Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 161.458
Filtrer
1.
Sci Transl Med ; 16(756): eadm8842, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39018366

RÉSUMÉ

Heart failure with preserved ejection fraction (HFpEF) is a complex syndrome associated with increased myocardial stiffness and cardiac filling abnormalities. Prior studies implicated increased α-tubulin detyrosination, which is catalyzed by the vasohibin enzymes, as a contributor to increased stabilization of the cardiomyocyte microtubule network (MTN) and stiffness in failing human hearts. We explored whether increased MTN detyrosination contributed to impaired diastolic function in the ZSF1 obese rat model of HFpEF and designed a small-molecule vasohibin inhibitor to ablate MTN detyrosination in vivo. Compared with ZSF1 lean and Wistar Kyoto rats, obese rats exhibited increased tubulin detyrosination concomitant with diastolic dysfunction, left atrial enlargement, and cardiac hypertrophy with a preserved left ventricle ejection fraction, consistent with an HFpEF phenotype. Ex vivo myocardial phenotyping assessed cardiomyocyte mechanics and contractility. Vasohibin inhibitor treatment of isolated cardiomyocytes from obese rats resulted in reduced stiffness and faster relaxation. Acute in vivo treatment with vasohibin inhibitor improved diastolic relaxation in ZSF1 obese rats compared with ZSF1 lean and Wistar Kyoto rats. Vasohibin inhibition also improved relaxation in isolated human cardiomyocytes from both failing and nonfailing hearts. Our data suggest the therapeutic potential for vasohibin inhibition to reduce myocardial stiffness and improve relaxation in HFpEF.


Sujet(s)
Modèles animaux de maladie humaine , Défaillance cardiaque , Myocytes cardiaques , Débit systolique , Animaux , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/physiopathologie , Défaillance cardiaque/anatomopathologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Débit systolique/effets des médicaments et des substances chimiques , Rats de lignée WKY , Rats , Mâle , Humains , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Diastole/effets des médicaments et des substances chimiques , Tubuline/métabolisme , Myocarde/anatomopathologie , Myocarde/métabolisme , Obésité/traitement médicamenteux , Obésité/physiopathologie
3.
Scand Cardiovasc J ; 58(1): 2373083, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39024033

RÉSUMÉ

OBJECTIVE: This paper was performed to decipher the serum microRNA (miR)-125b-5p expression in patients with dilated cardiomyopathy (DCM) combined with heart failure (HF) and its effect on myocardial fibrosis. METHODS: Serum miR-125b-5p expression, LVEDD, LVESD, LVEF, LVFS, and NT-proBNP levels were evaluated in clinical samples. A rat DCM model was established by continuous intraperitoneal injection of adriamycin and treated with miR-125b-5p agomir and its negative control. Cardiac function, serum TNF-α, hs-CRP, and NT-proBNP levels, pathological changes in myocardial tissues, cardiomyocyte apoptosis, and the expression levels of miR-125b-5p and fibrosis-related factors were detected in rats. RESULTS: In comparison to the control group, the case group had higher levels of LVEDD, LVESD, and NT-pro-BNP, and lower levels of LVEF, LVFS, and miR-125b-5p expression levels. Overexpression of miR-125b-5p effectively led to the improvement of cardiomyocyte hypertrophy and collagen arrangement disorder in DCM rats, the reduction of blue-stained collagen fibers in the interstitial myocardium, the reduction of the levels of TNF-α, hs-CRP, and NT-proBNP and the expression levels of TGF-1ß, Collagen I, and α-SMA, and the reduction of the number of apoptosis in cardiomyocytes. CONCLUSION: Overexpression of miR-125b-5p is effective in ameliorating myocardial fibrosis.


Sujet(s)
Apoptose , Cardiomyopathie dilatée , Modèles animaux de maladie humaine , Fibrose , Défaillance cardiaque , microARN , Myocarde , Fonction ventriculaire gauche , Animaux , Cardiomyopathie dilatée/génétique , Cardiomyopathie dilatée/sang , Cardiomyopathie dilatée/anatomopathologie , microARN/sang , microARN/génétique , microARN/métabolisme , Défaillance cardiaque/sang , Défaillance cardiaque/génétique , Défaillance cardiaque/métabolisme , Défaillance cardiaque/anatomopathologie , Mâle , Humains , Myocarde/anatomopathologie , Myocarde/métabolisme , Adulte d'âge moyen , Femelle , Études cas-témoins , Rat Sprague-Dawley , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/métabolisme , Peptide natriurétique cérébral/sang , Peptide natriurétique cérébral/génétique , Remodelage ventriculaire , Fragments peptidiques/sang , Adulte , MicroARN circulant/sang , MicroARN circulant/génétique , Sujet âgé , Débit systolique
4.
Front Immunol ; 15: 1371477, 2024.
Article de Anglais | MEDLINE | ID: mdl-39007149

