[1]沈鐲钰 黄丹 赵淑红 马振国.卡格列净对阿霉素所致心肌细胞损伤的作用及机制研究[J].心血管病学进展,2025,(3):272.[doi:10.16806/j.cnki.issn.1004-3934.2025.03.017]
 SHEN Zheyu,HUANG Dan,ZHAO Shuhong,et al.Canagliflozin’s Role in Ameliorating Doxorubicin-Induced Injury in Cardiomyocytes[J].Advances in Cardiovascular Diseases,2025,(3):272.[doi:10.16806/j.cnki.issn.1004-3934.2025.03.017]
点击复制

卡格列净对阿霉素所致心肌细胞损伤的作用及机制研究()
分享到:

《心血管病学进展》[ISSN:51-1187/R/CN:1004-3934]

卷:
期数:
2025年3期
页码:
272
栏目:
论著
出版日期:
2025-03-25

文章信息/Info

Title:
Canagliflozin’s Role in Ameliorating Doxorubicin-Induced Injury in Cardiomyocytes
作者:
沈鐲钰 黄丹 赵淑红 马振国
(武汉大学人民医院心血管内科 代谢与相关慢病湖北省重点实验室,湖北 武汉 430060 )
Author(s):
SHEN ZheyuHUANG DanZHAO ShuhongMA Zhengguo
(Department of Cardiology,Renmin Hospital of Wuhan University,Hubei Key Laboratory of Metabolic and Chronic Diseases ,Wuhan 430060,Hubei,China)
关键词:
卡格列净阿霉素炎症凋亡氧化应激蛋白激酶B
Keywords:
CanagliflozinDoxorubicinInflammationApoptosisOxidative stressProtein kinase B
DOI:
10.16806/j.cnki.issn.1004-3934.2025.03.017
摘要:
目的 研究卡格列净(CANA)对阿霉素(DOX)诱导的心肌细胞损伤的影响及其作用机制。方法 使用1 μmol/LDOX构建原代大鼠心肌细胞损伤模型,构建CANA浓度梯度(1、5、10、20和30 μmol/L)与DOX共刺激后,利用CCK-8法检测心肌细胞活力,结果显示当CANA浓度为10 μmol/L时细胞活力恢复最明显,后续实验CANA浓度均采用10 μmol/L;将原代大鼠心肌细胞随机分为4组,分别是对照组、CANA组、DOX组、DOX+CANA组;使用酶联免疫吸附试验检测乳酸脱氢酶、肌酸激酶同工酶、超氧化物歧化酶、过氧化氢酶和丙二醛含量,蛋白质印迹法检测B淋巴细胞瘤-2(Bcl-2)、Bcl-2相关X蛋白、裂解型半胱氨酸天冬氨酸蛋白酶-3、蛋白激酶B和糖原合成酶激酶-3β的表达,TUNEL染色验证细胞凋亡,实时荧光定量聚合酶链反应检测白细胞介素-1β、白细胞介素-6、肿瘤坏死因子-α转录水平。结果 和对照组比较,DOX组心肌细胞活力显著降低,且心肌损伤标志物水平显著升高(P<0.05);细胞凋亡增加,Bcl-2相关X蛋白、裂解型半胱氨酸天冬氨酸蛋白酶-3以及丙二醛水平明显升高(P<0.05),而Bcl-2水平、过氧化氢酶以及超氧化物歧化酶活性显著降低(P<0.05);炎症反应增强,白细胞介素-1β、白细胞介素-6及肿瘤坏死因子-α转录水平显著上调(P<0.05);蛋白激酶B/糖原合成酶激酶-3β表达被明显抑制(P<0.05)。与DOX组比较,DOX+CANA组心肌细胞活力明显增加,且心肌损伤标志物水平降低(P<0.05);细胞凋亡减少,Bcl-2相关X蛋白、裂解型半胱氨酸天冬氨酸蛋白酶-3以及丙二醛水平明显下降(P<0.05),而Bcl-2水平、过氧化氢酶以及超氧化物歧化酶活性显著增加(P<0.05);炎症反应减弱,白细胞介素-1β、白细胞介素-6及肿瘤坏死因子-α转录水平明显下调(P<0.05);蛋白激酶B/糖原合成酶激酶-3β表达水平显著升高(P<0.05)。 结论 CANA可通过抑制氧化应激、减轻炎症反应、抑制细胞凋亡以及调控蛋白激酶B信号通路来减轻DOX诱导的心肌细胞损伤。
Abstract:
Objective To investigate the effects of canagliflozin (CANA) on doxorubicin(DOX)-induced cardiomyocyte injury and its underlying mechanisms. Methods P rimary neonatal rat cardiomyoc yte inj ury model was established using 1 μmol/L DOX. A gradient of CANA concentrations (1 ,5,10,20,30 μmol/L) was co-administered with DOX,and cell viability was assessed by the CCK-8 assay, t he results indicated that cell viability was most prominently restored at a CANA concentration of 10 μmol/L, and henceforth, a concentration of 10 μmol/L was employed for CANA in all subsequent experiments. Primary neonatal rat cardiomyocytes were randomly divided into four groups :Control,CANA,DOX,and DOX+CANA. Enzyme linked immunosorbent assay was employed to measure lactate dehydro genase,creatine kinase isoenzyme,superoxide dismutase,catalase,and malondialdehyde levels. Western blotting was used to detect the expression of B-cell lymphoma-2( Bcl-2),Bcl-2-associated X,cleaved caspase-3,protein kinase B and glycogen synthase kinase-3β. TUNEL staining was used to confirm apoptosis,and real-time quantitative polymerase chain reaction was used to quantif y the transcription levels of i nterleukin-1β,interleukin-6,and tumor necrosis factor-α. Results Compared with the control group ,the DOX group showed significantly reduced cell viability and elevated levels of myocardial injury markers ( P<0.05). Increased apoptosis,along with heightened levels of Bcl-2-associated X,cleaved caspase-3,and malondialdehyde,was observed,while Bcl-2 levels ,catalase activity,and superoxide dismutase activity were notably reduced (P<0.05). Enhanced inflammatory response was evidenced by significantly upregulated transcription of i nterleukin-1β,interleukin-6,and tumor necrosis factor-α ( P<0.05),accompanied by suppressed protein kinase B/glycogen synthase kinase-3β expression ( P<0.05). In contrast,the DOX+CANA group demonstrated increased cell viability ,reduced myocardial injury marker levels,decreased apoptosis,lowered Bcl-2-associated X,cleaved caspase-3,and malondialdehyde levels,and elevated Bcl-2 levels ,catalase activity ,and superoxide dismutase activity compared to the DOX group ( P<0.05). The inflammatory response was attenuated,with transcription levels of interleukin-1β ,interleukin-6,and tumor necrosis factor-α significantly downregulated .The expression levels of protein kinase B/glycogen synthase kinase-3β are significantly increased ( P<0.05). Conclusion CANA alleviates DOX-induced cardiomyocyte damage through inhibiting oxidative stress,mitigating inflammation,suppressing apoptosis,and modulating the protein kinase B signaling pathway.

参考文献/References:

[1] Ghignatti PVDC,Nogueira LJ,Lehnen AM,et al. Cardioprotective effects of exercise training on doxorubicin-induced cardiomyopathy:a systematic review with meta-analysis of preclinical studies[J]. Sci Rep,2021,11(1):6330.

[2] Wu J,Feng A,Liu C,et al. Genistein alleviates doxorubicin-induced cardiomyocyte autophagy and apoptosis via ERK/STAT3/c-Myc signaling pathway in rat model[J]. Phytother Res,2024,38(8):3921-3934.

[3] Zhu W,Lian N,Wang J,et al. Liguzinediol potentiates the metabolic remodeling by activating the AMPK/SIRT3 pathway and represses Caspase-3/GSDME-mediated pyroptosis to ameliorate cardiotoxicity[J]. Chin Med,2024,19(1):85.

[4] Ge X,Zhang T,Yu X,et al. LIM-Nebulette reinforces podocyte structural integrity by linking actin and vimentin filaments [J]. J Am Soc Nephrol,2020,31(10):2372-2391.

[5] Steinberg GR,Hardie DG. New insights into activation and function of the AMPK[J]. Nat Rev Mol Cell Biol,2023,24(4):255-272.

[6] Zinman B,Wanner C,Lachin JM,et al. Empagliflozin ,cardiovascular outcomes,and mortality in Type 2 diabetes[J]. N Engl J Med,2015,373(22):2117-2128.

[7] Voors AA,Angermann CE,Teerlink JR,et al. The SGLT2 inhibitor empagliflozin in patients hospitalized for acute heart failure :a multinational randomized trial[J]. Nat Med,2022,28(3):568-574.

[8] Zhong J,Sun P,Xu N,et al. Canagliflozin inhibits P-gp function and early autophagy and improves the sensitivity to the antitumor effect of doxorubicin[J]. Biochem Pharmacol,2020,175:113856.

[9] Liu D,Ma Z,Di S,et al. AMPK/PGC1α activation by melatonin attenuates acute doxorubicin cardiotoxicity via alleviating mitochondrial oxidative damage and apoptosis[J]. Free Radic Biol Med,2018,129:59-72.

[10] Zhang X,Hu C,Zhang N,et al. Matrine attenuates pathological cardiac fibrosis via RPS5/p38 in mice[J]. Acta Pharmacol Sin,2021,42(4):573-584.

[11] Hu C,Zhang X,Zhang N,et al. Osteocrin attenuates inflammation,oxidative stress,apoptosis,and cardiac dysfunction in doxorubicin-induced cardiotoxicity[J]. Clin Transl Med,2020,10(3):e124.

[12] Vitale R,Marzocco S,Popolo A. Role of oxidative stress and inflammation in doxorubicin-induced cardiotoxicity:a brief account[J]. Int J Mol Sci,2024,25(13):7477.

[13] Zamorano JL,Lancellotti P,Rodriguez Mu?oz D,et al. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines:The Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC)[J]. Eur Heart J,2016,37(36):2768-2801.

[14] Kong CY,Guo Z,Song P,et al. Underlying the mechanisms of doxorubicin-induced acute cardiotoxicity:oxidative stress and cell death[J]. Int J Biol Sci,2022,18(2):760-770.

[15] Christidi E,Brunham LR. Regulated cell death pathways in doxorubicin-induced cardiotoxicity[J]. Cell Death Dis,2021,12(4):339.

[16] Liu P,Li J,Liu M,et al. Hesperetin modulates the Sirt1/Nrf2 signaling pathway in counteracting myocardial ischemia through suppression of oxidative stress,inflammation,and apoptosis[J]. Biomed Pharmacother,2021,139:111552.

[17] Zhang X,Huang C,Hou Y,et al. Research progress on the role and mechanism of Sirtuin family in doxorubicin cardiotoxicity[J]. Phytomedicine,2024,129:155673.

[18] Elmorsy EA,Saber S,Hamad RS,et al. Mechanistic insights into carvedilol’s potential protection against doxorubicin-induced cardiotoxicity[J]. Eur J Pharm Sci,2024,200:106849.

[19] Kitakata H,Endo J,Ikura H,et al. Therapeutic targets for DOX-induced cardiomyopathy:role of apoptosis vs. ferroptosis[J]. Int J Mol Sci,2022,23(3):1414.

[20] Zhang X,Hu C,Kong CY,et al. FNDC5 alleviates oxidative stress and cardiomyocyte apoptosis in doxorubicin-induced cardiotoxicity via activating AKT[J]. Cell Death Differ,2020,27(2):540-555.

[21] Li F,Zhan Z,Qian J,et al. Naringin attenuates rat myocardial ischemia/reperfusion injury via PI3K/Akt pathway-mediated inhibition of apoptosis,oxidative stress and autophagy[J]. Exp Ther Med,2021,22(2):811.

[22] Lee CT,Lin KD,Hsieh CF,et al. SGLT2 inhibitor canagliflozin alleviates high glucose-induced inflammatory toxicity in BV-2 microglia[J]. Biomedicines,2023,12(1):36.

相似文献/References:

[1]王菲 卢新政.卡格列净对2型糖尿病患者心血管保护作用的研究进展[J].心血管病学进展,2020,(3):231.[doi:10.16806/j.cnki.issn.1004-3934.2020.03.004]
 WANG Fei,LU Xinzheng.Cardiovascular Protection of Canagliflozin in Type 2 Diabetes Patients[J].Advances in Cardiovascular Diseases,2020,(3):231.[doi:10.16806/j.cnki.issn.1004-3934.2020.03.004]
[2]王鹏 王鑫 张瑶 张经泽 贺世豪 李瑾.内质网应激在阿霉素心脏毒性中作用的研究进展[J].心血管病学进展,2023,(4):341.[doi:10.16806/j.cnki.issn.1004-3934.2023.04.012]
 WANG Peng,WANG Xin,ZHANG Yao,et al.Role of Endoplasmic Reticulum Stress?n the Cardiotoxicity of Doxorubicin[J].Advances in Cardiovascular Diseases,2023,(3):341.[doi:10.16806/j.cnki.issn.1004-3934.2023.04.012]
[3]李登科 张伟 黄从新.SIRT1介导的信号通路在阿霉素诱导心脏毒性中的作用机制[J].心血管病学进展,2024,(3):257.[doi:10.16806/j.cnki.issn.1004-3934.2024.03.015]
 LI Dengke,ZHANG Wei,HUANG Congxin.The Mechanisms of SIRT1-Mediated Signal Pathway in Doxorubicin-Induced Cardiotoxicity[J].Advances in Cardiovascular Diseases,2024,(3):257.[doi:10.16806/j.cnki.issn.1004-3934.2024.03.015]
[4]李铭,高继先,吴冰,等.卡格列净减轻心肌梗死后细胞凋亡及炎症反应改善预后[J].心血管病学进展,2024,(4):368.[doi:10.16806/j.cnki.issn.1004-3934.2024.04.018]
 LI Ming,GAO Jixian,WU Bing,et al.Canagliflozin alleviates apoptosis and inflammation after myocardial infarction, and improves prognosis[J].Advances in Cardiovascular Diseases,2024,(3):368.[doi:10.16806/j.cnki.issn.1004-3934.2024.04.018]
[5]李登科 张伟 黄从新.黄芩苷通过调节Nrf2/HO-1信号通路减轻阿霉素诱导的H9c2细胞毒性[J].心血管病学进展,2024,(5):457.[doi:10.16806/j.cnki.issn.1004-3934.2024.05.017]
 LI Dengke,ZHANG Wei,HUANG Congxin.Baicalin Reduces Doxorubicin Induced H9c2 Cell Toxicity by Regulating the Nrf2/HO-1 Signaling Pathway[J].Advances in Cardiovascular Diseases,2024,(3):457.[doi:10.16806/j.cnki.issn.1004-3934.2024.05.017]

更新日期/Last Update: 2025-04-29