Advances of regulating mitochondrial function by microRNA in kidney diseases

Zhang Xiaolu, Li Shuzhen, Zhang Aihua

PDF(766 KB)
Chinese Journal of Nephrology ›› 2021, Vol. 37 ›› Issue (5) : 448-454. DOI: 10.3760/cma.j.cn441217-20200831-00068
Review

Advances of regulating mitochondrial function by microRNA in kidney diseases

Author information +
History +

Abstract

Cite this article

Download Citations
Zhang Xiaolu. , Li Shuzhen. , Zhang Aihua. Advances of regulating mitochondrial function by microRNA in kidney diseases[J]. Chinese Journal of Nephrology, 2021, 37(5): 448-454. DOI: 10.3760/cma.j.cn441217-20200831-00068.
肾脏是人体重要的高耗能器官之一,能量代谢效率仅次于心脏,丰富的线粒体为维持其正常功能提供能量,线粒体功能障碍会导致肾脏损伤[1]。影响线粒体功能障碍的分子机制较多,如动力学相关蛋白1(dynamin-related protein 1,Drp-1)缺乏[2]、过氧化物酶体增殖物激活受体γ辅激活因子1α(peroxisome proliferator-activated receptor gamma coactivator-1 alpha,PGC-1α)信号通路异常[3]、非编码RNA(miRNA、lncRNA)异常调控[4]、活性氧(reactive oxygen species,ROS)过度产生[5]等,其中微RNA(microRNA,miRNA)调控线粒体功能是近年的研究热点。miRNA通过影响细胞内氧化磷酸化等过程,参与肾脏疾病的发生及发展。本文将综述miRNA调控线粒体功能在肾脏衰老和肾脏疾病中作用的研究进展,探讨其在肾脏疾病诊断与治疗中的潜在价值。

一、 线粒体的特征及功能

线粒体是真核生物细胞内重要的细胞器,主要通过氧化磷酸化为生命活动提供能量,还参与其他生物学过程,如细胞生长、凋亡、自噬及氧化应激等[6]。细胞内的线粒体可经融合和分裂维持动态平衡,适应不同条件下的能量需求。线粒体的结构损伤、DNA损伤、动力学紊乱以及呼吸链受抑制均可致线粒体功能障碍。线粒体功能障碍与多种高耗能器官的疾病密切相关,如心脏病、肾脏疾病、肝炎、癌症等[7]。此外,线粒体与细胞内其他细胞器通过生物膜接触点进行动态交流,如内质网、溶酶体、过氧化物酶体等,调节多种细胞过程;线粒体与各细胞器的接触时间、接触数量及接触方式的差异,决定其接触点结构、功能及动力[8]。有研究发现线粒体TBC1D15促进Rab7 GTP酶在线粒体-溶酶体接触点处水解,使RAB7效应蛋白从溶酶体膜释放,发挥溶酶体动力作用[9],参与调节细胞自噬、细胞增殖与死亡、线粒体动力、物质运输等[10];内质网-线粒体接触点被发现参与完成脂质代谢、钙离子转运、线粒体DNA(mitochondrial DNA,mtDNA)合成与分裂、内质网应激、细胞代谢等过程[11-12];此外,过氧化物酶体和线粒体均在调节信号传递及细胞代谢中起到关键作用,近年,大量证据表明两者在特定条件下会紧密接触,并在脂肪酸氧化、免疫信号传导等过程中发挥协调作用[13]
1. 线粒体与能量供应: 线粒体内膜上的氧化呼吸链是ATP的合成部位,生物代谢物在氧化呼吸链上复合体Ⅰ(NADH-泛醌氧化还原酶)、复合体Ⅱ(琥珀酸-泛醌氧化还原酶)、复合体Ⅲ(泛醇-细胞色素c氧化还原酶)、复合体Ⅳ(细胞色素c氧化酶)、ATP合酶及多种辅酶的作用下,经一系列氧化还原反应、电子及质子的传递,驱动ADP磷酸化为ATP,为人体生命活动提供能量[6]
2. 线粒体与ROS产生: 氧化呼吸链中少数逸出电子与O2结合生成ROS,ROS是一类有高氧化活性的分子或离子,包括超氧自由基(O2-)、过氧化氢(H2O2)、羟自由基(OH)、过氧化脂质(LOOH)等。正常情况下,体内的抗氧化系统可清除氧化应激产生的ROS。若ROS的产生与清除失调,过度产生的ROS通过激活细胞信号转导途径,损害细胞内蛋白、核酸、脂质等物质,引起细胞损伤[7]
3. 线粒体动力学: 线粒体动力学包括融合和分裂两部分。融合及分裂间的动态平衡,在维持线粒体正常大小、形态、分布及功能中发挥重要作用。线粒体动力学紊乱可参与肾脏疾病、心血管疾病、癌症、神经退行性疾病等疾病的发生及发展[2]。(1)线粒体融合:线粒体融合包括外膜融合及内膜融合两部分,线粒体融合相关蛋白1和线粒体融合相关蛋白2是参与调控外膜融合的重要分子,视神经萎缩蛋白1(optic atrophy 1,OPA1)则参与调控内膜融合。线粒体融合在生长发育中发挥重要作用,线粒体基质蛋白及mtDNA通过融合进行交换与互补,降低突变型mtDNA的不良影响,延长细胞寿命,而线粒体融合受阻可导致细胞发育异常。此外,细胞内氧化磷酸化障碍、ROS过度产生、mtDNA缺失可抑制线粒体融合反应[2]。(2)线粒体分裂:在哺乳动物中,Drp-1和线粒体分裂蛋白1(mitochondrial fission protein1,Fis1)是参与线粒体分裂的重要分子。位于线粒体外膜的Fis1,可趋化胞质内Drp1移动至线粒体周围,两者在线粒体表面多聚化为螺旋状的Drp环,Drp环收缩时引起线粒体分裂。线粒体分裂有利于维持细胞生理活动,而线粒体过度分裂可诱导细胞凋亡[2],还可参与急、慢性肾损伤的发生。有研究发现,在急性肾损伤(acute kidney injury,AKI)小鼠模型的肾小管细胞及糖尿病小鼠模型的肾脏足细胞中,线粒体碎片明显增加,而特异性抑制细胞内线粒体分裂,可改善细胞凋亡及肾脏损伤[14-15]
4. mtDNA: 线粒体内具有独立的mtDNA,可单独完成复制、转录、合成蛋白质,故又称为半自主细胞器。人体mtDNA含37个基因,编码13种呼吸链复合体亚基、22种tRNA、2种rRNA。mtDNA受损及基因突变可引起线粒体功能障碍,导致多种临床疾病的发生[16]。此外,mtDNA与核基因组间存在信息交流,共同调控线粒体功能。研究发现,核基因编码的miRNA可进入线粒体调控mtDNA表达,影响线粒体结构及功能[17]
5. 线粒体与细胞凋亡: 细胞凋亡是由外源性及内源性信号诱导的细胞程序性死亡,以维持内环境稳定,其中内源性细胞凋亡途径由线粒体介导。当凋亡信号刺激时,线粒体膜的通透性发生改变,其通道转换孔开放可释放细胞色素c及凋亡因子等进入细胞质,激活各级胱天蛋白酶(caspase)并引发级联反应,介导caspase依赖途径的细胞凋亡;而线粒体膜间隙蛋白,例如凋亡诱导因子、核酸酶-核酸内切酶、丝氨酸蛋白酶Omi/HtrA2等,释放入细胞质后可转位至细胞核,通过非caspase依赖途径引起染色体浓缩、DNA片段断裂等,导致细胞凋亡[18]。参与调控线粒体途径的细胞凋亡机制较复杂,有研究发现miRNA可通过调节线粒体凋亡途径中的重要蛋白分子,参与调控细胞凋亡[19]
6. 线粒体自噬: 线粒体自噬是一种重要的线粒体质量控制机制,也是近年的研究热点。当细胞受到外界不良信号刺激时,细胞内的线粒体可发生去极化而引起损伤,同时细胞内还存在部分功能不完整的多余的线粒体;这类受损和多余的线粒体可被自噬泡特异性包裹形成自噬体,自噬体与溶酶体融合后,溶酶体酶可降解自噬体内受损及多余的线粒体,从而维持细胞内稳态,该过程称为线粒体自噬[20]。过度或不足的线粒体自噬与人类疾病的发生紧密相关,如癌症、神经退行性病变、心脏疾病、肝脏疾病、器官衰老等[20]。近年,miRNA在线粒体自噬中的作用研究日益增多,并有研究证实多种miRNA通过影响自噬相关分子NIX、PINK1、Parkin、FUNDC1等的表达,纠正几种重要信号通路对线粒体自噬的异常调控,发挥器官保护作用[21-22]

二、 线粒体miRNA的生物学特征

miRNA是一类由内源基因编码的高度保守的非编码RNA,通过干扰靶基因mRNA的翻译或稳定性来调节基因表达。人体中miRNA约2 000个,广泛分布于全身各组织器官,miRNA作为基因表达的重要调节剂,在多种细胞生物学过程及疾病进程中发挥重要作用。miRNA的前体(pre-miRNA)是长度约70个核苷酸且为茎环结构的非编码RNA小分子,经核糖核酸酶Ⅲ型Dicer酶及辅助因子剪切加工后,形成长度约19~25个核苷酸的成熟产物,成熟的单链miRNA分子与Argonaute2(AGO2)等蛋白组装成RNA诱导的沉默复合物,引起靶基因mRNA的降解并抑制其翻译[23]。有研究证实由核基因组编码的miRNA可进入细胞内多种细胞器中发挥调节作用,如内质网、核糖体、线粒体等[4]
研究者们把参与调控线粒体基因mRNA表达的miRNA命名为线粒体miRNA(mitomiR),如mmu-miR-1-3p、mmu-miR-378a-3p、has-miR-21-5p等。大部分mitomiR由核基因组编码,在细胞质内加工至成熟后,进入线粒体调节基因表达,生物信息学结果显示mtDNA中存在169个mitomiR的靶基因位点[4,17]。2011年Barrey等[17]发现mtDNA中存在25种pre-miRNA和33种潜在miRNA编码序列,如pre-let-7b、pre-miR-302a等,推测mtDNA也可直接编码mitomiR,在线粒体内加工成熟后直接作用于线粒体mRNA,调节线粒体功能。mitomiR可影响多种细胞内途径,如三羧酸循环、呼吸链传递、细胞内离子稳态、氧化应激、脂质及氨基酸代谢等,参与人体各类疾病的发生[4]。近年mitomiR在肾脏疾病中的调控作用得到关注,以下将总结mitomiR及其他参与调控线粒体能量代谢途径的miRNA在肾脏衰老、AKI、慢性肾脏病(chronic kidney disease,CKD)、多囊性肾病(polycystic kidney disease,PKD)中的作用研究。

