Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.542
Filtrer
1.
Bull Exp Biol Med ; 177(2): 190-196, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39096448

RÉSUMÉ

This study aimed to investigate the cardioprotective effect of quinacrine in an in vivo model of myocardial ischemia/reperfusion injury. A 30-min regional myocardial ischemia followed by a 2-h reperfusion was modeled in anesthetized Wistar rats. Starting at the last minute of ischemia and during the first 9 min of reperfusion the rats in the control (n=8) and experimental (n=9) groups were injected with 0.9% NaCl and quinacrine solution (5 mg/kg), respectively. The area at risk and infarct size were evaluated by "double staining" with Evans blue and triphenyltetrazolium chloride. To assess vascular permeability in the area at risk zone, indocyanine green (ICG) and thioflavin S (ThS) were injected intravenously at the 90th and 120th minutes of reperfusion, respectively, to assess the no-reflow zone. The images of ICG and ThS fluorescence in transverse sections of rat hearts were obtained using a FLUM multispectral fluorescence organoscope. HR tended to decrease by 13% after intravenous administration of quinacrine and then recovered within 50 min. Quinacrine reduced the size of the necrotic zone (p=0.01), vascular permeability in the necrosis region, and the no-reflow area (p=0.027); at the same time, the area at risk did not significantly differ between the groups. Intravenous administration of quinacrine at the beginning of reperfusion of the rat myocardium reduces no-reflow phenomenon and infarct size.


Sujet(s)
Cardiotoniques , Lésion de reperfusion myocardique , Mépacrine , Rat Wistar , Animaux , Mépacrine/pharmacologie , Mépacrine/usage thérapeutique , Lésion de reperfusion myocardique/traitement médicamenteux , Lésion de reperfusion myocardique/anatomopathologie , Rats , Mâle , Cardiotoniques/pharmacologie , Cardiotoniques/usage thérapeutique , Modèles animaux de maladie humaine , Perméabilité capillaire/effets des médicaments et des substances chimiques , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/anatomopathologie , Myocarde/anatomopathologie
2.
BMC Cardiovasc Disord ; 24(1): 415, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39123142

RÉSUMÉ

BACKGROUND: Ischemia reperfusion (IR) causes impaired myocardial function, and autophagy activation ameliorates myocardial IR injury. Isoliquiritigenin (ISO) has been found to protect myocardial tissues via AMPK, with exerting anti-tumor property through autophagy activation. This study aims to investigate ISO capacity to attenuate myocardial IR through autophagy activation mediated by AMPK/mTOR/ULK1 signaling. METHODS: ISO effects were explored by SD rats and H9c2 cells. IR rats and IR-induced H9c2 cell models were established by ligating left anterior descending (LAD) coronary artery and hypoxia/re-oxygenation, respectively, followed by low, medium and high dosages of ISO intervention (Rats: 10, 20, and 40 mg/kg; H9c2 cells: 1, 10, and 100 µmol/L). Myocardial tissue injury in rats was assessed by myocardial function-related index, HE staining, Masson trichrome staining, TTC staining, and ELISA. Autophagy of H9c2 cells was detected by transmission electron microscopy (TEM) and immunofluorescence. Autophagy-related and AMPK/mTOR/ULK1 pathway-related protein expressions were detected with western blot. RESULTS: ISO treatment caused myocardial function improvement, and inhibition of myocardial inflammatory infiltration, fibrosis, infarct area, oxidative stress, CK-MB, cTnI, and cTnT expression in IR rats. In IR-modeled H9c2 cells, ISO treatment lowered apoptosis rate and activated autophagy and LC3 fluorescence expression. In vivo and in vitro, ISO intervention exhibited enhanced Beclin1, LC3II/LC3I, and p-AMPK/AMPK levels, whereas inhibited P62, p-mTOR/mTOR and p-ULK1(S757)/ULK1 protein expression, activating autophagy and protecting myocardial tissues from IR injury. CONCLUSION: ISO treatment may induce autophagy by regulating AMPK/mTOR/ULK1 signaling, thereby improving myocardial IR injury, as a potential candidate for treatment of myocardial IR injury.


Sujet(s)
AMP-Activated Protein Kinases , Homologue de la protéine-1 associée à l'autophagie , Autophagie , Chalcones , Modèles animaux de maladie humaine , Lésion de reperfusion myocardique , Myocytes cardiaques , Rat Sprague-Dawley , Transduction du signal , Sérine-thréonine kinases TOR , Animaux , Homologue de la protéine-1 associée à l'autophagie/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Lésion de reperfusion myocardique/traitement médicamenteux , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/physiopathologie , Lésion de reperfusion myocardique/enzymologie , Lésion de reperfusion myocardique/prévention et contrôle , Autophagie/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Chalcones/pharmacologie , Sérine-thréonine kinases TOR/métabolisme , AMP-Activated Protein Kinases/métabolisme , Lignée cellulaire , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/enzymologie , Myocytes cardiaques/métabolisme , Mâle , Rats , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/métabolisme , Infarctus du myocarde/physiopathologie , Infarctus du myocarde/enzymologie , Apoptose/effets des médicaments et des substances chimiques , Fibrose
3.
FASEB J ; 38(16): e23890, 2024 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-39143722

RÉSUMÉ

Thromboinflammation is a complex pathology associated with inflammation and coagulation. In cases of cardiovascular disease, in particular ischemia-reperfusion injury, thromboinflammation is a common complication. Increased understanding of thromboinflammation depends on an improved concept of the mechanisms of cells and proteins at the axis of coagulation and inflammation. Among these elements are activated protein C and platelets. This review summarizes the complex interactions of activated protein C and platelets regulating thromboinflammation in cardiovascular disease. By unraveling the pathways of platelets and APC in the inflammatory and coagulation cascades, this review summarizes the role of these vital mediators in the development and perpetuation of heart disease and the thromboinflammation-driven complications of cardiovascular disease. Furthermore, this review emphasizes the significance of the counteracting effects of platelets and APC and their combined role in disease states.


Sujet(s)
Coagulation sanguine , Plaquettes , Inflammation , Lésion de reperfusion myocardique , Protéine C , Humains , Plaquettes/métabolisme , Plaquettes/anatomopathologie , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Inflammation/métabolisme , Inflammation/anatomopathologie , Coagulation sanguine/physiologie , Protéine C/métabolisme , Animaux
4.
J Cell Mol Med ; 28(14): e18558, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39048917

RÉSUMÉ

Myocardial ischemia-reperfusion injury (MIRI) represents a critical pathology in acute myocardial infarction (AMI), which is characterized by high mortality and morbidity. Cardiac microvascular dysfunction contributes to MIRI, potentially culminating in heart failure (HF). Pigment epithelium-derived factor (PEDF), which belongs to the non-inhibitory serpin family, exhibits several physiological effects, including anti-angiogenesis, anti-inflammatory and antioxidant properties. Our study aims to explore the impact of PEDF and its functional peptide 34-mer on both cardiac microvascular perfusion in MIRI rats and human cardiac microvascular endothelial cells (HCMECs) injury under hypoxia reoxygenation (HR). It has been shown that MIRI is accompanied by ferroptosis in HCMECs. Furthermore, we investigated the effect of PEDF and its 34-mer, particularly regarding the Nrf2/HO-1 signalling pathway. Our results demonstrated that PEDF 34-mer significantly ameliorated cardiac microvascular dysfunction following MIRI. Additionally, they exhibited a notable suppression of ferroptosis in HCMECs, and these effects were mediated through activation of Nrf2/HO-1 signalling. These findings highlight the therapeutic potential of PEDF and 34-mer in alleviating microvascular dysfunction and MIRI. By enhancing cardiac microvascular perfusion and mitigating endothelial ferroptosis, PEDF and its derivative peptide represent promising candidates for the treatment of AMI.


Sujet(s)
Cellules endothéliales , Protéines de l'oeil , Ferroptose , Lésion de reperfusion myocardique , Facteur-2 apparenté à NF-E2 , Facteurs de croissance nerveuse , Serpines , Transduction du signal , Serpines/pharmacologie , Serpines/métabolisme , Facteurs de croissance nerveuse/pharmacologie , Facteurs de croissance nerveuse/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/traitement médicamenteux , Lésion de reperfusion myocardique/anatomopathologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Humains , Protéines de l'oeil/métabolisme , Protéines de l'oeil/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Rats , Heme oxygenase-1/métabolisme , Mâle , Rat Sprague-Dawley , Microvaisseaux/effets des médicaments et des substances chimiques , Microvaisseaux/métabolisme , Microvaisseaux/anatomopathologie , Peptides/pharmacologie
5.
Nat Commun ; 15(1): 6279, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39060225

RÉSUMÉ

The molecular mechanisms by which FoxO transcription factors mediate diametrically opposite cellular responses, namely death and survival, remain unknown. Here we show that Mst1 phosphorylates FoxO1 Ser209/Ser215/Ser218/Thr228/Ser232/Ser243, thereby inhibiting FoxO1-mediated transcription of proapoptotic genes. On the other hand, Mst1 increases FoxO1-C/EBP-ß interaction and activates C/EBP-ß by phosphorylating it at Thr299, thereby promoting transcription of prosurvival genes. Myocardial ischemia/reperfusion injury is larger in cardiac-specific FoxO1 knockout mice than in control mice. However, the concurrent presence of a C/EBP-ß T299E phospho-mimetic mutation reduces infarct size in cardiac-specific FoxO1 knockout mice. The C/EBP-ß phospho-mimetic mutant exhibits greater binding to the promoter of prosurvival genes than wild type C/EBP-ß. In conclusion, phosphorylation of FoxO1 by Mst1 inhibits binding of FoxO1 to pro-apoptotic gene promoters but enhances its binding to C/EBP-ß, phosphorylation of C/EBP-ß, and transcription of prosurvival genes, which stimulate protective mechanisms in the heart.