RÉSUMÉ

Background: A high-fat diet (HFD) contributes to various metabolic disorders and obesity, which are major contributors to cardiovascular disease. As an essential regulator for heart homeostasis, cardiac resident macrophages may go awry and contribute to cardiac pathophysiology upon HFD. Thus, to better understand how HFD induced cardiac dysfunction, this study intends to explore the transcriptional and functional changes in cardiac resident macrophages of HFD mice. Methods: C57BL/6J female mice that were 6 weeks old were fed with HFD or normal chow diet (NCD) for 16 weeks. After an evaluation of cardiac functions by echocardiography, mouse hearts were harvested and cardiac resident CCR2- macrophages were sorted, followed by Smart sequencing. Bioinformatics analysis including GO, KEGG, and GSEA analyses were employed to elucidate transcriptional and functional changes. Results: Hyperlipidemia and obesity were observed easily upon HFD. The mouse hearts also displayed more severe fibrosis and diastolic dysfunction in HFD mice. Smart sequencing and functional analysis revealed metabolic dysfunctions, especially lipid-related genes and pathways. Besides this, antigen-presentation-related gene such as Ctsf and inflammation, particularly for NF-κB signaling and complement cascades, underwent drastic changes in cardiac resident macrophages. GO cellular compartment analysis was also performed and showed specific organelle enrichment trends of the involved genes. Conclusion: Dysregulated metabolism intertwines with inflammation in cardiac resident macrophages upon HFD feeding in mice, and further research on crosstalk among organelles could shed more light on potential mechanisms.


Sujet(s)
Alimentation riche en graisse , Macrophages , Souris de lignée C57BL , Myocarde , Animaux , Alimentation riche en graisse/effets indésirables , Souris , Macrophages/immunologie , Macrophages/métabolisme , Femelle , Myocarde/métabolisme , Myocarde/immunologie , Obésité/immunologie , Obésité/métabolisme , Hyperlipidémies/immunologie , Hyperlipidémies/métabolisme
5.
J Clin Invest ; 134(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38949027

RÉSUMÉ

Biological sex is an important modifier of physiology and influences pathobiology in many diseases. While heart disease is the number one cause of death worldwide in both men and women, sex differences exist at the organ and cellular scales, affecting clinical presentation, diagnosis, and treatment. In this Review, we highlight baseline sex differences in cardiac structure, function, and cellular signaling and discuss the contribution of sex hormones and chromosomes to these characteristics. The heart is a remarkably plastic organ and rapidly responds to physiological and pathological cues by modifying form and function. The nature and extent of cardiac remodeling in response to these stimuli are often dependent on biological sex. We discuss organ- and molecular-level sex differences in adaptive physiological remodeling and pathological cardiac remodeling from pressure and volume overload, ischemia, and genetic heart disease. Finally, we offer a perspective on key future directions for research into cardiac sex differences.


Sujet(s)
Caractères sexuels , Remodelage ventriculaire , Humains , Femelle , Mâle , Animaux , Cardiopathies/anatomopathologie , Cardiopathies/métabolisme , Cardiopathies/physiopathologie , Cardiopathies/génétique , Hormones sexuelles stéroïdiennes/métabolisme , Coeur/physiopathologie , Coeur/physiologie , Myocarde/anatomopathologie , Myocarde/métabolisme
6.
Georgian Med News ; (349): 54-59, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38963202

RÉSUMÉ

Doxorubicin is the common chemotherapeutic agent that has been harnessed for the treatment of various types of malignancy including the treatment of soft tissue and osteosarcoma and cancers of the vital organs like breast, ovary, bladder, and thyroid. It is also used to treat leukaemia and lymphoma, however, this is an obstacle because of their prominent side effects including cardiotoxicity and lung fibrosis, we do aim to determine the role of CoQ10 as an antioxidant on the impeding the deleterious impacts of doxorubicin on tissue degenerative effects. To do so, 27 rats were subdivided into 3 groups of 9 each; CoQ10 exposed group, Doxorubicin exposed group, and CoQ10 plus Doxorubicin group. At the end of the study, the animals were sacrificed and lungs with hearts were harvested, and slides were prepared for examination under a microscope. The results indicated that doxorubicin induced abnormal cellular structure resulting in damaging cellular structures of the lung and heart while CoQ10 impeded these damaging effects and nearly restoring normal tissue structure. As a result, CoQ10 will maintain normal tissue of the lung and heart.


Sujet(s)
Doxorubicine , Poumon , Ubiquinones , Animaux , Doxorubicine/effets indésirables , Ubiquinones/analogues et dérivés , Ubiquinones/pharmacologie , Rats , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Antibiotiques antinéoplasiques/effets indésirables , Antibiotiques antinéoplasiques/toxicité , Myocarde/anatomopathologie , Mâle , Antioxydants/pharmacologie , Cardiotoxicité/étiologie , Cardiotoxicité/anatomopathologie , Coeur/effets des médicaments et des substances chimiques
7.
Theranostics ; 14(9): 3486-3508, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948064

