation de l'expression des gènes
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.
Front Endocrinol (Lausanne) ; 15: 1370387, 2024.
Article de Anglais | MEDLINE | ID: mdl-38883603

RÉSUMÉ

Background: Diabetes mellitus is an independent risk factor for heart failure, and diabetes-induced heart failure severely affects patients' health and quality of life. Cuproptosis is a newly defined type of programmed cell death that is thought to be involved in the pathogenesis and progression of cardiovascular disease, but the molecular mechanisms involved are not well understood. Therefore, we aimed to identify biomarkers associated with cuproptosis in diabetes mellitus-associated heart failure and the potential pathological mechanisms in cardiomyocytes. Materials: Cuproptosis-associated genes were identified from the previous publication. The GSE26887 dataset was downloaded from the GEO database. Methods: The consistency clustering was performed according to the cuproptosis gene expression. Differentially expressed genes were identified using the limma package, key genes were identified using the weighted gene co-expression network analysis(WGCNA) method, and these were subjected to immune infiltration analysis, enrichment analysis, and prediction of the key associated transcription factors. Consistency clustering identified three cuproptosis clusters. The differentially expressed genes for each were identified using limma and the most critical MEantiquewhite4 module was obtained using WGCNA. We then evaluated the intersection of the MEantiquewhite4 output with the three clusters, and obtained the key genes. Results: There were four key genes: HSDL2, BCO2, CORIN, and SNORA80E. HSDL2, BCO2, and CORIN were negatively associated with multiple immune factors, while SNORA80E was positively associated, and T-cells accounted for a major proportion of this relationship with the immune system. Four enriched pathways were found to be associated: arachidonic acid metabolism, peroxisomes, fatty acid metabolism, and dorsoventral axis formation, which may be regulated by the transcription factor MECOM, through a change in protein structure. Conclusion: HSDL2, BCO2, CORIN, and SNORA80E may regulate cardiomyocyte cuproptosis in patients with diabetes mellitus-associated heart failure through effects on the immune system. The product of the cuproptosis-associated gene LOXL2 is probably involved in myocardial fibrosis in patients with diabetes, which leads to the development of cardiac insufficiency.


Sujet(s)
Biologie informatique , Défaillance cardiaque , Myocytes cardiaques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Humains , Défaillance cardiaque/génétique , Défaillance cardiaque/anatomopathologie , Défaillance cardiaque/métabolisme , Biologie informatique/méthodes , Analyse de profil d'expression de gènes , Réseaux de régulation génique , Ferroptose/génétique , Cardiomyopathies diabétiques/génétique , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/anatomopathologie
5.
Cell Death Dis ; 15(6): 406, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38858351

RÉSUMÉ

Diabetic cardiomyopathy (DCM) is a prevalent myocardial microvascular complication of the myocardium with a complex pathogenesis. Investigating the pathogenesis of DCM can significantly contribute to enhancing its prevention and treatment strategies. Our study revealed an upregulation of lysine acetyltransferase 2 A (Kat2a) expression in DCM, accompanied by a decrease in N6-methyladenosine (m6A) modified Kat2a mRNA levels. Our study revealed an upregulation of lysine acetyltransferase 2 A (Kat2a) expression in DCM, accompanied by a decrease in N6-methyladenosine (m6A) modified Kat2a mRNA levels. Functionally, inhibition of Kat2a effectively ameliorated high glucose-induced cardiomyocyte injury both in vitro and in vivo by suppressing ferroptosis. Mechanistically, Demethylase alkB homolog 5 (Alkbh5) was found to reduce m6A methylation levels on Kat2a mRNA, leading to its upregulation. YTH domain family 2 (Ythdf2) played a crucial role as an m6A reader protein mediating the degradation of Kat2a mRNA. Furthermore, Kat2a promoted ferroptosis by increasing Tfrc and Hmox1 expression via enhancing the enrichment of H3K27ac and H3K9ac on their promoter regions. In conclusion, our findings unveil a novel role for the Kat2a-ferroptosis axis in DCM pathogenesis, providing valuable insights for potential clinical interventions.