Sujet(s)
Protéine bêta de liaison aux séquences stimulatrices de type CCAAT , Protéine O1 à motif en tête de fourche , Lésion de reperfusion myocardique , Myocytes cardiaques , Animaux , Humains , Mâle , Souris , Rats , Apoptose , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/métabolisme , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/génétique , Protéine O1 à motif en tête de fourche/métabolisme , Protéine O1 à motif en tête de fourche/génétique , Facteur de croissance des hépatocytes/métabolisme , Souris knockout , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/génétique , Lésion de reperfusion myocardique/anatomopathologie , Lésion de reperfusion myocardique/prévention et contrôle , Myocytes cardiaques/métabolisme , Phosphorylation , Régions promotrices (génétique) , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protéines proto-oncogènes
6.
Sci Rep ; 14(1): 15246, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956068

RÉSUMÉ

This study aimed to explore the effects of peroxisome proliferator-activated receptor α (PPAR-α), a known inhibitor of ferroptosis, in Myocardial ischemia/reperfusion injury (MIRI) and its related mechanisms. In vivo and in vitro MIRI models were established. Our results showed that activation of PPAR-α decreased the size of the myocardial infarct, maintained cardiac function, and decreased the serum contents of creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH), and Fe2+ in ischemia/reperfusion (I/R)-treated mice. Additionally, the results of H&E staining, DHE staining, TUNEL staining, and transmission electron microscopy demonstrated that activation of PPAR-α inhibited MIRI-induced heart tissue and mitochondrial damage. It was also found that activation of PPAR-α attenuated MIRI-induced ferroptosis as shown by a reduction in malondialdehyde, total iron, and reactive oxygen species (ROS). In vitro experiments showed that intracellular contents of malondialdehyde, total iron, LDH, reactive oxygen species (ROS), lipid ROS, oxidized glutathione disulphide (GSSG), and Fe2+ were reduced by the activation of PPAR-α in H9c2 cells treated with anoxia/reoxygenation (A/R), while the cell viability and GSH were increased after PPAR-α activation. Additionally, changes in protein levels of the ferroptosis marker further confirmed the beneficial effects of PPAR-α activation on MIRI-induced ferroptosis. Moreover, the results of immunofluorescence and dual-luciferase reporter assay revealed that PPAR-α achieved its activity via binding to the 14-3-3η promoter, promoting its expression level. Moreover, the cardioprotective effects of PPAR-α could be canceled by pAd/14-3-3η-shRNA or Compound C11 (14-3-3η inhibitor). In conclusion, our results indicated that ferroptosis plays a key role in aggravating MIRI, and PPAR-α/14-3-3η pathway-mediated ferroptosis and mitochondrial injury might be an effective therapeutic target against MIRI.


Sujet(s)
Protéines 14-3-3 , Ferroptose , Lésion de reperfusion myocardique , Récepteur PPAR alpha , Animaux , Mâle , Souris , Rats , Protéines 14-3-3/métabolisme , Lignée cellulaire , Modèles animaux de maladie humaine , Ferroptose/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Récepteur PPAR alpha/métabolisme , Espèces réactives de l'oxygène/métabolisme , Régulation positive/effets des médicaments et des substances chimiques
7.
Cardiovasc Diabetol ; 23(1): 236, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970123

RÉSUMÉ

BACKGROUND: Owing to its unique location and multifaceted metabolic functions, epicardial adipose tissue (EAT) is gradually emerging as a new metabolic target for coronary artery disease risk stratification. Microvascular obstruction (MVO) has been recognized as an independent risk factor for unfavorable prognosis in acute myocardial infarction patients. However, the concrete role of EAT in the pathogenesis of MVO formation in individuals with ST-segment elevation myocardial infarction (STEMI) remains unclear. The objective of the study is to evaluate the correlation between EAT accumulation and MVO formation measured by cardiac magnetic resonance (CMR) in STEMI patients and clarify the underlying mechanisms involved in this relationship. METHODS: Firstly, we utilized CMR technique to explore the association of EAT distribution and quantity with MVO formation in patients with STEMI. Then we utilized a mouse model with EAT depletion to explore how EAT affected MVO formation under the circumstances of myocardial ischemia/reperfusion (I/R) injury. We further investigated the immunomodulatory effect of EAT on macrophages through co-culture experiments. Finally, we searched for new therapeutic strategies targeting EAT to prevent MVO formation. RESULTS: The increase of left atrioventricular EAT mass index was independently associated with MVO formation. We also found that increased circulating levels of DPP4 and high DPP4 activity seemed to be associated with EAT increase. EAT accumulation acted as a pro-inflammatory mediator boosting the transition of macrophages towards inflammatory phenotype in myocardial I/R injury through secreting inflammatory EVs. Furthermore, our study declared the potential therapeutic effects of GLP-1 receptor agonist and GLP-1/GLP-2 receptor dual agonist for MVO prevention were at least partially ascribed to its impact on EAT modulation. CONCLUSIONS: Our work for the first time demonstrated that excessive accumulation of EAT promoted MVO formation by promoting the polarization state of cardiac macrophages towards an inflammatory phenotype. Furthermore, this study identified a very promising therapeutic strategy, GLP-1/GLP-2 receptor dual agonist, targeting EAT for MVO prevention following myocardial I/R injury.


Sujet(s)
Tissu adipeux , Modèles animaux de maladie humaine , Récepteur du peptide-1 similaire au glucagon , Macrophages , Souris de lignée C57BL , Lésion de reperfusion myocardique , Péricarde , Infarctus du myocarde avec sus-décalage du segment ST , Animaux , Péricarde/métabolisme , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Mâle , Macrophages/métabolisme , Macrophages/anatomopathologie , Récepteur du peptide-1 similaire au glucagon/métabolisme , Récepteur du peptide-1 similaire au glucagon/agonistes , Infarctus du myocarde avec sus-décalage du segment ST/métabolisme , Infarctus du myocarde avec sus-décalage du segment ST/anatomopathologie , Infarctus du myocarde avec sus-décalage du segment ST/imagerie diagnostique , Tissu adipeux/métabolisme , Tissu adipeux/anatomopathologie , Humains , Femelle , Adulte d'âge moyen , Phénotype , Dipeptidyl peptidase 4/métabolisme , Sujet âgé , Techniques de coculture , Adiposité , Circulation coronarienne , Transduction du signal , Microcirculation , Vaisseaux coronaires/métabolisme , Vaisseaux coronaires/anatomopathologie , Vaisseaux coronaires/imagerie diagnostique , Incrétines/pharmacologie , Microvaisseaux/métabolisme , Microvaisseaux/anatomopathologie , Cellules cultivées , Souris ,
8.
Int Immunopharmacol ; 139: 112664, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39008937