三、 miRNA调控线粒体功能在肾脏衰老中的作用

随着年龄增长,肾脏呈现不同程度的生理和病理改变,器官功能逐渐衰退,出现肾脏衰老并影响机体健康,而肾脏衰老也是引起肾脏疾病的重要机制之一,延缓肾脏衰老可有效减少肾脏疾病的发生[24]。多种因素可导致细胞和组织衰老,如细胞程序性死亡、自由基损伤细胞、mtDNA突变等。早期研究发现,lin-4和lin-14通过调节胰岛素样生长因子1信号通路,影响秀丽隐杆线虫的衰老速度,提示miRNA可作为生物细胞发育及器官衰老的重要调节剂[25]。有研究者提出mtDNA突变在组织衰老中发挥重要作用,尤其对高能量需求的组织及器官产生衰老影响,而mitomiR参与调控mtDNA的表达,影响细胞能量代谢途径中重要蛋白的表达,可能在线粒体功能障碍所致的器官衰老中发挥调控作用[26]
近年Bai等[27]研究发现,与年幼大鼠相比,年老大鼠的肾脏组织出现偶发的局灶节段性肾小球硬化、肾间质纤维化、肾小管萎缩、炎性细胞浸润等衰老的病理改变,通过检测年幼及年老大鼠体内miRNA表达水平发现,18种参与调节细胞能量代谢、氧化应激、细胞增殖等途径的miRNA表达增高,如miR-335、miR-34a、miR-184、miR-224等,参与调节多个抗氧化相关靶基因表达及降低,如谷胱甘肽过氧化物酶(glutathione peroxidase,Gpx)、超氧化物歧化酶2(superoxide dismutase 2,SOD2)、硫氧还蛋白还原酶2(thioredoxin reductase 2,Txnrd2)、硫氧还蛋白样蛋白(thioredoxin-like protein 2,Txnl2)等;其中miR-335及miR-34a的表达水平在衰老的大鼠肾小球系膜细胞、肾小管上皮细胞、肾间质成纤维细胞中明显增高,故对其作用机制进行研究,发现在年老大鼠肾小球系膜细胞中,miR-335及miR-34a分别通过抑制SOD2和Txnrd2促进细胞衰老;反之,转染反义miR-335或miR-34a抑制剂后,年老大鼠肾小球系膜细胞中的SOD2及Txnrd2表达上调且ROS水平降低,细胞衰老状态得到改善,这提示miRNA可通过影响线粒体氧化应激过程调节肾脏的衰老。虽然目前关于miRNA在线粒体功能障碍所致的肾脏衰老中的机制研究不多,但为今后研究肾脏衰老提供了新的理论依据。

四、 miRNA调控线粒体功能在AKI中的作用

AKI是由缺血缺氧、感染、药物、脓毒症等因素引起的常见临床综合征,也是CKD发生和发展的重要危险因素。多项证据表明,线粒体功能损伤是诱发多种AKI及促进AKI进展至CKD的重要因素,改善线粒体稳态对恢复AKI后的肾功能具有潜在作用[28-30]。在急性缺血缺氧后,肾脏内皮细胞、足细胞、小管细胞中的线粒体可出现持续损伤,受损线粒体释放出的ROS、心磷脂等分子可激活炎性小体NLRP3,增强肾脏内炎性反应,促进肾纤维化的发生;AKI后尽早应用线粒体保护剂SS-31,可修复受损线粒体,抑制肾脏炎症和纤维化[28]。另有研究发现,在脓毒症早期,循环中mtDNA的水平升高,通过激活Toll样受体9(Toll-like receptor 9,TLR9)引起脓毒症AKI的发生,而在盲肠结扎诱导的脓毒症小鼠中,特异性敲除TLR9可逆转肾小管中线粒体的空泡化,减轻肾脏损伤的程度[30]。有临床研究表明,在脓毒症AKI患者血清中,差异表达的miRNA,如hsa-miR-23a-3p、hsa-miR-4456、hsa-miR-142-5p、hsa-miR-22-3p、hsa-miR-191-5p、miR-4270等,经GO及KEGG分析发现,大部分参与调控细胞代谢、细胞凋亡、线粒体氧化应激等信号途径,预测miRNA通过影响线粒体功能参与脓毒症AKI[31]。此外,有研究者对miRNA在AKI中调控线粒体功能的作用机制进行了研究。Wei等[32]研究发现,miR-668在缺血性AKI小鼠模型、肾小管细胞系及AKI患者肾组织中表达增高,在近端肾小管细胞中,缺氧诱导因子1(hypoxia-inducible factor-1,HIF-1)可诱导miR-668表达增加,从而抑制线粒体分裂蛋白MTP18,维持线粒体动力学稳定,从而阻止肾脏损伤。我们的研究发现[33]在顺铂诱导的AKI小鼠模型及AKI患者肾组织中,miR-709在近端肾小管细胞中表达增高;在小鼠肾小管细胞中过表达miR-709,可抑制线粒体转录因子A,引起线粒体功能障碍,导致细胞死亡,促进AKI的发生发展;而转染miR-709抑制剂,可阻止小鼠肾小管细胞mtDNA拷贝数减少与ROS产生,抑制细胞凋亡。

五、 miRNA调控线粒体功能在CKD中的作用

CKD是多种因素引起的肾脏结构及功能的长期不可逆损伤,进展至终末期可出现肾脏纤维化[34]。线粒体功能障碍是引起CKD的重要机制之一[35],并且氧化呼吸链障碍及ROS过度产生在CKD疾病进程中持续存在[34]。生理状态下肾脏内富含多种miRNA,如miR-192、miR-214、miR-204、miR-200等,在各类原发性和继发性CKD中,部分异常表达的肾特异性miRNA通过调控TGF-β/Smad、NF-κB、PI3K/Akt、Wnt/β-catenin等关键信号通路,参与肾小球硬化、肾小管间质纤维化的发生与发展[36]。Gomez等[37]发现在CKD患者肾组织、单侧输尿管梗阻(unilated ureteral obstruction,UUO)及缺血/再灌注(ischemia/reperfusion,I/R)引发的肾纤维化动物模型中,有24种miRNA表达明显增高,如miR-15b、miR-21、miR-199、miR-214、miR-25等;其中,miR-21及miR-214被发现可通过调节线粒体代谢途径中重要蛋白及线粒体基因的表达,影响线粒体相关功能,参与肾纤维化的进展[38-39]。以下将介绍数个通过调控线粒体功能在CKD中发挥作用的miRNA。
1. miR-214: 人体中miR-214位于染色体1q24.3臂上dynamin3基因的14号内含子内,具有多靶点特点,参与调节机体生物学过程,如细胞增殖、细胞凋亡、炎性反应、细胞代谢及氧化应激等[40]。miR-214在CKD患者的肾皮质中表达丰富,且被证实可作为肾脏内源性线粒体调节剂,靶向调控线粒体基因mt-Nd6mt-Nd4l,影响线粒体氧化磷酸化,参与CKD的发生[39]。我们研究发现[39],在肾小管上皮细胞中miR-214主要定位于线粒体内,尤其在CKD患者肾组织中表达显著增加。过表达miR-214可显著降低肾小管上皮细胞中mtDNA拷贝数、线粒体呼吸链复合酶活性,促进ROS产生及细胞凋亡;而在白蛋白超负荷、单侧I/R、UUO诱导的3种CKD模型中特异性敲除肾小管中的miR-214,可改善线粒体功能,减轻细胞凋亡、炎症及纤维化,发挥保护肾脏的作用。
2. miR-21: miR-21是一种广泛存在于全身各组织的miRNA,在正常肾脏组织中表达丰富。近期研究发现,肾脏上皮细胞中脂质及脂肪酸代谢产生的ATP是肾组织主要的能量来源,用于维持细胞的活性及生理功能[41]。Chau等[38]研究发现,在UUO及单侧I/R诱导的CKD小鼠模型中miR-21表达升高,过氧化物酶体增殖物激活受体α(peroxisome proliferators-activated receptorsα,PPARα)及MPV17L蛋白表达下降;通过检测发现,在过表达miR-21的CKD小鼠肾组织中,凋亡细胞、成纤维细胞、上皮细胞损伤标志物Kim1显著增高,而特异性敲除CKD小鼠体内的miR-21可抑制肾脏上皮胞外信号调节激酶(extracellular signal-regulated kinase,ERK)的激活,减弱ERK/MAPK信号通路调控的细胞活化,还可靶向激活PPARα介导的脂肪酸氧化过程,同时通过上调MPV17L的表达,减少ROS产生,从而减轻肾脏上皮细胞损伤及肾脏纤维化,发挥肾脏保护作用。
3. miR-9-5p: 既往miR-9-5p的相关研究多集中于肿瘤疾病,如参与调节癌细胞增殖、迁移及侵袭过程,促进或抑制癌症进展[42-43],仅少数研究与器官纤维化等慢性疾病有关,如肺纤维化、肾纤维化、腹膜纤维化等[44]。肾纤维化是多种因素所致的CKD的共同病理基础,寻找有效治疗靶点来改善肾纤维化,对延缓CKD患者的疾病进展至关重要。近年Fierro-Fernández等[45]研究发现,在UUO诱导的肾纤维化小鼠模型中过表达miR-9-5p,可抑制肾小管上皮细胞凋亡,减轻肾小管萎缩和细胞外基质的沉积,从而改善肾脏纤维化;经GO分析发现,miR-9-5p可阻止UUO模型中参与调节线粒体能量代谢途径基因的表达下调,如Cox5bATP5c1Ndufa9CPT1a等,并抑制促纤维化基因的表达,如ACTA2Col1α1FN-1TGF-β1等,提示miR-9-5p可能通过影响UUO诱导的线粒体代谢途径改变,发挥抗纤维化作用;此外,在TGF-β1诱导的肾小管上皮细胞(HKC-8细胞)中观察到线粒体呼吸链、糖酵解及ATP产生发生障碍,而过表达miR-9-5p则可改善能量代谢紊乱,维持细胞活性状态,这进一步提示miR-9-5p通过调节线粒体能量代谢发挥肾脏保护作用。
4. miR-30e: miR-30家族在肾脏中含量丰富,参与维持肾脏足细胞的稳态[46],我们的研究发现[47],miR-30e在醛固酮诱导的足细胞损伤模型中表达明显下降,而过表达miR-30e可通过抑制促细胞凋亡蛋白BNIP3L,改善足细胞线粒体内mtDNA拷贝数下降及ROS过度产生,减轻足细胞损伤。在另一项研究中,Jiang等[48]研究发现miR-30e在UUO诱导的CKD小鼠模型及TGF-β1诱导的肾小管上皮细胞(NRK-52E细胞)中表达下降,而解耦联蛋白2(uncoupling protein 2,UCP2)表达升高,UCP2是位于线粒体内膜的质子转运蛋白,当其被激活可引起质子的渗漏,导致氧化磷酸化解耦联,减少ATP合成和ROS产生;在TGF-β1诱导的NRK-52E细胞中过表达miR-30e,可通过抑制UCP2表达,减轻TGF-β1引起的肾小管细胞外基质沉积,改善肾脏纤维化,但目前UCP2在TGF-β1诱导的肾纤维化中的具体作用机制尚不明确,有待进一步研究。