Sujet(s)
Cardiomyopathies diabétiques , Ferroptose , Heme oxygenase-1 , Histone acetyltransferases , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/génétique , Animaux , Ferroptose/génétique , Humains , Heme oxygenase-1/métabolisme , Heme oxygenase-1/génétique , Souris , Histone acetyltransferases/métabolisme , Histone acetyltransferases/génétique , Mâle , Souris de lignée C57BL , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Adénosine/analogues et dérivés , Adénosine/métabolisme
6.
Crit Rev Eukaryot Gene Expr ; 34(5): 45-57, 2024.
Article de Anglais | MEDLINE | ID: mdl-38842203

RÉSUMÉ

Inflammation-mediated dysfunction of cardiomyocytes is the main cause of diabetic cardiomyopathy (DCM). The present study aimed to investigate the roles of siah E3 ubiquitin protein ligase 1 (SIAH1) in DCM. The online dataset GSE4172 was used to analyze the differentially expressed genes in myocardial inflammation of DCM patients. RT-qPCR was conducted to detect mRNA levels. Enzyme-Linked Immunosorbent Assay (ELISA) was performed to detect cytokine release. Western blot was used to detect protein expression. Lactate dehydrogenase (LDH) assay was used to determine cytotoxicity. In vitro ubiquitination assay was applied to determine the ubiquitination of nuclear factor kappa B inhibitor alpha (1κВ-α). Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was used to detect the death of cardiomyocytes. Flow cytometry was applied for determining cardiomyocyte pyroptosis. The results showed that SIAH1 was overexpressed in human inflammatory cardiomyopathy. High expression of SIAH1 was associated with inflammatory response. SIAH1 was also overexpressed lipopolysaccharide (LPS)-induced inflammatory cardiomyopathy model in vitro. However, SIAH1 knockdown suppressed the inflammatory-related pyroptosis of cardiomyocytes. SIAH1 promoted the ubiquitination of 1κВ-α and activated nuclear factor kappa В (NF-κВ) signaling, which promoted the pyroptosis of cardiomyocytes. In conclusion, SIAH1 exacerbated the progression of human inflammatory cardiomyopathy via inducing the ubiquitination of 1κВ-α and activation of NF-κВ signaling. Therefore, SIAHI/IκB-α/NF-κB signaling may be a potential target for human inflammatory cardiomyopathy.


Sujet(s)
Cardiomyopathies diabétiques , Myocytes cardiaques , Facteur de transcription NF-kappa B , Pyroptose , Transduction du signal , Ubiquitin-protein ligases , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/génétique , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Humains , Facteur de transcription NF-kappa B/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Inhibiteur alpha de NF-KappaB/métabolisme , Inhibiteur alpha de NF-KappaB/génétique , Ubiquitination , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/génétique
7.
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
8.
Gene ; 923: 148563, 2024 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-38754569

RÉSUMÉ

BACKGROUND: Diabetic cardiomyopathy (DCM) is a special type of cardiovascular disease, termed as a situation of abnormal myocardial structure and function that occurs in diabetic patients. However, the most fundamental mechanisms of DCM have not been fully explicated, and useful targets for the therapeutic strategies still need to be explored. METHODS: In the present study, we combined bioinformatics analysis and in vitro experiments throughout the process of DCM. Differentially Expressed Genes (DEGs) analysis was performed and the weighted gene co-expression network analysis (WGCNA) was constructed to determine the crucial genes that were tightly connected to DCM. Additionally, Functional enrichment analysis was conducted to define biological pathways. To identify the specific molecular mechanism, the human cardiomyocyte cell line (AC16) was stimulated by high glucose (HG, 50 mM D-glucose) and used to imitate DCM condition. Then, we tentatively examined the effect of high glucose on cardiomyocytes, the expression levels of crucial genes were further validated by in vitro experiments. RESULTS: Generally, NPPA, IGFBP5, SERPINE1, and C3 emerged as potential therapeutic targets. Functional enrichment analysis performed by bioinformatics indicated that the pathogenesis of DCM is mainly related to heart muscle contraction and calcium (Ca2+) release activation. In vitro, we discovered that high glucose treatment induced cardiomyocyte injury and exacerbated mitochondrial dysfunction remarkably. CONCLUSION: Our research defined four crucial genes, as well as determined that mitochondrial function impairment compromises calcium homeostasis ultimately resulting in contractile dysfunction is a central contributor to DCM progression. Hopefully, this study will offer more effective biomarkers for DCM diagnosis and treatment.


Sujet(s)
Cardiomyopathies diabétiques , Glucose , Myocytes cardiaques , Cardiomyopathies diabétiques/génétique , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/anatomopathologie , Humains , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Glucose/métabolisme , Glucose/pharmacologie , Lignée cellulaire , Inhibiteur-1 d'activateur du plasminogène/génétique , Inhibiteur-1 d'activateur du plasminogène/métabolisme , Biologie informatique/méthodes , Réseaux de régulation génique , Analyse de profil d'expression de gènes , Mitochondries/métabolisme , Mitochondries/génétique , Calcium/métabolisme
9.
Pharmacol Res ; 205: 107235, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38815879

RÉSUMÉ

Diabetic cardiomyopathy (DCM) is a major complication of diabetes and is characterized by left ventricular dysfunction. Currently, there is a lack of effective treatments for DCM. Ubiquitin-specific protease 7 (USP7) plays a key role in various diseases. However, whether USP7 is involved in DCM has not been established. In this study, we demonstrated that USP7 was upregulated in diabetic mouse hearts and NMCMs co-treated with HG+PA or H9c2 cells treated with PA. Abnormalities in diabetic heart morphology and function were reversed by USP7 silencing through conditional gene knockout or chemical inhibition. Proteomic analysis coupled with biochemical validation confirmed that PCG1ß was one of the direct protein substrates of USP7 and aggravated myocardial damage through coactivation of the PPARα signaling pathway. USP7 silencing restored the expression of fatty acid metabolism-related proteins and restored mitochondrial homeostasis by inhibiting mitochondrial fission and promoting fusion events. Similar effects were also observed in vitro. Our data demonstrated that USP7 promoted cardiometabolic metabolism disorders and mitochondrial homeostasis dysfunction via stabilizing PCG1ß and suggested that silencing USP7 may be a therapeutic strategy for DCM.


Sujet(s)
Diabète expérimental , Cardiomyopathies diabétiques , Homéostasie , Souris de lignée C57BL , Ubiquitin-specific peptidase 7 , Animaux , Ubiquitin-specific peptidase 7/métabolisme , Ubiquitin-specific peptidase 7/génétique , Diabète expérimental/métabolisme , Diabète expérimental/génétique , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/génétique , Mâle , Souris , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Mitochondries du myocarde/métabolisme , Lignée cellulaire , Souris knockout , Rats , Mitochondries/métabolisme , Humains
10.
Redox Biol ; 73: 103184, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38718533

RÉSUMÉ

RATIONALE: The disruption of the balance between fatty acid (FA) uptake and oxidation (FAO) leads to cardiac lipotoxicity, serving as the driving force behind diabetic cardiomyopathy (DbCM). Sirtuin 5 (Sirt5), a lysine de-succinylase, could impact diverse metabolic pathways, including FA metabolism. Nevertheless, the precise roles of Sirt5 in cardiac lipotoxicity and DbCM remain unknown. OBJECTIVE: This study aims to elucidate the role and underlying mechanism of Sirt5 in the context of cardiac lipotoxicity and DbCM. METHODS AND RESULTS: The expression of myocardial Sirt5 was found to be modestly elevated in diabetic heart failure patients and mice. Cardiac dysfunction, hypertrophy and lipotoxicity were exacerbated by ablation of Sirt5 but improved by forced expression of Sirt5 in diabetic mice. Notably, Sirt5 deficiency impaired FAO without affecting the capacity of FA uptake in the diabetic heart, leading to accumulation of FA intermediate metabolites, which mainly included medium- and long-chain fatty acyl-carnitines. Mechanistically, succinylomics analyses identified carnitine palmitoyltransferase 2 (CPT2), a crucial enzyme involved in the reconversion of fatty acyl-carnitines to fatty acyl-CoA and facilitating FAO, as the functional succinylated substrate mediator of Sirt5. Succinylation of Lys424 in CPT2 was significantly increased by Sirt5 deficiency, leading to the inactivation of its enzymatic activity and the subsequent accumulation of fatty acyl-carnitines. CPT2 K424R mutation, which mitigated succinylation modification, counteracted the reduction of enzymatic activity in CPT2 mediated by Sirt5 deficiency, thereby attenuating Sirt5 knockout-induced FAO impairment and lipid deposition. CONCLUSIONS: Sirt5 deficiency impairs FAO, leading to cardiac lipotoxicity in the diabetic heart through the succinylation of Lys424 in CPT2. This underscores the potential roles of Sirt5 and CPT2 as therapeutic targets for addressing DbCM.


Sujet(s)
Carnitine O-palmitoyltransferase , Cardiomyopathies diabétiques , Acides gras , Métabolisme lipidique , Myocytes cardiaques , Sirtuines , Animaux , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/génétique , Cardiomyopathies diabétiques/anatomopathologie , Carnitine O-palmitoyltransferase/métabolisme , Carnitine O-palmitoyltransferase/génétique , Sirtuines/métabolisme , Sirtuines/génétique , Souris , Acides gras/métabolisme , Myocytes cardiaques/métabolisme , Humains , Mâle , Oxydoréduction , Diabète expérimental/métabolisme , Diabète expérimental/complications
11.
Zhongguo Zhong Yao Za Zhi ; 49(7): 1905-1914, 2024 Apr.
Article de Chinois | MEDLINE | ID: mdl-38812203

RÉSUMÉ

This study aimed to explore the mechanism of Shexiang Tongxin Dropping Pills(STDP) in treating diabetic cardiomyopathy(DCM) based on network pharmacology, molecular docking, and animal experiments. BATMAN, TCMSP, and GeneCards were searched for the active ingredients and targets of STDP against DCM. STRING and Cytoscape were used to build the protein-protein interaction(PPI) network and "drug-active ingredient-target" network. Gene Ontology(GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes(KEGG) enrichment analysis of the targets were carried out based on DAVID. The molecular docking of key receptor proteins with corresponding active ingredients was performed using AutoDock Vina. The rat model of DCM was established by a high-fat diet combined with intraperitoneal injection of streptozotocin. Rats were assigned into control, model, low-(20 mg·kg~(-1)) and high-dose(40 mg·kg~(-1)) STDP, and metformin(200 mg·kg~(-1)) groups. After 8 weeks of continuous administration, the cardiac function, myocardial pathological changes, and myocardial collagen fiber deposition of rats in each group were detected by echocardiography, hematoxylin-eosin(HE) staining, and Sirius red staining, respectively. The myocardial hypertrophy was detected by WGA staining. The expression levels of p38 mitogen-activated protein kinase(p38), phosphorylation-p38(p-p38), c-Jun N-terminal kinase(JNK), phosphorylation-JNK(p-JNK), caspase-3, and C-caspase-3 in the myocardial tissue of rats in each group were measured by Western blot. The network pharmacology predicted 199 active ingredients and 1 655 targets of STDP and 463 targets of DCM. One hundred and thirty-four potential targets of STDP for treating DCM were obtained, and the AGE-RAGE signaling pathway in diabetic complications was screened out. Molecular docking results showed that miltirone, dehydromiltirone, and tryptanthrin had strong binding affinity with RAGE. The results of animal experiments confirmed that STDP effectively protected the cardiac function of DCM rats. Compared with the DCM model group, the STDP groups showed significantly down-regulated protein levels of p-p38, p-JNK, and C-caspase-3. To sum up, STDP may protect the cardiac function of DCM rats by regulating the AGE-RAGE signaling pathway.


Sujet(s)
Cardiomyopathies diabétiques , Médicaments issus de plantes chinoises , Simulation de docking moléculaire , Pharmacologie des réseaux , Animaux , Cardiomyopathies diabétiques/traitement médicamenteux , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/génétique , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Médicaments issus de plantes chinoises/administration et posologie , Rats , Mâle , Rat Sprague-Dawley , Humains
12.
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
13.
Cell Mol Life Sci ; 81(1): 228, 2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38777955

RÉSUMÉ

Diabetic cardiomyopathy (DCM) is a prevalent complication of type 2 diabetes (T2D). 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) is a glycolysis regulator. However, the potential effects of PFKFB3 in the DCM remain unclear. In comparison to db/m mice, PFKFB3 levels decreased in the hearts of db/db mice. Cardiac-specific PFKFB3 overexpression inhibited myocardial oxidative stress and cardiomyocyte apoptosis, suppressed mitochondrial fragmentation, and partly restored mitochondrial function in db/db mice. Moreover, PFKFB3 overexpression stimulated glycolysis. Interestingly, based on the inhibition of glycolysis, PFKFB3 overexpression still suppressed oxidative stress and apoptosis of cardiomyocytes in vitro, which indicated that PFKFB3 overexpression could alleviate DCM independent of glycolysis. Using mass spectrometry combined with co-immunoprecipitation, we identified optic atrophy 1 (OPA1) interacting with PFKFB3. In db/db mice, the knockdown of OPA1 receded the effects of PFKFB3 overexpression in alleviating cardiac remodeling and dysfunction. Mechanistically, PFKFB3 stabilized OPA1 expression by promoting E3 ligase NEDD4L-mediated atypical K6-linked polyubiquitination and thus prevented the degradation of OPA1 by the proteasomal pathway. Our study indicates that PFKFB3/OPA1 could be potential therapeutic targets for DCM.


Sujet(s)
Cardiomyopathies diabétiques , dGTPases , Myocytes cardiaques , Phosphofructokinase-2 , Ubiquitination , Phosphofructokinase-2/métabolisme , Phosphofructokinase-2/génétique , Animaux , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/génétique , Souris , dGTPases/métabolisme , dGTPases/génétique , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Mâle , Stress oxydatif , Apoptose/génétique , Myocarde/métabolisme , Myocarde/anatomopathologie , Souris de lignée C57BL , Diabète de type 2/métabolisme , Diabète de type 2/génétique , Glycolyse , Humains , Stabilité protéique
14.
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
15.
Clin Epigenetics ; 16(1): 52, 2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38581056

RÉSUMÉ

Diabetic cardiomyopathy (DCM) is a critical complication that poses a significant threat to the health of patients with diabetes. The intricate pathological mechanisms of DCM cause diastolic dysfunction, followed by impaired systolic function in the late stages. Accumulating researches have revealed the association between DCM and various epigenetic regulatory mechanisms, including DNA methylation, histone modifications, non-coding RNAs, and other epigenetic molecules. Recently, a profound understanding of epigenetics in the pathophysiology of DCM has been broadened owing to advanced high-throughput technologies, which assist in developing potential therapeutic strategies. In this review, we briefly introduce the epigenetics regulation and update the relevant progress in DCM. We propose the role of epigenetic factors and non-coding RNAs (ncRNAs) as potential biomarkers and drugs in DCM diagnosis and treatment, providing a new perspective and understanding of epigenomics in DCM.


Sujet(s)
Diabète , Cardiomyopathies diabétiques , Humains , Cardiomyopathies diabétiques/génétique , Méthylation de l'ADN , Épigénomique , Épigenèse génétique , Code histone , Diabète/génétique
16.
Free Radic Biol Med ; 218: 149-165, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38570171

RÉSUMÉ

Proper protein degradation is required for cellular protein homeostasis and organ function. Particularly, in post-mitotic cells, such as cardiomyocytes, unbalanced proteolysis due to inflammatory stimuli and oxidative stress contributes to organ dysfunction. To ensure appropriate protein turnover, eukaryotic cells exert two main degradation systems, the ubiquitin-proteasome-system and the autophagy-lysosome-pathway. It has been shown that proteasome activity affects the development of cardiac dysfunction differently, depending on the type of heart failure. Studies analyzing the inducible subtype of the proteasome, the immunoproteasome (i20S), demonstrated that the i20S plays a double role in diseased hearts. While i20S subunits are increased in cardiac hypertrophy, atrial fibrillation and partly in myocarditis, the opposite applies to diabetic cardiomyopathy and ischemia/reperfusion injury. In addition, the i20S appears to play a role in autophagy modulation depending on heart failure phenotype. This review summarizes the current literature on the i20S in different heart failure phenotypes, emphasizing the two faces of i20S in injured hearts. A selection of established i20S inhibitors is introduced and signaling pathways linking the i20S to autophagy are highlighted. Mapping the interplay of the i20S and autophagy in different types of heart failure offers potential approaches for developing treatment strategies against heart failure.


Sujet(s)
Autophagie , Défaillance cardiaque , Proteasome endopeptidase complex , Défaillance cardiaque/anatomopathologie , Défaillance cardiaque/métabolisme , Défaillance cardiaque/génétique , Défaillance cardiaque/immunologie , Humains , Proteasome endopeptidase complex/métabolisme , Animaux , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/métabolisme , Phénotype , Transduction du signal , Protéolyse , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/génétique , Myocardite/anatomopathologie , Myocardite/métabolisme , Myocardite/immunologie , Myocardite/génétique , Cardiomégalie/anatomopathologie , Cardiomégalie/métabolisme , Cardiomégalie/génétique
17.
Cardiovasc Diabetol ; 23(1): 139, 2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38664790

RÉSUMÉ

BACKGROUND: Diabetic cardiomyopathy (DCM) poses a growing health threat, elevating heart failure risk in diabetic individuals. Understanding DCM is crucial, with fibroblasts and endothelial cells playing pivotal roles in driving myocardial fibrosis and contributing to cardiac dysfunction. Advances in Multimodal single-cell profiling, such as scRNA-seq and scATAC-seq, provide deeper insights into DCM's unique cell states and molecular landscape for targeted therapeutic interventions. METHODS: Single-cell RNA and ATAC data from 10x Multiome libraries were processed using Cell Ranger ARC v2.0.1. Gene expression and ATAC data underwent Seurat and Signac filtration. Differential gene expression and accessible chromatin regions were identified. Transcription factor activity was estimated with chromVAR, and Cis-coaccessibility networks were calculated using Cicero. Coaccessibility connections were compared to the GeneHancer database. Gene Ontology analysis, biological process scoring, cell-cell communication analysis, and gene-motif correlation was performed to reveal intricate molecular changes. Immunofluorescent staining utilized various antibodies on paraffin-embedded tissues to verify the findings. RESULTS: This study integrated scRNA-seq and scATAC-seq data obtained from hearts of WT and DCM mice, elucidating molecular changes at the single-cell level throughout the diabetic cardiomyopathy progression. Robust and accurate clustering analysis of the integrated data revealed altered cell proportions, showcasing decreased endothelial cells and macrophages, coupled with increased fibroblasts and myocardial cells in the DCM group, indicating enhanced fibrosis and endothelial damage. Chromatin accessibility analysis unveiled unique patterns in cell types, with heightened transcriptional activity in myocardial cells. Subpopulation analysis highlighted distinct changes in cardiomyocytes and fibroblasts, emphasizing pathways related to fatty acid metabolism and cardiac contraction. Fibroblast-centered communication analysis identified interactions with endothelial cells, implicating VEGF receptors. Endothelial cell subpopulations exhibited altered gene expressions, emphasizing contraction and growth-related pathways. Candidate regulators, including Tcf21, Arnt, Stat5a, and Stat5b, were identified, suggesting their pivotal roles in DCM development. Immunofluorescence staining validated marker genes of cell subpopulations, confirming PDK4, PPARγ and Tpm1 as markers for metabolic pattern-altered cardiomyocytes, activated fibroblasts and endothelial cells with compromised proliferation. CONCLUSION: Our integrated scRNA-seq and scATAC-seq analysis unveils intricate cell states and molecular alterations in diabetic cardiomyopathy. Identified cell type-specific changes, transcription factors, and marker genes offer valuable insights. The study sheds light on potential therapeutic targets for DCM.


