Research progress on ferroptosis in acute kidney injury

Luo Lianxin, Zhou Fangfang, Luo Qun

PDF(717 KB)
Chinese Journal of Nephrology ›› 2021, Vol. 37 ›› Issue (4) : 375-379. DOI: 10.3760/cma.j.cn441217-20200713-00055
Review

Research progress on ferroptosis in acute kidney injury

Author information +
History +

Abstract

Cite this article

Download Citations
Luo Lianxin. , Zhou Fangfang. , Luo Qun. Research progress on ferroptosis in acute kidney injury[J]. Chinese Journal of Nephrology, 2021, 37(4): 375-379. DOI: 10.3760/cma.j.cn441217-20200713-00055.
急性肾损伤(acute kidney injury,AKI)是临床常见的急危重症,其发病率高、预后差,是慢性肾脏病的危险因素[1]。因此,了解AKI 的发病机制、早期识别、及时干预十分重要。AKI发病机制复杂,以往研究认为主要涉及血管收缩、氧化应激、凋亡、炎症、缺氧等。铁死亡(ferroptosis)是铁依赖的细胞程序性死亡,最近大量研究表明铁死亡与AKI相关,通过激活或抑制铁死亡可以干预AKI的发生发展。本文将近年来铁死亡在AKI中的研究进展作一综述。

一、铁死亡

(一)概述

铁作为人体最重要的必需微量元素之一,广泛参与机体多种代谢过程。2012年Dixon等[2]首次提出铁死亡,为一种非凋亡性过氧化诱导的细胞死亡。铁死亡主要是细胞内脂质活性氧生成与降解的平衡失调所致,在含铁的线粒体氧磷酸化过程中,细胞产生活性氧(reactive oxygen species,ROS)并产生ATP。当ROS水平超过细胞的抗氧化能力会导致氧化应激反应,直接或间接地损害蛋白质、核酸和脂类等大分子物质,导致细胞损伤或死亡[3]。区别于凋亡、自噬等其他调控性细胞死亡,铁死亡具有以下特点:(1)在形态上主要表现为细胞膜断裂和出泡,线粒体体积缩小、膜密度增高、线粒体嵴减少甚至消失,而无细胞皱缩、染色质凝聚、骨架解体及凋亡小体形成等凋亡的形态学特征[4];(2)与凋亡不同,铁死亡发生过程中无半胱天冬酶3(caspase 3)的活化,并且该过程不能被caspase抑制剂所逆转;(3)铁死亡不能被细胞凋亡、细胞焦亡、细胞自噬的抑制剂所抑制,却可以被铁螯合剂、抗氧化剂所抑制;(4)生物学特征为铁和ROS聚集,两者被认为是铁死亡的中心环节,通过减少胱氨酸的摄取、消耗谷胱甘肽(glutathione,GSH)、释放花生四烯酸等分子来抑制胱氨酸/谷氨酸反向转运体(cystine/glutamate antiporter system,System Xc-)和谷胱甘肽过氧化物酶4(glutathione peroxidase 4,GPX4)的活性[5];(5)在遗传学上可出现多种基因的异常表达,尤其是铁代谢相关的基因,比如转铁蛋白受体(transferrin receptor,TFRC)、二价金属转运体1(divalent metal transporter 1,DMT1)、铁蛋白重链1(ferritin heavy chain 1,FTH1)、核受体辅活化子4(nuclear receptor coactivator 4,NCOA4)等[6]

(二)铁死亡主要调节途径

铁死亡受细胞内多条信号途径调节,包括GSH-GPX4途径、铁稳态调节途径和电压依赖性阴离子通道途径等。
1. GSH-GPX4途径:GSH由谷氨酸、半胱氨酸及甘氨酸组成,是重要的自由基清除剂,对于保护细胞免受氧化应激反应的损害至关重要。System Xc-位于细胞膜表面,包括功能亚基SLC7A11和调节亚基SLC3A2,负责胞内谷氨酸和胞外胱氨酸的互换,胱氨酸进一步转化成半胱氨酸,后者是合成GSH的关键;在氧化应激情况下,同型半胱氨酸经胱硫醚β-合酶催化为胱硫醚,最后通过硫转移途径(sulphur-transfer pathway)转化为半胱氨酸,合成GSH,抑制脂质活性氧的合成。
GPX4是一种硒依赖性抗氧化酶,是铁死亡的核心调控蛋白[7]。它最早由Ursini和他的同事在猪肝中分离提纯,其抑制铁催化的脂质过氧化的能力已被证实[8]。GPX4可催化还原型GSH生成氧化型谷胱甘肽(glutathione oxidized,GSSG),同时将细胞内毒性脂质过氧化物还原为无毒脂醇,保护细胞膜结构稳定,避免细胞氧化性损伤。当GPX4活性被抑制时,细胞内的ROS蓄积,进而导致细胞死亡[9]。研究发现,GSH亦可调控GPX4的活性,恢复细胞内GSH水平,可诱导GPX4表达,抑制脂质过氧化和ROS蓄积,最终限制细胞铁死亡的发生,表明GSH-GPX4的调控是铁死亡过程中ROS产生的关键步骤。
2. 铁稳态调节途径:铁稳态对铁死亡的发生有重要影响,过量的游离二价铁离子通过芬顿反应产生ROS继而导致铁死亡。发生铁死亡时,调节铁稳态的蛋白也出现异常表达,主要表现为铁蛋白表达下降,TFRC1表达升高,DMT1表达下降,铁调节蛋白1和2则分别降低和升高等[10],而这些蛋白的异常表达也间接提示铁死亡的发生。近年来一些新的调节细胞内铁摄取、利用的关键蛋白被发现,如热休克蛋白B1、铁反应元件结合蛋白2、锌铁调控蛋白家族等[11]。研究发现,热休克蛋白B1可通过抑制转铁蛋白受体表达而降低细胞内铁离子浓度,过表达的热休克蛋白B1则阻止铁死亡;抑制铁代谢的主要转录因子铁反应元件结合蛋白2可减轻Erastin诱导的铁死亡[12]
3. 电压依赖性阴离子通道途径:电压依赖性阴离子通道(voltage-dependent anion channels,VDACs)是位于线粒体的一种膜孔蛋白,主要功能是转运离子和代谢产物。研究发现Erastin作用于VDACs,可引起线粒体功能障碍,外膜通透性改变,氧化性物质释放,最终导致细胞铁死亡[13]

二、铁死亡与AKI

AKI发病机制十分复杂,目前多种诱发或加重AKI的分子机制被提出,但ROS诱导的肾损伤被认为是AKI关键的机制之一。多项研究表明,铁死亡是一种很有前途的治疗靶点,特别是在以肾小管坏死为主的疾病中[14]。一项使用可诱导的GPX4缺陷小鼠的研究发现,敲除GPX4可导致AKI,进一步说明了铁死亡在AKI中的重要性[15]。最近,关于缺血再灌注、药物、横纹肌溶解所诱导的AKI模型的研究提供了额外的证据,支持铁死亡在AKI中所发挥的重要作用。
1. 铁死亡与缺血再灌注诱导的AKI:目前研究认为炎性反应、氧化应激、细胞内钙超载、肾素-血管紧张素激活、微循环障碍等是肾脏缺血再灌注损伤的主要病理生理机制。凋亡曾被认为是各种缺血损伤模型中主要的调控细胞死亡形式,是心脏和肾脏缺血性损伤的主要原因。然而,近年来研究发现,铁死亡可能是缺血性损伤的主要驱动因素[16]。使用铁螯合剂及降低铁相关氧化应激的抗氧化剂在一定程度上使缺血再灌注损伤后肾功能得到改善,间接表明缺血再灌注损伤存在铁代谢紊乱及其诱导的AKI[17]。Huang等[18]在缺血再灌注诱导的AKI体外模型中抑制肝再生增强因子(augmenter of liver regeneration,ALR)的表达,发现细胞铁死亡水平升高,同时ROS升高,线粒体损伤明显;此外用Erastin抑制System Xc-会促进细胞铁死亡,同时沉默了ALR的表达,提示缺血再灌注诱导的AKI可能由ALR所介导,并且该过程与谷胱甘肽-谷胱甘肽过氧化物酶(glutathione-glutathione peroxidase,GSH-GPx)系统相关。泛连接蛋白1(Pannexin1,Panx1)是ATP释放通路家族蛋白,Su等[19]在缺血再灌注诱导的AKI小鼠模型中发现,沉默Panx1表达显著减弱脂质过氧化和铁积累,主要通过丝裂原活化蛋白激酶/细胞外信号调节激酶(mitogen-activated protein kinase/extracellular signal-regulated kinase,MAPK/ERK)信号激活铁蛋白,调节NCAO4介导的铁蛋白噬铁和血红素加氧酶1(hemeoxygenase1,HO1)的表达。肾缺血再灌注损伤是心脏手术患者发生AKI的主要原因,Choi等[20]提出术中较低水平的铁结合蛋白间接反映体外循环过程中机体对催化铁的处理能力受损,从而导致肾脏损伤。这一结果强调了铁稳态在缺血再灌注损伤中的重要性,并提示在心脏手术相关的肾脏损伤或缺血再灌注诱导的AKI中,铁稳态可能是一个潜在的治疗靶点。机械通气是心脏术后发生AKI的独立危险因素之一,在缺血再灌注小鼠模型中,我们前期研究发现随着机械通气时间延长,铁死亡的关键调节因子GPX4的水平逐渐降低,肾脏脂质过氧化增加(4-羟基烯醛水平升高和超氧化物歧化酶2水平降低),提示机械通气时间延长可能通过铁死亡途径加重缺血再灌注所致的AKI;此外,机械通气时间延长可致血GSH和肾匀浆GSH水平逐渐降低,且两者呈正相关,提示机械通气时间延长可能导致血GSH耗竭,通过GSH-GPX4轴诱发肾脏铁死亡,但其与肾脏铁死亡之间更深入的内在关系尚需要今后进一步研究[21]
2. 铁死亡与药物诱导的AKI:顺铂是治疗多种实体肿瘤的重要化疗药物之一,但由于严重的不良反应,尤其是肾毒性限制了其临床应用。Baliga等[22]在1998年报道了顺铂诱导的体外细胞毒性模型和顺铂诱导的体内急性肾衰竭模型的研究,结果表明接触顺铂后,释放到培养基中的博莱霉素可检测到铁,即能催化自由基反应的铁显著增加,而铁螯合剂可显著降低顺铂诱导的细胞毒性,提示铁在其中发挥重要作用,但具体机制未能阐明。铁蛋白在铁代谢中起核心作用,研究发现在近端小管中,FTH敲除小鼠在顺铂给药后肾脏损伤较对照组更严重,这一结果强调了FTH在AKI中的保护作用[23]。肌醇加氧酶是一种近端肾小管酶,Deng等[24]发现其过表达可加重顺铂诱导AKI的细胞氧化还原损伤,主要通过铁蛋白吞噬和脂质过氧化作用促进铁死亡,还可能通过下调GPX4活性和细胞内GSH浓度来抑制“铁死亡终止系统”。Mishima等[25]在顺铂诱导的AKI的小鼠模型中发现细胞色素P450(cytochrome P450,CYP450)底物如异丙嗪、利福平等可通过清除脂质过氧化自由基减轻组织损伤和细胞死亡,但由于未建立铁死亡的具体标志物,损伤模型中可能存在多种细胞死亡途径,未能直接证实药物对铁死亡具有抑制作用。
在啮齿动物中,叶酸可导致AKI的发生,一定剂量的叶酸可在肾腔内形成晶体,高剂量的叶酸也可直接对肾小管上皮产生毒性。Martin-Sanchez等[26]研究证实,在叶酸诱导的小鼠AKI模型中存在典型的铁死亡特征——脂质过氧化和GSH下调。此外铁死亡抑制剂 Ferrostatin-1(Fer-1)通过抑制白细胞介素33(interleukin-33,IL-33)等趋化因子和细胞因子的上调,抑制巨噬细胞浸润及保护因子Klotho的下调,减少氧化应激和小管细胞死亡,减轻组织损伤,而caspase抑制剂无肾脏保护作用[26];研究还发现,受体结合丝氨酸/苏氨酸激酶3(receptor-interacting protein kinase 3,RIPK3)和混合系列蛋白激酶样结构域(mixed lineage domain-like protein,MLKL)的蛋白表达增加,但使用RIPK1抑制剂或RIPK3、MLKL基因缺陷并不能防止肾损伤,表明铁死亡在叶酸诱导的AKI中是最主要的细胞死亡通路[26]。FG-4592是低氧诱导因子(hypoxia-inducible factor,HIF)前体羟化酶的抑制剂,Li等[27]最近在叶酸诱导的AKI模型中发现FG-4592预处理的小鼠肾功能明显改善,同时组织中铁、丙二醛和4-羟基壬烯醛的含量降低,表明铁积累和脂质过氧化受到抑制;此外存在HIF-1α表达上调、核因子E2相关因子2(nuclear factor-E2-related factor 2,Nrf2)激活、下游蛋白高表达,包括HO1、GPX4、System Xc-、膜铁转运蛋白,进一步信号通路研究提示Nrf2激活受到蛋白激酶B/糖原合成酶激酶-3β(protein kinase B/glycogensynthesis kinase-3β,PKB/GSK-3β)调控,提示PKB/GSK-3β是潜在的铁死亡治疗靶点。在叶酸诱导的AKI小鼠模型中,Córdoba-David等[28]提出星状聚谷氨酸-姜黄素耦联物通过抑制B细胞核因子κ轻链增强子(nuclear factor kappa B,NF-κB)激活、下调铁死亡标志物的表达,同时保留Klotho的肾脏表达,最终发挥保护肾脏作用。
3. 铁死亡与横纹肌溶解诱导的AKI:据报道横纹肌溶解患者中约40%可发生AKI,严重AKI患者的病死率为50%[29]。既往研究证实,肌红蛋白在肾脏的积累是导致肾脏损伤的核心机制,其可引起肾小管阻塞、氧化损伤和血管收缩,最终导致AKI[30]。当发生横纹肌溶解时,大量的肌红蛋白被分解成球蛋白和高铁血红素,高铁血红素进一步分解成亚铁血红素和铁,在相对低血压、低灌注及细胞内ATP不足的情况下,导致肾小管细胞内铁超载,催化脂类、蛋白质及脱氧核苷酸过氧化,损伤肾小管上皮细胞,引起AKI[30]。当前研究发现,横纹肌溶解诱导AKI的机制可能是肌红蛋白代谢产生的二价铁离子通过芬顿反应,诱导脂质过氧化对近端小管造成直接损伤,其中FTH具有重要意义[31]。Zarjou等[23]发现,在横纹肌溶解诱导的AKI模型中FTH敲除小鼠比野生型小鼠有更高的死亡率和更严重的肾脏损伤,表明了FTH对肾小管损伤的保护作用及铁离子在AKI中的作用。Guerrero-Hue等[32]观察到,发生横纹肌溶解的小鼠体内铁积累增加、脂质过氧化作用增强;同时管状上皮细胞生存能力研究证实,与凋亡或坏死抑制剂相比,铁死亡抑制剂对肌红蛋白介导的细胞死亡具有更强的保护作用;此外横纹肌溶解所致肾功能障碍和组织学损害可通过姜黄素治疗得以减轻,其中有HO1的参与。这些研究有力地表明,铁死亡在横纹肌溶解诱导的AKI中起着重要的作用。
4. 其他:缺乏有效的药物治疗是AKI目前亟需解决的问题,而铁死亡参与AKI的发生和进展,抑制铁死亡有助于AKI的预防和治疗。Fer-1已被证明通过氧化还原反应减少脂质过氧化物来阻止膜脂损伤,从而抑制细胞死亡[26,33-34]。他汀类药物经常被报道减轻脂质过氧化物介导的组织损伤,包括AKI[35]。除了抗氧化剂和铁螯合剂(如维生素E),研究还发现去铁胺通过降低细胞内铁含量和减轻氧化损伤来抑制铁死亡[3]。Kagan等[36]提出抑制酰基辅酶A合成酶4来抑制酰基花生四烯基和肾上腺素基的酯化作用,可以预防铁死亡的发生。最近还发现一种独立于经典GPX4信号通路的新型铁死亡抑制因子1,其在细胞膜上利用还原型烟酰胺腺嘌呤二核苷酸磷酸(nicotinamide adenine dinucleotide phosphate,NADPH)将泛醌(coenzyme Q10,CoQ10)还原为泛醇(coenzyme Q10 H2,CoQ10 H2),减少细胞膜脂质过氧化,从而抑制铁死亡[37-38]。目前这些铁死亡抑制剂均在啮齿动物AKI模型或体外实验进行研究,尚缺乏临床应用证据。

三、总结与展望

铁死亡是一种独特的细胞死亡调控形式,已被报道参与多种肿瘤和神经系统疾病。越来越多的研究表明铁死亡在AKI中有着重要的作用,在缺血再灌注、药物、横纹肌溶解所诱导的AKI模型中可观察到铁死亡过程,而铁死亡抑制剂的应用可以起到肾脏保护作用。但目前仍有许多问题尚未解决,需进一步研究,包括:(1)铁与铁死亡的关系;(2)铁死亡与其他生理过程的联系;(3)参与铁坏死的信号通路与其他调节性细胞死亡信号通路是否存在交叉或重叠;(4)铁死亡中最关键执行分子是什么;(5)如何通过调控铁死亡来保护肾脏。未来我们尚需要对AKI的发病机制进行更为深入、全面的研究,进行预防和靶向治疗,最终减少其发病率,改善患者预后。

References

[1]
Ronco C, Bellomo R, Kellum JA. Acute kidney injury[J]. Lancet, 2019, 394(10212): 1949-1964. DOI: 10.1016/S0140-6736(19)32563-2.
Acute kidney injury (AKI) is defined by a rapid increase in serum creatinine, decrease in urine output, or both. AKI occurs in approximately 10-15% of patients admitted to hospital, while its incidence in intensive care has been reported in more than 50% of patients. Kidney dysfunction or damage can occur over a longer period or follow AKI in a continuum with acute and chronic kidney disease. Biomarkers of kidney injury or stress are new tools for risk assessment and could possibly guide therapy. AKI is not a single disease but rather a loose collection of syndromes as diverse as sepsis, cardiorenal syndrome, and urinary tract obstruction. The approach to a patient with AKI depends on the clinical context and can also vary by resource availability. Although the effectiveness of several widely applied treatments is still controversial, evidence for several interventions, especially when used together, has increased over the past decade.Copyright © 2019 Elsevier Ltd. All rights reserved.
[2]
Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death[J]. Cell, 2012, 149(5): 1060-1072. DOI: 10.1016/j.cell.2012.03.042.
Nonapoptotic forms of cell death may facilitate the selective elimination of some tumor cells or be activated in specific pathological states. The oncogenic RAS-selective lethal small molecule erastin triggers a unique iron-dependent form of nonapoptotic cell death that we term ferroptosis. Ferroptosis is dependent upon intracellular iron, but not other metals, and is morphologically, biochemically, and genetically distinct from apoptosis, necrosis, and autophagy. We identify the small molecule ferrostatin-1 as a potent inhibitor of ferroptosis in cancer cells and glutamate-induced cell death in organotypic rat brain slices, suggesting similarities between these two processes. Indeed, erastin, like glutamate, inhibits cystine uptake by the cystine/glutamate antiporter (system x(c)(-)), creating a void in the antioxidant defenses of the cell and ultimately leading to iron-dependent, oxidative death. Thus, activation of ferroptosis results in the nonapoptotic destruction of certain cancer cells, whereas inhibition of this process may protect organisms from neurodegeneration.Copyright © 2012 Elsevier Inc. All rights reserved.
[3]
Yu H, Guo P, Xie X, et al. Ferroptosis, a new form of cell death, and its relationships with tumourous diseases[J]. J Cell Mol Med, 2017, 21(4): 648-657. DOI: 10.1111/jcmm.13008.
Ferroptosis is a newly discovered type of cell death that differs from traditional apoptosis and necrosis and results from iron-dependent lipid peroxide accumulation. Ferroptotic cell death is characterized by cytological changes, including cell volume shrinkage and increased mitochondrial membrane density. Ferroptosis can be induced by two classes of small-molecule substances known as class 1 (system X inhibitors) and class 2 ferroptosis inducers [glutathione peroxidase 4 (GPx4) inhibitors]. In addition to these small-molecule substances, a number of drugs (e.g. sorafenib, artemisinin and its derivatives) can induce ferroptosis. Various factors, such as the mevalonate (MVA) and sulphur-transfer pathways, play pivotal roles in the regulation of ferroptosis. Ferroptosis plays an unneglectable role in regulating the growth and proliferation of some types of tumour cells, such as lymphocytoma, ductal cell cancer of the pancreas, renal cell carcinoma (RCC) and hepatocellular carcinoma (HCC). Here, we will first introduce the discovery of and research pertaining to ferroptosis; then summarize the induction mechanisms and regulatory pathways of ferroptosis; and finally, further elucidate the roles of ferroptosis in human tumourous diseases.© 2016 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine.
[4]
Mou Y, Wang J, Wu J, et al. Ferroptosis, a new form of cell death: opportunities and challenges in cancer[J]. J Hematol Oncol, 2019, 12(1): 34. DOI: 10.1186/s13045-019-0720-y.
[5]
Doll S, Conrad M. Iron and ferroptosis: a still ill-defined liaison[J]. IUBMB Life, 2017, 69(6): 423-434. DOI: 10.1002/iub.1616.
Ferroptosis is a recently described form of regulated necrotic cell death, which appears to contribute to a number of diseases, such as tissue ischemia/reperfusion injury, acute renal failure, and neurodegeneration. A hallmark of ferroptosis is iron-dependent lipid peroxidation, which can be inhibited by the key ferroptosis regulator glutathione peroxidase 4(Gpx4), radical trapping antioxidants and ferroptosis-specific inhibitors, such as ferrostatins and liproxstatins, as well as iron chelation. Although great strides have been made towards a better understanding of the proximate signals of distinctive lipid peroxides in ferroptosis, still little is known about the mechanistic implication of iron in the ferroptotic process. Hence, this review aims at summarizing recent advances in our understanding to what is known about enzymatic and nonenzymatic routes of lipid peroxidation, the involvement of iron in this process and the identification of novel players in ferroptotic cell death. Additionally, we review early works carried out long time before the term "ferroptosis" was actually introduced but which were instrumental in a better understanding of the role of ferroptosis in physiological and pathophysiological contexts. © 2017 IUBMB Life, 69(6):423-434, 2017.© 2017 International Union of Biochemistry and Molecular Biology.
[6]
Wang S, Luo J, Zhang Z, et al. Iron and magnetic: new research direction of the ferroptosis-based cancer therapy[J]. Am J Cancer Res, 2018, 8(10): 1933-1946.
Ferroptosis is an iron depend cell death which caused by lipid peroxidation. Abnormal iron metabolism and high intracellular iron content are the characteristics of most cancer cells. Iron is a promoter of cell growth and proliferation. However, iron also could take part in Fenton reaction to produce reactive oxygen species (ROS). The intercellular ROS could induce lipid peroxidation, which is necessary for ferroptosis. Iron metabolism mainly includes three parts: iron uptake, storage and efflux. Therefore, iron metabolism-related genes could regulate intercellular iron content and status, which can be involved ferroptosis. In recent years, the application of nanoparticles in cancer therapy research has become more and more extensive. The iron-based nanoparticles (iron-based NPs) can release ferrous (Fe) or ferric (Fe) in acidic lysosomes and inducing ferroptosis. Magnetic field is widely used in the targeted concentration of iron-based NPs related disease therapy. Furthermore, multiple studies showed that magnetic fields can inhibit cancer cell proliferation by promoting intracellular ROS production. Herein, we focus on the relationship of between ferroptosis and iron metabolism in cancer cells, the application of nanoparticles and magnetic field in inducing ferroptosis of cancer cells, and trying to provide new ideas for cancer treatment research.
[7]
Friedmann Angeli JP, Conrad M. Selenium and GPX4, a vital symbiosis[J]. Free Radic Biol Med, 2018, 127: 153-159. DOI: 10.1016/j.freeradbiomed.2018.03.001.
[8]
Hirschhorn T, Stockwell BR. The development of the concept of ferroptosis[J]. Free Radic Biol Med, 2019, 133: 130-143. DOI: 10.1016/j.freeradbiomed.2018.09.043.
[9]
Cao JY, Dixon SJ. Mechanisms of ferroptosis[J]. Cell Mol Life Sci, 2016, 73(11-12): 2195-2209. DOI: 10.1007/s00018-016-2194-1.
Ferroptosis is a non-apoptotic form of cell death that can be triggered by small molecules or conditions that inhibit glutathione biosynthesis or the glutathione-dependent antioxidant enzyme glutathione peroxidase 4 (GPX4). This lethal process is defined by the iron-dependent accumulation of lipid reactive oxygen species and depletion of plasma membrane polyunsaturated fatty acids. Cancer cells with high level RAS-RAF-MEK pathway activity or p53 expression may be sensitized to this process. Conversely, a number of small molecule inhibitors of ferroptosis have been identified, including ferrostatin-1 and liproxstatin-1, which can block pathological cell death events in brain, kidney and other tissues. Recent work has identified a number of genes required for ferroptosis, including those involved in lipid and amino acid metabolism. Outstanding questions include the relationship between ferroptosis and other forms of cell death, and whether activation or inhibition of ferroptosis can be exploited to achieve desirable therapeutic ends.
[10]
Masaldan S, Clatworthy S, Gamell C, et al. Iron accumulation in senescent cells is coupled with impaired ferritinophagy and inhibition of ferroptosis[J]. Redox Biol, 2018, 14: 100-115. DOI: 10.1016/j.redox.2017.08.015.
Cellular senescence is characterised by the irreversible arrest of proliferation, a pro-inflammatory secretory phenotype and evasion of programmed cell death mechanisms. We report that senescence alters cellular iron acquisition and storage and also impedes iron-mediated cell death pathways. Senescent cells, regardless of stimuli (irradiation, replicative or oncogenic), accumulate vast amounts of intracellular iron (up to 30-fold) with concomitant changes in the levels of iron homeostasis proteins. For instance, ferritin (iron storage) levels provided a robust biomarker of cellular senescence, for associated iron accumulation and for resistance to iron-induced toxicity. Cellular senescence preceded iron accumulation and was not perturbed by sustained iron chelation (deferiprone). Iron accumulation in senescent cells was driven by impaired ferritinophagy, a lysosomal process that promotes ferritin degradation and ferroptosis. Lysosomal dysfunction in senescent cells was confirmed through several markers, including the build-up of microtubule-associated protein light chain 3 (LC3-II) in autophagosomes. Impaired ferritin degradation explains the iron accumulation phenotype of senescent cells, whereby iron is effectively trapped in ferritin creating a perceived cellular deficiency. Accordingly, senescent cells were highly resistant to ferroptosis. Promoting ferritin degradation by using the autophagy activator rapamycin averted the iron accumulation phenotype of senescent cells, preventing the increase of TfR1, ferritin and intracellular iron, but failed to re-sensitize these cells to ferroptosis. Finally, the enrichment of senescent cells in mouse ageing hepatic tissue was found to accompany iron accumulation, an elevation in ferritin and mirrored our observations using cultured senescent cells.Copyright © 2017 The Authors. Published by Elsevier B.V. All rights reserved.
[11]
Zhang L, Ye Y, Tu H, et al. MicroRNA-related genetic variants in iron regulatory genes, dietary iron intake, microRNAs and lung cancer risk[J]. Ann Oncol, 2017, 28(5): 1124-1129. DOI: 10.1093/annonc/mdx046.
Genetic variations in MicroRNA (miRNA) binding sites may alter structural accessibility of miRNA binding sites to modulate risk of cancer. This large-scale integrative multistage study was aimed to evaluate the interplay of genetic variations in miRNA binding sites of iron regulatory pathway, dietary iron intake and lung cancer (LC) risk.The interplay of genetic variant, dietary iron intake and LC risk was assessed in large-scale case-control study. Functional characterization of the validated SNP and analysis of target miRNAs were performed.We found that the miRNA binding site SNP rs1062980 in 3' UTR of Iron-Responsive Element Binding protein 2 gene (IREB2) was associated with a 14% reduced LC risk (P value = 4.9×10 - 9). Comparing to AA genotype, GG genotype was associated with a 27% reduced LC risk. This association was evident in males and ever-smokers but not in females and never-smokers. Higher level of dietary iron intake was significantly associated with 39% reduced LC risk (P value = 2.0×10 - 8). This association was only present in individuals with AG + AA genotypes with a 46% reduced risk (P value = 1.0×10 - 10), but not in GG genotype. The eQTL-analysis showed that rs1062980 significantly alters IREB2 expression level. Rs1062980 is predicted to alter a miR-29 binding site on IREB2 and indeed the expression of miR-29 is inversely correlated with IREB2 expression. Further, we found that higher circulating miR-29a level was significantly associated with 78% increased LC risk.The miRNA binding site SNP rs1062980 in iron regulatory pathway, which may alter the expression of IREB2 potentially through modulating the binding of miR-29a, together with dietary iron intake may modify risk of LC both individually and jointly. These discoveries reveal novel pathway for understanding lung cancer tumorigenesis and risk stratification.© The Author 2017. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For Permissions, please email: journals.permissions@oup.com.
[12]
Sun X, Ou Z, Xie M, et al. HSPB1 as a novel regulator of ferroptotic cancer cell death[J]. Oncogene, 2015, 34(45): 5617-5625. DOI: 10.1038/onc.2015.32.
Ferroptosis is an iron-dependent form of non-apoptotic cell death, but its molecular mechanism remains largely unknown. Here, we demonstrate that heat shock protein beta-1 (HSPB1) is a negative regulator of ferroptotic cancer cell death. Erastin, a specific ferroptosis-inducing compound, stimulates heat shock factor 1 (HSF1)-dependent HSPB1 expression in cancer cells. Knockdown of HSF1 and HSPB1 enhances erastin-induced ferroptosis, whereas heat shock pretreatment and overexpression of HSPB1 inhibits erastin-induced ferroptosis. Protein kinase C-mediated HSPB1 phosphorylation confers protection against ferroptosis by reducing iron-mediated production of lipid reactive oxygen species. Moreover, inhibition of the HSF1-HSPB1 pathway and HSPB1 phosphorylation increases the anticancer activity of erastin in human xenograft mouse tumor models. Our findings reveal an essential role for HSPB1 in iron metabolism with important effects on ferroptosis-mediated cancer therapy.
[13]
Chen Y, Liu Y, Lan T, et al. Quantitative profiling of protein carbonylations in ferroptosis by an aniline-derived probe[J]. J Am Chem Soc, 2018, 140(13): 4712-4720. DOI: 10.1021/jacs.8b01462.
Ferroptosis is a regulated form of necrotic cell death implicated in carcinogenesis and neurodegeneration that is driven by phospholipid peroxidation. Lipid-derived electrophiles (LDEs) generated during this process can covalently modify proteins ("carbonylation") and affect their functions. Here we report the development of a quantitative chemoproteomic method to profile carbonylations in ferroptosis by an aniline-derived probe. Using the method, we established a global portrait of protein carbonylations in ferroptosis with >400 endogenously modified proteins and for the first time, identified >20 residue sites with endogenous LDE modifications in ferroptotic cells. Specifically, we discovered and validated a novel cysteine site of modification on voltage-dependent anion-selective channel protein 2 (VDAC2) that might play an important role in sensitizing LDE signals and mediating ferroptosis. Our results will contribute to the understanding of ferroptotic signaling and pathogenesis and provide potential biomarkers for ferroptosis detection.
[14]
Linkermann A. Nonapoptotic cell death in acute kidney injury and transplantation[J]. Kidney Int, 2016, 89(1): 46-57. DOI: 10.1016/j.kint.2015.10.008.
Acute tubular necrosis causes a loss of renal function, which clinically presents as acute kidney failure (AKI). The biochemical signaling pathways that trigger necrosis have been investigated in detail over the past 5 years. It is now clear that necrosis (regulated necrosis, RN) represents a genetically driven process that contributes to the pathophysiology of AKI. RN pathways such as necroptosis, ferroptosis, parthanatos, and mitochondrial permeability transition-induced regulated necrosis (MPT-RN) may be mechanistically distinct, and the relative contributions to overall organ damage during AKI in living organisms largely remain elusive. In a synchronized manner, some necrotic programs induce the breakdown of tubular segments and multicellular functional units, whereas others are limited to killing single cells in the tubular compartment. Importantly, the means by which a renal cell dies may have implications for the subsequent inflammatory response. In this review, the recent advances in the field of renal cell death in AKI and key enzymes that might serve as novel therapeutic targets will be discussed. As a consequence of the interference with RN, the immunogenicity of dying cells in AKI in renal transplants will be diminished, rendering inhibitors of RN indirect immunosuppressive agents.Copyright © 2016 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.
[15]
Friedmann Angeli JP, Schneider M, Proneth B, et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice[J]. Nat Cell Biol, 2014, 16(12): 1180-1191. DOI: 10.1038/ncb3064.
Ferroptosis is a non-apoptotic form of cell death induced by small molecules in specific tumour types, and in engineered cells overexpressing oncogenic RAS. Yet, its relevance in non-transformed cells and tissues is unexplored and remains enigmatic. Here, we provide direct genetic evidence that the knockout of glutathione peroxidase 4 (Gpx4) causes cell death in a pathologically relevant form of ferroptosis. Using inducible Gpx4(-/-) mice, we elucidate an essential role for the glutathione/Gpx4 axis in preventing lipid-oxidation-induced acute renal failure and associated death. We furthermore systematically evaluated a library of small molecules for possible ferroptosis inhibitors, leading to the discovery of a potent spiroquinoxalinamine derivative called Liproxstatin-1, which is able to suppress ferroptosis in cells, in Gpx4(-/-) mice, and in a pre-clinical model of ischaemia/reperfusion-induced hepatic damage. In sum, we demonstrate that ferroptosis is a pervasive and dynamic form of cell death, which, when impeded, promises substantial cytoprotection.
[16]
Tonnus W, Linkermann A. The in vivo evidence for regulated necrosis[J]. Immunol Rev, 2017, 277(1): 128-149. DOI: 10.1111/imr.12551.
Necrosis is a hallmark of several widespread diseases or their direct complications. In the past decade, we learned that necrosis can be a regulated process that is potentially druggable. RIPK3- and MLKL-mediated necroptosis represents by far the best studied pathway of regulated necrosis. During necroptosis, the release of damage-associated molecular patterns (DAMPs) drives a phenomenon referred to as necroinflammation, a common consequence of necrosis. However, most studies of regulated necrosis investigated cell lines in vitro in a cell autonomous manner, which represents a non-physiological situation. Conclusions based on such work might not necessarily be transferrable to disease states in which synchronized, non-cell autonomous effects occur. Here, we summarize the current knowledge of the pathophysiological relevance of necroptosis in vivo, and in light of this understanding, we reassess the morphological classification of necrosis that is generally used by pathologists. Along these lines, we discuss the paucity of data implicating necroptosis in human disease. Finally, the in vivo relevance of non-necroptotic forms of necrosis, such as ferroptosis, is addressed.© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.
[17]
Walker VJ, Agarwal A. Targeting iron homeostasis in acute kidney injury[J]. Semin Nephrol, 2016, 36(1): 62-70. DOI: 10.1016/j.semnephrol.2016.01.003.
Iron is an essential metal involved in several major cellular processes required to maintain life. Because of iron's ability to cause oxidative damage, its transport, metabolism, and storage is strictly controlled in the body, especially in the small intestine, liver, and kidney. Iron plays a major role in acute kidney injury and has been a target for therapeutic intervention. However, the therapies that have been effective in animal models of acute kidney injury have not been successful in human beings. Targeting iron trafficking via ferritin, ferroportin, or hepcidin may offer new insights. This review focuses on the biology of iron, particularly in the kidney, and its implications in acute kidney injury. Copyright © 2016. Published by Elsevier Inc.
[18]
Huang LL, Liao XH, Sun H, et al. Augmenter of liver regeneration protects the kidney from ischaemia-reperfusion injury in ferroptosis[J]. J Cell Mol Med, 2019, 23(6): 4153-4164. DOI: 10.1111/jcmm.14302.
Acute kidney injury (AKI) is a common and severe clinical condition with high morbidity and mortality. Ischaemia-reperfusion (I/R) injury remains the major cause of AKI in the clinic. Ferroptosis is a recently discovered form of programmed cell death (PCD) that is characterized by iron-dependent accumulation of reactive oxygen species (ROS). Compelling evidence has shown that renal tubular cell death involves ferroptosis, although the underlying mechanisms remain unclear. Augmenter of liver regeneration (ALR) is a widely distributed multifunctional protein that is expressed in many tissues. Our previous study demonstrated that ALR possesses an anti-oxidant function. However, the modulatory mechanism of ALR remains unclear and warrants further investigation. Here, to elucidate the role of ALR in ferroptosis, ALR expression was inhibited using short hairpin RNA lentivirals (shRNA) in vitro model of I/R-induced AKI. The results suggest that the level of ferroptosis is increased, particularly in the shRNA/ALR group, accompanied by increased ROS and mitochondrial damage. Furthermore, inhibition of system xc- with erastin aggravates ferroptosis, particularly silencing of the expression of ALR. Unexpectedly, we demonstrate a novel signalling pathway of ferroptosis. In summary, we show for the first time that silencing ALR aggravates ferroptosis in an in vitro model of I/R. Notably, we show that I/R induced kidney ferroptosis is mediated by ALR, which is linked to the glutathione-glutathione peroxidase (GSH-GPx) system.© 2019 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine.
[19]
Su L, Jiang X, Yang C, et al. Pannexin 1 mediates ferroptosis that contributes to renal ischemia/reperfusion injury[J]. J Biol Chem, 2019, 294(50): 19395-19404. DOI: 10.1074/jbc.RA119.010949.
Renal ischemia/reperfusion injury (IRI) is a significant challenge in perioperative medicine and is related to oxidative programmed cell death. However, the role of ferroptosis, a newly discovered form of oxidative cell death, has not been evaluated widely. Pannexin 1 (PANX1), an ATP-releasing pathway family protein, has pro-apoptotic effects during kidney injury. Here, we demonstrate that PANX1 deletion protects against renal IRI by regulating ferroptotic cell death. knockout mice subjected to renal IRI had decreased plasma creatinine, malondialdehyde (MDA) levels in kidney tissues, and tubular cell death (visible as decreased TUNEL-positive renal tubular cells) compared with WT mice. In cultured human kidney 2 (HK-2) cells, silenced expression significantly attenuated ferroptotic lipid peroxidation and iron accumulation induced by the ferroptosis inducer erastin. Moreover, the silencing significantly modulated ferroptosis-related protein expression. Furthermore, deletion induced the expression of a cytoprotective chaperone, heme oxygenase-1 (HO-1), and inhibited ferroptinophagy via the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway. In summary, deletion protects against renal IRI by attenuating MAPK/ERK activation in a ferroptotic pathway. Our findings provide critical insights into the role of PANX1 in ferroptotic cell death and highlight a potential therapeutic target for the management of acute kidney injury (AKI) during the perioperative period.© 2019 Su et al.
[20]
Choi N, Whitlock R, Klassen J, et al. Early intraoperative iron-binding proteins are associated with acute kidney injury after cardiac surgery[J]. J Thorac Cardiovasc Surg, 2019, 157(1): 287-297.e2. DOI: 10.1016/j.jtcvs.2018.06.091.
Iron regulation is an important modifier of renal ischemia-reperfusion injury, but the role of iron-binding proteins during cardiopulmonary bypass remains unclear. The goal was to characterize iron-binding proteins throughout ischemia-reperfusion injury to determine their association with acute kidney injury development.A prospective observational cohort of adult patients who underwent cardiac surgery (n = 301) was obtained, and acute kidney injury was defined by Kidney Disease Improving Global Outcomes. Serum ferritin, transferrin saturation, and urine hepcidin-25 were measured.Intraoperative serum ferritin was lower at the start of cardiopulmonary bypass (P = .005) and 1-hour cardiopulmonary bypass (P = .001) in patients with acute kidney injury versus patients without acute kidney injury. Lower serum ferritin and higher transferrin saturation at 1-hour cardiopulmonary bypass were independent predictors of acute kidney injury (serum ferritin odds ratio, 0.66; 95% confidence interval [CI], 0.48-0.91; transferrin saturation odds ratio, 1.26; 95% CI, 1.02-1.55) and improved model discrimination (area under the curve [AUC], 0.76; 95% CI, 0.67-0.85) compared with clinical prediction alone (AUC, 0.72; 95% CI, 0.62-0.81; ΔAUC and net reclassification index, P = .01). Lower ferritin, higher transferrin saturation at 1-hour cardiopulmonary bypass, and lower urine hepcidin-25 at postoperative day 1 were also independent predictors for acute kidney injury development, and this model demonstrated an AUC of 0.80 (0.72-0.87), which was superior to clinical prediction (ΔAUC P = .002, integrated discrimination improvement and net reclassification index P = .003).Our findings suggest that lower levels of intraoperative iron-binding proteins may reflect an impaired capacity to rapidly handle catalytic iron released during cardiopulmonary bypass, leading to kidney injury. These data highlight the importance of iron homeostasis in human ischemia-reperfusion injury and suggest it is a potentially modifiable risk during cardiac surgery. Intraoperative detection of incipient acute kidney injury may be feasible and could be used as an enrichment strategy for clinical trials.Copyright © 2018 The American Association for Thoracic Surgery. Published by Elsevier Inc. All rights reserved.
[21]
Zhou F, Yang Y, Luo L, et al. Impact of prolonged mechanical ventilation on ferroptosis in renal ischemia/reperfusion injury in rats[J]. Biomed Res Int, 2020, 2020: 6097516. DOI: 10.1155/2020/6097516.
[22]
Baliga R, Zhang Z, Baliga M, et al. In vitro and in vivo evidence suggesting a role for iron in cisplatin-induced nephrotoxicity[J]. Kidney Int, 1998, 53(2): 394-401. DOI: 10.1046/j.1523-1755.1998.00767.x.
Cisplatin is a widely used antineoplastic agent that has nephrotoxicity as a major side effect. The underlying mechanism of this nephrotoxicity is still not well known. Iron has been implicated to play an important role in several models of tissue injury, presumably through the generation of hydroxyl radicals via the Haber-Weiss reaction or other highly toxic free radicals. In the present study we examined the catalytic iron content and the effect of iron chelators in an in vitro model of cisplatin-induced cytotoxicity in LLC-PK1 cells (renal tubular epithelial cells) and in an in vivo model of cisplatin-induced acute renal failure in rats. Exposure of LLC-PK1 cells to cisplatin resulted in a significant increase in bleomycin-detectable iron (iron capable of catalyzing free radical reactions) released into the medium. Concurrent incubation of LLC-PK1 cells with iron chelators including deferoxamine and 1,10-phenanthroline significantly attenuated cisplatin-induced cytotoxicity as measured by lactate dehydrogenase (LDH) release. Bleomycin-detectable iron content was also markedly increased in the kidney of rats treated with cisplatin. Similarly, administration of deferoxamine in rats provided marked functional (as measured by blood urea nitrogen and creatinine) and histological protection against cisplatin-induced acute renal failure. In a separate study, we examined the role of hydroxyl radical in cisplatin-induced nephrotoxicity. Incubation of LLC-PK1 cells with cisplatin caused an increase in hydroxyl radical formation. Hydroxyl radical scavengers, dimethyl sulfoxide, mannitol and benzoic acid, significantly reduced cisplatin-induced cytotoxicity and, treatment with dimethyl sulfoxide or dimethylthiourea provided significant protection against cisplatin-induced acute renal failure. Taken together, our data strongly support a critical role for iron in mediating tissue injury via hydroxyl radical (or a similar oxidant) in this model of nephrotoxicity.
[23]
Zarjou A, Bolisetty S, Joseph R, et al. Proximal tubule H-ferritin mediates iron trafficking in acute kidney injury[J]. J Clin Invest, 2013, 123(10): 4423-4434. DOI: 10.1172/JCI67867.
Ferritin plays a central role in iron metabolism and is made of 24 subunits of 2 types: heavy chain and light chain. The ferritin heavy chain (FtH) has ferroxidase activity that is required for iron incorporation and limiting toxicity. The purpose of this study was to investigate the role of FtH in acute kidney injury (AKI) and renal iron handling by using proximal tubule-specific FtH-knockout mice (FtH(PT-/-) mice). FtH(PT-/-) mice had significant mortality, worse structural and functional renal injury, and increased levels of apoptosis in rhabdomyolysis and cisplatin-induced AKI, despite significantly higher expression of heme oxygenase-1, an antioxidant and cytoprotective enzyme. While expression of divalent metal transporter-1 was unaffected, expression of ferroportin (FPN) was significantly lower under both basal and rhabdomyolysis-induced AKI in FtH(PT-/-) mice. Apical localization of FPN was disrupted after AKI to a diffuse cytosolic and basolateral pattern. FtH, regardless of iron content and ferroxidase activity, induced FPN. Interestingly, urinary levels of the iron acceptor proteins neutrophil gelatinase-associated lipocalin, hemopexin, and transferrin were increased in FtH(PT-/-) mice after AKI. These results underscore the protective role of FtH and reveal the critical role of proximal tubule FtH in iron trafficking in AKI.
[24]
Deng F, Sharma I, Dai Y, et al. Myo-inositol oxygenase expression profile modulates pathogenic ferroptosis in the renal proximal tubule[J]. J Clin Invest, 2019, 129(11): 5033-5049. DOI: 10.1172/JCI129903.
Overexpression of myo-inositol oxygenase (MIOX), a proximal tubular enzyme, exacerbates cellular redox injury in acute kidney injury (AKI). Ferroptosis, a newly coined term associated with lipid hydroperoxidation, plays a critical role in the pathogenesis of AKI. Whether or not MIOX exacerbates tubular damage by accelerating ferroptosis in cisplatin-induced AKI remains elusive. Cisplatin-treated HK-2 cells exhibited notable cell death, which was reduced by ferroptosis inhibitors. Also, alterations in various ferroptosis metabolic sensors, including lipid hydroperoxidation, glutathione peroxidase 4 (GPX4) activity, NADPH and reduced glutathione (GSH) levels, and ferritinophagy, were observed. These perturbations were accentuated by MIOX overexpression, while ameliorated by MIOX knockdown. Likewise, cisplatin-treated CD1 mice exhibited tubular damage and derangement of renal physiological parameters, which were alleviated by ferrostatin-1, a ferroptosis inhibitor. To investigate the relevance of MIOX to ferroptosis, WT mice, MIOX-overexpressing transgenic (MIOX-Tg) mice, and MIOX-KO mice were subjected to cisplatin treatment. In comparison with cisplatin-treated WT mice, cisplatin-treated MIOX-Tg mice had more severe renal pathological changes and perturbations in ferroptosis metabolic sensors, which were minimal in cisplatin-treated MIOX-KO mice. In conclusion, these findings indicate that ferroptosis, an integral process in the pathogenesis of cisplatin-induced AKI, is modulated by the expression profile of MIOX.
[25]
Mishima E, Sato E, Ito J, et al. Drugs repurposed as antiferroptosis agents suppress organ damage, including AKI, by functioning as lipid peroxyl radical scavengers[J]. J Am Soc Nephrol, 2020, 31(2): 280-296. DOI: 10.1681/ASN.2019060570.
Ferroptosis, nonapoptotic cell death mediated by free radical reactions and driven by the oxidative degradation of lipids, is a therapeutic target because of its role in organ damage, including AKI. Ferroptosis-causing radicals that are targeted by ferroptosis suppressors have not been unequivocally identified. Because certain cytochrome P450 substrate drugs can prevent lipid peroxidation obscure mechanisms, we evaluated their antiferroptotic potential and used them to identify ferroptosis-causing radicals.Using a cell-based assay, we screened cytochrome P450 substrate compounds to identify drugs with antiferroptotic activity and investigated the underlying mechanism. To evaluate radical-scavenging activity, we used electron paramagnetic resonance-spin trapping methods and a fluorescence probe for lipid radicals, NBD-Pen, that we had developed. We then assessed the therapeutic potency of these drugs in mouse models of cisplatin-induced AKI and LPS/galactosamine-induced liver injury.We identified various US Food and Drug Administration-approved drugs and hormones that have antiferroptotic properties, including rifampicin, promethazine, omeprazole, indole-3-carbinol, carvedilol, propranolol, estradiol, and thyroid hormones. The antiferroptotic drug effects were closely associated with the scavenging of lipid peroxyl radicals but not significantly related to interactions with other radicals. The elevated lipid peroxyl radical levels were associated with ferroptosis onset, and known ferroptosis suppressors, such as ferrostatin-1, also functioned as lipid peroxyl radical scavengers. The drugs exerted antiferroptotic activities in various cell types, including tubules, podocytes, and renal fibroblasts. Moreover, in mice, the drugs ameliorated AKI and liver injury, with suppression of tissue lipid peroxidation and decreased cell death.Although elevated lipid peroxyl radical levels can trigger ferroptosis onset, some drugs that scavenge lipid peroxyl radicals can help control ferroptosis-related disorders, including AKI.Copyright © 2020 by the American Society of Nephrology.
[26]
Martin-Sanchez D, Ruiz-Andres O, Poveda J, et al. Ferroptosis, but not necroptosis, is important in nephrotoxic folic acid-induced AKI[J]. J Am Soc Nephrol, 2017, 28(1): 218-229. DOI: 10.1681/ASN.2015121376.
AKI is histologically characterized by necrotic cell death and inflammation. Diverse pathways of regulated necrosis have been reported to contribute to AKI, but the molecular regulators involved remain unclear. We explored the relative contributions of ferroptosis and necroptosis to folic acid (FA)-induced AKI in mice. FA-AKI in mice associates with lipid peroxidation and downregulation of glutathione metabolism proteins, features that are typical of ferroptotic cell death. We show that ferrostatin-1 (Fer-1), an inhibitor of ferroptosis, preserved renal function and decreased histologic injury, oxidative stress, and tubular cell death in this model. With respect to the immunogenicity of ferroptosis, Fer-1 prevented the upregulation of IL-33, an alarmin linked to necroptosis, and other chemokines and cytokines and prevented macrophage infiltration and Klotho downregulation. In contrast, the pancaspase inhibitor zVAD-fmk did not protect against FA-AKI. Additionally, although FA-AKI resulted in increased protein expression of the necroptosis mediators receptor-interacting protein kinase 3 (RIPK3) and mixed lineage domain-like protein (MLKL), targeting necroptosis with the RIPK1 inhibitor necrostatin-1 or genetic deficiency of RIPK3 or MLKL did not preserve renal function. Indeed, compared with wild-type mice, MLKL knockout mice displayed more severe AKI. However, RIPK3 knockout mice with AKI had less inflammation than their wild-type counterparts, and this effect associated with higher IL-10 concentration and regulatory T cell-to-leukocyte ratio in RIPK3 knockout mice. These data suggest that ferroptosis is the primary cause of FA-AKI and that immunogenicity secondary to ferroptosis may further worsen the damage, although necroptosis-related proteins may have additional roles in AKI.Copyright © 2016 by the American Society of Nephrology.
[27]
Li X, Zou Y, Xing J, et al. Pretreatment with roxadustat (FG-4592) attenuates folic acid-induced kidney injury through antiferroptosis via Akt/GSK-3β/Nrf2 pathway[J]. Oxid Med Cell Longev, 2020, 2020: 6286984. DOI: 10.1155/2020/6286984.
[28]
Córdoba-David G, Duro-Castano A, Castelo-Branco RC, et al. Effective nephroprotection against acute kidney injury with a star-shaped polyglutamate-curcuminoid conjugate[J]. Sci Rep, 2020, 10(1): 2056. DOI: 10.1038/s41598-020-58974-9.
[29]
McKenna MC, Kelly M, Boran G, et al. Spectrum of rhabdomyolysis in an acute hospital[J]. Ir J Med Sci, 2019, 188(4): 1423-1426. DOI: 10.1007/s11845-019-01968-y.
[30]
Panizo N, Rubio-Navarro A, Amaro-Villalobos JM, et al. Molecular mechanisms and novel therapeutic approaches to rhabdomyolysis-induced acute kidney injury[J]. Kidney Blood Press Res, 2015, 40(5): 520-532. DOI: 10.1159/000368528.
Rhabdomyolysis is a syndrome caused by injury to skeletal muscle that usually leads to acute kidney injury (AKI). Rhabdomyolysis has been linked to different conditions, including severe trauma and intense physical exercise. Myoglobin-induced renal toxicity plays a key role in rhabdomyolysis-associated kidney damage by increasing oxidative stress, inflammation, endothelial dysfunction, vasoconstriction, and apoptosis. New drugs that target the harmful effects of myoglobin have been recently developed, and some have been proven to be successful in animal models of acute renal failure secondary to rhabdomyolysis. This review aims to provide a comprehensive and updated overview of the pathological mechanisms of renal damage and describes new therapeutic approaches to this condition based on novel compounds that target key pathways involved in myoglobin-mediated kidney damage.
[31]
Xie Y, Hou W, Song X, et al. Ferroptosis: process and function[J]. Cell Death Differ, 2016, 23(3): 369-379. DOI: 10.1038/cdd.2015.158.
Ferroptosis is a recently recognized form of regulated cell death. It is characterized morphologically by the presence of smaller than normal mitochondria with condensed mitochondrial membrane densities, reduction or vanishing of mitochondria crista, and outer mitochondrial membrane rupture. It can be induced by experimental compounds (e.g., erastin, Ras-selective lethal small molecule 3, and buthionine sulfoximine) or clinical drugs (e.g., sulfasalazine, sorafenib, and artesunate) in cancer cells and certain normal cells (e.g., kidney tubule cells, neurons, fibroblasts, and T cells). Activation of mitochondrial voltage-dependent anion channels and mitogen-activated protein kinases, upregulation of endoplasmic reticulum stress, and inhibition of cystine/glutamate antiporter is involved in the induction of ferroptosis. This process is characterized by the accumulation of lipid peroxidation products and lethal reactive oxygen species (ROS) derived from iron metabolism and can be pharmacologically inhibited by iron chelators (e.g., deferoxamine and desferrioxamine mesylate) and lipid peroxidation inhibitors (e.g., ferrostatin, liproxstatin, and zileuton). Glutathione peroxidase 4, heat shock protein beta-1, and nuclear factor erythroid 2-related factor 2 function as negative regulators of ferroptosis by limiting ROS production and reducing cellular iron uptake, respectively. In contrast, NADPH oxidase and p53 (especially acetylation-defective mutant p53) act as positive regulators of ferroptosis by promotion of ROS production and inhibition of expression of SLC7A11 (a specific light-chain subunit of the cystine/glutamate antiporter), respectively. Misregulated ferroptosis has been implicated in multiple physiological and pathological processes, including cancer cell death, neurotoxicity, neurodegenerative diseases, acute renal failure, drug-induced hepatotoxicity, hepatic and heart ischemia/reperfusion injury, and T-cell immunity. In this review, we summarize the regulation mechanisms and signaling pathways of ferroptosis and discuss the role of ferroptosis in disease.
[32]
Guerrero-Hue M, García-Caballero C, Palomino-Antolín A, et al. Curcumin reduces renal damage associated with rhabdomyolysis by decreasing ferroptosis-mediated cell death[J]. FASEB J, 2019, 33(8): 8961-8975. DOI: 10.1096/fj.201900077R.
Acute kidney injury is a common complication of rhabdomyolysis. A better understanding of this syndrome may be useful to identify novel therapeutic targets because there is no specific treatment so far. Ferroptosis is an iron-dependent form of regulated nonapoptotic cell death that is involved in renal injury. In this study, we investigated whether ferroptosis is associated with rhabdomyolysis-mediated renal damage, and we studied the therapeutic effect of curcumin, a powerful antioxidant with renoprotective properties. Induction of rhabdomyolysis in mice increased serum creatinine levels, endothelial damage, inflammatory chemokines, and cytokine expression, alteration of redox balance (increased lipid peroxidation and decreased antioxidant defenses), and tubular cell death. Treatment with curcumin initiated before or after rhabdomyolysis induction ameliorated all these pathologic and molecular alterations. Although apoptosis or receptor-interacting protein kinase (RIPK)3-mediated necroptosis were activated in rhabdomyolysis, our results suggest a key role of ferroptosis. Thus, treatment with ferrostatin 1, a ferroptosis inhibitor, improved renal function in glycerol-injected mice, whereas no beneficial effects were observed with the pan-caspase inhibitor carbobenzoxy-valyl-alanyl-aspartyl-(-methyl)-fluoromethylketone or in RIPK3-deficient mice. In cultured renal tubular cells, myoglobin (Mb) induced ferroptosis-sensitive cell death that was also inhibited by curcumin. Mechanistic studies showed that curcumin reduced Mb-mediated inflammation and oxidative stress by inhibiting the TLR4/NF-κB axis and activating the cytoprotective enzyme heme oxygenase 1. Our findings are the first to demonstrate the involvement of ferroptosis in rhabdomyolysis-associated renal damage and its sensitivity to curcumin treatment. Therefore, curcumin may be a potential therapeutic approach for patients with this syndrome.-Guerrero-Hue, M., García-Caballero, C., Palomino-Antolín, A., Rubio-Navarro, A., Vázquez-Carballo, C., Herencia, C., Martín-Sanchez, D., Farré-Alins, V., Egea, J., Cannata, P., Praga, M., Ortiz, A., Egido, J., Sanz, A. B., Moreno, J. A. Curcumin reduces renal damage associated with rhabdomyolysis by decreasing ferroptosis-mediated cell death.
[33]
Xie Y, Song X, Sun X, et al. Identification of baicalein as a ferroptosis inhibitor by natural product library screening[J]. Biochem Biophys Res Commun, 2016, 473(4): 775-780. DOI: 10.1016/j.bbrc.2016.03.052.
[34]
Zilka O, Shah R, Li B, et al. On the mechanism of cytoprotection by ferrostatin-1 and liproxstatin-1 and the role of lipid peroxidation in ferroptotic cell death[J]. ACS Cent Sci, 2017, 3(3): 232-243. DOI: 10.1021/acscentsci.7b00028.
[35]
Conrad M, Angeli JP, Vandenabeele P, et al. Regulated necrosis: disease relevance and therapeutic opportunities[J]. Nat Rev Drug Discov, 2016, 15(5): 348-366. DOI: 10.1038/nrd.2015.6.
The discovery of regulated cell death presents tantalizing possibilities for gaining control over the life-death decisions made by cells in disease. Although apoptosis has been the focus of drug discovery for many years, recent research has identified regulatory mechanisms and signalling pathways for previously unrecognized, regulated necrotic cell death routines. Distinct critical nodes have been characterized for some of these alternative cell death routines, whereas other cell death routines are just beginning to be unravelled. In this Review, we describe forms of regulated necrotic cell death, including necroptosis, the emerging cell death modality of ferroptosis (and the related oxytosis) and the less well comprehended parthanatos and cyclophilin D-mediated necrosis. We focus on small molecules, proteins and pathways that can induce and inhibit these non-apoptotic forms of cell death, and discuss strategies for translating this understanding into new therapeutics for certain disease contexts.
[36]
Kagan VE, Mao G, Qu F, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis[J]. Nat Chem Biol, 2017, 13(1): 81-90. DOI: 10.1038/nchembio.2238.
Enigmatic lipid peroxidation products have been claimed as the proximate executioners of ferroptosis-a specialized death program triggered by insufficiency of glutathione peroxidase 4 (GPX4). Using quantitative redox lipidomics, reverse genetics, bioinformatics and systems biology, we discovered that ferroptosis involves a highly organized oxygenation center, wherein oxidation in endoplasmic-reticulum-associated compartments occurs on only one class of phospholipids (phosphatidylethanolamines (PEs)) and is specific toward two fatty acyls-arachidonoyl (AA) and adrenoyl (AdA). Suppression of AA or AdA esterification into PE by genetic or pharmacological inhibition of acyl-CoA synthase 4 (ACSL4) acts as a specific antiferroptotic rescue pathway. Lipoxygenase (LOX) generates doubly and triply-oxygenated (15-hydroperoxy)-diacylated PE species, which act as death signals, and tocopherols and tocotrienols (vitamin E) suppress LOX and protect against ferroptosis, suggesting a homeostatic physiological role for vitamin E. This oxidative PE death pathway may also represent a target for drug discovery.
[37]
Bersuker K, Hendricks JM, Li Z, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis[J]. Nature, 2019, 575(7784): 688-692. DOI: 10.1038/s41586-019-1705-2.
[38]
Doll S, Freitas FP, Shah R, et al. FSP1 is a glutathione-independent ferroptosis suppressor[J]. Nature, 2019, 575(7784): 693-698. DOI: 10.1038/s41586-019-1707-0.
PDF(717 KB)

3612

Accesses

0

Citation

Detail

Sections
Recommended

/