六、 miRNA调控线粒体功能在PKD中的作用

PKD是一种遗传性的肾脏囊性疾病,包括常染色体显性多囊肾病(autosomal dominant polycystic kidney disease,ADPKD)和常染色体隐性多囊肾病。ADPKD是引起终末期肾衰竭最常见的遗传性病因,其发病原因多与PKD1PKD2基因突变有关,该病表现为双侧肾脏出现多个大小不一且进行性增大的囊肿,导致肾脏结构和功能损害,最终引起肾衰竭[49]
Hajarnis等[50]研究发现,在PKD1PKD2基因敲除诱导的ADPKD小鼠模型中,部分miRNA异常表达,其中miR-17的表达量明显增高,且通过抑制PPARα表达,干扰线粒体氧化磷酸化,促进囊肿生长,从而加重肾脏损伤;反之,特异性敲除两种PKD小鼠模型中的miR-17,均可抑制囊肿增殖,延缓疾病进展;应用miR-17探针检测健康人及ADPKD患者的肾组织发现,miR-17在ADPKD患者的肾脏囊肿上皮中表达增高,而miR-17抑制剂可呈剂量依赖性地抑制体外培养的人类ADPKD囊肿原代细胞增长。因此,miR-17抑制剂有望通过改善线粒体功能为ADPKD患者提供新的治疗方向。

七、 miRNA调控线粒体功能在肾细胞癌(renal cell carcinoma,RCC)中的作用

RCC是起源于肾小管上皮的恶性肿瘤,占肾脏恶性肿瘤的90%,也是引起全球癌症患者死亡的主要原因之一[51]。有研究发现miRNA的异常表达与癌症的发生、侵袭、转移有关,研究者们把下调抑癌蛋白并参与癌细胞的细胞周期、凋亡、DNA损伤等过程的miRNA统称为癌基因miRNA(OncomiRNA)。近年来多项研究分析了OncomiRNA与RCC间的联系,对比健康志愿者及RCC患者血清中的miRNA后发现,miR-21、miR-193a、miR-210、miR-221、miR-362、miR-378、miR-1233等几十余种miRNA的表达明显增高,其中miR-210、miR-378、miR-1233可被当作RCC早期诊断的生物标志物[52]。此外,miR-19a、miR-183、miR-203、miR-486、miR-708等多种OncomiRNA通过影响与RCC发生发展相关的重要分子,如PTEN、HIF、VHL、TGF-β、mTOR、GSK-3β、DKK-3、BMI1等,促进肿瘤细胞的增殖及侵袭、抑制癌细胞凋亡[51-53]
肿瘤细胞快速增殖是大量耗能的过程,所以肿瘤细胞能量代谢依赖于线粒体糖酵解及氧化磷酸化,为其提供大量能量,以适应体内微环境,促进肿瘤细胞的生长、增殖、扩散[54]。与核DNA相比,mtDNA因缺乏保护性组蛋白及修复蛋白,更易出现突变;有研究发现几乎在全部肾脏、卵巢、乳腺、结直肠、胰腺肿瘤中都可观察到突变的mtDNA,这一发现提示mtDNA突变可能参与RCC的发生和发展[55]。目前聚焦于肿瘤细胞线粒体能量代谢及mtDNA突变的研究较多,也有少量研究提出miRNA在RCC能量代谢中的作用。研究发现miR-494可能靶向自噬蛋白LC3B增加细胞中脂滴的累积,并通过募集Drp-1抑制PINK表达,导致线粒体分裂,从而减弱769-P肾癌细胞系的生存活力[56];此前有证据表明miR-210在肾透明细胞癌中表达异常增高,一项最新研究发现,近端肾小管中的miR-210可靶向下调IscuNdufa4基因,激活无氧糖酵解和戊糖磷酸途径,利于新细胞的生长及增殖,提示miR-210可能在肾透明细胞癌早期通过影响线粒体能量代谢变化,促进癌细胞的快速生长[57]

八、 总结与展望

如前所述,miR-709[33]、miR-21[38]、miR-214[39]、miR-17[50]、miR-335、miR-34a[27]通过介导线粒体功能障碍或抑制ROS的清除,参与肾脏疾病及肾脏衰老;miR-668[32]、miR-9-5p[44]、miR-30e[47]通过改善细胞内能量代谢,发挥保护肾脏的作用;miR-494[56]、miR-210[57]则参与RCC中线粒体能量代谢改变,促进肿瘤细胞的生长及增殖。虽然miRNA在肾脏疾病中可靶向线粒体代谢途径发挥调控作用,但是应用于肾脏疾病的诊断与治疗仍存在一定局限性;基于细胞及动物水平的研究,部分miRNA在肾脏衰老、急慢性肾损伤、肾脏纤维化模型的肾组织中呈差异性表达,有望成为一种生物诊断标志物,但是肾活检是一种侵入性检查,在肾脏病早期阶段应用不广泛,故miRNA用于临床诊断的适用范围较局限。此外,诸多肾脏疾病的发生与线粒体功能障碍紧密相关,而mitomiR及其他miRNA可分别参与调节线粒体基因组及线粒体能量代谢途径中的重要蛋白的表达,影响细胞内代谢、氧化应激、氧化磷酸化等过程,干预疾病的发生发展,因此miRNA调控线粒体功能可为肾脏疾病的治疗提供新方向。
目前,miRNA调控线粒体功能在肾脏疾病中的作用研究仅停留于实验室水平,在顺利进入临床研究前,仍需克服一系列特异性及安全性问题;miRNA广泛分布于全身各器官,通过多种信号通路参与调节细胞凋亡、细胞自噬、炎性反应、氧化应激、上皮-间充质转化等生物学过程,若有研究可证实在肾脏病动物模型中,调节某种miRNA可特异地作用于细胞代谢途径,改善肾脏中的线粒体功能及整体肾功能,而不影响其他信号通路、细胞器及器官的正常功能,才有望进入临床研究。
综上所述,线粒体功能障碍是肾脏疾病的重要发病机制之一,miRNA可通过调控线粒体功能,影响细胞代谢的稳态,从而参与肾脏衰老、急慢性肾脏病、RCC的发生发展。目前miRNA调控线粒体功能在肾脏病中作用的研究仍处于起步阶段,还需进一步探究miRNA与线粒体功能障碍之间的具体调节机制,以及其在肾脏病中的作用,为临床中肾脏疾病的诊治提供新思路,为攻克难治性肾病带来希望。

References

[1]
Wang Z, Ying Z, Bosy-Westphal A, et al. Specific metabolic rates of major organs and tissues across adulthood: evaluation by mechanistic model of resting energy expenditure[J]. Am J Clin Nutr, 2010, 92(6): 1369-1377. DOI: 10.3945/ajcn.2010.29885.
The specific resting metabolic rates (K(i); in kcal · kg(-1 )· d(-1)) of major organs and tissues in adults were suggested by Elia (in Energy metabolism: tissue determinants and cellular corollaries. New York, NY: Raven Press, 1992) to be as follows: 200 for liver, 240 for brain, 440 for heart and kidneys, 13 for skeletal muscle, 4.5 for adipose tissue, and 12 for residual organs and tissues. However, Elia's K(i) values have never been fully evaluated.The objectives of the present study were to evaluate the applicability of Elia's K(i) values across adulthood and to explore the potential influence of age on the K(i) values.A new approach was developed to evaluate the K(i) values of major organs and tissues on the basis of a mechanistic model: REE = Σ(K(i) × T(i)), where REE is whole-body resting energy expenditure measured by indirect calorimetry, and T(i) is the mass of individual organs and tissues measured by magnetic resonance imaging. With measured REE and T(i), marginal 95% CIs for K(i) values were calculated by stepwise univariate regression analysis. An existing database of nonobese, healthy adults [n = 131; body mass index (in kg/m²) <30] was divided into 3 age groups: 21-30 y (young, n = 43), 31-50 y (middle-age, n = 51), and > 50 y (n = 37).Elia's K(i) values were within the range of 95% CIs in the young and middle-age groups. However, Elia's K(i) values were outside the right boundaries of 95% CIs in the >50-y group, which indicated that Elia's study overestimated K(i) values by 3% in this group. Age-adjusted K(i) values for adults aged >50 y were 194 for liver, 233 for brain, 426 for heart and kidneys, 12.6 for skeletal muscle, 4.4 for adipose tissue, and 11.6 for residuals.The general applicability of Elia's K(i) values was validated across adulthood, although age adjustment is appropriate for specific applications.
[2]
Tilokani L, Nagashima S, Paupe V, et al. Mitochondrial dynamics: overview of molecular mechanisms[J]. Essays Biochem, 2018, 62(3): 341-360. DOI: 10.1042/EBC20170104.
Mitochondria are highly dynamic organelles undergoing coordinated cycles of fission and fusion, referred as 'mitochondrial dynamics', in order to maintain their shape, distribution and size. Their transient and rapid morphological adaptations are crucial for many cellular processes such as cell cycle, immunity, apoptosis and mitochondrial quality control. Mutations in the core machinery components and defects in mitochondrial dynamics have been associated with numerous human diseases. These dynamic transitions are mainly ensured by large GTPases belonging to the Dynamin family. Mitochondrial fission is a multi-step process allowing the division of one mitochondrion in two daughter mitochondria. It is regulated by the recruitment of the GTPase Dynamin-related protein 1 (Drp1) by adaptors at actin- and endoplasmic reticulum-mediated mitochondrial constriction sites. Drp1 oligomerization followed by mitochondrial constriction leads to the recruitment of Dynamin 2 to terminate membrane scission. Inner mitochondrial membrane constriction has been proposed to be an independent process regulated by calcium influx. Mitochondrial fusion is driven by a two-step process with the outer mitochondrial membrane fusion mediated by mitofusins 1 and 2 followed by inner membrane fusion, mediated by optic atrophy 1. In addition to the role of membrane lipid composition, several members of the machinery can undergo post-translational modifications modulating these processes. Understanding the molecular mechanisms controlling mitochondrial dynamics is crucial to decipher how mitochondrial shape meets the function and to increase the knowledge on the molecular basis of diseases associated with morphology defects. This article will describe an overview of the molecular mechanisms that govern mitochondrial fission and fusion in mammals.© 2018 The Author(s).
[3]
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.
[4]
Macgregor-Das AM, Das S. A microRNA's journey to the center of the mitochondria[J]. Am J Physiol Heart Circ Physiol, 2018, 315(2): H206-H215. DOI: 10.1152/ajpheart.00714.2017.
MicroRNAs (miRNAs) are known as the master regulators of gene expression, and for the last two decades our knowledge of their functional reach keeps expanding. Recent studies have shown that a miRNA’s role in regulation extends to extracellular and intracellular organelles. Several studies have shown a role for miRNA in regulating the mitochondrial genome in normal and disease conditions. Mitochondrial dysfunction occurs in many human pathologies, such as cardiovascular disease, diabetes, cancer, and neurological diseases. These studies have shed some light on regulation of the mitochondrial genome as well as helped to explain the role of miRNA in altering mitochondrial function and the ensuing effects on cells. Although the field has grown in recent years, many questions still remain. For example, little is known about how nuclear-encoded miRNAs translocate to the mitochondrial matrix. Knowledge of the mechanisms of miRNA transport into the mitochondrial matrix is likely to provide important insights into our understanding of disease pathophysiology and could represent new targets for therapeutic intervention. For this review, our focus will be on the role of a subset of miRNAs, known as MitomiR, in mitochondrial function. We also discuss the potential mechanisms used by these nuclear-encoded miRNAs for import into the mitochondrial compartment.
[5]
Larosa V, Remacle C. Insights into the respiratory chain and oxidative stress[J]. Biosci Rep, 2018, 38(5): BSR20171492. DOI: 10.1042/BSR20171492.
Reactive oxygen species (ROS) are highly reactive reduced oxygen molecules that result from aerobic metabolism. The common forms are the superoxide anion (O2∙−) and hydrogen peroxide (H2O2) and their derived forms, hydroxyl radical (HO∙) and hydroperoxyl radical (HOO∙). Their production sites in mitochondria are reviewed. Even though being highly toxic products, ROS seem important in transducing information from dysfunctional mitochondria. Evidences of signal transduction mediated by ROS in mitochondrial deficiency contexts are then presented in different organisms such as yeast, mammals or photosynthetic organisms.
[6]
Van der Bliek AM, Sedensky MM, Morgan PG. Cell biology of the mitochondrion[J]. Genetics, 2017, 207(3): 843-871. DOI: 10.1534/genetics.117.300262.
Mitochondria are best known for harboring pathways involved in ATP synthesis through the tricarboxylic acid cycle and oxidative phosphorylation. Major advances in understanding these roles were made with mutants affecting key components of the metabolic pathways. These mutants have not only helped elucidate some of the intricacies of metabolism pathways, but they have also served as jumping off points for pharmacology, toxicology, and aging studies. The field of mitochondria research has also undergone a renaissance, with the increased appreciation of the role of mitochondria in cell processes other than energy production. Here, we focus on discoveries that were made using, with a few excursions into areas that were studied more thoroughly in other organisms, like mitochondrial protein import in yeast. Advances in mitochondrial biogenesis and membrane dynamics were made through the discoveries of novel functions in mitochondrial fission and fusion proteins. Some of these functions were only apparent through the use of diverse model systems, such as Studies of stress responses, exemplified by mitophagy and the mitochondrial unfolded protein response, have also benefitted greatly from the use of model organisms. Recent developments include the discoveries in of cell autonomous and nonautonomous pathways controlling the mitochondrial unfolded protein response, as well as mechanisms for degradation of paternal mitochondria after fertilization. The evolutionary conservation of many, if not all, of these pathways ensures that results obtained with are equally applicable to studies of human mitochondria in health and disease.Copyright © 2017 by the Genetics Society of America.
[7]
Supinski GS, Schroder EA, Callahan LA. Mitochondria and critical illness[J]. Chest, 2020, 157(2): 310-322. DOI: 10.1016/j.chest.2019.08.2182.
Classically, mitochondria have largely been believed to influence the development of illness by modulating cell metabolism and determining the rate of production of high-energy phosphate compounds (eg, adenosine triphosphate). It is now recognized that this view is simplistic and that mitochondria play key roles in many other processes, including cell signaling, regulating gene expression, modulating cellular calcium levels, and influencing the activation of cell death pathways (eg, caspase activation). Moreover, these multiple mitochondrial functional characteristics are now known to influence the evolution of cellular and organ function in many disease states, including sepsis, ICU-acquired skeletal muscle dysfunction, acute lung injury, acute renal failure, and critical illness-related immune function dysregulation. In addition, diseased mitochondria generate toxic compounds, most notably released mitochondrial DNA, which can act as danger-associated molecular patterns to induce systemic toxicity and damage multiple organs throughout the body. This article reviews these evolving concepts relating mitochondrial function and acute illness. The discussion is organized into four sections: (1) basics of mitochondrial physiology; (2) cellular mechanisms of mitochondrial pathophysiology; (3) critical care disease processes whose initiation and evolution are shaped by mitochondrial pathophysiology; and (4) emerging treatments for mitochondrial dysfunction in critical illness.Copyright © 2019 American College of Chest Physicians. Published by Elsevier Inc. All rights reserved.
[8]
Lackner LL. The expanding and unexpected functions of mitochondria contact sites[J]. Trends Cell Biol, 2019, 29(7): 580-590. DOI: 10.1016/j.tcb.2019.02.009.
Mitochondria make functionally relevant contacts with most, if not all, other organelles in the cell. These contacts impact on mitochondrial behavior and function as well as on a wide variety of cellular functions. Many recent advances have been made in the rapidly growing field of mitochondria contact site biology, and these advances have expanded the known functions of mitochondria contact sites in exciting and unexpected ways.Copyright © 2019 Elsevier Ltd. All rights reserved.
[9]
Wong YC, Ysselstein D, Krainc D. Mitochondria-lysosome contacts regulate mitochondrial fission via RAB7 GTP hydrolysis[J]. Nature, 2018, 554(7692): 382-386. DOI: 10.1038/nature25486.
[10]
Wong YC, Kim S, Peng W, et al. Regulation and function of mitochondria-lysosome membrane contact sites in cellular homeostasis[J]. Trends Cell Biol, 2019, 29(6): 500-513. DOI: 10.1016/j.tcb.2019.02.004.
Mitochondrial and lysosomal function are intricately related and critical for maintaining cellular homeostasis, as highlighted by multiple diseases linked to dysfunction of both organelles. Recent work using high-resolution microscopy demonstrates the dynamic formation of inter-organelle membrane contact sites between mitochondria and lysosomes, allowing for their direct interaction in a pathway distinct from mitophagy or lysosomal degradation of mitochondrial-derived vesicles. Mitochondria-lysosome contact site tethering is mechanistically regulated by mitochondrial proteins promoting Rab7 GTP hydrolysis, and allows for the bidirectional crosstalk between mitochondria and lysosomes and the regulation of their organelle network dynamics, including mitochondrial fission. In this review, we summarize recent advances in mitochondria-lysosome contact site regulation and function, and discuss their potential roles in cellular homeostasis and various human diseases.Copyright © 2019 Elsevier Ltd. All rights reserved.
[11]
Lewis SC, Uchiyama LF, Nunnari J. ER-mitochondria contacts couple mtDNA synthesis with mitochondrial division in human cells[J]. Science, 2016, 353(6296): aaf5549. DOI: 10.1126/science.aaf5549.
\n It has been unclear how mitochondrial DNA (mtDNA) replication is spatially controlled in mammalian cells and how the mitochondrial nucleoid—the protein-DNA structure that is the unit of mtDNA inheritance—is distributed at the cellular level. Lewis\n et al.\n now show that homeostatic mtDNA synthesis in mitochondrial nucleoids in mammalian cells is spatially linked to a small subset of endoplasmic reticulum (ER)-mitochondria contact sites that are specifically destined for mitochondrial division. Successive events of mtDNA replication, mitochondrial division, and mitochondrial motility function together to ensure the accurate distribution of mtDNA in cells. Furthermore, ER-mitochondria contacts coordinate the licensing of mtDNA replication with division to distribute newly replicated nucleoids to daughter mitochondria.\n
[12]
Szymański J, Janikiewicz J, Michalska B, et al. Interaction of mitochondria with the endoplasmic reticulum and plasma membrane in calcium homeostasis, lipid trafficking and mitochondrial structure[J]. Int J Mol Sci, 2017, 18(7): 1576. DOI: 10.3390/ijms18071576.
[13]
Fransen M, Lismont C, Walton P. The peroxisome-mitochondria connection: how and why?[J]. Int J Mol Sci, 2017, 18(6): 1126. DOI: 10.3390/ijms18061126.
[14]
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.
[15]
Ayanga BA, Badal SS, Wang Y, et al. Dynamin-related protein 1 deficiency improves mitochondrial fitness and protects against progression of diabetic nephropathy[J]. J Am Soc Nephrol, 2016, 27(9): 2733-2747. DOI: 10.1681/ASN.2015101096.
Mitochondrial fission has been linked to the pathogenesis of diabetic nephropathy (DN). However, how mitochondrial fission affects progression of DN in vivo is unknown. Here, we report the effect of conditional podocyte-specific deletion of dynamin-related protein 1 (Drp1), an essential component of mitochondrial fission, on the pathogenesis and progression of DN. Inducible podocyte-specific deletion of Drp1 in diabetic mice decreased albuminuria and improved mesangial matrix expansion and podocyte morphology. Ultrastructure analysis revealed a significant increase in fragmented mitochondria in the podocytes of wild-type diabetic mice but a marked improvement in mitochondrial structure in Drp1-null podocytes of diabetic mice. When isolated from diabetic mice and cultured in high glucose, Drp1-null podocytes had more elongated mitochondria and better mitochondrial fitness associated with enhanced oxygen consumption and ATP production than wild-type podocytes. Furthermore, administration of a pharmacologic inhibitor of Drp1, Mdivi1, significantly blunted mitochondrial fission and rescued key pathologic features of DN in mice. Taken together, these results provide novel correlations between mitochondrial morphology and the progression of DN and point to Drp1 as a potential therapeutic target in DN.Copyright © 2016 by the American Society of Nephrology.
[16]
Neupert W, Herrmann JM. Translocation of proteins into mitochondria[J]. Annu Rev Biochem, 2007, 76: 723-749. DOI: 10.1146/annurev.biochem.76.052705.163409.
About 10% to 15% of the nuclear genes of eukaryotic organisms encode mitochondrial proteins. These proteins are synthesized in the cytosol and recognized by receptors on the surface of mitochondria. Translocases in the outer and inner membrane of mitochondria mediate the import and intramitochondrial sorting of these proteins; ATP and the membrane potential are used as energy sources. Chaperones and auxiliary factors assist in the folding and assembly of mitochondrial proteins into their native, three-dimensional structures. This review summarizes the present knowledge on the import and sorting of mitochondrial precursor proteins, with a special emphasis on unresolved questions and topics of current research.
[17]
Barrey E, Saint-Auret G, Bonnamy B, et al. Pre-microRNA and mature microRNA in human mitochondria[J]. PLoS One, 2011, 6(5): e20220. DOI: 10.1371/journal.pone.0020220.
[18]
McManus MJ, Murphy MP, Franklin JL. Mitochondria-derived reactive oxygen species mediate caspase-dependent and -independent neuronal deaths[J]. Mol Cell Neurosci, 2014, 63: 13-23. DOI: 10.1016/j.mcn.2014.09.002.
Mitochondrial dysfunction and oxidative stress are implicated in many neurodegenerative diseases. Mitochondria-targeted drugs that effectively decrease oxidative stress, protect mitochondrial energetics, and prevent neuronal loss may therefore lend therapeutic benefit to these currently incurable diseases. To investigate the efficacy of such drugs, we examined the effects of mitochondria-targeted antioxidants MitoQ10 and MitoE2 on neuronal death induced by neurotrophin deficiency. Our results indicate that MitoQ10 blocked apoptosis by preventing increased mitochondria-derived reactive oxygen species (ROS) and subsequent cytochrome c release, caspase activation, and mitochondrial damage in nerve growth factor (NGF)-deprived sympathetic neurons, while MitoE2 was largely ineffective. In this paradigm, the most proximal point of divergence was the ability of MitoQ10 to scavenge mitochondrial superoxide (O2(-)). MitoQ10 also prevented caspase-independent neuronal death in these cells demonstrating that the mitochondrial redox state significantly influences both apoptotic and nonapoptotic pathways leading to neuronal death. We suggest that mitochondria-targeted antioxidants may provide tools for delineating the role and significance of mitochondrial ROS in neuronal death and provide a new therapeutic approach for neurodegenerative conditions involving trophic factor deficits and multiple modes of cell death. Copyright © 2014 Elsevier Inc. All rights reserved.
[19]
杨莹, 范秋灵, 李露露, 等. miRNA-148b靶向AMPKα1通过氧化应激介导高糖诱导的人肾小管上皮细胞凋亡[J]. 中华肾脏病杂志, 2019, 35(1): 43-47. DOI: 10.3760/cma.j.issn.1001-7097.2019.01.007.
目的 探讨微小RNA-148b(miRNA-148b)在高糖诱导肾小管损伤中表达变化及其作用机制。 方法 体外培养人肾小管上皮细胞(HK-2细胞),分为正常糖组、甘露醇高渗对照组、高糖组,培养48 h后,实时定量PCR法检测miRNA-148b表达;采用2',7'-二氯二氢荧光素二乙酸酯(DCFH-DA)在荧光显微镜下检测细胞内活性氧(ROS)水平;Western印迹检测HK-2细胞腺苷单磷酸活化蛋白激酶α1(AMPKα1)、NOX2、NOX4、Bcl-2、cleaved-caspase3的蛋白表达。 结果 培养48 h后,与正常糖组相比,高糖组、高渗组HK-2细胞内miRNA-148b表达上调(P<0.01),ROS产生增多(P<0.01),NOX2、NOX4蛋白表达增多(均P<0.01),AMPKα1蛋白和抗凋亡蛋白Bcl-2蛋白表达减少(均P<0.01),线粒体凋亡通路相关蛋白cleaved-caspase3蛋白表达增加(P<0.01),差异均有统计学意义。 结论 高糖上调体外培养HK-2细胞miRNA-148b的表达,靶向抑制AMPKα1的表达,促进NOX2、NOX4表达,活性氧产生增多,活化线粒体凋亡途径,激活caspase酶,诱导HK-2细胞凋亡。高糖的肾小管毒性部分是渗透压的影响。miRNA-148b可能参与了糖尿病肾病病理损伤的发生,有望成为糖尿病肾病新的治疗靶点。
[20]
Um JH, Yun J. Emerging role of mitophagy in human diseases and physiology[J]. BMB Rep, 2017, 50(6): 299-307. DOI: 10.5483/bmbrep.2017.50.6.056.
[21]
Li W, Zhang X, Zhuang H, et al. MicroRNA-137 is a novel hypoxia-responsive microRNA that inhibits mitophagy via regulation of two mitophagy receptors FUNDC1 and NIX[J]. J Biol Chem, 2014, 289(15): 10691-10701. DOI: 10.1074/jbc.M113.537050.
Mitophagy receptors mediate the selective recognition and targeting of damaged mitochondria by autophagosomes. The mechanism for the regulation of these receptors remains unknown. Here, we demonstrated that a novel hypoxia-responsive microRNA, microRNA-137 (miR-137), markedly inhibits mitochondrial degradation by autophagy without affecting global autophagy. miR-137 targets the expression of two mitophagy receptors NIX and FUNDC1. Impaired mitophagy in response to hypoxia caused by miR-137 is reversed by re-expression of FUNDC1 and NIX expression vectors lacking the miR-137 recognition sites at their 3' UTR. Conversely, miR-137 also suppresses the mitophagy induced by fundc1 (CDS+3'UTR) but not fundc1 (CDS) overexpression. Finally, we found that miR-137 inhibits mitophagy by reducing the expression of the mitophagy receptor thereby leads to inadequate interaction between mitophagy receptor and LC3. Our results demonstrated the regulatory role of miRNA to mitophagy receptors and revealed a novel link between miR-137 and mitophagy.
[22]
Kim J, Fiesel FC, Belmonte KC, et al. miR-27a and miR-27b regulate autophagic clearance of damaged mitochondria by targeting PTEN-induced putative kinase 1 (PINK1)[J]. Mol Neurodegener, 2016, 11(1): 55. DOI: 10.1186/s13024-016-0121-4.
Background: Loss-of-function mutations in PINK1 and PARKIN are the most common causes of autosomal recessive Parkinson's disease (PD). PINK1 is a mitochondrial serine/threonine kinase that plays a critical role in mitophagy, a selective autophagic clearance of damaged mitochondria. Accumulating evidence suggests mitochondrial dysfunction is one of central mechanisms underlying PD pathogenesis. Therefore, identifying regulatory mechanisms of PINK1 expression may provide novel therapeutic opportunities for PD. Although post-translational stabilization of PINK1 upon mitochondrial damage has been extensively studied, little is known about the regulation mechanism of PINK1 at the transcriptional or translational levels. Results: Here, we demonstrated that microRNA-27a (miR-27a) and miR-27b suppress PINK1 expression at the translational level through directly binding to the 3'-untranslated region (3'UTR) of its mRNA. Importantly, our data demonstrated that translation of PINK1 is critical for its accumulation upon mitochondrial damage. The accumulation of PINK1 upon mitochondrial damage was strongly regulated by expression levels of miR-27a and miR-27b. miR-27a and miR-27b prevent mitophagic influx by suppressing PINK1 expression, as evidenced by the decrease of ubiquitin phosphorylation, Parkin translocation, and LC3-II accumulation in damaged mitochondria. Consequently, miR-27a and miR-27b inhibit lysosomal degradation of the damaged mitochondria, as shown by the decrease of the delivery of damaged mitochondria to lysosome and the degradation of cytochrome c oxidase 2 (COX2), a mitochondrial marker. Furthermore, our data demonstrated that the expression of miR-27a and miR-27b is significantly induced under chronic mitophagic flux, suggesting a negative feedback regulation between PINK1-mediated mitophagy and miR-27a and miR-27b. Conclusions: We demonstrated that miR-27a and miR-27b regulate PINK1 expression and autophagic clearance of damaged mitochondria. Our data further support a novel negative regulatory mechanism of PINK1-mediated mitophagy by miR-27a and miR-27b. Therefore, our results considerably advance our understanding of PINK1 expression and mitophagy regulation and suggest that miR-27a and miR-27b may represent potential therapeutic targets for PD.
[23]
Fan PC, Chen CC, Chen YC, et al. MicroRNAs in acute kidney injury[J]. Hum Genomics, 2016, 10(1): 29. DOI: 10.1186/s40246-016-0085-z.
[24]
Melk A, Mansfield ES, Hsieh SC, et al. Transcriptional analysis of the molecular basis of human kidney aging using cDNA microarray profiling[J]. Kidney Int, 2005, 68(6): 2667-2679. DOI: 10.1111/j.1523-1755.2005.00738.x.
The molecular basis of renal aging is not completely understood.We used global gene expression monitoring by cDNA microarrays to identify age associated genes in human kidney samples. Our samples included young (8 weeks-8 years, N= 4), adult (31-46 years, N= 7), and old kidneys (71-88 years, N= 9).Old kidneys had more glomerulosclerosis, tubular atrophy, interstitial fibrosis, and fibrous intimal thickening in small arteries. We identified approximately 500 genes that were differentially expressed among the three age groups. Old kidneys appeared to have increased extracellular matrix turnover and a nonspecific inflammatory response, combined with a reduction in processes dependent on energy metabolism and mitochondrial function. Quantitative supervised bioinformatics analyses of adult and old kidney expression data correlated the expression of 255 gene profiles with renal pathology scores. Microarray class prediction analysis (PAM) identified 50 unique genes that segregated old kidneys into two distinct clusters: those more similar within age class (OO, N= 5) versus old kidneys more similar to adult kidneys (OA, N= 4). The expression of six functionally significant genes was further validated by quantitative reverse transcription-polymerase chain reaction (RT-PCR) (FN1, MMP7, TNC, SERPIN3A, BPHL, CSPG2) in the experiment group and, subsequently, confirmed independently in 17 additional old and adult age-stratified test kidney samples. The p53 inducible gene, CSPG2, performed best in separating OO kidneys from adults and OA samples in this analysis.The method described in this study using independent validation samples can be envisioned to test utility of the identified genes in assessing age-related changes that contribute to decline in renal function.
[25]
Boehm M, Slack F. A developmental timing microRNA and its target regulate life span in C. elegans[J]. Science, 2005, 310(5756): 1954-1957. DOI: 10.1126/science.1115596.
The microRNA lin-4 and its target, the putative transcription factor lin-14, control the timing of larval development in Caenorhabditis elegans. Here, we report that lin-4 and lin-14 also regulate life span in the adult. Reducing the activity of lin-4 shortened life span and accelerated tissue aging, whereas overexpressing lin-4 or reducing the activity of lin-14 extended life span. Lifespan extension conferred by a reduction in lin-14 was dependent on the DAF-16 and HSF-1 transcription factors, suggesting that the lin-4-lin-14 pair affects life span through the insulin/insulin-like growth factor-1 pathway. This work reveals a role for microRNAs and developmental timing genes in life-span regulation.
[26]
Zapico SC, Ubelaker DH. mtDNA mutations and their role in aging, diseases and forensic sciences[J]. Aging Dis, 2013, 4(6): 364-380. DOI: 10.14336/AD.2013.0400364.

Mitochondria are independent organelles with their own DNA. As a primary function, mitochondria produce the energy for the cell through Oxidative Phosphorylation (OXPHOS) in the Electron Transport Chain (ETC). One of the toxic products of this process is Reactive Oxygen Species (ROS), which can induce oxidative damage in macromolecules like lipids, proteins and DNA. Mitochondrial DNA (mtDNA) is less protected and has fewer reparation mechanisms than nuclear DNA (nDNA), and as such is more exposed to oxidative, mutation-inducing damage. This review analyzes the causes and consequences of mtDNA mutations and their relationship with the aging process. Neurodegenerative diseases, related with the aging, are consequences of mtDNA mutations resulting in a decrease in mitochondrial function. Also described are “mitochondrial diseases”, pathologies produced by mtDNA mutations and whose symptoms are related with mitochondrial dysfunction. Finally, mtDNA haplogroups are defined in this review; these groups are important for determination of geographical origin of an individual. Additionally, different haplogroups exhibit variably longevity and risk of certain diseases. mtDNA mutations in aging and haplogroups are of special interest to forensic science research. Therefore this review will help to clarify the key role of mtDNA mutations in these processes and support further research in this area.

[27]
Bai XY, Ma Y, Ding R, et al. miR-335 and miR-34a promote renal senescence by suppressing mitochondrial antioxidative enzymes[J]. J Am Soc Nephrol, 2011, 22(7): 1252-1261. DOI: 10.1681/ASN.2010040367.
[28]
Szeto HH. Pharmacologic approaches to improve mitochondrial function in AKI and CKD[J]. J Am Soc Nephrol, 2017, 28(10): 2856-2865. DOI: 10.1681/ASN.2017030247.
AKI is associated with high morbidity and mortality, and it predisposes to the development and progression of CKD. Novel strategies that minimize AKI and halt the progression of CKD are urgently needed. Normal kidney function involves numerous different cell types, such as tubular epithelial cells, endothelial cells, and podocytes, working in concert. This delicate balance involves many energy-intensive processes. Fatty acids are the preferred energy substrates for the kidney, and defects in fatty acid oxidation and mitochondrial dysfunction are universally involved in diverse causes of AKI and CKD. This review provides an overview of ATP production and energy demands in the kidney and summarizes preclinical and clinical evidence of mitochondrial dysfunction in AKI and CKD. New therapeutic strategies targeting mitochondria protection and cellular bioenergetics are presented, with emphasis on those that have been evaluated in animal models of AKI and CKD. Targeting mitochondrial function and cellular bioenergetics upstream of cellular damage may offer advantages compared with targeting downstream inflammatory and fibrosis processes.Copyright © 2017 by the American Society of Nephrology.
[29]
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.
[30]
Tsuji N, Tsuji T, Ohashi N, et al. Role of mitochondrial DNA in septic AKI via toll-like receptor 9[J]. J Am Soc Nephrol, 2016, 27(7): 2009-2020. DOI: 10.1681/ASN.2015040376.
Toll-like receptor 9 (TLR9) contributes to the development of polymicrobial septic AKI. However, the mechanisms that activate the TLR9 pathway and cause kidney injury during sepsis remain unknown. To determine the role of mitochondrial DNA (mtDNA) in TLR9-associated septic AKI, we established a cecal ligation and puncture (CLP) model of sepsis in wild-type (WT) and Tlr9-knockout (Tlr9KO) mice. We evaluated systemic circulation and peritoneal cavity dynamics and immune response and tubular mitochondrial dysfunction to determine upstream and downstream effects on the TLR9 pathway, respectively. CLP increased mtDNA levels in the plasma and peritoneal cavity of WT and Tlr9KO mice in the early phase, but the increase in the peritoneal cavity was significantly higher in Tlr9KO mice than in WT mice. Concomitantly, leukocyte migration to the peritoneal cavity increased, and plasma cytokine production and splenic apoptosis decreased in Tlr9KO mice compared with WT mice. Furthermore, CLP-generated renal mitochondrial oxidative stress and mitochondrial vacuolization in the proximal tubules in the early phase were reversed in Tlr9KO mice. To elucidate the effects of mtDNA on immune response and kidney injury, we intravenously injected mice with mitochondrial debris (MTD), including substantial amounts of mtDNA. MTD caused an immune response similar to that induced by CLP, including upregulated levels of plasma IL-12, splenic apoptosis, and mitochondrial injury, but this effect was attenuated by Tlr9KO. Moreover, MTD-induced renal mitochondrial injury was abolished by DNase pretreatment. These findings suggest that mtDNA activates TLR9 and contributes to cytokine production, splenic apoptosis, and kidney injury during polymicrobial sepsis. Copyright © 2016 by the American Society of Nephrology.
[31]
Ge QM, Huang CM, Zhu XY, et al. Differentially expressed miRNAs in sepsis-induced acute kidney injury target oxidative stress and mitochondrial dysfunction pathways[J]. PLoS One, 2017, 12(3): e0173292. DOI: 10.1371/journal.pone.0173292.
[32]
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.
[33]
Guo Y, Ni J, Chen S, et al. MicroRNA-709 mediates acute tubular injury through effects on mitochondrial function[J]. J Am Soc Nephrol, 2018, 29(2): 449-461. DOI: 10.1681/ASN.2017040381.
Mitochondrial dysfunction has important roles in the pathogenesis of AKI, yet therapeutic approaches to improve mitochondrial function remain limited. In this study, we investigated the pathogenic role of microRNA-709 (miR-709) in mediating mitochondrial impairment and tubular cell death in AKI. In a cisplatin-induced AKI mouse model and in biopsy samples of human AKI kidney tissue, miR-709 was significantly upregulated in the proximal tubular cells (PTCs). The expression of miR-709 in the renal PTCs of patients with AKI correlated with the severity of kidney injury. In cultured mouse PTCs, overexpression of miR-709 markedly induced mitochondrial dysfunction and cell apoptosis, and inhibition of miR-709 ameliorated cisplatin-induced mitochondrial dysfunction and cell injury. Further analyses showed that mitochondrial transcriptional factor A (TFAM) is a target gene of miR-709, and genetic restoration of TFAM attenuated mitochondrial dysfunction and cell injury induced by cisplatin or miR-709 overexpression Moreover, antagonizing miR-709 with an miR-709 antagomir dramatically attenuated cisplatin-induced kidney injury and mitochondrial dysfunction in mice. Collectively, our results suggest that miR-709 has an important role in mediating cisplatin-induced AKI negative regulation of TFAM and subsequent mitochondrial dysfunction. These findings reveal a pathogenic role of miR-709 in acute tubular injury and suggest a novel target for the treatment of AKI.Copyright © 2018 by the American Society of Nephrology.
[34]
Granata S, Dalla Gassa A, Tomei P, et al. Mitochondria: a new therapeutic target in chronic kidney disease[J]. Nutr Metab (Lond), 2015, 12: 49. DOI: 10.1186/s12986-015-0044-z.
[35]
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.
[36]
Trionfini P, Benigni A, Remuzzi G. MicroRNAs in kidney physiology and disease[J]. Nat Rev Nephrol, 2015, 11(1): 23-33. DOI: 10.1038/nrneph.2014.202.
MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate gene expression. They have important roles during kidney development, homeostasis and disease. In particular, miRNAs participate in the onset and progression of tubulointerstitial sclerosis and end-stage glomerular lesions that occur in various forms of chronic kidney disease (CKD). Therefore, miRNAs represent potential new therapeutic targets for a debilitating disease that continues to increase in prevalence worldwide and for which fully effective therapies are lacking. Several lines of research aimed at improving common CKD diagnostic tools and avoiding invasive kidney biopsies have also identified circulating miRNAs as possible diagnostic and even prognostic biomarkers of kidney disease. This Review discusses current understanding of the function of miRNAs in CKD, focusing on functions specifically involved in the transforming growth factor β1 pathway, which is activated in CKD. miRNAs that, according to available evidence, seem to be involved in diabetic nephropathy, IgA nephropathy, lupus nephritis, polycystic kidney disease and graft rejection, are also discussed.
[37]
Gomez IG, Nakagawa N, Duffield JS. MicroRNAs as novel therapeutic targets to treat kidney injury and fibrosis[J]. Am J Physiol Renal Physiol, 2016, 310(10): F931-F944. DOI: 10.1152/ajprenal.00523.2015.