Sujet(s)
Cardiomyopathies diabétiques , Analyse sur cellule unique , Transcriptome , Cardiomyopathies diabétiques/génétique , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/physiopathologie , Animaux , Analyse de profil d'expression de gènes , Chromatine/métabolisme , Chromatine/génétique , Souris de lignée C57BL , Réseaux de régulation génique , Assemblage et désassemblage de la chromatine , Modèles animaux de maladie humaine , Mâle , RNA-Seq , Régulation de l'expression des gènes , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibrose , Souris , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie
18.
Stem Cell Res Ther ; 15(1): 120, 2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38659015

RÉSUMÉ

BACKGROUND: Diabetic cardiomyopathy (DCM) is a serious health-threatening complication of diabetes mellitus characterized by myocardial fibrosis and abnormal cardiac function. Human umbilical cord mesenchymal stromal cells (hUC-MSCs) are a potential therapeutic tool for DCM and myocardial fibrosis via mechanisms such as the regulation of microRNA (miRNA) expression and inflammation. It remains unclear, however, whether hUC-MSC therapy has beneficial effects on cardiac function following different durations of diabetes and which mechanistic aspects of DCM are modulated by hUC-MSC administration at different stages of its development. This study aimed to investigate the therapeutic effects of intravenous administration of hUC-MSCs on DCM following different durations of hyperglycemia in an experimental male model of diabetes and to determine the effects on expression of candidate miRNAs, target mRNA and inflammatory mediators. METHODS: A male mouse model of diabetes was induced by multiple low-dose streptozotocin injections. The effects on severity of DCM of intravenous injections of hUC-MSCs and saline two weeks previously were compared at 10 and 18 weeks after diabetes induction. At both time-points, biochemical assays, echocardiography, histopathology, polymerase chain reaction (PCR), immunohistochemistry and enzyme-linked immunosorbent assays (ELISA) were used to analyze blood glucose, body weight, cardiac structure and function, degree of myocardial fibrosis and expression of fibrosis-related mRNA, miRNA and inflammatory mediators. RESULTS: Saline-treated diabetic male mice had impaired cardiac function and increased cardiac fibrosis after 10 and 18 weeks of diabetes. At both time-points, cardiac dysfunction and fibrosis were improved in hUC-MSC-treated mice. Pro-fibrotic indicators (α-SMA, collagen I, collagen III, Smad3, Smad4) were reduced and anti-fibrotic mediators (FGF-1, miRNA-133a) were increased in hearts of diabetic animals receiving hUC-MSCs compared to saline. Increased blood levels of pro-inflammatory cytokines (IL-6, TNF, IL-1ß) and increased cardiac expression of IL-6 were also observed in saline-treated mice and were reduced by hUC-MSCs at both time-points, but to a lesser degree at 18 weeks. CONCLUSION: Intravenous injection of hUC-MSCs ameliorated key functional and structural features of DCM in male mice with diabetes of shorter and longer duration. Mechanistically, these effects were associated with restoration of intra-myocardial expression of miRNA-133a and its target mRNA COL1AI as well as suppression of systemic and localized inflammatory mediators.


Sujet(s)
Diabète expérimental , Cardiomyopathies diabétiques , Fibrose , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , microARN , Myocarde , Cordon ombilical , Animaux , Humains , Mâle , Souris , Diabète expérimental/thérapie , Diabète expérimental/métabolisme , Cardiomyopathies diabétiques/thérapie , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/génétique , Fibrose/thérapie , Transplantation de cellules souches mésenchymateuses/méthodes , Cellules souches mésenchymateuses/métabolisme , Souris de lignée C57BL , microARN/génétique , microARN/métabolisme , Myocarde/métabolisme , Myocarde/anatomopathologie , Cordon ombilical/cytologie , Cordon ombilical/métabolisme
19.
Cell Biochem Funct ; 42(2): e3968, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38439590

RÉSUMÉ

Over the past decade, the prevalence of diabetes has increased significantly worldwide, leading to an increase in vascular complications of diabetes (VCD), such as diabetic cardiomyopathy (DCM), diabetic nephropathy (DN), and diabetic retinopathy (DR). Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs), long Noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), play a key role in cellular processes, including the pathophysiology of diabetes and VCD via pyroptosis. ncRNAs (e.g., miR-17, lnc-MEG3, and lnc-KCNQ1OT1) can regulate pyroptosis in pancreatic ß cells. Some ncRNAs are involved in VCD progression. For example, miR-21, lnc-KCNQ1OT1, lnc-GAS5, and lnc-MALAT1 were reported in DN and DCM, and lnc-MIAT was identified in DCM and DR. Herein, this review aimed to summarize recent research findings related to ncRNAs-mediated pyroptosis at the onset and progression of diabetes and VCD.


Sujet(s)
Diabète , Cardiomyopathies diabétiques , Néphropathies diabétiques , microARN , Humains , Pyroptose , Cardiomyopathies diabétiques/génétique , Néphropathies diabétiques/génétique , ARN non traduit/génétique , microARN/génétique , Diabète/génétique
20.
Front Endocrinol (Lausanne) ; 15: 1185062, 2024.
Article de Anglais | MEDLINE | ID: mdl-38469146

RÉSUMÉ

Background: Diabetic cardiomyopathy (DCM) lacks specific and sensitive biomarkers, and its diagnosis remains a challenge. Therefore, there is an urgent need to develop useful biomarkers to help diagnose and evaluate the prognosis of DCM. This study aims to find specific diagnostic markers for diabetic cardiomyopathy. Methods: Two datasets (GSE106180 and GSE161827) from the GEO database were integrated to identify differentially expressed genes (DEGs) between control and type 2 diabetic cardiomyopathy. We assessed the infiltration of immune cells and used weighted coexpression network analysis (WGCNA) to construct the gene coexpression network. Then we performed a clustering analysis. Finally, a diagnostic model was built by the least absolute shrinkage and selection operator (LASSO). Results: A total of 3066 DEGs in the GSE106180 and GSE161827 datasets. There were differences in immune cell infiltration. According to gene significance (GS) > 0.2 and module membership (MM) > 0.8, 41 yellow Module genes and 1474 turquoise Module genes were selected. Hub genes were mainly related to the "proteasomal protein catabolic process", "mitochondrial matrix" and "protein processing in endoplasmic reticulum" pathways. LASSO was used to construct a diagnostic model composed of OXCT1, CACNA2D2, BCL7B, EGLN3, GABARAP, and ACADSB and verified it in the GSE163060 and GSE175988 datasets with AUCs of 0.9333 (95% CI: 0.7801-1) and 0.96 (95% CI: 0.8861-1), respectively. H9C2 cells were verified, and the results were similar to the bioinformatics analysis. Conclusion: We constructed a diagnostic model of DCM, and OXCT1, CACNA2D2, BCL7B, EGLN3, GABARAP, and ACADSB were potential biomarkers, which may provide new insights for improving the ability of early diagnosis and treatment of diabetic cardiomyopathy.


Sujet(s)
Diabète , Cardiomyopathies diabétiques , Humains , Cardiomyopathies diabétiques/diagnostic , Cardiomyopathies diabétiques/génétique , Marqueurs biologiques , Aire sous la courbe , Analyse de regroupements , Biologie informatique , Facteurs de transcription
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...