目的 探讨岩藻黄素(fucoxanthin,Fx)对顺铂诱导小鼠肾损伤的修复作用及分子机制。方法 将50只小鼠分为对照组(n = 10)和实验组(n = 40),实验组小鼠每周腹腔注射一次7 mg/kg的顺铂,连续4w造成小鼠肾损伤;将实验组分为模型组、阳性药物氨磷汀组(10 mg/kg)、Fx低剂量组[50 mg/(kg·d)]和Fx高剂量组[100 mg/(kg·d))],正常组和模型组给予正常饲料,后三组给予相应干预治疗4 w;实验结束后处死小鼠,检测小鼠血清中肌酐(creatinine, CRE)、尿素氮(blood urea nitrogen, BUN)和尿酸(uric acid, UA)的水平,检测肾组织中的抗氧化物酶指标、肿瘤坏死因子-α(tumor necrosis factor-α, TNF-α)、白细胞介素-1β(interleukin-1β, IL-1β)、白细胞介素-6(interleukin-6, IL-6)及肾脏损伤分子-1(kidney injury molecule-1, KIM-1)和中性粒细胞明胶酶相关载脂蛋白(neutrophil gelatinase-associated lipocalin, NGAL)的水平,HE染色检查肾脏组织学变化以评估肾脏损伤。Western blot检测肾组织氧化应激通路中核因子-红细胞2相关因子2 (nuclear factor erythroid 2-related factor 2, Nrf2)家族成员和谷胱甘肽过氧化物酶4(glutathione peroxidase 4, GPX4)等铁死亡信号通路蛋白表达,并通过免疫组化法观察小鼠肾脏的Nrf2和GPX4蛋白的水平。结果 Fx干预降低了小鼠血清中CRE、BUN和UA的水平,改善了肾脏病理变化,使肾脏中TNF-α、IL-1β和IL-6水平下降,提高了抗氧化物酶活性,降低了KIM-1和NGAL水平;Western blot检测显示,Fx干预提高了Nrf2、血红素加氧酶1(heme oxygenase-1, HO-1)、烟酰胺醌氧化还原酶 1(NAD(P)H quinone dehydrogenase 1, NQO1)、 GPX4等蛋白的表达,免疫组化结果显示Fx干预后Nrf2和GPX4的蛋白表达增加。结论 Fx对顺铂诱导的肾损伤具有修复作用,其机制可能是通过减轻炎症反应和氧化应激,抑制依赖Nrf2/GPX4信号通路的铁死亡途径而实现的。
Abstract
Objective To investigate the protective action and molecular mechanisms of fucoxanthin (Fx) on cisplatin-induced kidney injury in mice. Methods Fifty mice were divided into a control group (n = 10) and an experimental group (n = 40). The experimental group received an intraperitoneal injection of cisplatin (7 mg/kg) once per week for four consecutive weeks to induce kidney injury. The experimental group was then further divided into four subgroups: model group, positive drug amifostine group (10 mg/kg), low-dose Fx group (50 mg/(kg·d)), and high-dose Fx group [100 mg/(kg·d)]. The normal and model groups received a standard diet, while the other three groups underwent respective interventions for another four weeks. At the end of the experiment, the mice were sacrificed to measure serum levels of creatinine (CRE), blood urea nitrogen (BUN), and uric acid (UA). Oxidative enzyme activity, inflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), as well as kidney injury markers kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL), were assessed in kidney tissues. Histopathological changes in the kidney were evaluated using hematoxylin and eosin (HE) staining. Western blot analysis was performed to detect the expressions of oxidative stress-related nuclear factor erythroid 2-related factor 2 (Nrf2) family members and ferroptosis-related glutathione peroxidase 4 (GPX4) proteins. Immunohistochemistry was also used to observe Nrf2 and GPX4 protein levels in kidney tissues. Results Fx intervention reduced serum CRE, BUN, and UA levels in mice, decreased TNF-α, IL-1β, and IL-6 levels in the kidney, increased antioxidant enzyme activity, and lowered KIM-1 and NGAL levels. Renal histopathological changes were also improved. Additionally, Fx increased the expressions of Nrf2, heme oxygenase-1 (HO-1), NAD(P)H quinone dehydrogenase 1 (NQO1), and GPX4 proteins. Immunohistochemical analysis showed that Fx treatment led to increased Nrf2 and GPX4 protein expression in kidney tissues. Conclusion Fx protests against cisplatin-induced kidney injury in mice. The mechanism may involve alleviation of inflammation and oxidative stress, and inhibition of ferroptosis via activation of the Nrf2/GPX4 signaling pathway.
关键词
肾脏损伤 /
岩藻黄素 /
顺铂 /
铁死亡
Key words
fucoxanthin /
kidney injury /
cisplatin /
ferroptosis
{{custom_sec.title}}
{{custom_sec.title}}
{{custom_sec.content}}
参考文献
[1] Aburto A, Barría A, Cárdenas A, et al. Pre-stimulation of the kallikrein system in cisplatin-induced acute renal injury: an approach to renoprotection[J]. Toxicol Appl Pharmacol, 2014, 280: 216–223.
[2] Youn CK, Kim J, Park JH, et al. Role of autophagy in cisplatin-induced ototoxicity[J]. Int J Pediatr Otorhinolaryngol, 2015, 79: 1814–1819.
[3] Dugbartey GJ, Peppone LJ, DE Graaf IA.An integrative view of cisplatin-induced renal and cardiac toxicities: molecular mechanisms, current treatment challenges and potential protective measures[J]. Toxicology, 2016, 371: 58–66.
[4] Liao YJ, Lu XQ, Lu CW, et al. Selection of agents for prevention of cisplatin-induced hepatotoxicity[J]. Pharmacol Res, 2008, 57: 125–131.
[5] Kouvaris JR, Kouloulias VE, Vlahos LJ.Amifostine: the first selective-target and broad-spectrum radioprotector[J]. Oncologist, 2007, 12: 738-747.
[6] Ji LH, Cui PF, Zhou SW, et al. Advances of amifostine in radiation protection: administration and delivery[J]. Mol Pharm, 2023, 20: 5383-5395.
[7] Almaghrabi OA.Molecular and biochemical investigations on the effect of quercetin on oxidative stress induced by cisplatin in rat kidney[J]. Saudi J Biol Sci, 2015, 22: 227-231.
[8] El-Naga RN.Pre-treatment with cardamonin protects against cisplatin-induced nephrotoxicity in rats: impact on NOX-1, inflammation and apoptosis[J]. Toxicol Appl Pharmacol, 2014, 274: 87-95.
[9] Mao RW, He SP, Lan JG, et al. Honokiol ameliorates cisplatin-induced acute kidney injury via inhibition of mitochondrial fission[J]. Br J Pharmacol, 2022, 179: 3886-3904.
[10] Naghizadeh B, Mansouri SMT, Mashhadian NV.Crocin attenuates cisplatin-induced renal oxidative stress in rats[J]. Food Chem Toxicol, 2010, 48: 2650-2655.
[11] Guo SS, Zhou L, Liu XQ, et al. Baicalein alleviates cisplatin-induced acute kidney injury by inhibiting ALOX12-dependent ferroptosis[J]. Phytomedicine, 2024, 130: 155757.
[12] Yang JH, Sun XL, Huang N, et al. Entacapone alleviates acute kidney injury by inhibiting ferroptosis[J]. FASEB J, 2022, 36: e22399.
[13] Niu CW, Wan J, Bian Y, et al. Baicalein and its underlying mechanism as a protector against liver injury induced by cisplatin in mice[J]. Biotechnol Equip, 2017, 31:193–199.
[14] Omar HA, Mohamed WR, Arab HH, et al. Tangeretin alleviates cisplatin-induced acute hepatic injury in rats: targeting MAPKs and apoptosis[J]. PLoS One, 2016, 11: e0151649.
[15] Sugawara T, Baskaran V, Tsuzuki W, et al. Brown algae fucoxanthin is hydrolyzed to fucoxanthinol during absorption by Caco-2 human intestinal cells and mice[J]. J Nutr, 2002, 132: 946–951.
[16] Sun XW, Zhao HL, Liu ZL, et al. Modulation of gut microbiota by fucoxanthin during alleviation of obesity in high-fat diet-fed mice[J]. J Agric Food Chem, 2020, 68: 5118–5128.
[17] Zheng JW, Tian XX, Zhang W, et al. Protective effects of fucoxanthin against alcoholic liver injury by activation of Nrf2-mediated antioxidant defense and inhibition of TLR4-mediated inflammation[J]. Mar Drugs, 2019, 17: 552.
[18] 任祥雨, 郑佳文, 田笑笑, 等. 岩藻黄素对小鼠非酒精性脂肪性肝病的修复作用[J]. 食品科学, 2024, 45: 42-52.
[19] Yang HY, Xing RG, Liu S, et al. Role of fucoxanthin towards cadmium-induced renal impairment with the antioxidant and anti-lipid peroxide activities[J]. Bioengineered, 2021, 12: 7235–7247.
[20] Manohar S, Leung N.Cisplatin nephrotoxicity: a review of the literature[J]. J Nephrol, 2018, 31: 15–25.
[21] Chen TH, Sue YM, Cheng CY, et al. Fucoxanthin reduces renal tubulointerstitial fibrosis by upregulating NHE1[J]. Nephrol Dial Transplant, 2017, 32: 1.
[22] Szumilas D, Owczarek AJ, Brzozowska A, et al. The value of urinary NGAL, KIM-1, and IL-18 measurements in the early detection of kidney injury in oncologic patients treated with cisplatin-based chemotherapy[J]. Int J Mol Sci, 2024, 25: 1074.
[23] Ichimura T, Hung CC, Yang SA, et al. Kidney injury molecule-1: a tissue and urinary biomarker for nephrotoxicant-induced renal injury[J]. Am J Physiol Renal Physiol, 2004, 286: F552–F563.
[24] Nickolas TL, Schmidt-Ott KM, Canetta P, et al. Diagnostic and prognostic stratification in the emergency department using urinary biomarkers of nephron damage: a multicenter prospective cohort study[J]. J Am Coll Cardiol, 2012, 59: 246–255.
[25] Hosohata K.Role of oxidative stress in drug-induced kidney injury[J]. Int J Mol Sci, 2016, 17: 1826.
[26] El-Awady el-SE, Moustafa YM, Abo-Elmatty DM, et al. Cisplatin-induced cardiotoxicity: mechanisms and cardioprotective strategies[J]. Eur J Pharmacol, 2011, 650: 335–341.
[27] 顾银银, 古送汗·买提尼亚孜, 夏淑芳. 姜黄素对高脂饲料诱导的小鼠脂肪肝的改善作用[J]. 营养学报, 2024, 46: 48–55.
[28] Kobayashi M, Yamamoto M.Nrf2-Keap1 regulation of cellular defense mechanisms against electrophiles and reactive oxygen species[J]. Adv Enzyme Regul, 2006, 46: 113-140.
[29] Shaw P, Chattopadhyay A.Nrf2-ARE signaling in cellular protection: mechanism of action and the regulatory mechanisms[J]. J Cell Physiol, 2020, 235: 3119-3130.
[30] Ungvari Z, Tarantini S, Nyúl-Tóth Á, et al. Nrf2 dysfunction and impaired cellular resilience to oxidative stressors in the aged vasculature: from increased cellular senescence to the pathogenesis of age-related vascular diseases[J]. GeroScience, 2019, 41: 727–738.
[31] Abdalkader M, Lampinen R, Kanninen KM, et al. Targeting Nrf2 to suppress ferroptosis and mitochondrial dysfunction in neurodegeneration[J]. Front Neurosci, 2018, 12: 466.
[32] Song XH, Long DX.Nrf2 and ferroptosis: a new research direction for neurodegenerative diseases[J]. Front Neurosci, 2020, 14: 267.
[33] Imai H, Matsuoka M, Kumagai T, et al. Lipid peroxidation-dependent cell death regulated by GPX4 and ferroptosis[J]. Curr Top Microbiol Immunol, 2017, 403: 143–170.
[34] Zhou JR, Xiao CC, Zheng SS, et al. MicroRNA-214-3p aggravates ferroptosis by targeting GPX4 in cisplatin-induced acute kidney injury[J]. Cell Stress Chaperones, 2022, 27: 325–336.
[35] Li SQ, Huang P, Lai FF, et al. Mechanisms of ferritinophagy and ferroptosis in diseases[J]. Mol Neurobiol, 2024, 61: 1605–1626.
[36] 崔丹丹, 张小强. 原花青素通过Nrf2抑制Erastin介导的细胞铁死亡[J]. 营养学报, 2024, 46: 40–47,55.
[37] 彭红发, 蒋静静, 王惠兰, 等. 氨磷汀和维生素E对顺铂导致大鼠卵巢功能损伤作用机制探讨[J]. 社区医学杂志, 2023, 21: 573–578.
[38] Bae M, Kim MB, Park YK, et al. Health benefits of fucoxanthin in the prevention of chronic diseases[J]. Biochim Biophys Acta Mol Cell Biol Lipids, 2020, 1865: 158618.
基金
浙江省医药卫生科技计划项目(No.2024KY1776,2023RC109)