Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 12.857
Filtrer
1.
Proc Natl Acad Sci U S A ; 121(28): e2403581121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38968108

RÉSUMÉ

Adverse cardiac outcomes in COVID-19 patients, particularly those with preexisting cardiac disease, motivate the development of human cell-based organ-on-a-chip models to recapitulate cardiac injury and dysfunction and for screening of cardioprotective therapeutics. Here, we developed a heart-on-a-chip model to study the pathogenesis of SARS-CoV-2 in healthy myocardium established from human induced pluripotent stem cell (iPSC)-derived cardiomyocytes and a cardiac dysfunction model, mimicking aspects of preexisting hypertensive disease induced by angiotensin II (Ang II). We recapitulated cytopathic features of SARS-CoV-2-induced cardiac damage, including progressively impaired contractile function and calcium handling, apoptosis, and sarcomere disarray. SARS-CoV-2 presence in Ang II-treated hearts-on-a-chip decreased contractile force with earlier onset of contractile dysfunction and profoundly enhanced inflammatory cytokines compared to SARS-CoV-2 alone. Toward the development of potential therapeutics, we evaluated the cardioprotective effects of extracellular vesicles (EVs) from human iPSC which alleviated the impairment of contractile force, decreased apoptosis, reduced the disruption of sarcomeric proteins, and enhanced beta-oxidation gene expression. Viral load was not affected by either Ang II or EV treatment. We identified MicroRNAs miR-20a-5p and miR-19a-3p as potential mediators of cardioprotective effects of these EVs.


Sujet(s)
Angiotensine-II , COVID-19 , Vésicules extracellulaires , Cellules souches pluripotentes induites , Myocytes cardiaques , SARS-CoV-2 , Humains , Angiotensine-II/pharmacologie , COVID-19/virologie , COVID-19/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/virologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Vésicules extracellulaires/métabolisme , Cellules souches pluripotentes induites/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Laboratoires sur puces , microARN/métabolisme , microARN/génétique , Cytokines/métabolisme
2.
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.
Cells ; 13(13)2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38995002

RÉSUMÉ

Doxorubicin (DOX) is an anthracycline anticancer agent that is highly effective in the treatment of solid tumors. Given the multiplicity of mechanisms involved in doxorubicin-induced cardiotoxicity, it is difficult to identify a precise molecular target for toxicity. The findings of a literature review suggest that natural products may offer cardioprotective benefits against doxorubicin-induced cardiotoxicity, both in vitro and in vivo. However, further confirmatory studies are required to substantiate this claim. It is of the utmost importance to direct greater attention towards the intricate signaling networks that are of paramount importance for the survival and dysfunction of cardiomyocytes. Notwithstanding encouraging progress made in preclinical studies of natural products for the prevention of DOX-induced cardiotoxicity, these have not yet been translated for clinical use. One of the most significant obstacles hindering the development of cardioprotective adjuvants based on natural products is the lack of adequate bioavailability in humans. This review presents an overview of current knowledge on doxorubicin DOX-induced cardiotoxicity, with a focus on the potential benefits of natural compounds and herbal preparations in preventing this adverse effect. As literature search engines, the browsers in the Scopus, PubMed, Web of Science databases and the ClinicalTrials.gov register were used.


Sujet(s)
Anthracyclines , Produits biologiques , Cardiotoxicité , Humains , Produits biologiques/usage thérapeutique , Produits biologiques/pharmacologie , Cardiotoxicité/prévention et contrôle , Anthracyclines/effets indésirables , Animaux , Doxorubicine/effets indésirables , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Cardiotoniques/pharmacologie , Cardiotoniques/usage thérapeutique
4.
ACS Appl Mater Interfaces ; 16(28): 36030-36046, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38951110

RÉSUMÉ

Differentiation of induced pluripotent stem cells (iPSCs) is an extremely complex process that has proven difficult to study. In this research, we utilized nanotopography to elucidate details regarding iPSC differentiation by developing a nanodot platform consisting of nanodot arrays of increasing diameter. Subjecting iPSCs cultured on the nanodot platform to a cardiomyocyte (CM) differentiation protocol revealed several significant gene expression profiles that were associated with poor differentiation. The observed expression trends were used to select existing small-molecule drugs capable of modulating differentiation efficiency. BRD K98 was repurposed to inhibit CM differentiation, while iPSCs treated with NSC-663284, carmofur, and KPT-330 all exhibited significant increases in not only CM marker expression but also spontaneous beating, suggesting improved CM differentiation. In addition, quantitative polymerase chain reaction was performed to determine the gene regulation responsible for modulating differentiation efficiency. Multiple genes involved in extracellular matrix remodeling were correlated with a CM differentiation efficiency, while genes involved in the cell cycle exhibited contrasting expression trends that warrant further studies. The results suggest that expression profiles determined via short time-series expression miner analysis of nanodot-cultured iPSC differentiation can not only reveal drugs capable of enhancing differentiation efficiency but also highlight crucial sets of genes related to processes such as extracellular matrix remodeling and the cell cycle that can be targeted for further investigation. Our findings confirm that the nanodot platform can be used to reveal complex mechanisms behind iPSC differentiation and could be an indispensable tool for optimizing iPSC technology for clinical applications.


Sujet(s)
Différenciation cellulaire , Cellules souches pluripotentes induites , Myocytes cardiaques , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Cellules souches pluripotentes induites/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Myocytes cardiaques/cytologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Humains , Nanoparticules/composition chimique , Cellules cultivées , Nanostructures/composition chimique
5.
Int J Nanomedicine ; 19: 6399-6425, 2024.
Article de Anglais | MEDLINE | ID: mdl-38952676

RÉSUMÉ

Myocardial infarction, usually caused by the rupture of atherosclerotic plaque, leads to irreversible ischemic cardiomyocyte death within hours followed by impaired cardiac performance or even heart failure. Current interventional reperfusion strategies for myocardial infarction still face high mortality with the development of heart failure. Nanomaterial-based therapy has made great progress in reducing infarct size and promoting cardiac repair after MI, although most studies are preclinical trials. This review focuses primarily on recent progress (2016-now) in the development of various nanomedicines in the treatment of myocardial infarction. We summarize these applications with the strategy of mechanism including anti-cardiomyocyte death strategy, activation of neovascularization, antioxidants strategy, immunomodulation, anti-cardiac remodeling, and cardiac repair.


Sujet(s)
Infarctus du myocarde , Nanomédecine , Infarctus du myocarde/thérapie , Humains , Animaux , Myocytes cardiaques/effets des médicaments et des substances chimiques , Antioxydants/usage thérapeutique , Nanostructures/usage thérapeutique , Nanostructures/composition chimique , Néovascularisation physiologique/effets des médicaments et des substances chimiques
6.
Theranostics ; 14(9): 3719-3738, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948070

RÉSUMÉ

Rationale: Autophagy dysregulation is known to be a mechanism of doxorubicin (DOX)-induced cardiotoxicity (DIC). Mitochondrial-Endoplasmic Reticulum Contacts (MERCs) are where autophagy initiates and autophagosomes form. However, the role of MERCs in autophagy dysregulation in DIC remains elusive. FUNDC1 is a tethering protein of MERCs. We aim to investigate the effect of DOX on MERCs in cardiomyocytes and explore whether it is involved in the dysregulated autophagy in DIC. Methods: We employed confocal microscopy and transmission electron microscopy to assess MERCs structure. Autophagic flux was analyzed using the mCherry-EGFP-LC3B fluorescence assay and western blotting for LC3BII. Mitophagy was studied through the mCherry-EGFP-FIS1 fluorescence assay and colocalization analysis between LC3B and mitochondria. A total dose of 18 mg/kg of doxorubicin was administrated in mice to construct a DIC model in vivo. Additionally, we used adeno-associated virus (AAV) to cardiac-specifically overexpress FUNDC1. Cardiac function and remodeling were evaluated by echocardiography and Masson's trichrome staining, respectively. Results: DOX blocked autophagic flux by inhibiting autophagosome biogenesis, which could be attributed to the downregulation of FUNDC1 and disruption of MERCs structures. FUNDC1 overexpression restored the blocked autophagosome biogenesis by maintaining MERCs structure and facilitating ATG5-ATG12/ATG16L1 complex formation without altering mitophagy. Furthermore, FUNDC1 alleviated DOX-induced oxidative stress and cardiomyocytes deaths in an autophagy-dependent manner. Notably, cardiac-specific overexpression of FUNDC1 protected DOX-treated mice against adverse cardiac remodeling and improved cardiac function. Conclusions: In summary, our study identified that FUNDC1-meditated MERCs exerted a cardioprotective effect against DIC by restoring the blocked autophagosome biogenesis. Importantly, this research reveals a novel role of FUNDC1 in enhancing macroautophagy via restoring MERCs structure and autophagosome biogenesis in the DIC model, beyond its previously known regulatory role as an mitophagy receptor.


Sujet(s)
Autophagie , Cardiotoxicité , Doxorubicine , Réticulum endoplasmique , Protéines membranaires , Protéines mitochondriales , Myocytes cardiaques , Animaux , Doxorubicine/effets indésirables , Doxorubicine/pharmacologie , Souris , Autophagie/effets des médicaments et des substances chimiques , Cardiotoxicité/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Réticulum endoplasmique/métabolisme , Réticulum endoplasmique/effets des médicaments et des substances chimiques , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitophagie/effets des médicaments et des substances chimiques , Mâle , Autophagosomes/métabolisme , Autophagosomes/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Modèles animaux de maladie humaine
7.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 630-634, 2024 May 20.
Article de Chinois | MEDLINE | ID: mdl-38948270

RÉSUMÉ

Objective: To investigate the effect of Sanshentongmai (SSTM) mixture on the regulation of oxidative damage to rat cardiomyocytes (H9C2) through microRNA-146a and its mechanism. Methods: H9C2 were cultured in vitro, H2O2 was used as an oxidant to create an oxidative damage model in H9C2 cells. SSTM intervention was administered to the H9C2 cells. Then, the changes in H2O2-induced oxidative damage in H9C2 cells and the expression of microRNA-146a were observed to explore the protective effect of SSTM on H9C2 and its mechanism. H9C2 cells cultured i n vitro were divided into 3 groups, including a control group, a model group of H2O2-induced oxidative damage (referred to hereafter as the model group), and a group given H2O2 modeling plus SSTM intervention at 500 µg/L for 72 h (referred to hereafter as the treatment group). The cell viability was measured by CCK8 assay. In addition, the levels of N-terminal pro-brain natriuretic peptide (Nt-proBNP), nitric oxide (NO), high-sensitivity C-reactive protein (Hs-CRP), and angiotensin were determined by enzyme-linked immunosorbent assay (ELISA). The expression level of microRNA-146a was determined by real-time PCR (RT-PCR). Result: H9C2 cells were pretreated with SSTM at mass concentrations ranging from 200 to 1500 µg/L. Then, CCK8 assay was performed to measure cell viability and the findings showed that the improvement in cell proliferation reached its peak when the mass concentration of SSTM was 500 µg/L, which was subsequently used as the intervention concentration. ELISA was performed to measure the indicators related to heart failure, including Nt-proBNP, NO, Hs-CRP, and angiotensin Ⅱ. Compared with those of the control group, the expressions of Nt-proBNP and angiotensin Ⅱ in the treatment group were up-regulated (P<0.05), while the expression of NO was down-regulated (P<0.05). There was no significant difference in the expression of Hs-CRP between the treatment group and the control group. These findings indicate that SSTM could effectively ameliorate oxidative damage in H9C2 rat cardiomyocytes. Finally, according to the RT-PCR findings for the expression of microRNA-146a in each group, H2O2 treatment at 15 µmol/L could significantly reduce the expression of microRNA-146a, and the expression of microRNA-146a in the treatment group was nearly doubled compared with that in the model group. There was no significant difference between the treatment group and the control group. Conclusion: SSTM can significantly resist the H2O2-induced oxidative damage of H9C2 cells and may play a myocardial protective role by upregulating microRNA-146a.


Sujet(s)
Médicaments issus de plantes chinoises , Peroxyde d'hydrogène , microARN , Myocytes cardiaques , Stress oxydatif , Régulation positive , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/cytologie , Animaux , microARN/métabolisme , microARN/génétique , Rats , Stress oxydatif/effets des médicaments et des substances chimiques , Peroxyde d'hydrogène/toxicité , Médicaments issus de plantes chinoises/pharmacologie , Régulation positive/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Association médicamenteuse
8.
Pharmacogenomics J ; 24(4): 21, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951505

RÉSUMÉ

There is a known genetic susceptibility to anthracycline-induced cardiac dysfunction in childhood cancer survivors, but this has not been adequately shown in adolescent and young adult (AYA) patients. Our aim was to determine if the previously identified variants associated with cardiac dysfunction in childhood cancer patients affect AYA cancer patients similarly. Forty-five variants were selected for analysis in 253 AYAs previously treated with anthracyclines. We identified four variants that were associated with cardiac dysfunction: SLC10A2:rs7319981 (p = 0.017), SLC22A17:rs4982753 (p = 0.019), HAS3:rs2232228 (p = 0.023), and RARG:rs2229774 (p = 0.050). HAS3:rs2232228 and SLC10A2:rs7319981 displayed significant effects in our AYA cancer survivor population that were in the opposite direction than that reported in childhood cancer survivors. Genetic variants in the host genes were further analyzed for additional associations with cardiotoxicity in AYA cancer survivors. The host genes were then evaluated in a panel of induced pluripotent stem cell-derived cardiomyocytes to assess changes in levels of expression when treated with doxorubicin. Significant upregulation of HAS3 and SLC22A17 expression was observed (p < 0.05), with non-significant anthracycline-responsivity observed for RARG. Our study demonstrates that there is a genetic influence on cardiac dysfunction in AYA cancer patients, but there may be a difference in the role of genetics between childhood and AYA cancer survivors.


Sujet(s)
Anthracyclines , Survivants du cancer , Cardiotoxicité , Prédisposition génétique à une maladie , Humains , Adolescent , Anthracyclines/effets indésirables , Jeune adulte , Mâle , Femelle , Cardiotoxicité/génétique , Adulte , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Polymorphisme de nucléotide simple/génétique , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Cardiopathies/induit chimiquement , Cardiopathies/génétique , Antibiotiques antinéoplasiques/effets indésirables , Facteurs de risque
9.
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
10.
Vascul Pharmacol ; 155: 107324, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38985581

RÉSUMÉ

Doxorubicin (DOX) is a highly effective chemotherapeutic agent whose clinical use is hindered by the onset of cardiotoxic effects, resulting in reduced ejection fraction within the first year from treatment initiation. Recently it has been demonstrated that DOX accumulates within mitochondria, leading to disruption of metabolic processes and energetic imbalance. We previously described that phosphoinositide 3-kinase γ (PI3Kγ) contributes to DOX-induced cardiotoxicity, causing autophagy inhibition and accumulation of damaged mitochondria. Here we intend to describe the maladaptive metabolic rewiring occurring in DOX-treated hearts and the contribution of PI3Kγ signalling to this process. Metabolomic analysis of DOX-treated WT hearts revealed an accumulation of TCA cycle metabolites due to a cycle slowdown, with reduced levels of pyruvate, unchanged abundance of lactate and increased Acetyl-CoA production. Moreover, the activity of glycolytic enzymes was upregulated, and fatty acid oxidation downregulated, after DOX, indicative of increased glucose oxidation. In agreement, oxygen consumption was increased in after pyruvate supplementation, with the formation of cytotoxic ROS rather than energy production. These metabolic changes were fully prevented in KD hearts. Interestingly, they failed to increase glucose oxidation in response to DOX even with autophagy inhibition, indicating that PI3Kγ likely controls the fuel preference after DOX through an autophagy-independent mechanism. In vitro experiments showed that inhibition of PI3Kγ inhibits pyruvate dehydrogenase (PDH), the key enzyme of Randle cycle regulating the switch from fatty acids to glucose usage, while decreasing DOX-induced mobilization of GLUT-4-carrying vesicles to the plasma membrane and limiting the ensuing glucose uptake. These results demonstrate that PI3Kγ promotes a maladaptive metabolic rewiring in DOX-treated hearts, through a two-pronged mechanism controlling PDH activation and GLUT-4-mediated glucose uptake.


Sujet(s)
Cardiotoxicité , Doxorubicine , Métabolisme énergétique , Acides gras , Glucose , Oxydoréduction , Animaux , Doxorubicine/toxicité , Glucose/métabolisme , Acides gras/métabolisme , Métabolisme énergétique/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases de classe Ib/métabolisme , Glycolyse/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Cycle citrique/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Cardiopathies/induit chimiquement , Cardiopathies/métabolisme , Cardiopathies/anatomopathologie , Cardiopathies/prévention et contrôle , Cardiopathies/physiopathologie , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Mitochondries du myocarde/anatomopathologie , Mitochondries du myocarde/enzymologie , Souris knockout , Modèles animaux de maladie humaine , Espèces réactives de l'oxygène/métabolisme , Transporteur de glucose de type 4/métabolisme , Antibiotiques antinéoplasiques/toxicité , Antibiotiques antinéoplasiques/effets indésirables
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE