肾小管上皮细胞能量代谢障碍在急性肾损伤中作用的研究进展

朱子敬, 陈星华, 丁国华

PDF(3777 KB)
中华肾脏病杂志 ›› 2020, Vol. 36 ›› Issue (12) : 965-969. DOI: 10.3760/cma.j.cn441217-20200325-00168
综述

肾小管上皮细胞能量代谢障碍在急性肾损伤中作用的研究进展

作者信息 +

Advances on the role of renal tubular epithelial cell energy dysmetabolism in acute kidney injury

Author information +
History +

摘要

肾小管上皮细胞(tubular epithelial cell,TEC)作为肾脏重吸收功能的主要承担者,是急性肾损伤(acute kidney injury,AKI)主要的损伤部位之一。近年来研究提示TEC线粒体受损伴随能量代谢障碍在AKI的发生发展中起着重要作用。在AKI中,线粒体功能障碍促使TEC对能量代谢底物的利用发生改变,通过重编程能量代谢而适应病理环境,本文就TEC在正常生理环境的能量代谢、AKI病理环境的能量代谢、TEC能量代谢重编程与AKI发展及病理转归的关系作一综述,为AKI的治疗及预防提供新的理论依据。

关键词

急性肾损伤 / 肾小管 / 上皮细胞 / 线粒体 / 能量代谢 / 脂肪酸氧化

编辑

彭苗

引用本文

导出引用
朱子敬 , 陈星华 , 丁国华. 肾小管上皮细胞能量代谢障碍在急性肾损伤中作用的研究进展[J]. 中华肾脏病杂志, 2020, 36(12): 965-969. DOI: 10.3760/cma.j.cn441217-20200325-00168.
Zhu Zijing , Chen Xinghua , Ding Guohua. Advances on the role of renal tubular epithelial cell energy dysmetabolism in acute kidney injury[J]. Chinese Journal of Nephrology, 2020, 36(12): 965-969. DOI: 10.3760/cma.j.cn441217-20200325-00168.
急性肾损伤(acute kidney injury,AKI)是一种常见的临床综合征,是高发病率和高病死率的全球公共卫生问题。AKI发病机制复杂,尽管进行了深入的研究,其发病机理仍未完全了解,且缺乏有效的临床治疗方法[1]。肾脏是线粒体丰富的高代谢器官,是仅次于心脏的人体第二大代谢器官,在正常生理情况下,维持其功能需要大量的能量[2]。AKI损伤的主要部位之一是肾小管上皮细胞(tubular epithelial cell,TEC),在缺血缺氧的环境下,TEC线粒体受损严重,氧化磷酸化受阻,导致能量代谢障碍[2]。此时TEC对代谢底物(如葡萄糖、氨基酸和脂肪酸)的利用也会发生改变,并通过改变其供能方式来适应AKI的病理环境[3]。对这些能量代谢改变的机制作进一步研究,不仅能深入认识AKI和肾脏修复机制,而且可能为该疾病的诊断和治疗提供新的策略。

一、 生理状态下TEC的能量代谢

肾脏具有十分复杂的重吸收功能,而肾小管作为重吸收功能的主要承担者。为了维持其复杂的重吸收(钠、水、葡萄糖及其他物质)功能,TEC需要大量的能量供应[4]。葡萄糖氧化和脂肪酸氧化(fatty acid oxidation,FAO)是机体常见的供能方式。葡萄糖通过糖酵解和氧化磷酸化,每个葡萄糖分子产生36个ATP分子,而氧化脂肪酸,如软脂酸,1分子软脂酸彻底氧化可产生106个ATP分子,可见利用脂肪酸产生的能量比葡萄糖更高效[5]。研究发现,TEC中线粒体分布十分密集[6],FAO相关的酶含量也十分丰富,而调节糖酵解的关键酶如己糖激酶、磷酸果糖激酶及丙酮酸激酶含量却很低[7]。TEC主要利用脂肪酸通过FAO来产生大量ATP,而葡萄糖主要用于糖酵解反应,较少经历氧化磷酸化,产生的能量有限[8]
1. FAO: CD36也称为清道夫受体B2,是一种多功能受体,主要介导细胞摄入脂质。CD36在TEC中高表达,TEC通过CD36摄取长链脂肪酸、氧化的脂质和磷脂[9]。脂肪酸被氧化前先活化为长链脂酰CoA。因脂肪酸氧化的酶主要分布于线粒体基质,且线粒体内膜对长链脂酰CoA不通透,长链脂酰CoA需肉碱协助转运进入线粒体基质。肉碱脂酰转移酶(carnitine palmitoyltransferase,CPT)1位于线粒体内膜,为FAO的限速酶,催化长链脂酰CoA与肉碱合成脂酰肉碱,后者在线粒体内膜肉碱-脂酰肉碱转位酶的作用下,通过线粒体内膜进入线粒体基质,同时将等分子肉碱转运出线粒体。脂酰肉碱进入线粒体基质后,在线粒体内膜CPT2的作用下,转变为脂酰CoA并释放肉碱,前者在线粒体经过脂肪酸氧化酶体系彻底氧化,生成大量乙酰CoA、还原型黄素腺嘌呤二核苷酸(FADH2)及还原型烟酰胺腺嘌呤二核苷酸(NADH)[10]
2. 氧化磷酸化: FADH2及NADH进入氧化呼吸链,经过氧化磷酸化产生ATP。氧化呼吸链中复合体Ⅰ和复合体Ⅱ将FADH2及NADH的电子传递给泛醌,泛醌将募集到的电子传递给复合体Ⅲ,后者再将电子传递给细胞色素c,复合体Ⅳ将细胞色素c的电子传递给氧,使其还原成H2O。随着电子通过复合物Ⅰ、Ⅲ和Ⅳ,质子从线粒体基质主动泵入线粒体膜间腔,形成的电位差促使质子通过ATP合酶回流,驱动ADP向ATP转化[10]
FAO的优点在于能在短时间内产生大量ATP,但此过程也需要大量氧气供应。TEC作为高耗氧量细胞,对氧气的易感性高于其他肾脏细胞,这可部分解释为何肾脏在失血、手术或其他创伤时表现出能量供应失衡,容易出现损伤[11]

二、 AKI状态下TEC出现能量代谢障碍

AKI的主要病理表型是肾小管损害,表现为TEC刷状缘消失、凋亡、坏死,从基底膜脱落[12]。线粒体是细胞的“能量工厂”,为维持细胞的基本功能和细胞修复及再生提供能量来源。TEC的FAO、三羧酸循环及氧化磷酸化主要发生在线粒体中,TEC的能量供应离不开功能正常的线粒体。研究发现,在AKI中,TEC中的线粒体出现不同程度的肿胀、断裂并伴随功能障碍,表现为线粒体生物合成、线粒体动力学及线粒体氧化磷酸异常,最终导致线粒体能量产生障碍[13]。线粒体受损是TEC能量代谢失衡的主要原因,也是TEC凋亡及坏死的重要原因之一,在AKI的发病机理中起重要作用[14]
1. TEC线粒体生物合成受损: 过氧化物酶体增殖物激活受体γ辅激活因子1α(PGC-1α)是线粒体生物合成的主要调节分子。PGC-1α通过刺激线粒体的生物合成替代功能障碍的线粒体,从而维持线粒体结构、功能和数量的稳定,并适应细胞能量增加的需要[15]。在肾脏中,TEC作为高代谢、高耗氧的细胞,PGC-1α高度表达[16]。在败血症诱导的AKI小鼠模型中,TEC中PGC-1α的转录水平及蛋白表达同时下调,线粒体数量减少,ATP产生下降,其表达水平与AKI的严重程度成反比,TEC特异性敲除PGC-1α加剧了AKI的肾脏损伤,并呈现出疾病持续进展[17]。在TEC过表达PGC-1α时,线粒体的数量和ATP产生水平恢复,AKI的疾病进展缓慢[18],提示PGC-1α的表达水平对AKI的病理转归发挥着一定的作用。另外,在缺血再灌注(ischemia/reperfusion,I/R)诱导的AKI小鼠模型中,PGC-1α表达受到抑制[19]。TEC过表达PGC-1α可降低小鼠的死亡率,减轻肾脏的脂质蓄积[19],进一步证明TEC的线粒体生物合成参与了AKI的发生发展。
2. TEC线粒体动力学紊乱: 线粒体是高度动态的细胞器,依赖不断地分裂、融合来维持其网状结构,控制线粒体数量和质量的稳定,适应能量变化的需要。线粒体动力学受多种蛋白调控,包括动力相关蛋白1(dynamin-related protein 1,Drpl)、线粒体裂变蛋白1(Fisl)、线粒体融合蛋白1/2(Mitofusin 1/2,Mfnl/2)及视萎缩蛋白(Opa1)[20]。近期研究发现在AKI中,TEC中异常的线粒体动力学导致能量代谢障碍,ATP产生减少,参与疾病的发生发展[21]。在I/R及顺铂诱导的AKI小鼠模型中,肾脏缺血30 min后,短暂再灌注引起近端肾小管细胞约30%~40%的线粒体断裂;体外研究通过顺铂刺激TEC,也同样观察到线粒体快速断裂[22]。对其机制研究发现,TEC中分裂蛋白Drp1去磷酸化并被激活,迅速移位到线粒体,从而促进线粒体裂变,Drp1的抑制剂Mdivi-1通过抑制线粒体断裂减轻AKI的肾脏损伤;而线粒体融合蛋白Opa1则通过蛋白水解而失活,从而导致线粒体融合失调,分裂和融合的紊乱共同导致了线粒体断裂[22-23],提示TEC线粒体动力学异常参与了AKI的发生发展。沉默信息调节因子(Sirtuins,SIRT)是一类高度保守的烟酰胺腺嘌呤二核苷酸(NAD+)依赖的组蛋白去乙酰化酶,其中SIRT3分布在线粒体上并在TEC中高度表达,研究证实SIRT3是调控线粒体动力学和能量代谢的关键分子[24]。在右肾切除+I/R诱导的AKI小鼠模型中,肾脏皮质SIRT3表达显著下降,伴随着融合蛋白Mfn1、Opa1表达下调及分裂蛋白Drp1上调,同时观察到线粒体呼吸链受损[25]。此外,通过构建TEC特异性Mfn2敲除小鼠,探讨融合蛋白Mfn2在肾脏应激中的作用,结果发现该模型小鼠肾单位比正常小鼠少了将近20%,但肾功能相对正常,分离出的初级近端肾小管细胞显示明显的线粒体断裂,在应激状态下肾脏损伤程度更重[26],进一步证明TEC线粒体动力学改变在AKI中发挥作用。

三、 AKI状态下TEC能量代谢重编程

在AKI中,TEC线粒体受损,FAO受阻,氧化磷酸化障碍。能量代谢可能从FAO向糖酵解转换,来响应AKI中TEC线粒体能量代谢障碍,以适应疾病环境[3]
1. TEC的FAO障碍: 在肾脏遭受缺血、缺氧、脓毒血症及肾毒性物质的刺激后,观察到肾脏TEC三酰甘油蓄积,ATP产生匮乏[27]。研究提示,TEC中FAO缺陷可能参与此过程。PPARα属于配体激活的核激素受体超家族的转录因子,已经证实PPARα在调节脂质和葡萄糖代谢及细胞生长和分化中起关键作用。PPARα主要在代谢活跃的组织中表达,如肝脏、心脏以及肾近端小管上皮细胞。PPARα通过编码线粒体FAO酶来激活FAO,同时还通过调控CD36调节脂肪酸进入细胞[28-29]。越来越多的证据表明,PPARα在能量稳态中起着重要的作用。在I/R及顺铂诱导的AKI中,PPARα转录活性降低[30],长链FAO障碍,导致脂肪在细胞积累并产生细胞毒性,从而诱导近端小管上皮细胞坏死[31-32],提示在AKI中TEC的FAO受阻。PPARα的激动剂贝特类药物可恢复AKI中的FAO功能,改善近端小管细胞的脂质蓄积和细胞坏死[33]。在I/R诱导的AKI小鼠模型中,肾脏FAO的关键酶CPT1活性下降,切断了脂肪酸进入线粒体的通路,并伴随着ATP明显减少,通过药理性恢复CPT1的活性可以逆转能量的丧失[34]。同样在I/R诱导的AKI小鼠模型中,肾脏FAO氧化酶体系活性在缺血和再灌注阶段均受到不同程度的抑制[35]。说明在AKI中,近端TEC脂肪酸在运输至线粒体及在线粒体氧化过程中均出现障碍。
2. TEC糖酵解增强: TEC糖酵解能力非常低,但是最近研究发现TEC可以通过增加糖酵解量产生ATP来适应线粒体功能障碍及FAO缺陷。在遭受缺血损伤后,肾脏皮质葡萄糖消耗大量增加,糖酵解限速酶丙酮酸激酶M2(PKM2)活性增加,糖酵解产物丙酮酸、乳酸生成增多,再灌注后糖酵解迅速恢复至基线水平。从正常小鼠分离的近端小管进行体外缺氧/复氧损伤观察发现,同样糖酵解增强[36-37]。提示在AKI中,TEC能量代谢向糖酵解增强。体外培养的近端小管在经历缺氧、线粒体解耦联剂处理和抑制复合体I的作用下,糖酵解代谢明显增加,代偿的糖酵解可部分逆转能量的失衡[38],这进一步证实了TEC通过能量代谢重编程以适应病理环境。在AKI中,增强的糖酵解不仅用于代偿ATP的产生,还具有抗氧化应激的作用。在败血症诱导的AKI小鼠模型,肾皮质糖酵解关键酶己糖激酶活性短时间提高约2倍,主要用于增强磷酸戊糖途径,而磷酸戊糖途径可用于生成还原型烟酰胺腺嘌呤二核苷酸磷酸(NADPH)以维持谷胱甘肽(GSH)的还原状态,进而对抗过氧化物对肾脏的损伤作用[39]。增强的糖酵解可能是TEC对抗损伤的一种方式,恢复的能量供应和抗氧化作用可部分维持TEC正常功能。缺氧诱导因子1α(HIF-1α)是调节细胞缺氧应答的关键转录因子,在缺氧情况下,HIF-1α会驱动糖酵解并同时抑制氧化磷酸化,从而调控细胞代谢重编程的作用。在癌细胞中,SIRT6是调控HIF-1α的关键分子,通过抑制HIF-1α的活性从而调控癌细胞的代谢重编程作用[40]。在I/R诱导的AKI小鼠模型中,TEC中SIRT6表达显著降低;体外实验通过低氧刺激HK-2细胞(人肾小管上皮细胞系),同样观察到SIRT6表达受抑制,HK-2细胞过表达SIRT6时可减轻低氧诱导的细胞损伤及上皮-间质转化(epithelial-to-mesenchymal transition,EMT)[41],初步推测可能是SIRT6\ HIF-1α轴调控的代谢重编程作用参与了此过程,提示SIRT6可能是调控TEC能量代谢的新靶点[3]

四、 TEC能量代谢重编程与AKI发生发展的关系

在AKI状态下,TEC的FAO缺陷不仅导致能量产生障碍,还可能促进细胞内脂质蓄积。短期内增强的糖酵解虽可代偿能量产生障碍,发挥一定的肾脏保护作用,但是研究发现,增强的糖酵解也可能有一定的促炎作用,TEC若长期无法恢复正常能量代谢可能影响AKI的疾病转归,导致肾脏纤维化[42]。说明AKI状态下,TEC能量代谢重编程作用虽然起到了适应AKI病理环境的作用,却有一定的局限性,不能忽视其在AKI发生发展及病理转归中的作用。
1. TEC糖酵解增强炎性反应: 在I/R及败血症诱导的AKI模型中,TEC不仅是炎性反应的“受害者”,也是炎性反应的“积极参与者”,其不仅分泌促炎因子,还能激活炎性细胞,参与肾脏炎性反应,促进肾脏纤维化[43-44]。已经证实免疫细胞(如巨噬细胞、树突状细胞和T细胞)为响应不断变化的炎症环境,也能发生类似的能量代谢重编程作用,以调节免疫功能。如巨噬细胞可通过增强糖酵解以增加吞噬能力,对入侵的病原体做出迅速反应[45]。但是Yang等[46]发现,巨噬细胞中增强的糖酵解也能引起过度炎性反应,在败血症中,PKM2能诱导巨噬细胞释放HMGB1(high mobility group box 1),后者可作为致死性败血症的“晚期炎性反应介质”,而PKM2抑制剂紫草素可降低HMGB1水平,保护小鼠免受致命性败血症的侵害,提示增强糖酵解具有促炎作用。在I/R及败血症诱导的AKI模型中,同样观察到TEC释放HMGB1,促进肾脏炎性反应[47-48],但是并没有直接的证据表明TEC糖酵解增强与肾脏炎性反应有直接联系,二者之间的关系还值得深入探究。
2. TEC的FAO缺陷与脂质蓄积: 在I/R及顺铂诱导的AKI模型中,TEC经常出现一定程度的脂质蓄积,导致细胞功能障碍及坏死,称为脂毒性[49]。TEC的FAO障碍与脂质蓄积是否存在因果关系尚无定论。一方面,在肾脏经历I/R期间,TEC脂质蓄积会干扰正常的线粒体功能,导致FAO障碍。体外培养近端小管在缺氧1 h后,细胞内非脂化脂肪酸(nonestesterified fatty acid,NEFA)显著上调,线粒体膜电位下降,ATP产生明显减少[50]。另一方面,在AKI病理环境中,TEC线粒体对脂肪酸的摄取及氧化均出现障碍,出现了脂肪酸利用与摄取关系的失衡,在一定程度上促进了细胞内脂质蓄积[51]。Kang等[8]通过体外实验发现,抑制TEC的FAO可促进细胞内的脂质蓄积。这提示TEC的FAO缺陷与脂质蓄积可能相互促进,共同参与AKI的发生发展。
3. TEC的FAO缺陷促进AKI向慢性肾脏病(CKD)转变: CKD的最终病理表现是肾脏纤维化,肾纤维化代表慢性及持续性损伤后肾脏组织自我修复失败,其特征是肾小球硬化、肾小管萎缩和间质纤维化[52]。有研究发现AKI中TEC的FAO缺陷和脂质蓄积可能促进EMT和炎性反应,导致肾脏纤维化的形成,从而介导AKI向CKD转变[53]。Simon等[53]认为,为应对AKI中TEC的FAO缺陷导致的能量产生障碍,TEC从耗能较高的上皮表型向耗能较低的间质表型转换,这可部分解释为何短暂的AKI也会增加CKD的风险。而Kang等[8]的研究更加证实了FAO缺陷与肾脏纤维化的关系,利用无差异的代谢组学及转录组学分析,发现在明显纤维化的肾脏中,调节FAO的限速酶及关键分子PPARα表达明显下降,并且下降水平与肾脏小管组织脂质蓄积呈正相关;同样在小鼠肾脏纤维化模型中,转录组学也显示了FAO限速酶及PPARα下调;在体外实验中,抑制小鼠肾脏小管上皮细胞FAO可导致细胞ATP产生减少,细胞去分化及脂质蓄积,而使用FAO激动剂如非诺贝特恢复脂肪酸氧化水平,可减轻小鼠小管间质纤维化。这提示早期恢复TEC的FAO可能是防治AKI甚至阻止肾脏纤维化的新方法。
综上所述,在AKI中,TEC线粒体生物合成受损、线粒体动力学紊乱、氧化磷酸化受阻和FAO缺陷,从而产生能量代谢障碍,促进了AKI的发生发展,并参与肾脏的纤维化过程。在此过程中,TEC可能经历能量代谢重编程,短期内增强的糖酵解不仅承担着能量供应的功能,也发挥着抗氧化应激的作用,从而减轻TEC受损,但是长期的能量代谢重编程也会促进AKI的发生发展,干扰疾病转归。在AKI中,通过药物干预受损的线粒体生物合成、动力学稳态及恢复FAO或可改善肾脏功能。总之,早期恢复TEC正常的能量代谢可能是治疗AKI和预防CKD的新策略。

参考文献

[1]
Hoste E, Kellum JA, Selby NM, et al. Global epidemiology and outcomes of acute kidney injury[J]. Nat Rev Nephrol, 2018, 14(10): 607-625. DOI: 10.1038/s41581-018-0052-0.
Acute kidney injury (AKI) is a commonly encountered syndrome associated with various aetiologies and pathophysiological processes leading to decreased kidney function. In addition to retention of waste products, impaired electrolyte homeostasis and altered drug concentrations, AKI induces a generalized inflammatory response that affects distant organs. Full recovery of kidney function is uncommon, which leaves these patients at risk of long-term morbidity and death. Estimates of AKI prevalence range from <1% to 66%. These variations can be explained by not only population differences but also inconsistent use of standardized AKI classification criteria. The aetiology and incidence of AKI also differ between high-income and low-to-middle-income countries. High-income countries show a lower incidence of AKI than do low-to-middle-income countries, where contaminated water and endemic diseases such as malaria contribute to a high burden of AKI. Outcomes of AKI are similar to or more severe than those of patients in high-income countries. In all resource settings, suboptimal early recognition and care of patients with AKI impede their recovery and lead to high mortality, which highlights unmet needs for improved detection and diagnosis of AKI and for efforts to improve care for these patients.
[2]
Bhargava P, Schnellmann RG. Mitochondrial energetics in the kidney[J]. Nat Rev Nephrol, 2017, 13(10): 629-646. DOI: 10.1038/nrneph.2017.107.
The kidney requires a large number of mitochondria to remove waste from the blood and regulate fluid and electrolyte balance. Mitochondria provide the energy to drive these important functions and can adapt to different metabolic conditions through a number of signalling pathways (for example, mechanistic target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) pathways) that activate the transcriptional co-activator peroxisome proliferator-activated receptor-γ co-activator 1α (PGC1α), and by balancing mitochondrial dynamics and energetics to maintain mitochondrial homeostasis. Mitochondrial dysfunction leads to a decrease in ATP production, alterations in cellular functions and structure, and the loss of renal function. Persistent mitochondrial dysfunction has a role in the early stages and progression of renal diseases, such as acute kidney injury (AKI) and diabetic nephropathy, as it disrupts mitochondrial homeostasis and thus normal kidney function. Improving mitochondrial homeostasis and function has the potential to restore renal function, and administering compounds that stimulate mitochondrial biogenesis can restore mitochondrial and renal function in mouse models of AKI and diabetes mellitus. Furthermore, inhibiting the fission protein dynamin 1-like protein (DRP1) might ameliorate ischaemic renal injury by blocking mitochondrial fission.
[3]
Gómez H, Kellum JA, Ronco C. Metabolic reprogramming and tolerance during sepsis-induced AKI[J]. Nat Rev Nephrol, 2017, 13(3): 143-151. DOI: 10.1038/nrneph.2016.186.
The host defence against infection is an adaptive response in which several mechanisms are deployed to decrease the pathogen load, limit tissue injury and restore homeostasis. In the past few years new evidence has suggested that the ability of the immune system to limit the microbial burden - termed resistance - might not be the only defence mechanism. In fact, the capacity of the host to decrease its own susceptibility to inflammation- induced tissue damage - termed tolerance - might be as important as resistance in determining the outcome of the infection. Metabolic adaptations are central to the function of the cellular immune response. Coordinated reprogramming of metabolic signalling enables cells to execute resistance and tolerance pathways, withstand injury, steer tissue repair and promote organ recovery. During sepsis-induced acute kidney injury, early reprogramming of metabolism can determine the extent of organ dysfunction, progression to fibrosis, and the development of chronic kidney disease. Here we discuss the mechanisms of tolerance that act in the kidney during sepsis, with particular attention to the role of metabolic responses in coordinating these adaptive strategies. We suggest a novel conceptual model of the cellular and organic response to sepsis that might lead to new avenues for targeted, organ-protective therapies.
[4]
Forbes JM, Thorburn DR. Mitochondrial dysfunction in diabetic kidney disease[J]. Nat Rev Nephrol, 2018, 14(5): 291-312. DOI: 10.1038/nrneph.2018.9.
Globally, diabetes is the leading cause of chronic kidney disease and end-stage renal disease, which are major risk factors for cardiovascular disease and death. Despite this burden, the factors that precipitate the development and progression of diabetic kidney disease (DKD) remain to be fully elucidated. Mitochondrial dysfunction is associated with kidney disease in nondiabetic contexts, and increasing evidence suggests that dysfunctional renal mitochondria are pathological mediators of DKD. These complex organelles have a broad range of functions, including the generation of ATP. The kidneys are mitochondrially rich, highly metabolic organs that require vast amounts of ATP for their normal function. The delivery of metabolic substrates for ATP production, such as fatty acids and oxygen, is altered by diabetes. Changes in metabolic fuel sources in diabetes to meet ATP demands result in increased oxygen consumption, which contributes to renal hypoxia. Inherited factors including mutations in genes that impact mitochondrial function and/or substrate delivery may also be important risk factors for DKD. Hence, we postulate that the diabetic milieu and inherited factors that underlie abnormalities in mitochondrial function synergistically drive the development and progression of DKD.
[5]
Rich PR. The molecular machinery of Keilin's respiratory chain[J]. Biochem Soc Trans, 2003, 31(Pt 6): 1095-1105. DOI: 10.1042/bst0311095.
Keilin's classic paper of 1925 [Keilin (1925) Proc. R. Soc. London Ser. B 100, 129–151], achieved with simple, but elegant, techniques, describes the cytochrome components of the respiratory chain and their roles in intracellular respiration and oxygen consumption. Since that time, a tremendous amount of work has clarified the intricate details of the prosthetic groups, cofactors and proteins that comprise the respiratory chain and associated machinery for ATP synthesis. The work has culminated in advanced crystallographic and spectroscopic methods that provide structural and mechanistic details of this mitochondrial molecular machinery, in many instances to atomic level. I review here the current state of understanding of the mitochondrial respiratory chain in terms of structures and dynamics of the component proteins and their roles in the biological electron and proton transfer processes that result in ATP synthesis. These advances, together with emerging evidence of further diverse roles of mitochondria in health and disease, have prompted a new era of interest in mitochondrial function.
[6]
Brinkkoetter PT, Bork T, Salou S, et al. Anaerobic glycolysis maintains the glomerular filtration barrier independent of mitochondrial metabolism and dynamics[J]. Cell Rep, 2019, 27(5): 1551-1566.e5. DOI: 10.1016/j.celrep.2019.04.012.
The cellular responses induced by mitochondrial dysfunction remain elusive. Intrigued by the lack of almost any glomerular phenotype in patients with profound renal ischemia, we comprehensively investigated the primary sources of energy of glomerular podocytes. Combining functional measurements of oxygen consumption rates, glomerular metabolite analysis, and determination of mitochondrial density of podocytes in vivo, we demonstrate that anaerobic glycolysis and fermentation of glucose to lactate represent the key energy source of podocytes. Under physiological conditions, we could detect neither a developmental nor late-onset pathological phenotype in podocytes with impaired mitochondrial biogenesis machinery, defective mitochondrial fusion-fission apparatus, or reduced mtDNA stability and transcription caused by podocyte-specific deletion of Pgc-1α, Drp1, or Tfam, respectively. Anaerobic glycolysis represents the predominant metabolic pathway of podocytes. These findings offer a strategy to therapeutically interfere with the enhanced podocyte metabolism in various progressive kidney diseases, such as diabetic nephropathy or focal segmental glomerulosclerosis (FSGS).Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.
[7]
Guder WG, Ross BD. Enzyme distribution along the nephron[J]. Kidney Int, 1984, 26(2): 101-111. DOI: 10.1038/ki.1984.143.
[8]
Kang HM, Ahn SH, Choi P, et al. Defective fatty acid oxidation in renal tubular epithelial cells has a key role in kidney fibrosis development[J]. Nat Med, 2015, 21(1): 37-46. DOI: 10.1038/nm.3762.
Renal fibrosis is the histological manifestation of a progressive, usually irreversible process causing chronic and end-stage kidney disease. We performed genome-wide transcriptome studies of a large cohort (n = 95) of normal and fibrotic human kidney tubule samples followed by systems and network analyses and identified inflammation and metabolism as the top dysregulated pathways in the diseased kidneys. In particular, we found that humans and mouse models with tubulointerstitial fibrosis had lower expression of key enzymes and regulators of fatty acid oxidation (FAO) and higher intracellular lipid deposition compared to controls. In vitro experiments indicated that inhibition of FAO in tubule epithelial cells caused ATP depletion, cell death, dedifferentiation and intracellular lipid deposition, phenotypes observed in fibrosis. In contrast, restoring fatty acid metabolism by genetic or pharmacological methods protected mice from tubulointerstitial fibrosis. Our results raise the possibility that correcting the metabolic defect in FAO may be useful for preventing and treating chronic kidney disease.
[9]
Yang X, Okamura DM, Lu X, et al. CD36 in chronic kidney disease: novel insights and therapeutic opportunities[J]. Nat Rev Nephrol, 2017, 13(12): 769-781. DOI: 10.1038/nrneph.2017.126.
CD36 (also known as scavenger receptor B2) is a multifunctional receptor that mediates the binding and cellular uptake of long-chain fatty acids, oxidized lipids and phospholipids, advanced oxidation protein products, thrombospondin and advanced glycation end products, and has roles in lipid accumulation, inflammatory signalling, energy reprogramming, apoptosis and kidney fibrosis. Renal CD36 is mainly expressed in tubular epithelial cells, podocytes and mesangial cells, and is markedly upregulated in the setting of chronic kidney disease (CKD). As fatty acids are the preferred energy source for proximal tubule cells, a reduction in fatty acid oxidation in CKD affects kidney lipid metabolism by disrupting the balance between fatty acid synthesis, uptake and consumption. The outcome is intracellular lipid accumulation, which has an important role in the pathogenesis of kidney fibrosis. In experimental models, antagonist blockade or genetic knockout of CD36 prevents kidney injury, suggesting that CD36 could be a novel target for therapy. Here, we discuss the regulation and post-translational modification of CD36, its role in renal pathophysiology and its potential as a biomarker and as a therapeutic target for the prevention of kidney fibrosis.
[10]
Lodish H, Berk A, Zipursky SL, et al. Molecular cell biology[M]. 4th ed. New York: W.H. Freeman and Company, 2000.
[11]
Forbes JM. Mitochondria-power players in kidney function?[J]. Trends Endocrinol Metab, 2016, 27(7): 441-442. DOI: 10.1016/j.tem.2016.05.002.
[12]
Basile DP, Anderson MD, Sutton TA. Pathophysiology of acute kidney injury[J]. Compr Physiol, 2012, 2(2): 1303-1353. DOI: 10.1002/cphy.c110041.
Acute kidney injury (AKI) is the leading cause of nephrology consultation and is associated with high mortality rates. The primary causes of AKI include ischemia, hypoxia, or nephrotoxicity. An underlying feature is a rapid decline in glomerular filtration rate (GFR) usually associated with decreases in renal blood flow. Inflammation represents an important additional component of AKI leading to the extension phase of injury, which may be associated with insensitivity to vasodilator therapy. It is suggested that targeting the extension phase represents an area potential of treatment with the greatest possible impact. The underlying basis of renal injury appears to be impaired energetics of the highly metabolically active nephron segments (i.e., proximal tubules and thick ascending limb) in the renal outer medulla, which can trigger conversion from transient hypoxia to intrinsic renal failure. Injury to kidney cells can be lethal or sublethal. Sublethal injury represents an important component in AKI, as it may profoundly influence GFR and renal blood flow. The nature of the recovery response is mediated by the degree to which sublethal cells can restore normal function and promote regeneration. The successful recovery from AKI depends on the degree to which these repair processes ensue and these may be compromised in elderly or chronic kidney disease (CKD) patients. Recent data suggest that AKI represents a potential link to CKD in surviving patients. Finally, earlier diagnosis of AKI represents an important area in treating patients with AKI that has spawned increased awareness of the potential that biomarkers of AKI may play in the future.© 2012 American Physiological Society. Compr Physiol 2:1303-1353, 2012.
[13]
Duann P, Lin PH. Mitochondria damage and kidney disease[J]. Adv Exp Med Biol, 2017, 982: 529-551. DOI: 10.1007/978-3-319-55330-6_27.
The kidney is a vital organ that demands an extraordinary amount of energy to actively maintain the body's metabolism, plasma hemodynamics, electrolytes and water homeostasis, nutrients reabsorption, and hormone secretion. Kidney is only second to the heart in mitochondrial count and oxygen consumption. As such, the health and status of the energy power house, the mitochondria, is pivotal to the health and proper function of the kidney. Mitochondria are heterogeneous and highly dynamic organelles and their functions are subject to complex regulations through modulation of its biogenesis, bioenergetics, dynamics and clearance within cell. Kidney diseases, either acute kidney injury (AKI) or chronic kidney disease (CKD), are important clinical issues and global public health concerns with high mortality rate and socioeconomic burden due to lack of effective therapeutic strategies to cure or retard the progression of the diseases. Mitochondria-targeted therapeutics has become a major focus for modern research with the belief that maintaining mitochondria homeostasis can prevent kidney pathogenesis and disease progression. A better understanding of the cellular and molecular events that govern mitochondria functions in health and disease will potentially lead to improved therapeutics development.
[14]
Ishimoto Y, Inagi R. Mitochondria: a therapeutic target in acute kidney injury[J]. Nephrol Dial Transplant, 2016, 31(7): 1062-1069. DOI: 10.1093/ndt/gfv317.
[15]
Liang H, Ward WF. PGC-1alpha: a key regulator of energy metabolism[J]. Adv Physiol Educ, 2006, 30(4): 145-151. DOI: 10.1152/advan.00052.2006.
Peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1alpha is a member of a family of transcription coactivators that plays a central role in the regulation of cellular energy metabolism. It is strongly induced by cold exposure, linking this environmental stimulus to adaptive thermogenesis. PGC-1alpha stimulates mitochondrial biogenesis and promotes the remodeling of muscle tissue to a fiber-type composition that is metabolically more oxidative and less glycolytic in nature, and it participates in the regulation of both carbohydrate and lipid metabolism. It is highly likely that PGC-1alpha is intimately involved in disorders such as obesity, diabetes, and cardiomyopathy. In particular, its regulatory function in lipid metabolism makes it an inviting target for pharmacological intervention in the treatment of obesity and Type 2 diabetes.
[16]
Fontecha-Barriuso M, Martin-Sanchez D, Martinez-Moreno JM, et al. The role of PGC-1α and mitochondrial biogenesis in kidney diseases[J]. Biomolecules, 2020, 10(2): 347. DOI: 10.3390/biom10020347.
Chronic kidney disease (CKD) is one of the fastest growing causes of death worldwide, emphasizing the need to develop novel therapeutic approaches. CKD predisposes to acute kidney injury (AKI) and AKI favors CKD progression. Mitochondrial derangements are common features of both AKI and CKD and mitochondria-targeting therapies are under study as nephroprotective agents. PGC-1α is a master regulator of mitochondrial biogenesis and an attractive therapeutic target. Low PGC-1α levels and decreased transcription of its gene targets have been observed in both preclinical AKI (nephrotoxic, endotoxemia, and ischemia-reperfusion) and in experimental and human CKD, most notably diabetic nephropathy. In mice, PGC-1α deficiency was associated with subclinical CKD and predisposition to AKI while PGC-1α overexpression in tubular cells protected from AKI of diverse causes. Several therapeutic strategies may increase kidney PGC-1α activity and have been successfully tested in animal models. These include AMP-activated protein kinase (AMPK) activators, phosphodiesterase (PDE) inhibitors, and anti-TWEAK antibodies. In conclusion, low PGC-1α activity appears to be a common feature of AKI and CKD and recent characterization of nephroprotective approaches that increase PGC-1α activity may pave the way for nephroprotective strategies potentially effective in both AKI and CKD.
[17]
Tran M, Tam D, Bardia A, et al. PGC-1α promotes recovery after acute kidney injury during systemic inflammation in mice[J]. J Clin Invest, 2011, 121(10): 4003-4014. DOI: 10.1172/JCI58662.
Sepsis-associated acute kidney injury (AKI) is a common and morbid condition that is distinguishable from typical ischemic renal injury by its paucity of tubular cell death. The mechanisms underlying renal dysfunction in individuals with sepsis-associated AKI are therefore less clear. Here we have shown that endotoxemia reduces oxygen delivery to the kidney, without changing tissue oxygen levels, suggesting reduced oxygen consumption by the kidney cells. Tubular mitochondria were swollen, and their function was impaired. Expression profiling showed that oxidative phosphorylation genes were selectively suppressed during sepsis-associated AKI and reactivated when global function was normalized. PPARγ coactivator-1α (PGC-1α), a major regulator of mitochondrial biogenesis and metabolism, not only followed this pattern but was proportionally suppressed with the degree of renal impairment. Furthermore, tubular cells had reduced PGC-1α expression and oxygen consumption in response to TNF-α; however, excess PGC-1α reversed the latter effect. Both global and tubule-specific PGC-1α-knockout mice had normal basal renal function but suffered persistent injury following endotoxemia. Our results demonstrate what we believe to be a novel mechanism for sepsis-associated AKI and suggest that PGC-1α induction may be necessary for recovery from this disorder, identifying a potential new target for future therapeutic studies.
[18]
Rasbach KA, Schnellmann RG. PGC-1alpha over-expression promotes recovery from mitochondrial dysfunction and cell injury[J]. Biochem Biophys Res Commun, 2007, 355(3): 734-739. DOI: 10.1016/j.bbrc.2007.02.023.
[19]
Tran MT, Zsengeller ZK, Berg AH, et al. PGC1α drives NAD biosynthesis linking oxidative metabolism to renal protection[J]. Nature, 2016, 531(7595): 528-532. DOI: 10.1038/nature17184.
[20]
Chan DC. Fusion and fission: interlinked processes critical for mitochondrial health[J]. Annu Rev Genet, 2012, 46: 265-287. DOI: 10.1146/annurev-genet-110410-132529.
Mitochondria are dynamic organelles that continually undergo fusion and fission. These opposing processes work in concert to maintain the shape, size, and number of mitochondria and their physiological function. Some of the major molecules mediating mitochondrial fusion and fission in mammals have been discovered, but the underlying molecular mechanisms are only partially unraveled. In particular, the cast of characters involved in mitochondrial fission needs to be clarified. By enabling content mixing between mitochondria, fusion and fission serve to maintain a homogeneous and healthy mitochondrial population. Mitochondrial dynamics has been linked to multiple mitochondrial functions, including mitochondrial DNA stability, respiratory capacity, apoptosis, response to cellular stress, and mitophagy. Because of these important functions, mitochondrial fusion and fission are essential in mammals, and even mild defects in mitochondrial dynamics are associated with disease. A better understanding of these processes likely will ultimately lead to improvements in human health.
[21]
Zhan M, Brooks C, Liu F, et al. Mitochondrial dynamics: regulatory mechanisms and emerging role in renal pathophysiology[J]. Kidney Int, 2013, 83(4): 568-581. DOI: 10.1038/ki.2012.441.
Mitochondria are a class of dynamic organelles that constantly undergo fission and fusion. Mitochondrial dynamics is governed by a complex molecular machinery and finely tuned by regulatory proteins. During cell injury or stress, the dynamics is shifted to fission, resulting in mitochondrial fragmentation, which contributes to mitochondrial damage and consequent cell injury and death. Emerging evidence has suggested a role of mitochondrial fragmentation in the pathogenesis of renal diseases including acute kidney injury and diabetic nephropathy. A better understanding of the regulation of mitochondrial dynamics and its pathogenic changes may unveil novel therapeutic strategies.
[22]
Brooks C, Wei Q, Cho SG, et al. Regulation of mitochondrial dynamics in acute kidney injury in cell culture and rodent models[J]. J Clin Invest, 2009, 119(5): 1275-1285. DOI: 10.1172/JCI37829.
The mechanism of mitochondrial damage, a key contributor to renal tubular cell death during acute kidney injury, remains largely unknown. Here, we have demonstrated a striking morphological change of mitochondria in experimental models of renal ischemia/reperfusion and cisplatin-induced nephrotoxicity. This change contributed to mitochondrial outer membrane permeabilization, release of apoptogenic factors, and consequent apoptosis. Following either ATP depletion or cisplatin treatment of rat renal tubular cells, mitochondrial fragmentation was observed prior to cytochrome c release and apoptosis. This mitochondrial fragmentation was inhibited by Bcl2 but not by caspase inhibitors. Dynamin-related protein 1 (Drp1), a critical mitochondrial fission protein, translocated to mitochondria early during tubular cell injury, and both siRNA knockdown of Drp1 and expression of a dominant-negative Drp1 attenuated mitochondrial fragmentation, cytochrome c release, caspase activation, and apoptosis. Further in vivo analysis revealed that mitochondrial fragmentation also occurred in proximal tubular cells in mice during renal ischemia/reperfusion and cisplatin-induced nephrotoxicity. Notably, both tubular cell apoptosis and acute kidney injury were attenuated by mdivi-1, a newly identified pharmacological inhibitor of Drp1. This study demonstrates a rapid regulation of mitochondrial dynamics during acute kidney injury and identifies mitochondrial fragmentation as what we believe to be a novel mechanism contributing to mitochondrial damage and apoptosis in vivo in mouse models of disease.
[23]
Wei Q, Sun H, Song S, et al. MicroRNA-668 represses MTP18 to preserve mitochondrial dynamics in ischemic acute kidney injury[J]. J Clin Invest, 2018, 128(12): 5448-5464. DOI: 10.1172/JCI121859.
The pathogenesis of ischemic diseases remains unclear. Here we demonstrate the induction of microRNA-668 (miR-668) in ischemic acute kidney injury (AKI) in human patients, mice, and renal tubular cells. The induction was HIF-1 dependent, as HIF-1 deficiency in cells and kidney proximal tubules attenuated miR-668 expression. We further identified a functional HIF-1 binding site in the miR-668 gene promoter. Anti-miR-668 increased apoptosis in renal tubular cells and enhanced ischemic AKI in mice, whereas miR-668 mimic was protective. Mechanistically, anti-miR-668 induced mitochondrial fragmentation, whereas miR-668 blocked mitochondrial fragmentation during hypoxia. We analyzed miR-668 target genes through immunoprecipitation of microRNA-induced silencing complexes followed by RNA deep sequencing and identified 124 protein-coding genes as likely targets of miR-668. Among these genes, only mitochondrial protein 18 kDa (MTP18) has been implicated in mitochondrial dynamics. In renal cells and mouse kidneys, miR-668 mimic suppressed MTP18, whereas anti-miR-668 increased MTP18 expression. Luciferase microRNA target reporter assay further verified MTP18 as a direct target of miR-668. In renal tubular cells, knockdown of MTP18 suppressed mitochondrial fragmentation and apoptosis. Together, the results suggest that miR-668 is induced via HIF-1 in ischemic AKI and that, upon induction, miR-668 represses MTP18 to preserve mitochondrial dynamics for renal tubular cell survival and kidney protection.
[24]
Morigi M, Perico L, Rota C, et al. Sirtuin 3-dependent mitochondrial dynamic improvements protect against acute kidney injury[J]. J Clin Invest, 2015, 125(2): 715-726. DOI: 10.1172/JCI77632.
Acute kidney injury (AKI) is a public health concern with an annual mortality rate that exceeds those of breast and prostate cancer, heart failure, and diabetes combined. Oxidative stress and mitochondrial damage are drivers of AKI-associated pathology; however, the pathways that mediate these events are poorly defined. Here, using a murine cisplatin-induced AKI model, we determined that both oxidative stress and mitochondrial damage are associated with reduced levels of renal sirtuin 3 (SIRT3). Treatment with the AMPK agonist AICAR or the antioxidant agent acetyl-l-carnitine (ALCAR) restored SIRT3 expression and activity, improved renal function, and decreased tubular injury in WT animals, but had no effect in Sirt3-/- mice. Moreover, Sirt3-deficient mice given cisplatin experienced more severe AKI than WT animals and died, and neither AICAR nor ALCAR treatment prevented death in Sirt3-/- AKI mice. In cultured human tubular cells, cisplatin reduced SIRT3, resulting in mitochondrial fragmentation, while restoration of SIRT3 with AICAR and ALCAR improved cisplatin-induced mitochondrial dysfunction. Together, our results indicate that SIRT3 is protective against AKI and suggest that enhancing SIRT3 to improve mitochondrial dynamics has potential as a strategy for improving outcomes of renal injury.
[25]
程玲莉, 杨定平, 刘杰, 等. SIRT3在小鼠缺血再灌注致急性肾损伤早期纤维化中的作用[J]. 武汉大学学报(医学版), 2019, 40(3): 350-355. DOI: 10.14188/j.1671-8852.2018.0717.
[26]
Gall JM, Wang Z, Liesa M, et al. Role of mitofusin 2 in the renal stress response[J]. PLoS One, 2012, 7(1): e31074. DOI: 10.1371/journal.pone.0031074.
[27]
Zager RA, Johnson AC, Hanson SY. Renal tubular triglyercide accumulation following endotoxic, toxic, and ischemic injury[J]. Kidney Int, 2005, 67(1): 111-121. DOI: 10.1111/j.1523-1755.2005.00061.x.
Cholesterol accumulates in renal cortical proximal tubules in response to diverse forms of injury or physiologic stress. However, the fate of triglycerides after acute renal insults is poorly defined. This study sought new insights into this issue.CD-1 mice were subjected to three diverse models of renal stress: (1) endotoxemia [Escherichia coli lipopolysaccharide (LPS), injection]; (2) ischemia/reperfusion (I/R); or (3) glycerol-induced rhabdomyolysis. Renal cortical, or isolated proximal tubule, triglyceride levels were measured approximately 18 hours later. To gain mechanistic insights, triglyceride levels were determined in (1) proximal tubules following exogenous phospholipase A(2) (PLA(2)) treatment; (2) cultured HK-2 cells after mitochondrial blockade (antimycin A) +/- serum; or (3) HK-2 cells following "septic" (post-LPS) serum, or exogenous fatty acid (oleate) addition.Each form of in vivo injury evoked three-to fourfold triglyceride increases in renal cortex and/or proximal tubules. PLA(2) treatment of proximal tubules evoked acute, dose-dependent, triglyceride formation. HK-2 cell triglyceride levels rose with antimycin A. With serum present, antimycin A induced an exaggerated triglyceride loading state (vs. serum alone or antimycin A alone). "Septic" serum stimulated HK-2 triglyceride formation (compared to control serum). Oleate addition caused striking HK-2 cell triglyceride accumulation. Following oleate washout, HK-2 cells were sensitized to adenosine triphosphate (ATP) depletion or oxidant attack.Diverse forms of renal injury induce dramatic triglyceride loading in proximal tubules/renal cortex, suggesting that this is a component of a cell stress response. PLA(2) activity, increased triglyceride/triglyceride substrate (e.g., fatty acid) uptake, and possible systemic cytokine (e.g., from LPS) stimulation, may each contribute to this result. Finally, in addition to being a marker of prior cell injury, accumulation of triglyceride (or of its constituent fatty acids) may predispose tubules to superimposed ATP depletion or oxidant attack.
[28]
Chung KW, Lee EK, Lee MK, et al. Impairment of PPARα and the fatty acid oxidation pathway aggravates renal fibrosis during aging[J]. J Am Soc Nephrol, 2018, 29(4): 1223-1237. DOI: 10.1681/ASN.2017070802.
Defects in the renal fatty acid oxidation (FAO) pathway have been implicated in the development of renal fibrosis. Although, compared with young kidneys, aged kidneys show significantly increased fibrosis with impaired kidney function, the mechanisms underlying the effects of aging on renal fibrosis have not been investigated. In this study, we investigated peroxisome proliferator-activated receptor (PPAR) and the FAO pathway as regulators of age-associated renal fibrosis. The expression of PPAR and the FAO pathway-associated proteins significantly decreased with the accumulation of lipids in the renal tubular epithelial region during aging in rats. In particular, decreased PPAR protein expression associated with increased expression of PPAR-targeting microRNAs. Among the microRNAs with increased expression during aging, miR-21 efficiently decreased PPAR expression and impaired FAO when ectopically expressed in renal epithelial cells. In cells pretreated with oleic acid to induce lipid stress, miR-21 treatment further enhanced lipid accumulation. Furthermore, treatment with miR-21 significantly exacerbated the TGF--induced fibroblast phenotype of epithelial cells. We verified the physiologic importance of our findings in a calorie restriction model. Calorie restriction rescued the impaired FAO pathway during aging and slowed fibrosis development. Finally, compared with kidneys of aged littermate controls, kidneys of aged PPAR mice showed exaggerated lipid accumulation, with decreased activity of the FAO pathway and a severe fibrosis phenotype. Our results suggest that impaired renal PPAR signaling during aging aggravates renal fibrosis development, and targeting PPAR is useful for preventing age-associated CKD.Copyright © 2018 by the American Society of Nephrology.
[29]
Lakhia R, Yheskel M, Flaten A, et al. PPARα agonist fenofibrate enhances fatty acid β-oxidation and attenuates polycystic kidney and liver disease in mice[J]. Am J Physiol Renal Physiol, 2018, 314(1): F122-F131. DOI: 10.1152/ajprenal.00352.2017.
Peroxisome proliferator-activated receptor α (PPARα) is a nuclear hormone receptor that promotes fatty acid β-oxidation (FAO) and oxidative phosphorylation (OXPHOS). We and others have recently shown that PPARα and its target genes are downregulated, and FAO and OXPHOS are impaired in autosomal dominant polycystic kidney disease (ADPKD). However, whether PPARα and FAO/OXPHOS are causally linked to ADPKD progression is not entirely clear. We report that expression of PPARα and FAO/OXPHOS genes is downregulated, and in vivo β-oxidation rate of3H-labeled triolein is reduced in Pkd1RC/RCmice, a slowly progressing orthologous model of ADPKD that closely mimics the human ADPKD phenotype. To evaluate the effects of upregulating PPARα, we conducted a 5-mo, randomized, preclinical trial by treating Pkd1RC/RCmice with fenofibrate, a clinically available PPARα agonist. Fenofibrate treatment resulted in increased expression of PPARα and FAO/OXPHOS genes, upregulation of peroxisomal and mitochondrial biogenesis markers, and higher β-oxidation rates in Pkd1RC/RCkidneys. MRI-assessed total kidney volume and total cyst volume, kidney-weight-to-body-weight ratio, cyst index, and serum creatinine levels were significantly reduced in fenofibrate-treated compared with untreated littermate Pkd1RC/RCmice. Moreover, fenofibrate treatment was associated with reduced kidney cyst proliferation and infiltration by inflammatory cells, including M2-like macrophages. Finally, fenofibrate treatment also reduced bile duct cyst number, cyst proliferation, and liver inflammation and fibrosis. In conclusion, our studies suggest that promoting PPARα activity to enhance mitochondrial metabolism may be a useful therapeutic strategy for ADPKD.
[30]
Portilla D, Dai G, McClure T, et al. Alterations of PPARalpha and its coactivator PGC-1 in cisplatin-induced acute renal failure[J]. Kidney Int, 2002, 62(4): 1208-1218. DOI: 10.1111/j.1523-1755.2002.kid553.x.
In this study we examined whether a recently characterized coactivator of Peroxisome proliferator activated receptor alpha (PPARalpha), Peroxisome proliferator activated receptor-gamma-coactivator-1 (PGC-1) plays a role in the regulation of fatty acid oxidation during cisplatin-induced nephrotoxicity.Studies in mouse kidneys used quantitative reverse transcription-polymerase chain reaction (RT-PCR) to measure peroxisomal acyl coenzyme A (acyl-CoA) and PGC-1 mRNA levels and in situ hybridization to localize PGC-1 mRNA. Studies in LLCPK1 cells used quantitative RT-PCR and biochemical assays to measure mRNA levels and enzyme activities of peroxisomal acyl-CoA, mitochondrial carnitine palmitoyl transferase (CPT) and PGC-1. Eletrophoretic mobility shift assays (EMSA) and Western blot analysis of nuclear extracts, and transient transfection of PGC-1 were used to examine the effect of cisplatin on PPARalpha-regulated fatty acid oxidation.Cisplatin decreased mRNA levels of peroxisomal acyl-CoA enzyme in mouse kidney and also reduced the mRNA levels and enzyme activities of acyl-CoA and mitochondrial CPT-1 in LLCPK1 cells. DNA-protein binding studies demonstrated that exposure to cisplatin reduces PPARalpha/retinoid X receptor (RXRalpha) binding activity. Immunoblotting studies demonstrated that cisplatin had no effect on nuclear levels of PPARalpha or RXRalpha protein. In situ hybridization studies in mouse kidney demonstrated the localization of PGC-1 mRNA to proximal tubules and thick ascending limb of Henley (TALH) cells. Cisplatin diminished the expression of PGC-1 mRNA levels in mouse kidney and also in LLCPK1 cells. Transient expression of PGC-1 shows the nuclear localization of PGC-1 protein and increased PPARalpha transcriptional activity in LLCPK1 cells.These results demonstrate that cisplatin deactivates PPARalpha by reducing its DNA binding activity and the availability of its tissue specific coactivator PGC-1.
[31]
Portilla D, Dai G, Peters JM, et al. Etomoxir-induced PPARalpha-modulated enzymes protect during acute renal failure[J]. Am J Physiol Renal Physiol, 2000, 278(4): F667-F675. DOI: 10.1152/ajprenal.2000.278.4.F667.
Regulation of fatty acid β-oxidation (FAO) represents an important mechanism for a sustained balance of energy production/utilization in kidney tissue. To examine the role of stimulated FAO during ischemia, Etomoxir (Eto), clofibrate, and WY-14,643 compounds were given 5 days prior to the induction of ischemia/reperfusion (I/R) injury. Compared with rats administered vehicle, Eto-, clofibrate-, and WY-treated rats had lower blood urea nitrogen and serum creatinines following I/R injury. Histological analysis confirmed a significant amelioration of acute tubular necrosis. I/R injury led to a threefold reduction of mRNA and protein levels of acyl CoA oxidase (AOX) and cytochrome P4A1, as well as twofold inhibition of their enzymatic activities. Eto treatment prevented the reduction of mRNA and protein levels and the inhibition of the enzymatic activities of these two peroxisome proliferator-activated receptor-α (PPARα) target genes during I/R injury. PPARα null mice subjected to I/R injury demonstrated significantly enhanced cortical necrosis and worse kidney function compared with wild-type controls. These results suggest that upregulation of PPARα-modulated FAO genes has an important role in the observed cytoprotection during I/R injury.
[32]
Li S, Nagothu KK, Desai V, et al. Transgenic expression of proximal tubule peroxisome proliferator-activated receptor-alpha in mice confers protection during acute kidney injury[J]. Kidney Int, 2009, 76(10): 1049-1062. DOI: 10.1038/ki.2009.330.
Our previous studies suggest that peroxisome proliferator-activated receptor-alpha (PPARalpha) plays a critical role in regulating fatty acid beta-oxidation in kidney tissue and this directly correlated with preservation of kidney morphology and function during acute kidney injury. To further study this, we generated transgenic mice expressing PPARalpha in the proximal tubule under the control of the promoter of KAP2 (kidney androgen-regulated protein 2). Segment-specific upregulation of PPARalpha expression by testosterone treatment of female transgenic mice improved kidney function during cisplatin or ischemia-reperfusion-induced acute kidney injury. Ischemia-reperfusion injury or treatment with cisplatin in wild-type mice caused inhibition of fatty-acid oxidation, reduction of mitochondrial genes of oxidative phosphorylation, mitochondrial DNA, fatty-acid metabolism, and the tricarboxylic acid cycle. Similar injury in testosterone-treated transgenic mice resulted in amelioration of these effects. Similarly, there were increases in the levels of 4-hydroxy-2-hexenal-derived lipid peroxidation products in wild-type mice, which were also reduced in the transgenic mice. Similarly, necrosis of the S3 segment was reduced in the two injury models in transgenic mice compared to wild type. Our results suggest proximal tubule PPARalpha activity serves as a metabolic sensor. Its increased expression without the use of an exogenous PPARalpha ligand in the transgenic mice is sufficient to protect kidney function and morphology, and to prevent abnormalities in lipid metabolism associated with acute kidney injury.
[33]
Li S, Wu P, Yarlagadda P, et al. PPAR alpha ligand protects during cisplatin-induced acute renal failure by preventing inhibition of renal FAO and PDC activity[J]. Am J Physiol Renal Physiol, 2004, 286(3): F572-F580. DOI: 10.1152/ajprenal.00190.2003.
Previous studies demonstrated that during cisplatin-induced acute renal failure, there is a significant reduction in proximal tubule fatty acid oxidation. We now report on the effects of peroxisome proliferator-activated receptor-α (PPARα) ligand Wy-14643 (WY) on the abnormalities of medium chain fatty acid oxidation and pyruvate dehydrogenase complex (PDC) activity in kidney tissue of cisplatin-treated mice. Cisplatin causes a significant reduction in mRNA levels and enzyme activity of mitochondrial medium chain acyl-CoA dehydrogenase (MCAD). PPARα ligand WY ameliorated cisplatin-induced acute renal failure and prevented cisplatin-induced reduction of mRNA levels and enzyme activity of MCAD. In contrast, in cisplatin-treated PPARα null mice, WY did not protect kidney function and did not reverse cisplatin-induced decreased expression of MCAD. Cisplatin inhibited renal PDC activity before the development of acute tubular necrosis, and PDC inhibition was reversed by pretreatment with PPARα agonist WY. Cisplatin also induced increased mRNA and protein levels of pyruvate dehydrogenase kinase-4 (PDK4), and PPARα ligand WY prevented cisplatin-induced increased expression of PDK4 protein levels in wild-type mice. We conclude that PPARα agonists have therapeutic potential for cisplatin-induced acute renal failure. Use of PPARα ligands prevents acute tubular necrosis by ameliorating cisplatin-induced inhibition of two distinct metabolic processes, MCAD-mediated fatty acid oxidation and PDC activity.
[34]
Idrovo JP, Yang WL, Nicastro J, et al. Stimulation of carnitine palmitoyltransferase 1 improves renal function and attenuates tissue damage after ischemia/reperfusion[J]. J Surg Res, 2012, 177(1): 157-164. DOI: 10.1016/j.jss.2012.05.053.
[35]
Ruidera E, Irazu CE, Rajagopalan PR, et al. Fatty acid metabolism in renal ischemia[J]. Lipids, 1988, 23(9): 882-884. DOI: 10.1007/BF02536209.
The increase in free fatty acids in the ischemic tissue is a consistent observation and these free fatty acids are considered to play a role in the cellular toxicity. To elucidate the cause of higher levels of free fatty acids in ischemic tissue, we examined the catabolism of fatty acids. The beta-oxidation of lignoceric (24:0), palmitic (16:0) and octanoic (8:0) acids and the peroxidation of fatty acids were measured at different times of renal ischemia in whole kidney homogenate. The enzymatic activities for the oxidation of fatty acids decreased with the increase in ischemia time. However, the lipid peroxide levels increased 2.5-fold of control with ischemic injury. Sixty min of ischemia reduced the rate of oxidation of octanoic, palmitic and lignoceric acids by 57, 59 and 69%, respectively. Almost similar loss of fatty acid oxidation activity was observed in the peroxisomes and mitochondria. These data suggest that loss of mitochondrial and peroxisomal fatty acid beta-oxidation enzyme activities from ischemic injury may be one of the factors responsible for the higher levels of free fatty acids.
[36]
Zager RA, Johnson AC, Becker K. Renal cortical pyruvate depletion during AKI[J]. J Am Soc Nephrol, 2014, 25(5): 998-1012. DOI: 10.1681/ASN.2013070791.
Pyruvate is a key intermediary in energy metabolism and can exert antioxidant and anti-inflammatory effects. However, the fate of pyruvate during AKI remains unknown. Here, we assessed renal cortical pyruvate and its major determinants (glycolysis, gluconeogenesis, pyruvate dehydrogenase [PDH], and H2O2 levels) in mice subjected to unilateral ischemia (15-60 minutes; 0-18 hours of vascular reflow) or glycerol-induced ARF. The fate of postischemic lactate, which can be converted back to pyruvate by lactate dehydrogenase, was also addressed. Ischemia and glycerol each induced persistent pyruvate depletion. During ischemia, decreasing pyruvate levels correlated with increasing lactate levels. During early reperfusion, pyruvate levels remained depressed, but lactate levels fell below control levels, likely as a result of rapid renal lactate efflux. During late reperfusion and glycerol-induced AKI, pyruvate depletion corresponded with increased gluconeogenesis (pyruvate consumption). This finding was underscored by observations that pyruvate injection increased renal cortical glucose content in AKI but not normal kidneys. AKI decreased PDH levels, potentially limiting pyruvate to acetyl CoA conversion. Notably, pyruvate therapy mitigated the severity of AKI. This renoprotection corresponded with increases in cytoprotective heme oxygenase 1 and IL-10 mRNAs, selective reductions in proinflammatory mRNAs (e.g., MCP-1 and TNF-α), and improved tissue ATP levels. Paradoxically, pyruvate increased cortical H2O2 levels. We conclude that AKI induces a profound and persistent depletion of renal cortical pyruvate, which may induce additional injury.Copyright © 2014 by the American Society of Nephrology.
[37]
Lan R, Geng H, Singha PK, et al. Mitochondrial pathology and glycolytic shift during proximal tubule atrophy after ischemic AKI[J]. J Am Soc Nephrol, 2016, 27(11): 3356-3367. DOI: 10.1681/ASN.2015020177.
During recovery by regeneration after AKI, proximal tubule cells can fail to redifferentiate, undergo premature growth arrest, and become atrophic. The atrophic tubules display pathologically persistent signaling increases that trigger production of profibrotic peptides, proliferation of interstitial fibroblasts, and fibrosis. We studied proximal tubules after ischemia-reperfusion injury (IRI) to characterize possible mitochondrial pathologies and alterations of critical enzymes that govern energy metabolism. In rat kidneys, tubules undergoing atrophy late after IRI but not normally recovering tubules showed greatly reduced mitochondrial number, with rounded profiles, and large autophagolysosomes. Studies after IRI of kidneys in mice, done in parallel, showed large scale loss of the oxidant-sensitive mitochondrial protein Mpv17L. Renal expression of hypoxia markers also increased after IRI. During early and late reperfusion after IRI, kidneys exhibited increased lactate and pyruvate content and hexokinase activity, which are indicators of glycolysis. Furthermore, normally regenerating tubules as well as tubules undergoing atrophy exhibited increased glycolytic enzyme expression and inhibitory phosphorylation of pyruvate dehydrogenase. TGF-β antagonism prevented these effects. Our data show that the metabolic switch occurred early during regeneration after injury and was reversed during normal tubule recovery but persisted and became progressively more severe in tubule cells that failed to redifferentiate. In conclusion, irreversibility of the metabolic switch, taking place in the context of hypoxia, high TGF-β signaling and depletion of mitochondria characterizes the development of atrophy in proximal tubule cells and may contribute to the renal pathology after AKI.Copyright © 2016 by the American Society of Nephrology.
[38]
Dickman KG, Mandel LJ. Differential effects of respiratory inhibitors on glycolysis in proximal tubules[J]. Am J Physiol, 1990, 258(6 Pt 2): F1608-F1615. DOI: 10.1152/ajprenal.1990.258.6.F1608.
[39]
Smith JA, Stallons LJ, Schnellmann RG. Renal cortical hexokinase and pentose phosphate pathway activation through the EGFR/Akt signaling pathway in endotoxin-induced acute kidney injury[J]. Am J Physiol Renal Physiol, 2014, 307(4): F435-F444. DOI: 10.1152/ajprenal.00271.2014.
While disruption of energy production is an important contributor to renal injury, metabolic alterations in sepsis-induced AKI remain understudied. We assessed changes in renal cortical glycolytic metabolism in a mouse model of sepsis-induced AKI. A specific and rapid increase in hexokinase (HK) activity (∼2-fold) was observed 3 h after LPS exposure and maintained up to 18 h, in association with a decline in renal function as measured by blood urea nitrogen (BUN). LPS-induced HK activation occurred independently of HK isoform expression or mitochondrial localization. No other changes in glycolytic enzymes were observed. LPS-mediated HK activation was not sufficient to increase glycolytic flux as indicated by reduced or unchanged pyruvate and lactate levels in the renal cortex. LPS-induced HK activation was associated with increased glucose-6-phosphate dehydrogenase activity but not glycogen production. Mechanistically, LPS-induced HK activation was attenuated by pharmacological inhibitors of the EGF receptor (EGFR) and Akt, indicating that EGFR/phosphatidylinositol 3-kinase/Akt signaling is responsible. Our findings reveal LPS rapidly increases renal cortical HK activity in an EGFR- and Akt-dependent manner and that HK activation is linked to increased pentose phosphate pathway activity.
[40]
Zwaans BM, Lombard DB. Interplay between sirtuins, MYC and hypoxia-inducible factor in cancer-associated metabolic reprogramming[J]. Dis Model Mech, 2014, 7(9): 1023-1032. DOI: 10.1242/dmm.016287.
In the early twentieth century, Otto Heinrich Warburg described an elevated rate of glycolysis occurring in cancer cells, even in the presence of atmospheric oxygen (the Warburg effect). Despite the inefficiency of ATP generation through glycolysis, the breakdown of glucose into lactate provides cancer cells with a number of advantages, including the ability to withstand fluctuations in oxygen levels, and the production of intermediates that serve as building blocks to support rapid proliferation. Recent evidence from many cancer types supports the notion that pervasive metabolic reprogramming in cancer and stromal cells is a crucial feature of neoplastic transformation. Two key transcription factors that play major roles in this metabolic reprogramming are hypoxia inducible factor-1 (HIF1) and MYC. Sirtuin-family deacetylases regulate diverse biological processes, including many aspects of tumor biology. Recently, the sirtuin SIRT6 has been shown to inhibit the transcriptional output of both HIF1 and MYC, and to function as a tumor suppressor. In this Review, we highlight the importance of HIF1 and MYC in regulating tumor metabolism and their regulation by sirtuins, with a main focus on SIRT6. © 2014. Published by The Company of Biologists Ltd.
[41]
Gao Z, Chen X, Fan Y, et al. Sirt6 attenuates hypoxia-induced tubular epithelial cell injury via targeting G2/M phase arrest[J]. J Cell Physiol, 2020, 235(4): 3463-3473. DOI: 10.1002/jcp.29235.
Acute kidney injury (AKI) is a condition that has a high incidence and death rate. Unfortunately, the kidney may not recover completely after AKI, which then develops to chronic kidney disease (CKD). Therefore, it is necessary to identify potential curative targets to avoid its development to CKD. As an NAD -dependent deacetylase, sirtuin 6 (Sirt6) has been linked to different types of biological processes. In the present work, our group investigated the role of Sirt6 in tubular epithelial cells (TECs) under hypoxic stress. Sirt6 expression was examined in mouse kidney following ischemia/reperfusion (IR) injury and hypoxia-challenged TECs. Using Sirt6 plasmid and small interfering RNA, we also investigated how, in regard to inflammation and epithelial-to-mesenchymal transition, Sirt6 affects hypoxia-triggered injury. In addition, cell cycle was detected in hypoxia-challenged TECs. Sirt6 was downregulated in the kidney of mice with IR injury and hypoxia-challenged TECs. Consequently, Sirt6 depletion aggravated hypoxia-induced injury and G2/M phase arrest. Sirt6 overexpression attenuated hypoxia-triggered damage and G2/M phase arrest in TECs. Sirt6 prevented hypoxia-triggered TEC damage via suppressing G2/M phase arrest. Thus, Sirt6 is a possible candidate for alleviating the effects of kidney injury.© 2019 Wiley Periodicals, Inc.
[42]
汪知玉, 张文. 脂肪酸氧化在缺血再灌注急性肾损伤发病及转归中的作用[J]. 中华肾脏病杂志, 2019, 35(10): 784-789. DOI: 10.3760/cma.j.issn.1001-7097.2019.10.011.
急性肾损伤(acute kidney injury,AKI)是临床常见危重症,组织缺血缺氧是各种原因引起AKI 的共同病理生理基础,肾脏缺血再灌注(ischemia reperfusion,IR)损伤是常用的AKI 模型。脂肪酸氧化是肾小管上皮细胞(renal tubular epithelial cells,RTECs)能量的主要来源,脂肪酸氧化减少是IR肾损伤主要的病理生理改变之一。本文回顾了近年来脂肪酸氧化在IR 肾损伤中的相关研究,对RTECs 脂肪酸氧化过程和异常脂肪酸氧化在IR 肾损伤发病及转归中的作用机制进行综述。目前研究发现,再灌注早期脂肪酸氧化水平下降可能减少ROS生成并减轻线粒体氧化应激水平,而长期脂肪酸氧化障碍则会造成RTECs能量生成不足和脂肪酸超载,不利于肾小管结构和功能恢复,持续性RTECs 脂肪酸氧化不足可进一步促进IR 肾损伤后肾小管萎缩和肾纤维化。改善RTECs 的脂肪酸氧化水平可能是阻断AKI 后肾纤维化发生发展的治疗新思路,但靶向调控脂肪酸氧化药物在IR肾损伤早期治疗中的价值尚需研究进一步探讨。
[43]
Bonventre JV, Zuk A. Ischemic acute renal failure: an inflammatory disease?[J]. Kidney Int, 2004, 66(2): 480-485. DOI: 10.1111/j.1523-1755.2004.761_2.x.
Inflammation plays a major role in the pathophysiology of acute renal failure resulting from ischemia. In this review, we discuss the contribution of endothelial and epithelial cells and leukocytes to this inflammatory response. The roles of cytokines/chemokines in the injury and recovery phase are reviewed. The ability of the mouse kidney to be protected by prior exposure to ischemia or urinary tract obstruction is discussed as a potential model to emulate as we search for pharmacologic agents that will serve to protect the kidney against injury. Understanding the inflammatory response prevalent in ischemic kidney injury will facilitate identification of molecular targets for therapeutic intervention.
[44]
Cantaluppi V, Quercia AD, Dellepiane S, et al. Interaction between systemic inflammation and renal tubular epithelial cells[J]. Nephrol Dial Transplant, 2014, 29(11): 2004-2011. DOI: 10.1093/ndt/gfu046.
[45]
Cheng SC, Quintin J, Cramer RA, et al. mTOR- and HIF-1α-mediated aerobic glycolysis as metabolic basis for trained immunity[J]. Science, 2014, 345(6204): 1250684. DOI: 10.1126/science.1250684.
\n To determine the epigenetic mechanisms that direct blood cells to develop into the many components of our immune system, the BLUEPRINT consortium examined the regulation of DNA and RNA transcription to dissect the molecular traits that govern blood cell differentiation. By inducing immune responses, Saeed\n et al.\n document the epigenetic changes in the genome that underlie immune cell differentiation. Cheng\n et al.\n demonstrate that trained monocytes are highly dependent on the breakdown of sugars in the presence of oxygen, which allows cells to produce the energy needed to mount an immune response. Chen\n et al.\n examine RNA transcripts and find that specific cell lineages use RNA transcripts of different length and composition (isoforms) to form proteins. Together, the studies reveal how epigenetic effects can drive the development of blood cells involved in the immune system.\n
[46]
Yang L, Xie M, Yang M, et al. PKM2 regulates the Warburg effect and promotes HMGB1 release in sepsis[J]. Nat Commun, 2014, 5: 4436. DOI: 10.1038/ncomms5436.
Increasing evidence suggests the important role of metabolic reprogramming in the regulation of the innate inflammatory response, but the underlying mechanism remains unclear. Here we provide evidence to support a novel role for the pyruvate kinase M2 (PKM2)-mediated Warburg effect, namely aerobic glycolysis, in the regulation of high-mobility group box 1 (HMGB1) release. PKM2 interacts with hypoxia-inducible factor 1 alpha (HIF1 alpha) and activates the HIF-1 alpha-dependent transcription of enzymes necessary for aerobic glycolysis in macrophages. Knockdown of PKM2, HIF1 alpha and glycolysis-related genes uniformly decreases lactate production and HMGB1 release. Similarly, a potential PKM2 inhibitor, shikonin, reduces serum lactate and HMGB1 levels, and protects mice from lethal endotoxemia and sepsis. Collectively, these findings shed light on a novel mechanism for metabolic control of inflammation by regulating HMGB1 release and highlight the importance of targeting aerobic glycolysis in the treatment of sepsis and other inflammatory diseases.
[47]
Lau A, Wang S, Liu W, et al. Glycyrrhizic acid ameliorates HMGB1-mediated cell death and inflammation after renal ischemia reperfusion injury[J]. Am J Nephrol, 2014, 40(1): 84-95. DOI: 10.1159/000364908.
Renal ischemia reperfusion injury (IRI) leads to acute kidney injury (AKI) and the death of tubular epithelial cells (TEC). The release of high-mobility group box-1 (HMGB1) and other damage-associated molecular pattern moieties from dying cells may promote organ dysfunction and inflammation by effects on TEC. Glycyrrhizic acid (GZA) is a functional inhibitor of HMGB1, but its ability to attenuate the HMGB1-mediated injury of TEC has not been tested.In vitro, hypoxia and cytokine treatment killed TEC and resulted in the progressive release of HMGB1 into the supernatant. GZA reduced the hypoxia-induced TEC death as measured by annexin-V and propidium iodide. Hypoxia increased the expression of MCP-1 and CXCL1 in TEC, which was reduced by GZA in a dose-dependent manner. Similarly, the HMGB1 activation of effector NK cells was inhibited by GZA. To test the effect of HMGB1 neutralization by GZA in vivo, mice were subjected to renal IRI. HMGB1 protein expression increased progressively in kidneys from 4 to 24 h after ischemia and was detected in tubular cells by 4 h using immunohistochemistry. GZA preserved renal function after IRI and reduced tubular necrosis and neutrophil infiltration by histological analyses and ethidium homodimer staining.Importantly, these data demonstrate for the first time that AKI following hypoxia and renal IRI may be promoted by HMGB1 release, which can reduce the survival of TEC and augment inflammation. Inhibition of the interaction of HMGB1 with TEC through GZA may represent a therapeutic strategy for the attenuation of renal injury following IRI and transplantation.© 2014 S. Karger AG, Basel.
[48]
Abdulmahdi W, Patel D, Rabadi MM, et al. HMGB1 redox during sepsis[J]. Redox Biol, 2017, 13: 600-607. DOI: 10.1016/j.redox.2017.08.001.
During sepsis, the alarmin HMGB1 is released from tissues and promotes systemic inflammation that results in multi-organ damage, with the kidney particularly susceptible to injury. The severity of inflammation and pro-damage signaling mediated by HMGB1 appears to be dependent on the alarmin's redox state. Therefore, we examined HMGB1 redox in kidney cells during sepsis. Using intravital microscopy, CellROX labeling of kidneys in live mice indicated increased ROS generation in the kidney perivascular endothelium and tubules during lipopolysaccharide (LPS)-induced sepsis. Subsequent CellROX and MitoSOX labeling of LPS-stressed endothelial and kidney proximal tubule cells demonstrated increased ROS generation in these cells as sepsis worsens. Consequently, HMGB1 oxidation increased in the cytoplasm of kidney cells during its translocation from the nucleus to the circulation, with the degree of oxidation dependent on the severity of sepsis, as measured in in vivo mouse samples using a thiol assay and mass spectrometry (LC-MS/MS). The greater the oxidation of HMGB1, the greater the ability of the alarmin to stimulate pro-inflammatory cyto-/chemokine release (measured by Luminex Multiplex) and alter mitochondrial ATP generation (Luminescent ATP Detection Assay). Administration of glutathione and thioredoxin inhibitors to cell cultures enhanced HMGB1 oxidation during sepsis in endothelial and proximal tubule cells, respectively. In conclusion, as sepsis worsens, ROS generation and HMGB1 oxidation increases in kidney cells, which enhances HMGB1's pro-inflammatory signaling. Conversely, the glutathione and thioredoxin systems work to maintain the protein in its reduced state.Copyright © 2017 The Authors. Published by Elsevier B.V. All rights reserved.
[49]
Erpicum P, Rowart P, Defraigne JO, et al. What we need to know about lipid-associated injury in case of renal ischemia-reperfusion[J]. Am J Physiol Renal Physiol, 2018, 315(6): F1714-F1719. DOI: 10.1152/ajprenal.00322.2018.
Renal segmental metabolism is reflected by the complex distribution of the main energy pathways along the nephron, with fatty acid oxidation preferentially used in the cortex area. Ischemia/reperfusion injury (IRI) is due to the restriction of renal blood flow, rapidly leading to a metabolic switch toward anaerobic conditions. Subsequent unbalance between energy demand and oxygen/nutrient delivery compromises kidney cell functions, resulting in a complex inflammatory cascade including the production of reactive oxygen species (ROS). Renal IRI especially involves lipid accumulation. Lipid peroxidation is one of the major events of ROS-associated tissue injury. Here, we briefly review the current knowledge of renal cell lipid metabolism in normal and ischemic conditions. Next, we focus on renal lipid-associated injury, with emphasis on its mechanisms and consequences during the course of IRI. Finally, we discuss preclinical observations aiming at preventing and/or attenuating lipid-associated IRI.
[50]
Feldkamp T, Kribben A, Roeser NF, et al. Accumulation of nonesterified fatty acids causes the sustained energetic deficit in kidney proximal tubules after hypoxia-reoxygenation[J]. Am J Physiol Renal Physiol, 2006, 290(2): F465-F477. DOI: 10.1152/ajprenal.00305.2005.
Kidney proximal tubules exhibit decreased ATP and reduced, but not absent, mitochondrial membrane potential (Δψm) during reoxygenation after severe hypoxia. This energetic deficit, which plays a pivotal role in overall cellular recovery, cannot be explained by loss of mitochondrial membrane integrity, decreased electron transport, or compromised F1F0-ATPase and adenine nucleotide translocase activities. Addition of oleate to permeabilized tubules produced concentration-dependent decreases of Δψmmeasured by safranin O uptake (threshold for oleate = 0.25 μM, 1.6 nmol/mg protein; maximal effect = 4 μM, 26 nmol/mg) that were reversed by delipidated BSA (dBSA). Cell nonesterified fatty acid (NEFA) levels increased from &lt;1 to 17.4 nmol/mg protein during 60- min hypoxia and remained elevated at 7.6 nmol/mg after 60 min reoxygenation, at which time ATP had recovered to only 10% of control values. Safranin O uptake in reoxygenated tubules, which was decreased 85% after 60-min hypoxia, was normalized by dBSA, which improved ATP synthesis as well. dBSA also almost completely normalized Δψmwhen the duration of hypoxia was increased to 120 min. In intact tubules, the protective substrate combination of α-ketoglutarate + malate (α-KG/MAL) increased ATP three- to fourfold, limited NEFA accumulation during hypoxia by 50%, and lowered NEFA during reoxygenation. Notably, dBSA also improved ATP recovery when added to intact tubules during reoxygenation and was additive to the effect of α-KG/MAL. We conclude that NEFA overload is the primary cause of energetic failure of reoxygenated proximal tubules and lowering NEFA substantially contributes to the benefit from supplementation with α-KG/MAL.
[51]
Jang HS, Noh MR, Kim J, et al. Defective mitochondrial fatty acid oxidation and lipotoxicity in kidney diseases[J]. Front Med (Lausanne), 2020, 7: 65. DOI: 10.3389/fmed.2020.00065.
[52]
Webster AC, Nagler EV, Morton RL, et al. Chronic kidney disease[J]. Lancet, 2017, 389(10075): 1238-1252. DOI: 10.1016/S0140-6736(16)32064-5.
The definition and classification of chronic kidney disease (CKD) have evolved over time, but current international guidelines define this condition as decreased kidney function shown by glomerular filtration rate (GFR) of less than 60 mL/min per 1·73 m, or markers of kidney damage, or both, of at least 3 months duration, regardless of the underlying cause. Diabetes and hypertension are the main causes of CKD in all high-income and middle-income countries, and also in many low-income countries. Incidence, prevalence, and progression of CKD also vary within countries by ethnicity and social determinants of health, possibly through epigenetic influence. Many people are asymptomatic or have non-specific symptoms such as lethargy, itch, or loss of appetite. Diagnosis is commonly made after chance findings from screening tests (urinary dipstick or blood tests), or when symptoms become severe. The best available indicator of overall kidney function is GFR, which is measured either via exogenous markers (eg, DTPA, iohexol), or estimated using equations. Presence of proteinuria is associated with increased risk of progression of CKD and death. Kidney biopsy samples can show definitive evidence of CKD, through common changes such as glomerular sclerosis, tubular atrophy, and interstitial fibrosis. Complications include anaemia due to reduced production of erythropoietin by the kidney; reduced red blood cell survival and iron deficiency; and mineral bone disease caused by disturbed vitamin D, calcium, and phosphate metabolism. People with CKD are five to ten times more likely to die prematurely than they are to progress to end stage kidney disease. This increased risk of death rises exponentially as kidney function worsens and is largely attributable to death from cardiovascular disease, although cancer incidence and mortality are also increased. Health-related quality of life is substantially lower for people with CKD than for the general population, and falls as GFR declines. Interventions targeting specific symptoms, or aimed at supporting educational or lifestyle considerations, make a positive difference to people living with CKD. Inequity in access to services for this disease disproportionally affects disadvantaged populations, and health service provision to incentivise early intervention over provision of care only for advanced CKD is still evolving in many countries.Copyright © 2017 Elsevier Ltd. All rights reserved.
[53]
Simon N, Hertig A. Alteration of fatty acid oxidation in tubular epithelial cells: from acute kidney injury to renal fibrogenesis[J]. Front Med(Lausanne), 2015, 2(52): 1-7. DOI: 10.3389/fmed.2015.00052.

利益冲突

所有作者均声明不存在利益冲突

基金

国家自然科学基金(81770687)
国家自然科学基金(81500516)
PDF(3777 KB)

4375

Accesses

0

Citation

Detail

段落导航
相关文章

/