RÉSUMÉ

Rationale: Device implantation frequently triggers cardiac remodeling and fibrosis, with monocyte-driven inflammatory responses precipitating arrhythmias. This study investigates the role of m6A modification enzymes METTL3 and METTL14 in these responses and explores a novel therapeutic strategy targeting these modifications to mitigate cardiac remodeling and fibrosis. Methods: Peripheral blood mononuclear cells (PBMCs) were collected from patients with ventricular septal defects (VSD) who developed conduction blocks post-occluder implantation. The expression of METTL3 and METTL14 in PBMCs was measured. METTL3 and METTL14 deficiencies were induced to evaluate their effect on angiotensin II (Ang II)-induced myocardial inflammation and fibrosis. m6A modifications were analyzed using methylated RNA immunoprecipitation followed by quantitative PCR. NF-κB pathway activity and levels of monocyte migration and fibrogenesis markers (CXCR2 and TGF-ß1) were assessed. An erythrocyte microvesicle-based nanomedicine delivery system was developed to target activated monocytes, utilizing the METTL3 inhibitor STM2457. Cardiac function was evaluated via echocardiography. Results: Significant upregulation of METTL3 and METTL14 was observed in PBMCs from patients with VSD occluder implantation-associated persistent conduction block. Deficiencies in METTL3 and METTL14 significantly reduced Ang II-induced myocardial inflammation and fibrosis by decreasing m6A modification on MyD88 and TGF-ß1 mRNAs. This disruption reduced NF-κB pathway activation, lowered CXCR2 and TGF-ß1 levels, attenuated monocyte migration and fibrogenesis, and alleviated cardiac remodeling. The erythrocyte microvesicle-based nanomedicine delivery system effectively targeted inflamed cardiac tissue, reducing inflammation and fibrosis and improving cardiac function. Conclusion: Inhibiting METTL3 and METTL14 in monocytes disrupts the NF-κB feedback loop, decreases monocyte migration and fibrogenesis, and improves cardiac function. Targeting m6A modifications of monocytes with STM2457, delivered via erythrocyte microvesicles, reduces inflammation and fibrosis, offering a promising therapeutic strategy for cardiac remodeling associated with device implantation.


Sujet(s)
Fibrose , Methyltransferases , Monocytes , Facteur de transcription NF-kappa B , Humains , Methyltransferases/métabolisme , Methyltransferases/génétique , Monocytes/métabolisme , Mâle , Animaux , Facteur de transcription NF-kappa B/métabolisme , Érythrocytes/métabolisme , Adénosine/analogues et dérivés , Adénosine/métabolisme , Femelle , Méthylation , Souris , Facteur de croissance transformant bêta-1/métabolisme , Microparticules membranaires/métabolisme , Agranulocytes/métabolisme , Angiotensine-II/métabolisme , Récepteurs à l'interleukine-8B/métabolisme , Récepteurs à l'interleukine-8B/génétique , Remodelage ventriculaire , Myocarde/métabolisme , Myocarde/anatomopathologie , Nanomédecine/méthodes
8.
Nat Commun ; 15(1): 4632, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951500

RÉSUMÉ

ANKRD11 (Ankyrin Repeat Domain 11) is a chromatin regulator and a causative gene for KBG syndrome, a rare developmental disorder characterized by multiple organ abnormalities, including cardiac defects. However, the role of ANKRD11 in heart development is unknown. The neural crest plays a leading role in embryonic heart development, and its dysfunction is implicated in congenital heart defects. We demonstrate that conditional knockout of Ankrd11 in the murine embryonic neural crest results in persistent truncus arteriosus, ventricular dilation, and impaired ventricular contractility. We further show these defects occur due to aberrant cardiac neural crest cell organization leading to outflow tract septation failure. Lastly, knockout of Ankrd11 in the neural crest leads to impaired expression of various transcription factors, chromatin remodelers and signaling pathways, including mTOR, BMP and TGF-ß in the cardiac neural crest cells. In this work, we identify Ankrd11 as a regulator of neural crest-mediated heart development and function.


Sujet(s)
Cardiopathies congénitales , Coeur , Souris knockout , Crête neurale , Protéines de répression , Animaux , Crête neurale/métabolisme , Crête neurale/embryologie , Souris , Coeur/embryologie , Protéines de répression/métabolisme , Protéines de répression/génétique , Cardiopathies congénitales/génétique , Cardiopathies congénitales/métabolisme , Cardiopathies congénitales/anatomopathologie , Régulation de l'expression des gènes au cours du développement , Chromatine/métabolisme , Transduction du signal , Myocarde/métabolisme , Femelle
9.
Commun Biol ; 7(1): 795, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951640

RÉSUMÉ

The peroxisome is a versatile organelle that performs diverse metabolic functions. PEX3, a critical regulator of the peroxisome, participates in various biological processes associated with the peroxisome. Whether PEX3 is involved in peroxisome-related redox homeostasis and myocardial regenerative repair remains elusive. We investigate that cardiomyocyte-specific PEX3 knockout (Pex3-KO) results in an imbalance of redox homeostasis and disrupts the endogenous proliferation/development at different times and spatial locations. Using Pex3-KO mice and myocardium-targeted intervention approaches, the effects of PEX3 on myocardial regenerative repair during both physiological and pathological stages are explored. Mechanistically, lipid metabolomics reveals that PEX3 promotes myocardial regenerative repair by affecting plasmalogen metabolism. Further, we find that PEX3-regulated plasmalogen activates the AKT/GSK3ß signaling pathway via the plasma membrane localization of ITGB3. Our study indicates that PEX3 may represent a novel therapeutic target for myocardial regenerative repair following injury.


Sujet(s)
Membrane cellulaire , Intégrine bêta3 , Souris knockout , Régénération , Animaux , Souris , Intégrine bêta3/métabolisme , Intégrine bêta3/génétique , Membrane cellulaire/métabolisme , Myocytes cardiaques/métabolisme , Mâle , Acétalphosphatides/métabolisme , Transduction du signal , Myocarde/métabolisme , Myocarde/anatomopathologie , Souris de lignée C57BL , Lésions traumatiques du coeur/métabolisme , Lésions traumatiques du coeur/anatomopathologie , Lésions traumatiques du coeur/génétique , Prolifération cellulaire , Protéines membranaires/métabolisme , Protéines membranaires/génétique
10.
J Nanobiotechnology ; 22(1): 385, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951822

RÉSUMÉ

BACKGROUND: Numerous studies have confirmed the involvement of extracellular vesicles (EVs) in various physiological processes, including cellular death and tissue damage. Recently, we reported that EVs derived from ischemia-reperfusion heart exacerbate cardiac injury. However, the role of EVs from healthy heart tissue (heart-derived EVs, or cEVs) on myocardial ischemia-reperfusion (MI/R) injury remains unclear. RESULTS: Here, we demonstrated that intramyocardial administration of cEVs significantly enhanced cardiac function and reduced cardiac damage in murine MI/R injury models. cEVs treatment effectively inhibited ferroptosis and maintained mitochondrial homeostasis in cardiomyocytes subjected to ischemia-reperfusion injury. Further results revealed that cEVs can transfer ATP5a1 into cardiomyocytes, thereby suppressing mitochondrial ROS production, alleviating mitochondrial damage, and inhibiting cardiomyocyte ferroptosis. Knockdown of ATP5a1 abolished the protective effects of cEVs. Furthermore, we found that the majority of cEVs are derived from cardiomyocytes, and ATP5a1 in cEVs primarily originates from cardiomyocytes of the healthy murine heart. Moreover, we demonstrated that adipose-derived stem cells (ADSC)-derived EVs with ATP5a1 overexpression showed much better efficacy on the therapy of MI/R injury compared to control ADSC-derived EVs. CONCLUSIONS: These findings emphasized the protective role of cEVs in cardiac injury and highlighted the therapeutic potential of targeting ATP5a1 as an important approach for managing myocardial damage induced by MI/R injury.


Sujet(s)
Vésicules extracellulaires , Souris de lignée C57BL , Mitochondrial Proton-Translocating ATPases , Lésion de reperfusion myocardique , Myocytes cardiaques , Animaux , Vésicules extracellulaires/métabolisme , Souris , Lésion de reperfusion myocardique/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Mâle , Mitochondrial Proton-Translocating ATPases/métabolisme , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Myocarde/métabolisme , Myocarde/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
11.
J Nanobiotechnology ; 22(1): 382, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951872

RÉSUMÉ

Reperfusion therapy is critical for saving heart muscle after myocardial infarction, but the process of restoring blood flow can itself exacerbate injury to the myocardium. This phenomenon is known as myocardial ischemia-reperfusion injury (MIRI), which includes oxidative stress, inflammation, and further cell death. microRNA-146a (miR-146a) is known to play a significant role in regulating the immune response and inflammation, and has been studied for its potential impact on the improvement of heart function after myocardial injury. However, the delivery of miR-146a to the heart in a specific and efficient manner remains a challenge as extracellular RNAs are unstable and rapidly degraded. Milk exosomes (MEs) have been proposed as ideal delivery platform for miRNA-based therapy as they can protect miRNAs from RNase degradation. In this study, the effects of miR-146a containing MEs (MEs-miR-146a) on improvement of cardiac function were examined in a rat model of MIRI. To enhance the targeting delivery of MEs-miR-146a to the site of myocardial injury, the ischemic myocardium-targeted peptide IMTP was modified onto the surfaces, and whether the modified MEs-miR-146a could exert a better therapeutic role was examined by echocardiography, myocardial injury indicators and the levels of inflammatory factors. Furthermore, the expressions of miR-146a mediated NF-κB signaling pathway-related proteins were detected by western blotting and qRT-PCR to further elucidate its mechanisms. MiR-146 mimics were successfully loaded into the MEs by electroporation at a square wave 1000 V voltage and 0.1 ms pulse duration. MEs-miR-146a can be up-taken by cardiomyocytes and protected the cells from oxygen glucose deprivation/reperfusion induced damage in vitro. Oral administration of MEs-miR-146a decreased myocardial tissue apoptosis and the expression of inflammatory factors and improved cardiac function after MIRI. The miR-146a level in myocardium tissues was significantly increased after the administration IMTP modified MEs-miR-146a, which was higher than that of the MEs-miR-146a group. In addition, intravenous injection of IMTP modified MEs-miR-146a enhanced the targeting to heart, improved cardiac function, reduced myocardial tissue apoptosis and suppressed inflammation after MIRI, which was more effective than the MEs-miR-146a treatment. Moreover, IMTP modified MEs-miR-146a reduced the protein levels of IRAK1, TRAF6 and p-p65. Therefore, IMTP modified MEs-miR-146a exerted their anti-inflammatory effect by inhibiting the IRAK1/TRAF6/NF-κB signaling pathway. Taken together, our findings suggested miR-146a containing MEs may be a promising strategy for the treatment of MIRI with better outcome after modification with ischemic myocardium-targeted peptide, which was expected to be applied in clinical practice in future.


Sujet(s)
Exosomes , microARN , Lésion de reperfusion myocardique , Facteur de transcription NF-kappa B , Rat Sprague-Dawley , Transduction du signal , Animaux , microARN/métabolisme , microARN/génétique , Lésion de reperfusion myocardique/métabolisme , Exosomes/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Rats , Mâle , Lait/composition chimique , Myocarde/métabolisme , Cardiotoniques/pharmacologie , Myocytes cardiaques/métabolisme
12.
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
13.
Clin Cardiol ; 47(7): e24307, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38953367

RÉSUMÉ

BACKGROUND: We aim to provide a comprehensive review of the current state of knowledge of myocardial viability assessment in patients undergoing coronary artery bypass grafting (CABG), with a focus on the clinical markers of viability for each imaging modality. We also compare mortality between patients with viable myocardium and those without viability who undergo CABG. METHODS: A systematic database search with meta-analysis was conducted of comparative original articles (both observations and randomized controlled studies) of patients undergoing CABG with either viable or nonviable myocardium, in EMBASE, MEDLINE, Cochrane database, and Google Scholar, from inception to 2022. Imaging modalities included were dobutamine stress echocardiography (DSE), cardiac magnetic resonance (CMR), single-photon emission computed tomography (SPECT), and positron emission tomography (PET). RESULTS: A total of 17 studies incorporating a total of 2317 patients were included. Across all imaging modalities, the relative risk of death post-CABG was reduced in patients with versus without viability (random-effects model: odds ratio: 0.42; 95% confidence interval: 0.29-0.61; p < 0.001). Imaging for myocardial viability has significant clinical implications as it can affect the accuracy of the diagnosis, guide treatment decisions, and predict patient outcomes. Generally, based on local availability and expertise, either SPECT or DSE should be considered as the first step in evaluating viability, while PET or CMR would provide further evaluation of transmurality, perfusion metabolism, and extent of scar tissue. CONCLUSION: The assessment of myocardial viability is an essential component of preoperative evaluation in patients with ischemic heart disease undergoing surgical revascularization. Careful patient selection and individualized assessment of viability remain paramount.


Sujet(s)
Pontage aortocoronarien , Ischémie myocardique , Fonction ventriculaire gauche , Humains , Cardiomyopathies/physiopathologie , Cardiomyopathies/chirurgie , Cardiomyopathies/diagnostic , Cardiomyopathies/étiologie , Pontage aortocoronarien/effets indésirables , Maladie des artères coronaires/chirurgie , Maladie des artères coronaires/physiopathologie , Maladie des artères coronaires/diagnostic , Maladie des artères coronaires/complications , Échocardiographie de stress/méthodes , Ischémie myocardique/physiopathologie , Ischémie myocardique/chirurgie , Ischémie myocardique/diagnostic , Ischémie myocardique/complications , Myocarde/anatomopathologie , Survie tissulaire , Tomographie par émission monophotonique , Dysfonction ventriculaire gauche/physiopathologie , Dysfonction ventriculaire gauche/étiologie , Fonction ventriculaire gauche/physiologie
14.
Development ; 151(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38984541

RÉSUMÉ

The cardiac extracellular matrix (cECM) is fundamental for organ morphogenesis and maturation, during which time it undergoes remodeling, yet little is known about whether mechanical forces generated by the heartbeat regulate this remodeling process. Using zebrafish as a model and focusing on stages when cardiac valves and trabeculae form, we found that altering cardiac contraction impairs cECM remodeling. Longitudinal volumetric quantifications in wild-type animals revealed region-specific dynamics: cECM volume decreases in the atrium but not in the ventricle or atrioventricular canal. Reducing cardiac contraction resulted in opposite effects on the ventricular and atrial ECM, whereas increasing the heart rate affected the ventricular ECM but had no effect on the atrial ECM, together indicating that mechanical forces regulate the cECM in a chamber-specific manner. Among the ECM remodelers highly expressed during cardiac morphogenesis, we found one that was upregulated in non-contractile hearts, namely tissue inhibitor of matrix metalloproteinase 2 (timp2). Loss- and gain-of-function analyses of timp2 revealed its crucial role in cECM remodeling. Altogether, our results indicate that mechanical forces control cECM remodeling in part through timp2 downregulation.


Sujet(s)
Matrice extracellulaire , Coeur , Inhibiteur tissulaire de métalloprotéinase-2 , Danio zébré , Animaux , Danio zébré/embryologie , Danio zébré/métabolisme , Matrice extracellulaire/métabolisme , Inhibiteur tissulaire de métalloprotéinase-2/métabolisme , Inhibiteur tissulaire de métalloprotéinase-2/génétique , Coeur/embryologie , Protéines de poisson-zèbre/métabolisme , Protéines de poisson-zèbre/génétique , Contraction myocardique/physiologie , Myocarde/métabolisme , Morphogenèse , Atrium du coeur/embryologie , Atrium du coeur/métabolisme , Phénomènes biomécaniques , Régulation de l'expression des gènes au cours du développement , Ventricules cardiaques/métabolisme , Ventricules cardiaques/embryologie
16.
Pancreas ; 53(7): e588-e594, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38986079

RÉSUMÉ

OBJECTIVE: It was targeted to assess the efficacy of certolizumab on pancreas and target organs via biochemical parameters and histopathologic scores in experimental acute pancreatitis (AP). MATERIALS AND METHODS: Forty male Sprague Dawley rats were divided into the following 5 equal groups: group 1 (sham group), group 2 (AP group), group 3 (AP + low-dose certolizumab group), group 4 (AP + high-dose certolizumab group), and group 5 (placebo group). Rats in all groups were sacrificed 24 hours after the last injection and amylase, tumor necrosis factor α, transforming growth factor ß, interleukin 1ß, malondialdehyde, superoxide dismutase, and glutathione peroxidase levels were studied in blood samples. Histopathological investigation of both the pancreas and target organs (lungs, liver, heart, kidneys) was performed by a pathologist blind to the groups. In silico analysis were also accomplished. RESULTS: The biochemical results in the certolizumab treatment groups were identified to be significantly favorable compared to the AP group (P < 0.001). The difference between the high-dose group (group 4) and low-dose treatment group (group 3) was found to be significant in terms of biochemical parameters and histopathological scores (P < 0.001). In terms of the effect of certolizumab treatment on the target organs (especially on lung tissue), the differences between the low-dose treatment group (group 3) and high-dose treatment group (group 4) with the AP group (group 2) were significant. CONCLUSIONS: Certolizumab has favorable protective effects on pancreas and target organs in AP. It may be a beneficial agent for AP treatment and may prevent target organ damage.


Sujet(s)
Amylases , Poumon , Pancréas , Pancréatite , Rat Sprague-Dawley , Facteur de nécrose tumorale alpha , Animaux , Mâle , Pancréatite/prévention et contrôle , Pancréatite/induit chimiquement , Pancréatite/anatomopathologie , Pancréatite/traitement médicamenteux , Pancréas/effets des médicaments et des substances chimiques , Pancréas/anatomopathologie , Pancréas/métabolisme , Amylases/sang , Maladie aigüe , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/sang , Certolizumab pégol/pharmacologie , Malonaldéhyde/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Foie/métabolisme , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/métabolisme , Interleukine-1 bêta/sang , Interleukine-1 bêta/métabolisme , Superoxide dismutase/métabolisme , Glutathione peroxidase/métabolisme , Myocarde/anatomopathologie , Myocarde/métabolisme , Facteur de croissance transformant bêta/métabolisme , Rats , Modèles animaux de maladie humaine , Stress oxydatif/effets des médicaments et des substances chimiques
17.
FASEB J ; 38(14): e23818, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-38989572

RÉSUMÉ

The association between cardiac fibrosis and galectin-3 was evaluated in patients with acute myocardial infarction (MI). The role of galectin-3 and its association with endoplasmic reticulum (ER) stress activation in the progression of cardiovascular fibrosis was also evaluated in obese-infarcted rats. The inhibitor of galectin-3 activity, modified citrus pectin (MCP; 100 mg/kg/day), and the inhibitor of the ER stress activation, 4-phenylbutyric acid (4-PBA; 500 mg/kg/day), were administered for 4 weeks after MI in obese rats. Overweight-obese patients who suffered a first MI showed higher circulating galectin-3 levels, higher extracellular volume, and LV infarcted size, as well as lower E/e'ratio and LVEF compared with normal-weight patients. A correlation was observed between galectin-3 levels and extracellular volume. Obese-infarcted animals presented cardiac hypertrophy and reduction in LVEF, and E/A ratio as compared with control animals. They also showed an increase in galectin-3 gene expression, as well as cardiac fibrosis and reduced autophagic flux. These alterations were associated with ER stress activation characterized by enhanced cardiac levels of binding immunoglobulin protein, which were correlated with those of galectin-3. Both MCP and 4-PBA not only reduced cardiac fibrosis, oxidative stress, galectin-3 levels, and ER stress activation, but also prevented cardiac functional alterations and ameliorated autophagic flux. These results show the relevant role of galectin-3 in the development of diffuse fibrosis associated with MI in the context of obesity in both the animal model and patients. Galectin-3 in tandem with ER stress activation could modulate different downstream mechanisms, including inflammation, oxidative stress, and autophagy.


Sujet(s)
Stress du réticulum endoplasmique , Galectine -3 , Obésité , Animaux , Galectine -3/métabolisme , Obésité/métabolisme , Obésité/complications , Mâle , Rats , Humains , Pectine/pharmacologie , Adulte d'âge moyen , Infarctus du myocarde/métabolisme , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/complications , Femelle , Fibrose , Rat Wistar , Ischémie myocardique/métabolisme , Ischémie myocardique/anatomopathologie , Phénylbutyrates/pharmacologie , Autophagie , Myocarde/métabolisme , Myocarde/anatomopathologie , Galectines/métabolisme , Sujet âgé , Protéines du sang/métabolisme
18.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 36(6): 624-629, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-38991962

RÉSUMÉ

OBJECTIVE: To explore the protective effect of methylene blue (MB) on myocardial injury in sepsis and its possible signaling pathway. METHODS: A total of 32 female Wistar rats were randomly divided into sham operation group, sepsis model group, MB prevention group, and MB treatment group, with 8 rats in each group. The MB prevention group was injected with 15 mg/kg MB in the peritoneal cavity 6 hours before modeling; the other 3 groups were injected with 4 mL/kg saline in the peritoneal cavity. The sepsis model was established by cecal ligation puncture (CLP); the sham operation group was only subjected to an exploratory incision without ligation or puncture of the caecum. The MB treatment group was injected with 15 mg/kg MB in the peritoneal cavity 0.5 hours after modeling; the other 3 groups were injected with 4 mL/kg saline in the peritoneal cavity. Peripheral blood and myocardial tissue were collected from each group at 6 hours and 12 hours after modeling. Histological changes in the myocardial tissue were observed under the microscope; the levels of serum cardiac troponin I (cTnI), MB isoenzyme of creatine kinase (CK-MB), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) were detected by enzyme-linked immunosorbent assay (ELISA); and the expressions of inducible nitric oxide synthase (iNOS), light chain 3 (LC3), and p62 in the myocardial tissue were detected by Western blotting. RESULTS: Under light microscopy, no obvious abnormalities were found in the myocardium of the sham operation group; the myocardium of the sepsis model group showed obvious inflammatory changes; the myocardium of the MB prevention group showed mild inflammatory changes at 6 hours after modeling, severe inflammatory changes at 12 hours but less severe than the sepsis model group; the myocardium of the MB treatment group showed more obvious inflammatory changes at 6 hours after modeling but less severe than the MB prevention group at 12 hours after modeling, and the inflammatory changes at 12 hours after modeling were alleviated but more severe than the 6 hours after modeling in MB prevention group. Compared with the sham operation group, the levels of cTnI, CK-MB, TNF-α and IL-6 in the MB prevention group at 6 hours and 12 hours after modeling were not significantly changed; compared with the sepsis model group, the cTnI, CK-MB, TNF-α and IL-6 levels in the MB treatment group at 6 hours and 12 hours after modeling were significantly lower [cTnI (ng/L): 175.03±12.26, 411.24±21.20 vs. 677.79±43.95 at 6 hours of modeling, 159.52±6.44, 412.46±32.94 vs. 687.61±55.09 at 12 hours of modeling; CK-MB (ng/L): 8.38±0.49, 16.87±1.41 vs. 24.87±1.74 at 6 hours of modeling, 7.94±0.30, 16.66±2.03 vs. 25.02±7.29 at 12 hours of modeling; TNF-α (ng/L): 26.98±3.31, 46.95±3.74 vs. 112.60±6.64 at 6 hours of modeling, 31.31±5.83, 90.97±5.14 vs. 149.30±4.67 at 12 hours of modeling; IL-6 (ng/L): 40.86±4.48, 128.90±3.14 vs. 248.90±12.76 at 6 hours of modeling, 80.13±7.94, 190.40±9.56 vs. 288.90±6.01 at 12 hours of modeling; all P < 0.05]. Western blotting showed that compared with the sham operation group, the protein expressions of iNOS, LC3, and p62 in the sepsis model group were significantly higher at 6 hours and 12 hours after modeling; compared with the sepsis model group, the protein expressions of iNOS, LC3, and p62 in the MB treatment group and MB prevention group were significantly lower at 6 hours and 12 hours after modeling (iNOS/GAPDH: 0.38±0.04, 0.60±0.04 vs. 0.77±0.04 at 6 hours of modeling; 0.38±0.02, 0.66±0.04 vs. 0.79±0.05 at 12 hours of modeling; LC3/GAPDH: 0.13±0.07, 0.42±0.07 vs. 1.05±0.16 at 6 hours of modeling; 0.08±0.02, 0.25±0.03 vs. 0.48±0.09 at 12 hours of modeling; p62/GAPDH: 0.17±0.05, 0.44±0.10 vs. 1.19±0.07 at 6 hours of modeling; 0.07±0.00, 0.28±0.08 vs. 0.69±0.02 at 12 hours of modeling; all P < 0.05). CONCLUSIONS: MB can reduce myocardial oxidative stress by inhibiting iNOS expression and mitochondrial autophagy in septic rats, thereby alleviating myocardial damage in sepsis, and has protective effect on myocardial damage in sepsis.


Sujet(s)
Interleukine-6 , Bleu de méthylène , Myocarde , Rat Wistar , Sepsie , Troponine I , Facteur de nécrose tumorale alpha , Animaux , Sepsie/traitement médicamenteux , Sepsie/complications , Rats , Femelle , Interleukine-6/métabolisme , Myocarde/métabolisme , Myocarde/anatomopathologie , Facteur de nécrose tumorale alpha/métabolisme , Troponine I/sang , Bleu de méthylène/pharmacologie , Modèles animaux de maladie humaine , MB Creatine kinase/sang , Nitric oxide synthase type II/métabolisme
19.
Cells ; 13(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38994928

RÉSUMÉ

Activation of the CXCL12/CXCR4/ACKR3 axis is known to aid myocardial repair through ischemia-triggered hypoxia-inducible factor-1α (HIF-1α). To enhance the upregulation of HIF-1α, we administered roxadustat, a novel prolyl hydroxylase inhibitor (PHI) clinically approved by the European Medicines Agency 2021 for the treatment of renal anemia, with the purpose of improving LV function and attenuating ischemic cardiomyopathy. METHODS: We evaluated roxadustat's impact on HIF-1 stimulation, cardiac remodeling, and function after MI. Therefore, we analyzed nuclear HIF-1 expression, the mRNA and protein expression of key HIF-1 target genes (RT-PCR, Western blot), inflammatory cell infiltration (immunohistochemistry), and apoptosis (TUNEL staining) 7 days after MI. Additionally, we performed echocardiography in male and female C57BL/6 mice 28 days post-MI. RESULTS: We found a substantial increase in nuclear HIF-1, associated with an upregulation of HIF-1α target genes like CXCL12/CXCR4/ACKR3 at the mRNA and protein levels. Roxadustat increased the proportion of myocardial reparative M2 CD206+ cells, suggesting beneficial alterations in immune cell migration and a trend towards reduced apoptosis. Echocardiography showed that roxadustat treatment significantly preserved ejection fraction and attenuated subsequent ventricular dilatation, thereby reducing adverse remodeling. CONCLUSIONS: Our findings suggest that roxadustat is a promising clinically approved treatment option to preserve myocardial function by attenuating adverse remodeling.


Sujet(s)
Glycine , Sous-unité alpha du facteur-1 induit par l'hypoxie , Isoquinoléines , Souris de lignée C57BL , Infarctus du myocarde , Remodelage ventriculaire , Animaux , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/métabolisme , Souris , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Glycine/analogues et dérivés , Glycine/pharmacologie , Glycine/usage thérapeutique , Mâle , Femelle , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Isoquinoléines/pharmacologie , Isoquinoléines/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Chimiokine CXCL12/métabolisme , Chimiokine CXCL12/génétique , Myocarde/anatomopathologie , Myocarde/métabolisme
20.
Int J Mol Sci ; 25(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-39000372

RÉSUMÉ

Circadian disruption increases the development of cardiovascular disease and diabetes. We found that circadian disruption causes glucose intolerance, cardiac fibrosis and adipocyte tissue dysfunction in male sand rats, Psammomys obesus. Whether these effects occur in female P. obesus is unknown. Male and female P. obesus were fed a high energy diet and exposed to a neutral (12 light:12 dark, control) or short (5 light:19 dark, circadian disruption) photoperiod for 20 weeks. Circadian disruption impaired glucose tolerance in males but not females. It also increased cardiac perivascular fibrosis and cardiac expression of inflammatory marker Ccl2 in males, with no effect in females. Females had reduced proapoptotic Bax mRNA and cardiac Myh7:Myh6 hypertrophy ratio. Cardiac protection in females occurred despite reductions in the clock gene Per2. Circadian disruption increased adipocyte hypertrophy in both males and females. This was concomitant with a reduction in adipocyte differentiation markers Pparg and Cebpa in males and females, respectively. Circadian disruption increased visceral adipose expression of inflammatory mediators Ccl2, Tgfb1 and Cd68 and reduced browning marker Ucp1 in males. However, these changes were not observed in females. Collectively, our study show that sex differentially influences the effects of circadian disruption on glucose tolerance, cardiac function and adipose tissue dysfunction.


Sujet(s)
Adipocytes , Fibrose , Gerbillinae , Intolérance au glucose , Animaux , Femelle , Adipocytes/métabolisme , Adipocytes/anatomopathologie , Mâle , Intolérance au glucose/métabolisme , Myocarde/métabolisme , Myocarde/anatomopathologie , Rythme circadien
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE