Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.411
Filtrer
1.
J Ovarian Res ; 17(1): 135, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38943148

RÉSUMÉ

BACKGROUND: This study aimed to investigate the mitigating effect of N-acetylcysteine (NAC) on doxorubicin (DOX)-induced ovarian and uterine toxicity in rats using laboratory tests, ultrasonographic (US) imaging, and histopathology analysis. METHODS: Forty-eight rats were divided into six groups (n = 8) as follows: Group A (control) (0.5 mL saline administered intraperitoneally [IP]), Group B (a single 10 mg/kg dose of DOX administered IP on day 1), Group C (a single 10 mg/kg dose of DOX administered IP 24 h before sacrifice), Group D (100 mg/kg of NAC administered IP for 21 days), Group E ( a single 10 mg/kg dose of DOX administered IP on day 1 and 100 mg/kg of NAC administered IP for 21 days), and Group F (100 mg/kg of NAC administered IP for 21 days and a single 10 mg/kg dose of DOX administered IP 24 h before sacrifice). The ovaries were examined using B-mode US on days 1, 14, and 21, and the histopathological examinations of the ovaries and the uterus were undertaken after sacrifice on day 22. RESULTS: Histomorphological analyses showed that ovarian weight decreased after DOX administration in Group B but not in Group E. US revealed a transient increase in ovarian size in Group B and E, reverting to baseline levels over time, as well as a progressive increase in peritoneal fluid in Groups B and E. Group B exhibited a significant decrease in the thickness of the endometrium and myometrium and uterine cornual length, which was not observed in Group E. Histopathological examination showed that DOX caused a decline in follicular count, especially in primordial, secondary, and Graafian follicles, and resulted in follicular atresia, predominantly in Group B. Destructive degeneration/necrosis and vascular changes were most prominently seen in the corpus luteum of Groups C and B. In NAC-treated rats (Groups E and F), although germ cell damage was present, atretic follicles and vascular changes, such as hyperemia and congestion, were reduced. The anti-müllerian hormone (AMH) level was the highest in Group F. CONCLUSIONS: NAC, an antioxidant, attenuated DOX-induced gonadotoxicity in rats.


Sujet(s)
Acétylcystéine , Doxorubicine , Ovaire , Échographie , Utérus , Animaux , Femelle , Doxorubicine/toxicité , Acétylcystéine/pharmacologie , Acétylcystéine/usage thérapeutique , Rats , Ovaire/effets des médicaments et des substances chimiques , Ovaire/anatomopathologie , Ovaire/imagerie diagnostique , Utérus/effets des médicaments et des substances chimiques , Utérus/anatomopathologie , Utérus/imagerie diagnostique , Antibiotiques antinéoplasiques/toxicité , Antibiotiques antinéoplasiques/effets indésirables
2.
Biomed Pharmacother ; 176: 116759, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38788603

RÉSUMÉ

BACKGROUND: Sodium-glucose cotransporter-2 inhibitors, such as empagliflozin, are pivotal therapies for heart failure. However, the effect of empagliflozin on doxorubicin-related cardiac dysfunction remains unclear. METHODS: Human induced pluripotent stem cell- and embryonic stem cell-derived cardiomyocytes were used to investigate the direct effect of empagliflozin on human cardiomyocytes. Then, the c-Jun amino-terminal kinases (JNK) inhibitor SP600125 was administered to the doxorubicin cardiotoxicity model in vitro and in vivo to investigate the role of JNK in empagliflozin. RESULTS: In human stem cell-derived cardiomyocytes, pretreatment with empagliflozin attenuated doxorubicin-induced cleavage of caspase 3 and other apoptosis markers. Empagliflozin significantly attenuated doxorubicin-induced phosphorylation of JNK and p38. Inhibiting the phosphorylation of JNK (SP600125) or STAT3 attenuated doxorubicin-induced apoptosis, but inhibiting the phosphorylation of p38 did not. SP600125 inhibits the phosphorylation of STAT3 (S727), and a STAT3 (Y705) inhibitor also inhibits the phosphorylation of JNK. Empagliflozin and SP600125 attenuated doxorubicin-induced increases in reactive oxygen species (ROS) and decreases in oxidized nicotinamide adenine dinucleotide (NAD+). In animal studies, empagliflozin and SP600125 attenuated doxorubicin-induced cardiac dysfunction and fibrosis. CONCLUSIONS: Empagliflozin attenuated doxorubicin-induced apoptosis by inhibiting the phosphorylation of JNK and its downstream signaling pathways, including ROS and NAD+.


Sujet(s)
Apoptose , Composés benzhydryliques , Cardiotoxicité , Doxorubicine , Glucosides , Myocytes cardiaques , Glucosides/pharmacologie , Composés benzhydryliques/pharmacologie , Doxorubicine/toxicité , Doxorubicine/effets indésirables , Cardiotoxicité/traitement médicamenteux , Cardiotoxicité/prévention et contrôle , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Humains , Animaux , Apoptose/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Mâle , Espèces réactives de l'oxygène/métabolisme , Anthracènes/pharmacologie , JNK Mitogen-Activated Protein Kinases/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Souris , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Cellules souches pluripotentes induites/métabolisme , Souris de lignée C57BL
3.
Food Chem Toxicol ; 189: 114741, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38759714

RÉSUMÉ

An impact of donepezil against doxorubicin-induced gut barrier disruption and gut dysbiosis has never been investigated. Twenty-four male Wistar rats were divided into three groups. Each group was treated with either vehicle as a control, doxorubicin, or doxorubicin-cotreated with donepezil. Heart rate variability was assessed to reflect the impact of doxorubicin and donepezil. Then, animals were euthanized, and the ileum and its contents were collected in each case to investigate the gut barrier and gut microbiota, respectively. The microbiota-derived endotoxin, trimethylamine N-oxide (TMAO), and short-chain fatty acids (SCFAs) in the serum were determined. An increase in the sympathetic tone, endotoxins, and TMAO levels with disruption of the gut barrier and a decrease in SCFAs levels were observed in doxorubicin-treated rats. Gut microbiota of doxorubicin-treated rats was significantly different from that of the control group. Donepezil treatment significantly decreased the sympathetic tone, restored the gut barrier, and reduced endotoxin and TMAO levels in doxorubicin-treated rats. Nonetheless, donepezil administration did not alter the gut microbiota profile and levels of SCFAs in doxorubicin-treated rats. Doxorubicin impaired the autonomic balance and the gut barrier, and induced gut dysbiosis, resulting in gut toxicity. Donepezil partially improved the doxorubicin-induced gut toxicity through balancing the autonomic disturbance.


Sujet(s)
Donépézil , Doxorubicine , Microbiome gastro-intestinal , Rat Wistar , Animaux , Donépézil/pharmacologie , Doxorubicine/toxicité , Mâle , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Rats , Acides gras volatils/métabolisme , Dysbiose/induit chimiquement , Méthylamines , Endotoxines/toxicité
4.
Exp Neurol ; 378: 114818, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38782352

RÉSUMÉ

Doxorubicin (DOX) is a highly effective anthracycline antibiotic used to treat a wide variety of cancers including breast cancer, leukemia and lymphoma. Unfortunately, clinical use of DOX is limited due to adverse off-target effects resulting in fatigue, respiratory muscle weakness and dyspnea. The diaphragm is the primary muscle of inspiration and respiratory insufficiency is likely the result of both muscle weakness and neural impairment. However, the contribution of neuropathology to DOX-induced respiratory muscle dysfunction is unclear. We hypothesized that diaphragm weakness following acute DOX exposure is associated with neurotoxicity and that exercise preconditioning is sufficient to improve diaphragm muscle contractility by maintaining neuromuscular integrity. Adult female Sprague-Dawley rats were randomized into four experimental groups: 1) sedentary-saline, 2) sedentary-DOX, 3) exercise-saline or 4) exercise-DOX. Endurance exercise preconditioning consisted of treadmill running for 1 h/day at 30 m/min for 10 days. Twenty-four hours after the last bout of exercise, animals were treated with DOX (20 mg/kg, I.P.) or saline (equal volume). Our results demonstrate that 48-h following DOX administration diaphragm muscle specific force is reduced in sedentary-DOX rats in response to both phrenic nerve and direct diaphragm stimulation. Importantly, endurance exercise preconditioning in DOX-treated rats attenuated the decrease in diaphragm contractile function, reduced neuromuscular transmission failure and altered phrenic nerve morphology. These changes were associated with an exercise-induced reduction in circulating biomarkers of inflammation, nerve injury and reformation. Therefore, the results are consistent with exercise preconditioning as an effective way of reducing respiratory impairment via preservation of phrenic-diaphragm neuromuscular conduction.


Sujet(s)
Muscle diaphragme , Doxorubicine , Conditionnement physique d'animal , Rat Sprague-Dawley , Animaux , Muscle diaphragme/effets des médicaments et des substances chimiques , Muscle diaphragme/innervation , Doxorubicine/toxicité , Femelle , Rats , Conditionnement physique d'animal/physiologie , Antibiotiques antinéoplasiques/toxicité , Transmission synaptique/effets des médicaments et des substances chimiques , Transmission synaptique/physiologie , Nerf phrénique/effets des médicaments et des substances chimiques , Contraction musculaire/effets des médicaments et des substances chimiques , Contraction musculaire/physiologie , Jonction neuromusculaire/effets des médicaments et des substances chimiques
5.
J Am Heart Assoc ; 13(9): e033700, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38700005

RÉSUMÉ

BACKGROUND: The only clinically approved drug that reduces doxorubicin cardiotoxicity is dexrazoxane, but its application is limited due to the risk of secondary malignancies. So, exploring alternative effective molecules to attenuate its cardiotoxicity is crucial. Colchicine is a safe and well-tolerated drug that helps reduce the production of reactive oxygen species. High doses of colchicine have been reported to block the fusion of autophagosomes and lysosomes in cancer cells. However, the impact of colchicine on the autophagy activity within cardiomyocytes remains inadequately elucidated. Recent studies have highlighted the beneficial effects of colchicine on patients with pericarditis, postprocedural atrial fibrillation, and coronary artery disease. It remains ambiguous how colchicine regulates autophagic flux in doxorubicin-induced heart failure. METHODS AND RESULTS: Doxorubicin was administered to establish models of heart failure both in vivo and in vitro. Prior studies have reported that doxorubicin impeded the breakdown of autophagic vacuoles, resulting in damaged mitochondria and the accumulation of reactive oxygen species. Following the administration of a low dose of colchicine (0.1 mg/kg, daily), significant improvements were observed in heart function (left ventricular ejection fraction: doxorubicin group versus treatment group=43.75%±3.614% versus 57.07%±2.968%, P=0.0373). In terms of mechanism, a low dose of colchicine facilitated the degradation of autolysosomes, thereby mitigating doxorubicin-induced cardiotoxicity. CONCLUSIONS: Our research has shown that a low dose of colchicine is pivotal in restoring the autophagy activity, thereby attenuating the cardiotoxicity induced by doxorubicin. Consequently, colchicine emerges as a promising therapeutic candidate to improve doxorubicin cardiotoxicity.


Sujet(s)
Autophagie , Cardiotoxicité , Colchicine , Doxorubicine , Lysosomes , Myocytes cardiaques , Colchicine/toxicité , Colchicine/pharmacologie , Doxorubicine/toxicité , Cardiotoxicité/prévention et contrôle , Autophagie/effets des médicaments et des substances chimiques , Lysosomes/effets des médicaments et des substances chimiques , Lysosomes/métabolisme , Animaux , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Modèles animaux de maladie humaine , Mâle , Défaillance cardiaque/induit chimiquement , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/métabolisme , Antibiotiques antinéoplasiques/toxicité , Espèces réactives de l'oxygène/métabolisme , Souris , Souris de lignée C57BL , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques
6.
J Am Heart Assoc ; 13(9): e032067, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38700010

RÉSUMÉ

BACKGROUND: Doxorubicin and other anthracyclines are crucial cancer treatment drugs. However, they are associated with significant cardiotoxicity, severely affecting patient care and limiting dosage and usage. Previous studies have shown that low carbon monoxide (CO) concentrations protect against doxorubicin toxicity. However, traditional methods of CO delivery pose complex challenges for daily administration, such as dosing and toxicity. To address these challenges, we developed a novel oral liquid drug product containing CO (HBI-002) that can be easily self-administered by patients with cancer undergoing doxorubicin treatment, resulting in CO being delivered through the upper gastrointestinal tract. METHODS AND RESULTS: HBI-002 was tested in a murine model of doxorubicin cardiotoxicity in the presence and absence of lung or breast cancer. The mice received HBI-002 twice daily before doxorubicin administration and experienced increased carboxyhemoglobin levels from a baseline of ≈1% to 7%. Heart tissue from mice treated with HBI-002 had a 6.3-fold increase in CO concentrations and higher expression of the cytoprotective enzyme heme oxygenase-1 compared with placebo control. In both acute and chronic doxorubicin toxicity scenarios, HBI-002 protected the heart from cardiotoxic effects, including limiting tissue damage and cardiac dysfunction and improving survival. In addition, HBI-002 did not compromise the efficacy of doxorubicin in reducing tumor volume, but rather enhanced the sensitivity of breast 4T1 cancer cells to doxorubicin while simultaneously protecting cardiac function. CONCLUSIONS: These findings strongly support using HBI-002 as a cardioprotective agent that maintains the therapeutic benefits of doxorubicin cancer treatment while mitigating cardiac damage.


Sujet(s)
Antibiotiques antinéoplasiques , Monoxyde de carbone , Cardiotoxicité , Doxorubicine , Protéines membranaires , Animaux , Doxorubicine/toxicité , Monoxyde de carbone/métabolisme , Antibiotiques antinéoplasiques/toxicité , Femelle , Administration par voie orale , Souris , Heme oxygenase-1/métabolisme , Cardiopathies/induit chimiquement , Cardiopathies/prévention et contrôle , Cardiopathies/métabolisme , Cardiopathies/anatomopathologie , Modèles animaux de maladie humaine , Souris de lignée C57BL , Carboxyhémoglobine/métabolisme , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Humains
7.
FASEB J ; 38(10): e23677, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38775792

RÉSUMÉ

Although the use of Doxorubicin (Dox) is extensive in the treatment of malignant tumor, the toxic effects of Dox on the heart can cause myocardial injury. Therefore, it is necessary to find an alternative drug to alleviate the Dox-induced cardiotoxicity. Dihydroartemisinin (DHA) is a semisynthetic derivative of artemisinin, which is an active ingredient of Artemisia annua. The study investigates the effects of DHA on doxorubicin-induced cardiotoxicity and ferroptosis, which are related to the activation of Nrf2 and the regulation of autophagy. Different concentrations of DHA were administered by gavage for 4 weeks in mice. H9c2 cells were pretreated with different concentrations of DHA for 24 h in vitro. The mechanism of DHA treatment was explored through echocardiography, biochemical analysis, real-time quantitative PCR, western blotting analysis, ROS/DHE staining, immunohistochemistry, and immunofluorescence. In vivo, DHA markedly relieved Dox-induced cardiac dysfunction, attenuated oxidative stress, alleviated cardiomyocyte ferroptosis, activated Nrf2, promoted autophagy, and improved the function of lysosomes. In vitro, DHA attenuated oxidative stress and cardiomyocyte ferroptosis, activated Nrf2, promoted clearance of autophagosomes, and reduced lysosomal destruction. The changes of ferroptosis and Nrf2 depend on selective degradation of keap1 and recovery of lysosome. We found for the first time that DHA could protect the heart from the toxic effects of Dox-induced cardiotoxicity. In addition, DHA significantly alleviates Dox-induced ferroptosis through the clearance of autophagosomes, including the selective degradation of keap1 and the recovery of lysosomes.


Sujet(s)
Artémisinines , Autophagie , Cardiotoxicité , Doxorubicine , Ferroptose , Myocytes cardiaques , Facteur-2 apparenté à NF-E2 , Artémisinines/pharmacologie , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Doxorubicine/effets indésirables , Doxorubicine/toxicité , Souris , Ferroptose/effets des médicaments et des substances chimiques , Cardiotoxicité/étiologie , Cardiotoxicité/prévention et contrôle , Cardiotoxicité/métabolisme , Mâle , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Lignée cellulaire , Rats
8.
Int J Mol Sci ; 25(10)2024 May 12.
Article de Anglais | MEDLINE | ID: mdl-38791311

RÉSUMÉ

Doxorubicin (DOX), widely used as a chemotherapeutic agent for various cancers, is limited in its clinical utility by its cardiotoxic effects. Despite its widespread use, the precise mechanisms underlying DOX-induced cardiotoxicity at the cellular and molecular levels remain unclear, hindering the development of preventive and early detection strategies. To characterize the cytotoxic effects of DOX on isolated ventricular cardiomyocytes, focusing on the expression of specific microRNAs (miRNAs) and their molecular targets associated with endogenous cardioprotective mechanisms such as the ATP-sensitive potassium channel (KATP), Sirtuin 1 (SIRT1), FOXO1, and GSK3ß. We isolated Guinea pig ventricular cardiomyocytes by retrograde perfusion and enzymatic dissociation. We assessed cell morphology, Reactive Oxygen Species (ROS) levels, intracellular calcium, and mitochondrial membrane potential using light microscopy and specific probes. We determined the miRNA expression profile using small RNAseq and validated it using stem-loop qRT-PCR. We quantified mRNA levels of some predicted and validated molecular targets using qRT-PCR and analyzed protein expression using Western blot. Exposure to 10 µM DOX resulted in cardiomyocyte shortening, increased ROS and intracellular calcium levels, mitochondrial membrane potential depolarization, and changes in specific miRNA expression. Additionally, we observed the differential expression of KATP subunits (ABCC9, KCNJ8, and KCNJ11), FOXO1, SIRT1, and GSK3ß molecules associated with endogenous cardioprotective mechanisms. Supported by miRNA gene regulatory networks and functional enrichment analysis, these findings suggest that DOX-induced cardiotoxicity disrupts biological processes associated with cardioprotective mechanisms. Further research must clarify their specific molecular changes in DOX-induced cardiac dysfunction and investigate their diagnostic biomarkers and therapeutic potential.


Sujet(s)
Cardiotoxicité , Doxorubicine , microARN , Myocytes cardiaques , Espèces réactives de l'oxygène , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Animaux , Doxorubicine/effets indésirables , Doxorubicine/toxicité , Cardiotoxicité/étiologie , microARN/génétique , microARN/métabolisme , Espèces réactives de l'oxygène/métabolisme , Cochons d'Inde , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Ventricules cardiaques/effets des médicaments et des substances chimiques , Ventricules cardiaques/métabolisme , Ventricules cardiaques/cytologie , Mâle , Calcium/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques
9.
Biomed Pharmacother ; 175: 116748, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38776683

RÉSUMÉ

Doxorubicin (DOX) is a commonly used anthracycline in cancer chemotherapy. The clinical application of DOX is constrained by its cardiotoxicity. Myricetin (MYR) is a natural flavonoid widely present in many plants with antioxidant and anti-inflammatory properties. However, MYR's beneficial effects and mechanisms in alleviating DOX-induced cardiotoxicity (DIC) remain unknown. C57BL/6 mice were injected with 15 mg/kg of DOX to establish the DIC, and MYR solutions were administrated by gavage to investigate its cardioprotective potentials. Histopathological analysis, physiological indicators assessment, transcriptomics analysis, and RT-qPCR were used to elucidate the potential mechanism of MYR in DIC treatment. MYR reduced cardiac injury produced by DOX, decreased levels of cTnI, AST, LDH, and BNP, and improved myocardial injury and fibrosis. MYR effectively prevented DOX-induced oxidative stress, such as lowered MDA levels and elevated SOD, CAT, and GSH activities. MYR effectively suppressed NLRP3 and ASC gene expression levels to inhibit pyroptosis while regulating Caspase1 and Bax levels to reduce cardiac cell apoptosis. According to the transcriptomic analysis, glucose and fatty acid metabolism were associated with differential gene expression. KEGG pathway analysis revealed differential gene enrichment in PPAR and AMPK pathways, among others. Following validation, MYR was found to alleviate DIC by regulating glycolipid metabolism and AMPK pathway-related genes. Our findings demonstrated that MYR could mitigate DIC by regulating the processes of oxidative stress, apoptosis, and pyroptosis. MYR is critical in improving DOX-induced myocardial energy metabolism abnormalities mediated by the AMPK signaling pathway. In conclusion, MYR holds promise as a therapeutic strategy for DIC.


Sujet(s)
Cardiotoxicité , Doxorubicine , Flavonoïdes , Analyse de profil d'expression de gènes , Souris de lignée C57BL , Stress oxydatif , Animaux , Doxorubicine/toxicité , Flavonoïdes/pharmacologie , Cardiotoxicité/prévention et contrôle , Mâle , Souris , Stress oxydatif/effets des médicaments et des substances chimiques , Analyse de profil d'expression de gènes/méthodes , Cardiotoniques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Transcriptome/effets des médicaments et des substances chimiques
10.
Bratisl Lek Listy ; 125(5): 281-288, 2024.
Article de Anglais | MEDLINE | ID: mdl-38624052

RÉSUMÉ

AIM: We aimed to investigate the possible cardioprotective effects of paricalcitol (PR), its vitamin D receptor agonist, and vitamin D3 (VIT-D3) on an experimental model of doxorubicin (DX) cardiotoxicity by 99mTc-PYP scintigraphy, electrocardiographic (ECG) and biochemical methods. METHOD: Forty-two male Wistar/Albino rats (250‒300 g; aged 10‒12 weeks) were randomly separated into six groups, namely into control (CN), doxorubicin (DX), paricalcitol (PR), vitamin D3 (VIT-D3), paricalcitol + doxorubicin (PR+DX), and vitamin D3 + doxorubicin (VIT-D3+DX) groups. Cardiotoxicity was induced by three doses of DX (18 mg/kg, i.p.) at 24-hour intervals on days 18, 19 and 20. PR (0.5 ug/ kg, i.p) and VIT-D3 (5,000 IU/kg, i.p) were injected for 20 days before and after the application of DX (18 mg/kg, i.p.). On day 21 of the experiment, biochemical parameters [tumor necrosis factor TNF-alpha (TNF-α); interleukin-6 (IL-6), nitric oxide (NO), and cardiac troponin T (cTnT)], as well as ECG and scintigraphic (99mTc-PYP) features were assessed. RESULTS: Compared to CN, DX significantly raised TNF-α, IL-6, and NO in heart tissue, cTnT in serum, 99mTc-PYP uptake in the myocardium, and ECG parameters, specifically QRS complex duration, QT interval duration, and ST-segment amplitude, while also reducing heart rate (p<0.001). Pretreatment with PR and VIT-D3 mitigated these abnormalities produced by DX in the heart (p<0.001). CONCLUSION: Results show that vitamin D receptor agonist paricalcitol and vitamin D protect against DX-induced cardiotoxicity through anti-inflammatory and antioxidant effects (Fig. 4, Ref. 59). Text in PDF www.elis.sk Keywords: paricalcitol, doxorubicin, vitamin D, ECG, 99mTc-PYP scintigraphy, cardiotoxicity, inflammation.


Sujet(s)
Cardiotoxicité , Ergocalciférol , Récepteur calcitriol , Rats , Mâle , Animaux , Cardiotoxicité/traitement médicamenteux , Cardiotoxicité/prévention et contrôle , Récepteur calcitriol/usage thérapeutique , Rat Wistar , Cholécalciférol/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Interleukine-6 , Électrocardiographie , Doxorubicine/toxicité , Antioxydants/pharmacologie , Scintigraphie , Stress oxydatif
11.
PLoS One ; 19(4): e0300261, 2024.
Article de Anglais | MEDLINE | ID: mdl-38568919

RÉSUMÉ

Doxorubicin (DOX) is a broad-spectrum, highly effective antitumor agent; however, its cardiotoxicity has greatly limited its use. Hydrogen sulfide (H2S) is an endogenous gaseous transmitter that exerts cardioprotective effects via the regulation of oxidative stress and apoptosis and maintenance of mitochondrial function, among other mechanisms. AP39 is a novel mitochondria-targeted H2S donor that, at appropriate concentrations, attenuates intracellular oxidative stress damage, maintains mitochondrial function, and ameliorates cardiomyocyte injury. In this study, DOX-induced cardiotoxicity models were established using H9c2 cells and Sprague-Dawley rats to evaluate the protective effect of AP39 and its mechanisms of action. Both in vivo and in vitro experiments showed that DOX induces oxidative stress injury, apoptosis, and mitochondrial damage in cardiomyocytes and decreases the expression of p-AMPK/AMPK and UCP2. All DOX-induced changes were attenuated by AP39 treatment. Furthermore, the protective effect of AP39 was significantly attenuated by the inhibition of AMPK and UCP2. The results suggest that AP39 ameliorates DOX-induced cardiotoxicity by regulating the expression of AMPK/UCP2.


Sujet(s)
Sulfure d'hydrogène , Rats , Animaux , Sulfure d'hydrogène/pharmacologie , Sulfure d'hydrogène/métabolisme , Cardiotoxicité/traitement médicamenteux , Cardiotoxicité/étiologie , Cardiotoxicité/prévention et contrôle , AMP-Activated Protein Kinases/métabolisme , Rat Sprague-Dawley , Lignée cellulaire , Doxorubicine/toxicité , Myocytes cardiaques/métabolisme , Stress oxydatif , Mitochondries/métabolisme , Apoptose
12.
J Biochem Mol Toxicol ; 38(4): e23702, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38567888

RÉSUMÉ

Doxorubicin (DOX) is widely used in cancer treatment but the dose-related toxicity of DOX on organs including the liver limit its use. Therefore, there is great interest in combining DOX with natural compounds with antioxidant properties to reduce toxicity and increase drug efficacy. Esculetin is a natural coumarin derivative with biological properties encompassing anti-inflammatory and antioxidant activities. In light of these properties, this study was meticulously crafted to investigate the potential of esculetin in preventing doxorubicin (DOX)-induced hepatotoxicity in Sprague-Dawley rats. The rats were divided into a total of six groups: control group, DOX group (administered DOX at a cumulative dose of 5 mg/kg intraperitoneally every other day for 2 weeks), E50 group (administered 50 mg/kg of esculetin intraperitoneally every day), E100 group (administered 100 mg/kg of esculetin intraperitoneally every day) and combined groups (DOX + E50 and DOX + E100) in which esculetin was administered together with DOX. The treatments, both with DOX alone and in combination with E50, manifested a reduction in catalase (CAT mRNA) levels in comparison to the control group. Notably, the enzymatic activities of superoxide dismutase (SOD), CAT, and glutathione peroxidase (GPx) witnessed significant decreases in the liver of rats treated with DOX. Moreover, DOX treatment induced a statistically significant elevation in malondialdehyde (MDA) levels, coupled with a concurrent decrease in glutathione (GSH) levels. Additionally, molecular docking studies were conducted. However, further studies are needed to confirm the hepatoprotective properties of esculetin and to precisely elucidate its mechanisms of action.


Sujet(s)
Antioxydants , Doxorubicine , Ombelliférones , Rats , Animaux , Antioxydants/pharmacologie , Rat Sprague-Dawley , Simulation de docking moléculaire , Doxorubicine/toxicité , Stress oxydatif , Glutathion/métabolisme , Foie/métabolisme , Antibiotiques antinéoplasiques/pharmacologie
13.
Environ Toxicol ; 39(7): 3872-3882, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38558324

RÉSUMÉ

Platycodi radix is a widely used herbal medicine that contains numerous phytochemicals beneficial to health. The health and biological benefits of P. radix have been found across various diseases. The utilization of umbilical cord stromal stem cells, derived from Wharton's jelly of the human umbilical cord, has emerged as a promising approach for treating degenerative diseases. Nevertheless, growing evidence indicates that the function of stem cells declines with age, thereby limiting their regenerative capacity. The primary objective in this study is to investigate the beneficial effects of P. radix in senescent stem cells. We conducted experiments to showcase that diminished levels of Lamin B1 and Sox-2, along with an elevation in p21, which serve as indicative markers for the senescent stem cells. Our findings revealed the loss of Lamin B1 and Sox-2, coupled with an increase in p21, in umbilical cord stromal stem cells subjected to a low-dose (0.1 µM) doxorubicin (Dox) stimulation. However, P. radix restored the Dox-damage in the umbilical cord stromal stem cells. P. radix reversed the senescent conditions when the umbilical cord stromal stem cells exposed to Dox-induced reactive oxygen species (ROS) and mitochondrial membrane potential are significantly changed. In Dox-challenged aged umbilical cord stromal stem cells, P. radix reduced senescence, increased longevity, prevented mitochondrial dysfunction and ROS and protected against senescence-associated apoptosis. This study suggests that P. radix might be as a therapeutic and rescue agent for the aging effect in stem cells. Inhibition of cell death, mitochondrial dysfunction and aging-associated ROS with P. radix provides additional insights into the underlying molecular mechanisms.


Sujet(s)
Vieillissement de la cellule , Doxorubicine , Mitochondries , Extraits de plantes , Espèces réactives de l'oxygène , Cordon ombilical , Humains , Espèces réactives de l'oxygène/métabolisme , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Cordon ombilical/cytologie , Cordon ombilical/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Doxorubicine/toxicité , Doxorubicine/pharmacologie , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Platycodon/composition chimique , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules cultivées
14.
Pharmacol Rep ; 76(3): 612-621, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38668812

RÉSUMÉ

BACKGROUND: Podocytes have a remarkable ability to recover from injury; however, little is known about the recovery mechanisms involved in this process. We recently showed that formoterol, a long-acting ß2-adrenergic receptor (ß2-AR) agonist, induced mitochondrial biogenesis (MB) in podocytes and led to renoprotection in mice. However, it is not clear whether this effect was mediated by formoterol acting through the ß2-AR or if it occurred through "off-target" effects. METHODS: We genetically deleted the ß2-AR specifically in murine podocytes and used these mice to determine whether formoterol acting through the podocyte ß2-AR alone is sufficient for recovery of renal filtration function following injury. The podocyte-specific ß2-AR knockout mice (ß2-ARfl/fl/PodCre) were generated by crossing ß2-AR floxed mice with podocin Cre (B6.Cg-Tg(NPHS2-cre)295Lbh/J) mice. These mice were then subjected to both acute and chronic glomerular injury using nephrotoxic serum (NTS) and adriamycin (ADR), respectively. The extent of injury was evaluated by measuring albuminuria and histological and immunostaining analysis of the murine kidney sections. RESULTS: A similar level of injury was observed in ß2-AR knockout and control mice; however, the ß2-ARfl/fl/PodCre mice failed to recover in response to formoterol. Functional evaluation of the ß2-ARfl/fl/PodCre mice following injury plus formoterol showed similar albuminuria and glomerular injury to control mice that were not treated with formoterol. CONCLUSIONS: These results indicate that the podocyte ß2-AR is a critical component of the recovery mechanism and may serve as a novel therapeutic target for treating podocytopathies.


Sujet(s)
Agonistes des récepteurs béta-2 adrénergiques , Doxorubicine , Fumarate de formotérol , Souris knockout , Podocytes , Récepteurs bêta-2 adrénergiques , Animaux , Podocytes/métabolisme , Podocytes/effets des médicaments et des substances chimiques , Podocytes/anatomopathologie , Récepteurs bêta-2 adrénergiques/métabolisme , Souris , Fumarate de formotérol/pharmacologie , Agonistes des récepteurs béta-2 adrénergiques/pharmacologie , Doxorubicine/pharmacologie , Doxorubicine/toxicité , Mâle , Souris de lignée C57BL , Albuminurie/métabolisme , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie
15.
Pharmacol Res ; 203: 107165, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38561112

RÉSUMÉ

The clinical use of the DNA damaging anticancer drug doxorubicin (DOX) is limited by irreversible cardiotoxicity, which depends on the cumulative dose. The RAS-homologous (RHO) small GTPase RAC1 contributes to DOX-induced DNA damage formation and cardiotoxicity. However, the pathophysiological relevance of other RHO GTPases than RAC1 and different cardiac cell types (i.e., cardiomyocytes, non-cardiomyocytes) for DOX-triggered cardiac damage is unclear. Employing diverse in vitro and in vivo models, we comparatively investigated the level of DOX-induced DNA damage in cardiomyocytes versus non-cardiomyocytes (endothelial cells and fibroblasts), in the presence or absence of selected RHO GTPase inhibitors. Non-cardiomyocytes exhibited the highest number of DOX-induced DNA double-strand breaks (DSB), which were efficiently repaired in vitro. By contrast, rather low levels of DSB were formed in cardiomyocytes, which however remained largely unrepaired. Moreover, DOX-induced apoptosis was detected only in non-cardiomyocytes but not in cardiomyocytes. Pharmacological inhibitors of RAC1 and CDC42 most efficiently attenuated DOX-induced DNA damage in all cell types examined in vitro. Consistently, immunohistochemical analyses revealed that the RAC1 inhibitor NSC23766 and the pan-RHO GTPase inhibitor lovastatin reduced the level of DOX-induced residual DNA damage in both cardiomyocytes and non-cardiomyocytes in vivo. Overall, we conclude that endothelial cells, fibroblasts and cardiomyocytes contribute to the pathophysiology of DOX-induced cardiotoxicity, with RAC1- and CDC42-regulated signaling pathways being especially relevant for DOX-stimulated DSB formation and DNA damage response (DDR) activation. Hence, we suggest dual targeting of RAC1/CDC42-dependent mechanisms in multiple cardiac cell types to mitigate DNA damage-dependent cardiac injury evoked by DOX-based anticancer therapy.


Sujet(s)
Aminoquinoléines , Doxorubicine , Cellules endothéliales , Fibroblastes , Myocytes cardiaques , Pyrimidines , Protéine G cdc42 , Protéine G rac1 , Protéine G rac1/métabolisme , Protéine G rac1/antagonistes et inhibiteurs , Protéine G rac1/génétique , Animaux , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Protéine G cdc42/métabolisme , Doxorubicine/toxicité , Doxorubicine/effets indésirables , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/anatomopathologie , Cellules endothéliales/métabolisme , Cardiotoxicité , Antibiotiques antinéoplasiques/toxicité , Souris , Apoptose/effets des médicaments et des substances chimiques , Mâle , Humains , Souris de lignée C57BL , Cassures double-brin de l'ADN/effets des médicaments et des substances chimiques , Neuropeptides/métabolisme , Altération de l'ADN/effets des médicaments et des substances chimiques , Cellules cultivées
16.
J Ethnopharmacol ; 329: 118156, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38583729

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Throughout Chinese history, Hydrangea paniculata Siebold has been utilized as a traditional medicinal herb to treat a variety of ailments associated to inflammation. In a number of immune-mediated kidney disorders, total coumarins extracted from Hydrangea paniculata (HP) have demonstrated a renal protective effect. AIM OF THE STUDY: To investigate renal beneficial effect of HP on experimental Adriamycin nephropathy (AN), and further clarify whether reversing lipid metabolism abnormalities by HP contributes to its renoprotective effect and find out the underlying critical pathways. MATERIALS AND METHODS: After establishment of rat AN model, HP was orally administrated for 6 weeks. Biochemical indicators related to kidney injury were determined. mRNAs sequencing using kidney tissues were performed to clarify the underlying mechanism. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis, western blot, molecular docking, and drug affinity responsive target stability (DARTS) assay was carried out to further explore and confirm pivotal molecular pathways and possible target by which HP and 7-hydroxylcoumarin (7-HC) played their renal protection effect via modulating lipid metabolism. RESULTS: HP could significantly improve renal function, and restore renal tubular abnormal lipid metabolism and interstitial fibrosis in AN. In vitro study demonstrated that HP and its main metabolite 7-HC could reduce ADR-induced intracellular lipid deposition and fibrosis characteristics in renal tubular cells. Mechanically, HP and 7-HC can activate AMP-activated protein kinase (AMPK) via direct interaction, which contributes to its lipid metabolism modulation effect. Moreover, HP and 7-HC can inhibit fibrosis by inhibiting CCAAT/enhancer binding protein beta (C/EBPß) expression in renal tubular cells. Normalization of lipid metabolism by HP and 7-HC further provided protection of mitochondrial structure integrity and inhibited the nuclear factor kappa-B (NF-κB) pathway. Long-term toxicity using beagle dogs proved the safety of HP after one-month administration. CONCLUSION: Coumarin derivates from HP alleviate adriamycin-induced lipotoxicity and fibrosis in kidney through activating AMPK and inhibiting C/EBPß.


Sujet(s)
AMP-Activated Protein Kinases , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT , Coumarines , Doxorubicine , Hydrangea , Animaux , Doxorubicine/toxicité , Coumarines/pharmacologie , Coumarines/isolement et purification , Mâle , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/métabolisme , AMP-Activated Protein Kinases/métabolisme , Rats , Hydrangea/composition chimique , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Rein/anatomopathologie , Rat Sprague-Dawley , Maladies du rein/induit chimiquement , Maladies du rein/traitement médicamenteux , Maladies du rein/métabolisme , Maladies du rein/prévention et contrôle , Simulation de docking moléculaire , Métabolisme lipidique/effets des médicaments et des substances chimiques , Lignée cellulaire , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Ombelliférones
17.
Biol Direct ; 19(1): 32, 2024 Apr 29.
Article de Anglais | MEDLINE | ID: mdl-38685056

RÉSUMÉ

BACKGROUND: Doxorubicin (Dox) is associated with various liver injuries, limiting its clinical utility. This study investigates whether NSUN2 participates in Dox-induced liver injury and the associated molecular mechanism. METHODS: In vivo and in vitro liver cell injury models were constructed based on Dox therapy. The protein levels of NSUN2 and oxidative stress indicators Nrf2, HO-1, and NQO1 were evaluated by Western blot. The RNA binding potential was detected by RNA methylation immunoprecipitation (RIP). Additionally, the effect of NSUN2 on Nrf2 mRNA synthesis and localization was evaluated using an RNA fluorescence probe. RESULTS: NSUN2 was downregulated, and liver tissue suffered significant pathological damage in the Dox group. The levels of ALT and AST significantly increased. NSUN2 interference exacerbated Dox-induced liver cell damage, which was reversed by NSUN2 overexpression. RIP demonstrated that NSUN2 recognized and bound to Nrf2 mRNA. Western blot analysis showed the protein level of Nrf2 in the NSUN2-WT group was significantly higher than that of the control group, whereas there was no significant change in Nrf2 level in the mutant NSUN2 group. Luciferase analysis demonstrated that NSUN2 could recognize and activate the Nrf2 5'UTR region of LO2 cells. In addition, RIP analysis revealed that ALYREF could recognize and bind to Nrf2 mRNA and that ALYREF controls the regulatory effect of NSUN2 on Nrf2. CONCLUSION: NSUN2 regulates Dox-induced liver cell damage by increasing Nrf2 mRNA m5C methylation to inhibit inhibiting antioxidant stress. The regulatory effect of NSUN2 on Nrf2 depends on ALYREF.


Sujet(s)
Carboxylic ester hydrolases , Doxorubicine , Facteur-2 apparenté à NF-E2 , Stress oxydatif , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Doxorubicine/toxicité , Doxorubicine/effets indésirables , Stress oxydatif/effets des médicaments et des substances chimiques , Animaux , Souris , Lésions hépatiques dues aux substances/métabolisme , Mâle , Humains , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques
18.
Free Radic Biol Med ; 218: 94-104, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38582228

RÉSUMÉ

Lamin A/C, essential inner nuclear membrane proteins, have been linked to progeria, a disease of accelerated aging, and many other diseases, which include cardiac disorder. Lamin A/C mutation and its phosphorylation are associated with altering nuclear shape and size. The role of lamin A/C in regulating normal cardiac function was reported earlier. In the present study, we hypothesized that Doxorubicin (Dox) may alter total lamin A/C expression and phosphorylation, thereby taking part in cardiac injury. An in vitro cellular injury model was generated with Dox (0.1-10.0 µM) treatment on cardiomyoblast cells (H9c2) to prove our hypothesis. Increased size and irregular (ameboid) nucleus shape were observed in H9c2 cells after Dox treatment. Similarly, we have observed a significant increase in cell death on increasing the Dox concentration. The expression of lamin A/C and its phosphorylation at serine 22 significantly decreased and increased, respectively in H9c2 cells and rat hearts after Dox exposure. Phosphorylation led to depolymerization of the lamin A/C in the inner nuclear membrane and was evidenced by their presence throughout the nucleoplasm as observed by immunocytochemistry techniques. Thinning and perforation on the walls of the nuclear membrane were observed in Dox-treated H9c2 cells. LMNA-overexpression in H9c2 protected the cells from Dox-induced cell death, reversing all changes described above. Further, improvement of lamin A/C levels was observed in Dox-treated H9c2 cells when treated with Purvalanol A, a CDK1 inhibitor and N-acetylcysteine, an antioxidant. The study provides new insight regarding Dox-induced cardiac injury with the involvement of lamin A/C and alteration of inner nuclear membrane structure.


Sujet(s)
Cardiotoxicité , Doxorubicine , Lamine A , Enveloppe nucléaire , Doxorubicine/toxicité , Lamine A/métabolisme , Lamine A/génétique , Animaux , Phosphorylation/effets des médicaments et des substances chimiques , Enveloppe nucléaire/métabolisme , Enveloppe nucléaire/effets des médicaments et des substances chimiques , Rats , Cardiotoxicité/métabolisme , Cardiotoxicité/anatomopathologie , Cardiotoxicité/étiologie , Lignée cellulaire , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Antibiotiques antinéoplasiques/toxicité , Mâle , Rat Sprague-Dawley
19.
Biochem Biophys Res Commun ; 710: 149910, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38593619

RÉSUMÉ

Ginsenoside Rb1 (Rb1), an active component isolated from traditional Chinese medicine Ginseng, is beneficial to many cardiovascular diseases. However, whether it can protect against doxorubicin induced cardiotoxicity (DIC) is not clear yet. In this study, we aimed to investigate the role of Rb1 in DIC. Mice were injected with a single dose of doxorubicin (20 mg/kg) to induce acute cardiotoxicity. Rb1 was given daily gavage to mice for 7 days. Changes in cardiac function, myocardium histopathology, oxidative stress, cardiomyocyte mitochondrion morphology were studied to evaluate Rb1's function on DIC. Meanwhile, RNA-seq analysis was performed to explore the potential underline molecular mechanism involved in Rb1's function on DIC. We found that Rb1 treatment can improve survival rate and body weight in Dox treated mice group. Rb1 can attenuate Dox induced cardiac dysfunction and myocardium hypertrophy and interstitial fibrosis. The oxidative stress increase and cardiomyocyte mitochondrion injury were improved by Rb1 treatment. Mechanism study found that Rb1's beneficial role in DIC is through suppressing of autophagy and ferroptosis. This study shown that Ginsenoside Rb1 can protect against DIC by regulating autophagy and ferroptosis.


Sujet(s)
Cardiotoxicité , Ferroptose , Ginsénosides , Animaux , Souris , Apoptose/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Cardiotoxicité/traitement médicamenteux , Cardiotoxicité/métabolisme , Cardiotoxicité/prévention et contrôle , Doxorubicine/effets indésirables , Doxorubicine/toxicité , Ginsénosides/pharmacologie , Myocytes cardiaques/métabolisme , Stress oxydatif
20.
Arch Toxicol ; 98(6): 1781-1794, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38573338

RÉSUMÉ

Doxorubicin (DOX) is one of the most frequently used chemotherapeutic drugs belonging to the class of anthracyclines. However, the cardiotoxic effects of anthracyclines limit their clinical use. Recent studies have suggested that ferroptosis is the main underlying pathogenetic mechanism of DOX-induced cardiomyopathy (DIC). BTB-and-CNC homology 1 (Bach1) acts as a key role in the regulation of ferroptosis. However, the mechanistic role of Bach1 in DIC remains unclear. Therefore, this study aimed to investigate the underlying mechanistic role of Bach1 in DOX-induced cardiotoxicity using the DIC mice in vivo (DOX at cumulative dose of 20 mg/kg) and the DOX-treated H9c2 cardiomyocytes in vitro (1 µM). Our results show a marked upregulation in the expression of Bach1 in the cardiac tissues of the DOX-treated mice and the DOX-treated cardiomyocytes. However, Bach1-/- mice exhibited reduced lipid peroxidation and less severe cardiomyopathy after DOX treatment. Bach1 knockdown protected against DOX-induced ferroptosis in both in vivo and in vitro models. Ferrostatin-1 (Fer-1), a potent inhibitor of ferroptosis, significantly alleviated DOX-induced cardiac damage. However, the cardioprotective effects of Bach1 knockdown were reversed by pre-treatment with Zinc Protoporphyrin (ZnPP), a selective inhibitor of heme oxygenase-1(HO-1). Taken together, these findings demonstrated that Bach1 promoted oxidative stress and ferroptosis through suppressing the expression of HO-1. Therefore, Bach1 may present as a promising new therapeutic target for the prevention and early intervention of DOX-induced cardiotoxicity.


Sujet(s)
Facteurs de transcription à motif basique et à glissière à leucines , Cardiomyopathies , Doxorubicine , Ferroptose , Heme oxygenase-1 , Souris de lignée C57BL , Souris knockout , Myocytes cardiaques , Stress oxydatif , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Doxorubicine/toxicité , Stress oxydatif/effets des médicaments et des substances chimiques , Cardiomyopathies/induit chimiquement , Cardiomyopathies/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Facteurs de transcription à motif basique et à glissière à leucines/métabolisme , Facteurs de transcription à motif basique et à glissière à leucines/génétique , Mâle , Souris , Heme oxygenase-1/métabolisme , Heme oxygenase-1/génétique , Lignée cellulaire , Rats , Cardiotoxicité , Antibiotiques antinéoplasiques/toxicité , Peroxydation lipidique/effets des médicaments et des substances chimiques , Protoporphyrines/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Cyclohexylamines , Protéines membranaires , Phénylènediamines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...