RÉSUMÉ

PANoptosis is a newly discovered type of cell death characterized by pyroptosis, apoptosis and/or necroptosis and has been implicated in the inflammatory response. Piezo1 is a mechanosensitive ion channel that plays important roles in physiological development and various diseases. However, whether cardiomyocytes undergo PANoptosis during myocardial ischaemia/reperfusion (I/R) injury and the role of Piezo1 in this process remain largely unexplored. In this study, our results revealed that the expression levels of the main components of the PANoptosome, including caspase-8, caspase-3, NLRP3, caspase-1, GSDMD, RIPK1, RIPK3 and MLKL, were significantly upregulated in I/R heart tissues over time, indicating the occurrence of PANoptosis in I/R hearts. Accordingly, Piezo1 expression was significantly upregulated in I/R-injured hearts and hypoxia/reoxygenation (H/R)-treated cardiomyocytes. In contrast, pharmacological inhibition of Piezo1 by the inhibitor GsMTx4 in mice markedly attenuated the I/R-mediated decline in cardiac contractile function and increases in infarct size, apoptosis, oxidative stress and inflammation accompanied by the inhibition of PANoptosis-related mediators in I/R hearts. Consistently, the effects of Piezo1 on calcium influx and PANoptosis were further verified by GsMTx4 and Piezo1 activator Yoda1 in H/R-treated cardiomyocytes in vitro. Moreover, caspase-8 rather than calcium influx was required for H/R-induced PANoptosis in vitro. Mechanistically, Piezo1 interacts with caspase-8, a key initial activator of the PANoptosome complex, which subsequently activates cardiomyocyte PANoptosis, leading to cardiac dysfunction. In summary, these data suggest that Piezo1 is a new cardiac mechanosensor that promotes cardiac I/R injury possibly through the caspase-8-mediated activation of cardiomyocyte PANoptosis and highlight that Piezo1 may represent a new target for treating ischaemic heart disease.


Sujet(s)
Caspase 8 , Canaux ioniques , Souris de lignée C57BL , Lésion de reperfusion myocardique , Myocytes cardiaques , Animaux , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Caspase 8/métabolisme , Caspase 8/génétique , Canaux ioniques/métabolisme , Canaux ioniques/génétique , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Souris , Mâle , Nécroptose , Apoptose , Oligopeptides/pharmacologie , Venins d'araignée , Protéines et peptides de signalisation intercellulaire
9.
Oxid Med Cell Longev ; 2024: 3534104, 2024.
Article de Anglais | MEDLINE | ID: mdl-38957586

RÉSUMÉ

Myocardial infarction (MI) is irreversible damage to the myocardial tissue caused by prolonged ischemia/hypoxia, subsequently leading to loss of contractile function and myocardial damage. However, after a perilous period, ischemia-reperfusion (IR) itself causes the generation of oxygen free radicals, disturbance in cation homeostasis, depletion of cellular energy stores, and activation of innate and adaptive immune responses. The present study employed Abatacept (ABT), which is an anti-inflammatory drug, originally used as an antirheumatic response agent. To investigate the cardioprotective potential of ABT, primarily, the dose was optimized in a chemically induced model of myocardial necrosis. Thereafter, ABT optimized the dose of 5 mg/kg s.c. OD was investigated for its cardioprotective potential in a surgical model of myocardial IR injury, where animals (n = 30) were randomized into five groups: Sham, IR-C, Telmi10 + IR (Telmisartan, 10 mg/kg oral OD), ABT5 + IR, ABT perse. ABT and telmisartan were administered for 21 days. On the 21st day, animals were subjected to LAD coronary artery occlusion for 60 min, followed by reperfusion for 45 min. Further, the cardioprotective potential was assessed through hemodynamic parameters, oxidant-antioxidant biochemical enzymatic parameters, cardiac injury, inflammatory markers, histopathological analysis, TUNEL assay, and immunohistochemical evaluation, followed by immunoblotting to explore signaling pathways. The statistics were performed by one-way analysis of variance, followed by the Tukey comparison post hoc tests. Noteworthy, 21 days of ABT pretreatment amended the hemodynamic and ventricular functions in the rat models of MI. The cardioprotective potential of ABT is accompanied by inhibiting MAP kinase signaling and modulating Nrf-2/HO-1 proteins downstream signaling cascade. Overall, the present work bolsters the previously known anti-inflammatory role of ABT in MI and contributes a mechanistic insight and application of clinically approved drugs in averting the activation of inflammatory response.


Sujet(s)
Abatacept , Modèles animaux de maladie humaine , Inflammation , Infarctus du myocarde , Animaux , Rats , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/anatomopathologie , Mâle , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Abatacept/pharmacologie , Abatacept/usage thérapeutique , Rat Wistar , Lésion de reperfusion myocardique/traitement médicamenteux , Lésion de reperfusion myocardique/anatomopathologie
10.
PeerJ ; 12: e17333, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948204

RÉSUMÉ

Acute heart attack is the primary cause of cardiovascular-related death worldwide. A common treatment is reperfusion of ischemic tissue, which can cause irreversible damage to the myocardium. The number of mitochondria in cardiomyocytes is large, which generate adenosine triphosphate (ATP) to sustain proper cardiac contractile function, and mitochondrial dysfunction plays a crucial role in cell death during myocardial ischemia-reperfusion, leading to an increasing number of studies investigating the impact of mitochondria on ischemia-reperfusion injury. The disarray of mitochondrial dynamics, excessive Ca2+ accumulation, activation of mitochondrial permeable transition pores, swelling of mitochondria, ultimately the death of cardiomyocyte are the consequences of ischemia-reperfusion injury. κ-opioid receptors can alleviate mitochondrial dysfunction, regulate mitochondrial dynamics, mitigate myocardial ischemia-reperfusion injury, exert protective effects on myocardium. The mechanism of κ-OR activation during myocardial ischemia-reperfusion to regulate mitochondrial dynamics and reduce myocardial ischemia-reperfusion injury will be discussed, so as to provide theoretical basis for the protection of ischemic myocardium.


Sujet(s)
Lésion de reperfusion myocardique , Myocytes cardiaques , Récepteur kappa , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Récepteur kappa/métabolisme , Humains , Animaux , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/anatomopathologie , Dynamique mitochondriale/physiologie , Calcium/métabolisme
11.
J Vasc Res ; 61(4): 179-196, 2024.
Article de Anglais | MEDLINE | ID: mdl-38952123

RÉSUMÉ

INTRODUCTION: The comorbidities of ischemic heart disease (IHD) and diabetes mellitus (DM) compromise the protection of the diabetic heart from ischemia/reperfusion (I/R) injury. We hypothesized that manipulation of reperfusion injury salvage kinase (RISK) and survivor activating factor enhancement (SAFE) pathways might protect the diabetic heart, and intervention of these pathways could be a new avenue for potentially protecting the diabetic heart. METHODS: All hearts were subjected to 30-min ischemia and 30-min reperfusion. During reperfusion, hearts were exposed to molecules proven to protect the heart from I/R injury. The hemodynamic data were collected using suitable software. The infarct size, troponin T levels, and protein levels in hearts were evaluated. RESULTS: Both cyclosporine-A and nitric oxide donor (SNAP) infusion at reperfusion protected 4-week diabetic hearts from I/R injury. However, 6-week diabetic hearts were protected only by SNAP, but not cyclosporin-A. These treatments significantly (p < 0.05) improved cardiac hemodynamics and decreased infarct size. CONCLUSIONS: The administration of SNAP to diabetic hearts protected both 4- and 6-week diabetic hearts; however, cyclosporine-A protected only the 4-week diabetic hearts. The eNOS/GLUT-4 pathway executed the SNAP-mediated cardioprotection.


Sujet(s)
Ciclosporine , Diabète expérimental , Lésion de reperfusion myocardique , Myocarde , Donneur d'oxyde nitrique , Monoxyde d'azote , Transduction du signal , Animaux , Lésion de reperfusion myocardique/prévention et contrôle , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Lésion de reperfusion myocardique/physiopathologie , Monoxyde d'azote/métabolisme , Diabète expérimental/complications , Mâle , Ciclosporine/pharmacologie , Donneur d'oxyde nitrique/pharmacologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/métabolisme , Infarctus du myocarde/prévention et contrôle , Glycémie/métabolisme , Glycémie/effets des médicaments et des substances chimiques , Facteurs temps , Rat Sprague-Dawley , Troponine T/métabolisme , Hyperglycémie/métabolisme , Hyperglycémie/complications , Transporteur de glucose de type 4
13.
Tissue Cell ; 89: 102428, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38878657

RÉSUMÉ

BACKGROUND: Myocardial ischemia-reperfusion (MI/R) occurs due to temporary or permanent interruptions in the coronary and circulatory system, indirectly affecting kidney function through reduced cardiac output for metabolic needs. In this study, the aim was to explore the indirect effects of using human amniotic membrane mesenchymal stem cells (hAMSCs) with the PGS-co-PCL/PGC/PPy/Gelatin scaffold in male rats with renal failure induced by miocardial ischemia-reperfusion. METHODS: MI/R injury was induced in 48 male Wistar rats through left anterior descending artery ligation, divided into four groups (n=12); control group, cell group, scaffold group, and celss+scaffold group. Evaluations were conducted at two and thirty days post MI/R injury, encompassing echocardiography, biochemical, inflammatory markers analysis, and histological assessment. RESULTS: Echocardiographic findings exhibited notable enhancement in ejection fraction, fractional shortening, and stroke volume of treated groups compared to controls after 30 days (P< 0.05). Serum creatinine (P< 0.001) and urea (P< 0.05) levels significantly decreased in the scaffold+cells group) compared to the control group. The treated cells+ scaffold group displayed improved kidney structure, evidenced by larger glomeruli and reduced Bowman's space compared to the control group (P< 0.01). Immunohistochemical analysis indicated reduced TNF-α protein in the scaffold+ cells group (P< 0.05) in contrast to the control group (P< 0.05). Inflammatory factors IL-6, TNF-α, and AKT gene expression in renal tissues were improved in scaffold+ cells-treated animals. CONCLUSION: Our research proposes the combination of hAMSCs and the PGS-co-PCL/PGC/PPy/Gelatin scaffold in MI/R injured rats appears to enhance renal function and reduce kidney inflammation by improving cardiac output.


Sujet(s)
Rat Wistar , Structures d'échafaudage tissulaires , Animaux , Mâle , Rats , Structures d'échafaudage tissulaires/composition chimique , Humains , Gélatine/pharmacologie , Lésion de reperfusion myocardique/anatomopathologie , Insuffisance rénale/anatomopathologie , Insuffisance rénale/étiologie , Insuffisance rénale/thérapie , Cellules souches mésenchymateuses/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Rein/anatomopathologie
14.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38892398

RÉSUMÉ

Myocardial infarction activates an intense fibro-inflammatory reaction that is essential for cardiac remodeling and heart failure (HF). Bioactive peptide galanin plays a critical role in regulating cardiovascular homeostasis; however, its specific functional relevance in post-infarction fibro-inflammatory reprogramming remains obscure. Here, we show that galanin coordinates the fibro-inflammatory trajectory and mitochondrial integrity in post-infarction reperfusion injury. Aberrant deposition of collagen was associated with a marked increase in CD68-positive macrophage infiltration in cardiac tissue in mice subjected to myocardial ischemia/reperfusion (I/R) for 14 days compared to sham controls. Furthermore, we found that the myocardial expression level of a specific marker of M2 macrophages, CD206, was significantly down-regulated in I/R-challenged mice. In contrast, galanin treatment started during the reperfusion phase blunted the fibro-inflammatory responses and promoted the expression of CD206 in I/R-remodeled hearts. In addition, we found that the anti-apoptotic and anti-hypertrophic effects of galanin were associated with the preservation of mitochondrial integrity and promotion of mitochondrial biogenesis. These findings depict galanin as a key arbitrator of fibro-inflammatory responses to cardiac I/R injury and offer a promising therapeutic trajectory for the treatment of post-infarct cardiovascular complications.


Sujet(s)
Galanine , Macrophages , Lésion de reperfusion myocardique , Animaux , Galanine/métabolisme , Galanine/pharmacologie , Souris , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Macrophages/métabolisme , Mâle , Infarctus du myocarde/métabolisme , Infarctus du myocarde/anatomopathologie , Mitochondries/métabolisme , Souris de lignée C57BL , Récepteurs de surface cellulaire/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Récepteur du mannose , Lectines de type C/métabolisme , Myocarde/métabolisme , Myocarde/anatomopathologie , Lectines liant le mannose/métabolisme , Modèles animaux de maladie humaine , Apoptose
15.
J Physiol Investig ; 67(3): 129-138, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38902960

RÉSUMÉ

Ischemia-reperfusion (IR) injury remains a pivotal contributor to myocardial damage following acute coronary events and revascularization procedures. Phosphoinositide 3-kinase (PI3K), a key mediator of cell survival signaling, plays a central role in regulating inflammatory responses and cell death mechanisms. Trans-chalcone (Tch), a natural compound known for its anti-inflammatory activities, has shown promise in various disease models. The aim of the current study was to investigate the potential protective effects of Tch against myocardial injury induced by ischemia and reperfusion challenges by targeting the PI3K-inflammasome interaction. Experimental models utilizing male rats subjected to an in vivo model of IR injury and myocardial infarction were employed. Administration of Tch (100 µg/kg, intraperitoneally) significantly reduced myocardial injury, as indicated by limited infarct size and decreased levels of the myocardial enzyme troponin. Mechanistically, Tch upregulated PI3K expression, thereby inhibiting the activity of the NOD-like receptor protein 3 inflammasome followed by the activation of pro-inflammatory cytokines interleukin-1ß (IL-1ß) and IL-18. Moreover, it mitigated oxidative stress and suppressed vascular-intercellular adhesion molecules, contributing to its cardioprotective effects. The PI3K/Akt pathway inhibitor LY294002 considerably attenuated the beneficial effects of Tch. These findings highlight the therapeutic potential of Tch in ameliorating myocardial injury associated with IR insults through its modulation of the PI3K/Akt-inflammasome axis. The multifaceted mechanisms underlying its protective effects signify Tch as a promising candidate for further exploration in developing targeted therapies aimed at mitigating ischemic heart injury and improving clinical outcomes in cardiovascular diseases characterized by IR injury.


Sujet(s)
Lésion de reperfusion myocardique , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Rat Sprague-Dawley , Animaux , Mâle , Lésion de reperfusion myocardique/traitement médicamenteux , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/prévention et contrôle , Lésion de reperfusion myocardique/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Rats , Phosphatidylinositol 3-kinases/métabolisme , Inflammasomes/métabolisme , Inflammasomes/effets des médicaments et des substances chimiques , Chalcone/pharmacologie , Chalcone/analogues et dérivés , Chalcone/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme
16.
Biomed Pharmacother ; 176: 116869, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38850665

RÉSUMÉ

OBJECTIVE: Integrated stress response (ISR) is activated to promote cell survival by maintaining the phosphorylation of eukaryotic translation initiation factor 2 (eIF2α). We investigated whether Sephin1 enhances ISR and attenuates myocardial ischemia-reperfusion (MIR) injury. METHODS: Male C57BL/6 J mice were injected with Sephin1 (2 mg/kg,i.p.) 30 min before surgery to establish a model of MIR with 45 min ischemia and 180 min reperfusion. In vitro, the H9C2 cell line with hypoxia-reoxygenation (H/R) was used to simulate MIR. Myocardial injury was evaluated by echocardiography, histologic observation after staining with TTC and H&E and electron microscopy. ISR, autophagy and apoptosis in vivo and in vitro were evaluated by immunoblotting, immunohistochemistry, immunofluorescence, and flow cytometry, respectively. Global protein synthesis was determined using a non-radioactive SUnSET Assay based on the puromycin method. Autophinib, an autophagy-specific inhibitor, was used to investigate the correlation between autophagy and apoptosis in the presence of Sephin1. RESULTS: In vivo, Sephin1 significantly reduced myocardial injury and improved the cardiac function in MIR mice. Sephin1 administration prolonged ISR, reduced cell apoptosis, and promoted autophagy. In vitro, Sephin1 increased the number of stress granules (SGs) and autophagic vesicles, enhanced ISR and related protein synthesis suppression, and reduced cell apoptosis. Autophinib partly reversed autophagosome formation and apoptosis in H9c2 cells. CONCLUSIONS: Sephin1 enhances ISR and related protein synthesis suppression, ameliorates myocardial apoptosis, and promotes autophagy during MIR stress. Sephin1 could act as a noval ISR enhancer for managing acute myocardial ischemia disease.


Sujet(s)
Apoptose , Autophagie , Souris de lignée C57BL , Lésion de reperfusion myocardique , Animaux , Autophagie/effets des médicaments et des substances chimiques , Mâle , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Lésion de reperfusion myocardique/traitement médicamenteux , Souris , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Modèles animaux de maladie humaine , Érythropoïétine , Fragments peptidiques
17.
Biomed Pharmacother ; 176: 116927, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38870633

RÉSUMÉ

Echinops plants have received great attention for the treatment of many diseases due to pharmacological properties such as their antidiabetic, antioxidant, and anti-inflammatory characteristics. The major purpose of the present study was to investigate the cardioprotective benefits of Echinops cephalotes (Ech) against myocardial ischemia-reperfusion (MI/R) injury. Male Wistar rats were randomly allocated to three groups: sham, MI, and MI + Ech. The left coronary artery (LAD) was blocked for 30 minutes to induce MI. In the treatment group, rats were given 150 mg/kg/day of Ech extract for 28 days. Aqueous extracts were made from Echinops plants. To study heart function, fibrosis, cardiac damage indicators, and oxidative stress factors, echocardiography, Masson's trichrome staining, and biochemical tests were used. The expression of matrix metalloproteinase 2 and 9 (MMP2 and MMP-9) and tissue inhibitor of metalloproteinase (TIMP) was determined using Western blotting. Tissue damage was assessed using hematoxylin and eosin staining. MI group exhibited significantly reduced ejection fraction (EF) and fractional shortening (FS), enhanced levels of lactate dehydrogenase (LDH), creatine kinase MB (CK-MB), cardiac Troponin I (cTnI), and malondialdehyde (MDA), as well as a decrease in the Glutathione (GSH) tissue content, reduced activity of superoxide dismutase (SOD), increasing fibrosis, upregulations of MMP-2 and MMP-9, and reduction of TIMP compared to the sham group. The findings suggest that Ech in particular, could be a promising therapeutic agent to reduce the damage in MI by targeting oxidative stress and modulating the activities of matrix metalloproteinases and their tissue inhibitors.


Sujet(s)
Cardiotoniques , Matrix metalloproteinase 2 , Matrix metalloproteinase 9 , Lésion de reperfusion myocardique , Stress oxydatif , Extraits de plantes , Rat Wistar , Animaux , Mâle , Stress oxydatif/effets des médicaments et des substances chimiques , Matrix metalloproteinase 2/métabolisme , Extraits de plantes/pharmacologie , Extraits de plantes/isolement et purification , Matrix metalloproteinase 9/métabolisme , Lésion de reperfusion myocardique/traitement médicamenteux , Lésion de reperfusion myocardique/anatomopathologie , Lésion de reperfusion myocardique/métabolisme , Cardiotoniques/pharmacologie , Cardiotoniques/isolement et purification , Rats , Myocarde/anatomopathologie , Myocarde/métabolisme , Inhibiteur tissulaire des métalloprotéinases/métabolisme , Fibrose , Eau/composition chimique , Antioxydants/pharmacologie
18.
Clin Exp Pharmacol Physiol ; 51(8): e13904, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38923060

RÉSUMÉ

Myocardial ischemia-reperfusion injury (MIRI) is a common clinic scenario that occurs in the context of reperfusion therapy for acute myocardial infarction. It has been shown that cocaine and amphetamine-regulated transcript (CART) can ameliorate cerebral ischemia-reperfusion (I/R) injury, but the effect of CART on MIRI has not been studied yet. Here, we revealed that CART protected the heart during I/R process by inhibiting apoptosis and excessive autophagy, indicating that CART would be a potential drug candidate for the treatment of MIRI. Further analysis showed that CART upregulated the activation of phospho-AKT, leading to downregulation of lactate dehydrogenase (LDH) release, apoptosis, oxidative stress and excessive autophagy after I/R, which was inhibited by PI3K inhibitor, LY294002. Collectively, CART attenuated MIRI through inhibition of cardiomyocytes apoptosis and excessive autophagy, and the protective effect was dependent on PI3K/AKT signalling pathway.


Sujet(s)
Apoptose , Autophagie , Lésion de reperfusion myocardique , Protéines de tissu nerveux , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Lésion de reperfusion myocardique/traitement médicamenteux , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Protéines de tissu nerveux/métabolisme , Mâle , Autophagie/effets des médicaments et des substances chimiques , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Rats , Stress oxydatif/effets des médicaments et des substances chimiques , Rat Sprague-Dawley
19.
Basic Res Cardiol ; 119(4): 691-697, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38864895

RÉSUMÉ

The mitochondrial metabolite succinate is a key driver of ischemia/reperfusion injury (IRI). Targeting succinate metabolism by inhibiting succinate dehydrogenase (SDH) upon reperfusion using malonate is an effective therapeutic strategy to achieve cardioprotection in the short term (< 24 h reperfusion) in mouse and pig in vivo myocardial infarction (MI) models. We aimed to assess whether inhibiting IRI with malonate given upon reperfusion could prevent post-MI heart failure (HF) assessed after 28 days. Male C57BL/6 J mice were subjected to 30 min left anterior coronary artery (LAD) occlusion, before reperfusion for 28 days. Malonate or without-malonate control was infused as a single dose upon reperfusion. Cardiac function was assessed by echocardiography and fibrosis by Masson's trichrome staining. Reperfusion without malonate significantly reduced ejection fraction (~ 47%), fractional shortening (~ 23%) and elevated collagen deposition 28 days post-MI. Malonate, administered as a single infusion (16 mg/kg/min for 10 min) upon reperfusion, gave a significant cardioprotective effect, with ejection fraction (~ 60%) and fractional shortening (~ 30%) preserved and less collagen deposition. Using an acidified malonate formulation, to enhance its uptake into cardiomyocytes via the monocarboxylate transporter 1, both 1.6 and 16 mg/kg/min 10 min infusion led to robust long-term cardioprotection with preserved ejection fraction (> 60%) and fractional shortening (~ 30%), as well as significantly less collagen deposition than control hearts. Malonate administration upon reperfusion prevents post-MI HF. Acidification of malonate enables lower doses of malonate to also achieve long-term cardioprotection post-MI. Therefore, the administration of acidified malonate upon reperfusion is a promising therapeutic strategy to prevent IRI and post-MI HF.


Sujet(s)
Modèles animaux de maladie humaine , Défaillance cardiaque , Malonates , Souris de lignée C57BL , Infarctus du myocarde , Lésion de reperfusion myocardique , Animaux , Malonates/pharmacologie , Mâle , Infarctus du myocarde/métabolisme , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/prévention et contrôle , Infarctus du myocarde/physiopathologie , Lésion de reperfusion myocardique/prévention et contrôle , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Lésion de reperfusion myocardique/physiopathologie , Défaillance cardiaque/prévention et contrôle , Défaillance cardiaque/métabolisme , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/physiopathologie , Défaillance cardiaque/étiologie , Souris , Myocarde/métabolisme , Myocarde/anatomopathologie , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Fibrose , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Facteurs temps
20.
Basic Res Cardiol ; 119(4): 509-544, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38878210

RÉSUMÉ

Despite recent progress, ischemic heart disease poses a persistent global challenge, driving significant morbidity and mortality. The pursuit of therapeutic solutions has led to the emergence of strategies such as ischemic preconditioning, postconditioning, and remote conditioning to shield the heart from myocardial ischemia/reperfusion injury (MIRI). These ischemic conditioning approaches, applied before, after, or at a distance from the affected organ, inspire future therapeutic strategies, including pharmacological conditioning. Gasotransmitters, comprising nitric oxide, hydrogen sulfide, sulfur dioxide, and carbon monoxide, play pivotal roles in physiological and pathological processes, exhibiting shared features such as smooth muscle relaxation, antiapoptotic effects, and anti-inflammatory properties. Despite potential risks at high concentrations, physiological levels of gasotransmitters induce vasorelaxation and promote cardioprotective effects. Noble gases, notably argon, helium, and xenon, exhibit organ-protective properties by reducing cell death, minimizing infarct size, and enhancing functional recovery in post-ischemic organs. The protective role of noble gases appears to hinge on their modulation of molecular pathways governing cell survival, leading to both pro- and antiapoptotic effects. Among noble gases, helium and xenon emerge as particularly promising in the field of cardioprotection. This overview synthesizes our current understanding of the roles played by gasotransmitters and noble gases in the context of MIRI and cardioprotection. In addition, we underscore potential future developments involving the utilization of noble gases and gasotransmitter donor molecules in advancing cardioprotective strategies.


Sujet(s)
Gazotransmetteurs , Lésion de reperfusion myocardique , Gaz rares , Humains , Gazotransmetteurs/métabolisme , Gazotransmetteurs/usage thérapeutique , Animaux , Lésion de reperfusion myocardique/prévention et contrôle , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Lésion de reperfusion myocardique/physiopathologie , Gaz rares/métabolisme , Préconditionnement ischémique myocardique , Transduction du signal , Cardiotoniques/pharmacologie , Cardiotoniques/usage thérapeutique , Ischémie myocardique/métabolisme , Ischémie myocardique/physiopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE