您的位置:山东大学 -> 科技期刊社 -> 《山东大学学报(医学版)》

山东大学学报 (医学版) ›› 2022, Vol. 60 ›› Issue (8): 14-22.doi: 10.6040/j.issn.1671-7554.0.2022.0250

• 基础医学 • 上一篇    下一篇

葛根素对索拉非尼心肌毒性的保护及作用机制

姜卉1,魏甜2,李建平1,王聪3   

  1. 1.山东大学第二医院儿内科, 山东 济南 250033;2.山东大学第二医院胃肠外科 山东大学齐鲁医学院, 山东 济南 250033;3.山东第一医科大学第一附属医院心血管内科, 山东 济南 250015
  • 发布日期:2022-07-27
  • 通讯作者: 王聪. E-mail:congwang87@outlook.com
  • 基金资助:
    山东省自然科学基金青年基金(ZR2021QH026)

Protective effects and molecular mechanism of puerarin on sorafenib-induced cardiotoxicity

JIANG Hui1, WEI Tian2, LI Jianping1, WANG Cong3   

  1. 1. Department of Pediatrics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, Shandong, China;
    2. Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, Shandong, China;
    3. Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong, China
  • Published:2022-07-27

摘要: 目的 探讨葛根素通过抑制心肌铁死亡对索拉非尼心肌毒性的保护及其分子作用机制。 方法 细胞实验:分离培养大鼠乳鼠原代心肌细胞,给予不同的药物处理,分为二甲基亚砜(DMSO)组、索拉非尼组、索拉非尼+Ferrostatin-1(Fer-1)组、索拉非尼+去铁胺组、索拉非尼+葛根素组,检测各组细胞活力和乳酸脱氢酶(LDH)释放,观察光镜下细胞形态,并检测脂质活性氧(ROS)水平以及脂质过氧化代谢产物丙二醛(MDA)水平;采用Western blotting法检测上述分组以及索拉非尼处理心肌细胞0、12、24、36、48、60、72 h后内质网应激标志物葡萄糖调节蛋白(GRP78)水平。给予不同的药物处理,将心肌细胞分为索拉非尼组、索拉非尼+葛根素组、索拉非尼+毒胡萝卜素组以及索拉非尼+葛根素+毒胡萝卜素,检测各组细胞活力、LDH释放、MDA水平,采用Western blotting法检测GRP78水平。小鼠实验:将40只C57BL/6J小鼠随机分为对照组、葛根素组、索拉非尼组、索拉非尼+葛根素组,采用ELISA法检测血浆肌酸激酶同工酶(CK-MB)水平,测算心脏质量/胫骨长度(HW/TL)、心肌组织切片Masson染色后测量心肌细胞横截面积(CSA),检测心肌组织ROS、MDA水平,采用Western blotting法检测心肌组织GRP78水平。 结果 细胞实验中索拉非尼组较DMSO组细胞活力降低、LDH释放增加,细胞空泡化,脂质ROS及MDA水平升高,GRP78蛋白上调,差异均有统计学意义(P均<0.01);与索拉非尼组相比,索拉非尼+ Fer-1组、索拉非尼+去铁胺组以及索拉非尼+葛根素组细胞活力升高、LDH释放减少、空泡化减轻,脂质ROS及MDA水平降低,且GRP78蛋白下调,差异均有统计学意义(P均<0.05);内质网应激诱导剂毒胡萝卜素和葛根素对GRP78水平、细胞活力、LDH释放、MDA水平的交互作用差异有统计学意义(P均<0.05)。小鼠实验中索拉非尼组较对照组CK-MB水平升高,HW/TL下降,CSA升高,脂质ROS和MDA水平升高,GRP78蛋白上调,差异均有统计学意义(P<0.01);葛根素与索拉非尼间的交互作用差异有统计学意义(P均<0.05);与索拉非尼组相比,索拉非尼+葛根素组小鼠CK-MB水平下降、HW/TL升高,CSA下降,脂质ROS和MDA水平降低,GRP78蛋白下调,差异均有统计学意义(P均<0.05)。 结论 葛根素可以抑制索拉非尼引发的心肌铁死亡,对索拉非尼的心肌毒性具有良好的保护作用,毒胡萝卜素可拮抗葛根素的细胞保护和抑制脂质过氧化作用,葛根素通过清除脂质ROS抑制内质网应激可能是其背后主要的分子机制。

关键词: 索拉非尼, 心肌毒性, 葛根素, 铁死亡, 内质网应激

Abstract: Objective To elucidate the protective effects and molecular mechanism of puerarin on sorafenib-induced cardiotoxicity via inhibiting the ferroptosis of cardiomyocytes. Methods In the cell study, primary cardiomyocytes were isolated and treated with dimethyl sulfoxide(DMSO), sorafenib, sorafenib+deferoxamine, sorafenib+ferrostatin-1(Fer-1), and sorafenib+puerarin, respectively. The cell viability, lactate dehydrogenase(LDH)release, lipid ROS, and malondialdehyde(MDA)level were measured. The cell morphology was observed with microscopy. At 0, 12, 24, 36, 48, 60 and 72 h after treatment, cardiomyocytes were harvested and the classic marker of endoplasmic reticulum(ER)stress, glucose-regulated protein 78(GRP78)protein was detected with Western blotting. Cardiomyocytes were treated with sorafenib, sorafenib+puerarin, sorafenib+puerarin+thapsigargin, and sorafenib+puerarin+thapsigargin, respectively. The cell viability, LDH release, lipid ROS, MDA level and GRP78 level were measured. In the mice study, 40 C57BL/6J mice were divided into the control, puerarin, sorafenib, and sorafenib+puerarin groups. The level of serum creatine kinase MB isoenzyme(CK-MB)was determined with ELISA. The heart weight/tibia length(HW/TL)was evaluated. The cross-sectional area(CSA)was measured after Masson staining. The ROS, MDA and GRP78 levels of cardiac tissue were measured. Results In the cell study, compared to the DMSO group, the sorafenib group had decreased cell viability, increased LDH release, increased lipid ROS, MDA and GRP78 levels(all P<0.01). Compared with the sorafenib group, the sorafenib+Fer-1 group, sorafenib+deferroamine group and sorafenib+puerarin group had increased cell viability, decreased LDH release, decreased vacuolation, decreased lipid ROS, MDA and GRP78 levels(all P<0.05). The interactions between ER stress inducers thapsigargin and puerarin on GRP78 level, cell viability, LDH release and MDA level were statistically significant(all P<0.05). In the mice study, sorafenib administration caused elevated CK-MB level, decreased HW/TL, larger CSA of surviving cardiomyocytes, increased lipid ROS level and MDA content, and upregulated level of GRP78, which could all be inhibited by puerarin(all P<0.05). Compared with the sorafenib group, the sorafenib+puerarin group had decreased level of CK-MB, increased HW/TL, decreased CSA, decreased ROS, MDA and GRP78 levels(all P<0.05). Conclusion Puerarin inhibits sorafenib-induced ferroptosis of cardiomyocytes via inhibiting ER stress, which might be triggered by lipid ROS. Puerarin has a promising role in alleviating sorafenib-related cardiotoxicity.

Key words: Sorafenib, Cardiotoxicity, Puerarin, Ferroptosis, Endoplasmic reticulum stress

中图分类号: 

  • R541.9
[1] Fan G, Wei X, Xu X. Is the era of sorafenib over? A review of the literature [J]. Ther Adv Med Oncol, 2020, 12: 1758835920927602. doi: 10.1177/1758835920927602.
[2] Carballo-Folgoso L, álvarez-Velasco R, Lorca R, et al. Evaluation of cardiovascular events in patients with hepatocellular carcinoma treated with sorafenib in the clinical practice. The CARDIO-SOR study [J]. Liver Int, 2021, 41(9): 2200-2211.
[3] Ma WZ, Liu M, Liang FF, et al. Cardiotoxicity of sorafenib is mediated through elevation of ROS level and CaMKII activity and dysregulation of calcium homoeostasis [J]. Basic Clin Pharmacol Toxicol, 2020, 126(2): 166-180.
[4] Hasinoff BB, Patel D. Mechanisms of myocyte cytotoxicity induced by the multikinase inhibitor sorafenib [J]. Cardiovasc Toxicol, 2010, 10(1): 1-8.
[5] Duran JM, Makarewich CA, Trappanese D, et al. Sorafenib cardiotoxicity increases mortality after myocardial infarction [J]. Circ Res, 2014, 114(11): 1700-1712.
[6] Dixon SJ, Patel DN, Welsch M, et al. Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic Reticulum stress and ferroptosis [J]. eLife, 2014, 3: e02523. doi: 10.7554/eLife.02523.
[7] Liu MR, Zhu WT, Pei DS. System Xc^-: a key regulatory target of ferroptosis in cancer[J]. Investig New Drugs, 2021, 39(4): 1123-1131.
[8] Stockwell BR, Jiang XJ. The chemistry and biology of ferroptosis [J]. Cell Chem Biol, 2020, 27(4): 365-375.
[9] Jiang Z, Cui X, Qu P, et al. Roles and mechanisms of puerarin on cardiovascular disease:a review [J]. Biomed Pharmacother, 2022, 147: 112655. doi: 10.1016/j.biopha.2022.112655.
[10] 蔡依梦, 翟昌林, 钱钢. 葛根素通过lncRNA TUG1或MIAT1抑制心肌肥厚进程的研究[J]. 中国中医药科技, 2022, 29(1): 25-29. CAI Yimeng, ZHAI Changlin, QIAN Gang. Inhibition effects of puerarin on cardiac hypertrophy process by lncRNA TUG1 or MIAT1 [J]. Chinese Journal of Traditional Medical Science and Technology, 2022, 29(1): 25-29.
[11] Zhou YX, Zhang H, Peng C. Effects of puerarin on the prevention and treatment of cardiovascular diseases [J]. Front Pharmacol, 2021, 12: 771793. doi: 10.3389/fphar.2021.771793.
[12] Burchert A, Bug G, Fritz LV, et al. Sorafenib maintenance after allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia with FLT3-internal tandem duplication mutation(SORMAIN)[J]. J Clin Oncol, 2020, 38(26): 2993-3002.
[13] Gounder MM, Mahoney MR, van Tine BA, et al. Sorafenib for advanced and refractory desmoid tumors [J]. N Engl J Med, 2018, 379(25): 2417-2428.
[14] Scheulen ME, Kaempgen E, Keilholz U, et al. Stream: A randomized discontinuation, blinded, placebo-controlled phase ii study of sorafenib(s)treatment of chemonaïve patients(pts)with metastatic uveal melanoma(mum)[J]. J Clin Oncol, 2017, 35(15_suppl): 9511.
[15] Schmidinger M, Zielinski CC, Vogl UM, et al. Cardiac toxicity of sunitinib and sorafenib in patients with metastatic renal cell carcinoma [J]. J Clin Oncol, 2008, 26(32): 5204-5212.
[16] Herrmann J. Adverse cardiac effects of cancer therapies: cardiotoxicity and arrhythmia [J]. Nat Rev Cardiol, 2020, 17(8): 474-502.
[17] Jiang XJ, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease [J]. Nat Rev Mol Cell Biol, 2021, 22(4): 266-282.
[18] Wu XG, Li Y, Zhang SC, et al. Ferroptosis as a novel therapeutic target for cardiovascular disease [J]. Theranostics, 2021, 11(7): 3052-3059.
[19] Liu B, Zhao CX, Li HK, et al. Puerarin protects against heart failure induced by pressure overload through mitigation of ferroptosis [J]. Biochem Biophys Res Commun, 2018, 497(1): 233-240.
[20] Almanza A, Carlesso A, Chintha C, et al. Endoplasmic Reticulum stress signalling - from basic mechanisms to clinical applications [J]. Febs J, 2019, 286(2): 241-278.
[21] Lee SH, Hadipour-Lakmehsari S, Murthy HR, et al. Reep5 depletion causes sarco-endoplasmic reticulum vacuolization and cardiac functional defects [J]. Nat Commun, 2020, 11(1): 965.
[22] Choy KW, Murugan D, Mustafa MR. Natural products targeting er stress pathway for the treatment of cardiovascular diseases [J]. Pharmacol Res, 2018, 132: 119-129. doi: 10.1016/j.phrs.2018.04.013.
[23] Lee YS, Lee DH, Choudry HA, et al. Ferroptosis-induced endoplasmic reticulum stress: Cross-talk between ferroptosis and apoptosis [J]. Mol Cancer Res, 2018, 16(7): 1073-1067.
[24] Feng HZ, Stockwell BR. Unsolved mysteries: how does lipid peroxidation cause ferroptosis? [J]. PLoS Biol, 2018, 16(5): e2006203. doi: 10.1371/journal.pbio.2006203.
[25] Abdullahi A, Stanojcic M, Parousis A, et al. Modeling acute ER stress in vivo and in vitro [J]. Shock Augusta Ga, 2017, 47(4): 506-513.
[1] 张高瑞,张玉婷,赵雨萱,王方青,于德新. MnFe2O4@CNS纳米探针在胰腺癌诊疗一体化中的价值[J]. 山东大学学报 (医学版), 2021, 59(4): 48-55.
[2] 温雪彬,傅志俭. 内质网应激在医用臭氧导致的大鼠脊髓神经毒性机制中的作用[J]. 山东大学学报 (医学版), 2019, 57(3): 7-12.
[3] 张亮,徐敏,庄向华,娄福臣,娄能俊,吕丽,郭文娟,郑凤杰,陈诗鸿. 内质网应激与凋亡在糖尿病周围神经病变中的表达变化[J]. 山东大学学报(医学版), 2017, 55(8): 13-17.
[4] 项春红,吕丽,江蓓,肖晓燕,胡昭. EGCG抑制内质网应激减轻高糖致HK-2细胞凋亡的作用[J]. 山东大学学报 (医学版), 2017, 55(12): 1-6.
[5] 贺武斌,苏荣健. 白头翁皂苷D联合索拉非尼对人肝癌细胞侵袭与转移的影响[J]. 山东大学学报(医学版), 2016, 54(7): 18-22.
[6] 李懋禹,唐诗,杜怡峰. TRB3在APP/PS1转基因小鼠脑内的表达及其意义[J]. 山东大学学报(医学版), 2016, 54(10): 1-5.
[7] 郝丽静,葛树卿,王淑芬,郑文娇,张斌. 索拉非尼对顺铂耐药性舌癌Tca8113/DDP细胞增殖及凋亡影响的体外研究[J]. 山东大学学报(医学版), 2016, 54(1): 17-21.
[8] 李梅, 孟庆慧, 蔡巧英, 徐雁, 范晓婷. 乙酰葛根素对Aβ25-35诱导BV-2小胶质细胞Caspase-3表达的影响[J]. 山东大学学报(医学版), 2015, 53(10): 32-36.
[9] 关慧,刘慧敏,贾晓青,罗争,阎明. 酒精性肝病中蛋白酶体活性抑制与内质网应激相关性的体外实验[J]. 山东大学学报(医学版), 2011, 49(8): 52-56.
[10] 蒋欣桐1,蔡可丽1,郭媛媛1,王荣2. 牛磺酸熊脱氧胆酸对高糖环境下晶状体上皮细胞的保护作用[J]. 山东大学学报(医学版), 2010, 48(3): 74-77.
[11] 杨雪莉,蔡可丽,高雪,吴晓莉,卓建,杨博,刘德荣,王荣 . 未折叠蛋白应答在晶状体上皮细胞凋亡中的作用[J]. 山东大学学报(医学版), 2008, 46(1): 52-56.
[12] 王群,鹿庆华,蒋卫东,刘玉胜,杜贻萌,董兆强,王欣,郝琳,秦爱琼,徐冬玲,盛林. 葛根素对球囊损伤后再狭窄过程中组织蛋白酶S及胱蛋白C表达的影响[J]. 山东大学学报(医学版), 2007, 45(11): 1122-1126.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed   
[1] 马青源,蒲沛东,韩飞,王超,朱洲均,王维山,史晨辉. miR-27b-3p调控SMAD1对骨肉瘤细胞增殖、迁移和侵袭作用的影响[J]. 山东大学学报 (医学版), 2020, 1(7): 32 -37 .
[2] 索东阳,申飞,郭皓,刘力畅,杨惠敏,杨向东. Tim-3在药物性急性肾损伤动物模型中的表达及作用机制[J]. 山东大学学报 (医学版), 2020, 1(7): 1 -6 .
[3] 龙婷婷,谢明,周璐,朱俊德. Noggin蛋白对小鼠脑缺血再灌注损伤后学习和记忆能力与齿状回结构的影响[J]. 山东大学学报 (医学版), 2020, 1(7): 15 -23 .
[4] 李宁,李娟,谢艳,李培龙,王允山,杜鲁涛,王传新. 长链非编码RNA AL109955.1在80例结直肠癌组织中的表达及对细胞增殖与迁移侵袭的影响[J]. 山东大学学报 (医学版), 2020, 1(7): 38 -46 .
[5] 张宝文,雷香丽,李瑾娜,罗湘俊,邹容. miR-21-5p靶向调控TIMP3抑制2型糖尿病肾病小鼠肾脏系膜细胞增殖及细胞外基质堆积[J]. 山东大学学报 (医学版), 2020, 1(7): 7 -14 .
[6] 付洁琦,张曼,张晓璐,李卉,陈红. Toll样受体4抑制过氧化物酶体增殖物激活受体γ加重血脂蓄积的分子机制[J]. 山东大学学报 (医学版), 2020, 1(7): 24 -31 .
[7] 丁祥云,于清梅,张文芳,庄园,郝晶. 胰岛素样生长因子II在84例多囊卵巢综合征患者颗粒细胞中的表达和促排卵结局的相关性[J]. 山东大学学报 (医学版), 2020, 1(7): 60 -66 .
[8] 徐玉香,刘煜东,张蓬,段瑞生. 101例脑小血管病患者脑微出血危险因素的回顾性分析[J]. 山东大学学报 (医学版), 2020, 1(7): 67 -71 .
[9] 史爽,李娟,米琦,王允山,杜鲁涛,王传新. 胃癌miRNAs预后风险评分模型的构建与应用[J]. 山东大学学报 (医学版), 2020, 1(7): 47 -52 .
[10] 肖娟,肖强,丛伟,李婷,丁守銮,张媛,邵纯纯,吴梅,刘佳宁,贾红英. 两种甲状腺超声数据报告系统诊断效能的比较[J]. 山东大学学报 (医学版), 2020, 1(7): 53 -59 .