MicroRNAs (miRs), a class of small noncoding RNAs that act as post-transcriptional regulators of gene expression, have attracted increasing attention as critical regulators of organogenesis, cancer, and disease. Interest has been spurred by development of a novel class of synthetic RNA oligonucleotides with excellent drug-like properties that hybridize to a specific miR, preventing its action. In kidney disease, a small number of miRs are dysregulated. These overlap with regulated miRs in nephrogenesis and kidney cancers. Several dysregulated miRs have been identified in fibrotic diseases of other organs, representing a “fibrotic signature,” and some of these fibrotic miRs contribute remarkably to the pathogenesis of kidney disease. Chronic kidney disease, affecting ∼10% of the population, leads to kidney failure, with few treatment options. Here, we will explore the pathological mechanism of miR-21, whose pre-eminent role in amplifying kidney disease and fibrosis by suppressing mitochondrial biogenesis and function is established. Evolving roles for miR-214, -199, -200, -155, -29, -223, and -126 in kidney disease will be discussed, and we will demonstrate how studying functions of distinct miRs has led to new mechanistic insights for kidney disease progression. Finally, the utility of anti-miR oligonucleotides as potential novel therapeutics to treat chronic disease will be highlighted.
[38]
Chau BN, Xin C, Hartner J, et al. MicroRNA-21 promotes fibrosis of the kidney by silencing metabolic pathways[J]. Sci Transl Med, 2012, 4(121): 121ra18. DOI: 10.1126/scitranslmed.3003205.
[39]
Bai M, Chen H, Ding D, et al. MicroRNA-214 promotes chronic kidney disease by disrupting mitochondrial oxidative phosphorylation[J]. Kidney Int, 2019, 95(6): 1389-1404. DOI: 10.1016/j.kint.2018.12.028.
Mitochondria are critical in determining a cell's energy homeostasis and fate, and mitochondrial dysfunction has been implicated in the pathogenesis of chronic kidney disease (CKD). We sought to identify causative mitochondrial microRNAs. A microarray screen of kidney tissue from healthy mice identified 97 microRNAs that were enriched in the mitochondrial fraction. We focused on microRNA-214-3p (miR-214) because of a very high ratio of mitochondrial to cytoplasmic expression in the kidney compared to other organs. Tubular expression of miR-214 was more abundant in kidney tissue from patients with CKD than from healthy controls, and was positively correlated with the degree of proteinuria and kidney fibrosis. Expression of miR-214 was also increased in the kidney of mouse models of CKD induced by obstruction, ischemia/reperfusion, and albumin overload. Proximal tubule-specific deletion of miR-214 attenuated apoptosis, inflammation, fibrosis, and mitochondrial damage in these CKD models. Pharmacologic inhibition of miR-214 had a similar effect in the albumin overload model of CKD. In vitro, overexpressing miR-214 in proximal tubular cell lines induced apoptosis and disrupted mitochondrial oxidative phosphorylation, while miR-214 expression was upregulated in response to a variety of insults. The mitochondrial genes mt-Nd6 and mt-Nd4l were identified as the specific targets of miR-214 in the kidney. Together, these results demonstrate a pathogenic role of miR-214 in CKD through the disruption of mitochondrial oxidative phosphorylation, and suggest the potential for miR-214 to serve as a therapeutic target and diagnostic biomarker for CKD.Copyright © 2019 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.
[40]
Sun Y, Kuek V, Liu Y, et al. MiR-214 is an important regulator of the musculoskeletal metabolism and disease[J]. J Cell Physiol, 2018, 234(1): 231-245. DOI: 10.1002/jcp.26856.
MiR-214 belongs to a family of microRNA (small, highly conserved noncoding RNA molecules) precursors that play a pivotal role in biological functions, such as cellular function, tissue development, tissue homeostasis, and pathogenesis of diseases. Recently, miR-214 emerged as a critical regulator of musculoskeletal metabolism. Specifically, miR-214 can mediate skeletal muscle myogenesis and vascular smooth muscle cell proliferation, migration, and differentiation. MiR-214 also modulates osteoblast function by targeting specific molecular pathways and the expression of various osteoblast-related genes; promotes osteoclast activity by targeting phosphatase and tensin homolog (Pten); and mediates osteoclast-osteoblast intercellular crosstalk via an exosomal miRNA paracrine mechanism. Importantly, dysregulation in miR-214 expression is associated with pathological bone conditions such as osteoporosis, osteosarcoma, multiple myeloma, and osteolytic bone metastasis of breast cancer. This review discusses the cellular targets of miR-214 in bone, the molecular mechanisms governing the activities of miR-214 in the musculoskeletal system, and the putative role of miR-214 in skeletal diseases. Understanding the biology of miR-214 could potentially lead to the development of miR-214 as a possible biomarker and a therapeutic target for musculoskeletal diseases.© 2018 Wiley Periodicals, Inc.
[41]
Portilla D. Energy metabolism and cytotoxicity[J]. Semin Nephrol, 2003, 23(5): 432-438. DOI: 10.1016/s0270-9295(03)00088-3.
Fatty acids constitute a major source of metabolic fuel for energy production in kidney tissue. During acute renal failure (ARF) injury to the proximal tubule and medullary thick ascending limb leads to structural and functional alterations that result in reduced expression and activity of mitochondrial and peroxisomal fatty acid oxidation (FAO) enzymes. Reduced DNA binding activity of peroxisome proliferator activated receptor-alpha (PPARalpha) to its target genes and decreased expression of its tissue-specific coactivator PPAR-gamma-coactivator-1 (PGC-1) in the mouse proximal tubule and the medullary thick ascending limb, represent 2 potential mechanisms that account for the observed alterations of FAO during ARF. Pretreatment with PPARalpha ligands restores the expression and activity of renal FAO enzymes, and this metabolic alteration leads to amelioration of acute tubular necrosis caused by ischemia/reperfusion or cisplatin-induced ARF. More studies are needed to examine further the cellular mechanisms of substrate inhibition, and to determine if metabolic pathways, in addition to the recovery of FAO, account for the protective effect (s) of PPARalpha ligands during acute renal failure.
[42]
Wang WX, Yu HL, Liu X. MiR-9-5p suppresses cell metastasis and epithelial-mesenchymal transition through targeting FOXP2 and predicts prognosis of colorectal carcinoma[J]. Eur Rev Med Pharmacol Sci, 2019, 23(15): 6467-6477. DOI: 10.26355/eurrev_201908_18530.
[43]
Wang M, Gao Q, Chen Y, et al. PAK4, a target of miR-9-5p, promotes cell proliferation and inhibits apoptosis in colorectal cancer[J]. Cell Mol Biol Lett, 2019, 24: 58. DOI: 10.1186/s11658-019-0182-9.
Colorectal cancer (CRC) is a leading cause of cancer-related death worldwide. P21-activated kinase 4 (PAK4) and miR-9-5p have emerged as attractive therapeutic targets in several tumor types, but in CRC, the regulation of their biological function and their target association remain unclear.The expression of PAK4 in CRC tissues was determined using quantitative real-time PCR and immunohistochemistry analyses. The targeted regulation between miR-9-5p and PAK4 was predicted and confirmed with bioinformatics analysis and the dual-luciferase reporter assay. Functional experiments, including the MTT assay and flow cytometry, were performed to investigate the impact of PAK4 knockdown and miR-9-5p overexpression on cell proliferation and apoptosis in CRC cells.We found that the expression of PAK4 was upregulated in CRC tissues. PAK4 knockdown significantly suppressed cell proliferation and promoted apoptosis in cells of the CRC cell lines HCT116 and SW1116. We also found that miR-9-5p directly targeted the 3'-UTR of PAK4 mRNA and negatively regulated its expression. The degree of downregulation of miR-9-5p inversely correlated with PAK4 expression. Intriguingly, enforced expression of miR-9-5p suppressed cell proliferation and promoted apoptosis. This could be partially reversed by PAK4 overexpression.These results suggest that miR-9-5p targeting of PAK4 could have therapeutic potential for CRC treatment.© The Author(s) 2019.
[44]
Fierro-Fernández M, Busnadiego Ó, Sandoval P, et al. miR-9-5p suppresses pro-fibrogenic transformation of fibroblasts and prevents organ fibrosis by targeting NOX4 and TGFBR2[J]. EMBO Rep, 2015, 16(10): 1358-1377. DOI: 10.15252/embr.201540750.
Uncontrolled extracellular matrix (ECM) production by fibroblasts in response to injury contributes to fibrotic diseases, including idiopathic pulmonary fibrosis (IPF). Reactive oxygen species (ROS) generation is involved in the pathogenesis of IPF. Transforming growth factor-β1 (TGF-β1) stimulates the production of NADPH oxidase 4 (NOX4)-dependent ROS, promoting lung fibrosis (LF). Dysregulation of microRNAs (miRNAs) has been shown to contribute to LF. To identify miRNAs involved in redox regulation relevant for IPF, we performed arrays in human lung fibroblasts exposed to ROS. miR-9-5p was selected as the best candidate and we demonstrate its inhibitory effect on TGF-β receptor type II (TGFBR2) and NOX4 expression. Increased expression of miR-9-5p abrogates TGF-β1-dependent myofibroblast phenotypic transformation. In the mouse model of bleomycin-induced LF, miR-9-5p dramatically reduces fibrogenesis and inhibition of miR-9-5p and prevents its anti-fibrotic effect both in vitro and in vivo. In lung specimens from patients with IPF, high levels of miR-9-5p are found. In omentum-derived mesothelial cells (MCs) from patients subjected to peritoneal dialysis (PD), miR-9-5p also inhibits mesothelial to myofibroblast transformation. We propose that TGF-β1 induces miR-9-5p expression as a self-limiting homeostatic response.© 2015 The Authors.
[45]
Fierro-Fernández M, Miguel V, Márquez-Expósito L, et al. MiR-9-5p protects from kidney fibrosis by metabolic reprogramming[J]. FASEB J, 2020, 34(1): 410-431. DOI: 10.1096/fj.201901599RR.
MicroRNAs (miRNAs) regulate gene expression posttranscriptionally and control biological processes (BPs), including fibrogenesis. Kidney fibrosis remains a clinical challenge and miRNAs may represent a valid therapeutic avenue. We show that miR-9-5p protected from renal fibrosis in the mouse model of unilateral ureteral obstruction (UUO). This was reflected in reduced expression of pro-fibrotic markers, decreased number of infiltrating monocytes/macrophages, and diminished tubular epithelial cell injury and transforming growth factor-beta 1 (TGF-β1)-dependent de-differentiation in human kidney proximal tubular (HKC-8) cells. RNA-sequencing (RNA-Seq) studies in the UUO model revealed that treatment with miR-9-5p prevented the downregulation of genes related to key metabolic pathways, including mitochondrial function, oxidative phosphorylation (OXPHOS), fatty acid oxidation (FAO), and glycolysis. Studies in human tubular epithelial cells demonstrated that miR-9-5p impeded TGF-β1-induced bioenergetics derangement. The expression of the FAO-related axis peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α)-peroxisome proliferator-activated receptor alpha (PPARα) was reduced by UUO, although preserved by the administration of miR-9-5p. We found that in mice null for the mitochondrial master regulator PGC-1α, miR-9-5p was unable to promote a protective effect in the UUO model. We propose that miR-9-5p elicits a protective response to chronic kidney injury and renal fibrosis by inducing reprogramming of the metabolic derangement and mitochondrial dysfunction affecting tubular epithelial cells.© 2019 Federation of American Societies for Experimental Biology.
[46]
Shi S, Yu L, Zhang T, et al. Smad2-dependent downregulation of miR-30 is required for TGF-β-induced apoptosis in podocytes[J]. PLoS One, 2013, 8(9): e75572. DOI: 10.1371/journal.pone.0075572.
[47]
Guo Y, Deng X, Chen S, et al. MicroRNA-30e targets BNIP3L to protect against aldosterone-induced podocyte apoptosis and mitochondrial dysfunction[J]. Am J Physiol Renal Physiol, 2017, 312(4): F589-F598. DOI: 10.1152/ajprenal.00486.2016.
MicroRNAs are essential for the maintenance of podocyte homeostasis. Emerging evidence has demonstrated a protective role of microRNA-30a (miR-30a), a member of the miR-30 family, in podocyte injury. However, the roles of other miR-30 family members in podocyte injury are unclear. The present study was undertaken to investigate the contribution of miR-30e to the pathogenesis of podocyte injury induced by aldosterone (Aldo), as well as the underlying mechanism. After Aldo treatment, miR-30e was reduced in a dose-and time-dependent manner. Notably, overexpression of miR-30e markedly attenuated Aldo-induced apoptosis in podocytes. In agreement with this finding, miR-30e silencing led to significant podocyte apoptosis. Mitochondrial dysfunction (MtD) has been shown to be an early event in Aldo-induced podocyte injury. Here we found that overexpression of miR-30e improved Aldo-induced MtD while miR-30e silencing resulted in MtD. Next, we found that miR-30e could directly target the BCL2/adenovirus E1B-interacting protein 3-like (BNIP3L) gene. Aldo markedly enhanced BNIP3L expression in podocytes, and silencing of BNIP3L largely abolished Aldo-induced MtD and cell apoptosis. On the contrary, overexpression of BNIP3L induced MtD and apoptosis in podocytes. Together, these findings demonstrate that miR-30e protects mitochondria and podocytes from Aldo challenge by targeting BNIP3L.
[48]
Jiang L, Qiu W, Zhou Y, et al. A microRNA-30e/mitochondrial uncoupling protein 2 axis mediates TGF-β1-induced tubular epithelial cell extracellular matrix production and kidney fibrosis[J]. Kidney Int, 2013, 84(2): 285-296. DOI: 10.1038/ki.2013.80.
Mitochondria dysfunction has been reported in various kidney diseases but how it leads to kidney fibrosis and how this is regulated is unknown. Here we found that mitochondrial uncoupling protein 2 (UCP2) was induced in kidney tubular epithelial cells after unilateral ureteral obstruction in mice and that mice with ablated UCP2 resisted obstruction-induced kidney fibrosis. We tested this association further in cultured NRK-52E cells and found that TGF-β1 remarkably induced UCP2 expression. Knockdown of UCP2 largely abolished the effect of TGF-β1, whereas overexpression of UCP2 promoted tubular cell phenotype changes. Analysis using a UCP2 mRNA-3'-untranslated region luciferase construct showed that UCP2 mRNA is a direct target of miR-30e. MiR-30e was downregulated in tubular cells from fibrotic kidneys and TGF-β1-treated NRK-52E cells. A miR-30e mimic significantly inhibited TGF-β1-induced tubular-cell epithelial-mesenchymal transition, whereas a miR-30e inhibitor imitated TGF-β1 effects. Finally, genipin, an aglycone UCP2 inhibitor, significantly ameliorated kidney fibrosis in mice. Thus, the miR-30e/UCP2 axis has an important role in mediating TGF-β1-induced epithelial-mesenchymal transition and kidney fibrosis. Targeting this pathway may shed new light for the future of fibrotic kidney disease therapy.
[49]
Bergmann C, Guay-Woodford LM, Harris PC, et al. Polycystic kidney disease[J]. Nat Rev Dis Primers, 2018, 4(1): 50. DOI: 10.1038/s41572-018-0047-y.
Cystic kidneys are common causes of end-stage renal disease, both in children and in adults. Autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD) are cilia-related disorders and the two main forms of monogenic cystic kidney diseases. ADPKD is a common disease that mostly presents in adults, whereas ARPKD is a rarer and often more severe form of polycystic kidney disease (PKD) that usually presents perinatally or in early childhood. Cell biological and clinical research approaches have expanded our knowledge of the pathogenesis of ADPKD and ARPKD and revealed some mechanistic overlap between them. A reduced 'dosage' of PKD proteins is thought to disturb cell homeostasis and converging signalling pathways, such as Ca2+, cAMP, mechanistic target of rapamycin, WNT, vascular endothelial growth factor and Hippo signalling, and could explain the more severe clinical course in some patients with PKD. Genetic diagnosis might benefit families and improve the clinical management of patients, which might be enhanced even further with emerging therapeutic options. However, many important questions about the pathogenesis of PKD remain. In this Primer, we provide an overview of the current knowledge of PKD and its treatment.
[50]
Hajarnis S, Lakhia R, Yheskel M, et al. MicroRNA-17 family promotes polycystic kidney disease progression through modulation of mitochondrial metabolism[J]. Nat Commun, 2017, 8: 14395. DOI: 10.1038/ncomms14395.
Autosomal dominant polycystic kidney disease (ADPKD) is the most frequent genetic cause of renal failure. Here we identify miR-17 as a target for the treatment of ADPKD. We report that miR-17 is induced in kidney cysts of mouse and human ADPKD. Genetic deletion of the miR-17 similar to 92 cluster inhibits cyst proliferation and PKD progression in four orthologous, including two long-lived, mouse models of ADPKD. Anti-miR-17 treatment attenuates cyst growth in short-term and long-term PKD mouse models. miR-17 inhibition also suppresses proliferation and cyst growth of primary ADPKD cysts cultures derived from multiple human donors. Mechanistically, c-Myc upregulates miR-17B92 in cystic kidneys, which in turn aggravates cyst growth by inhibiting oxidative phosphorylation and stimulating proliferation through direct repression of Ppar alpha. Thus, miR-17 family is a promising drug target for ADPKD, and miR-17-mediated inhibition of mitochondrial metabolism represents a potential new mechanism for ADPKD progression.
[51]
Mukhadi S, Hull R, Mbita Z, et al. The role of microRNAs in kidney disease[J]. Noncoding RNA, 2015, 1(3): 192-221. DOI: 10.3390/ncrna1030192.
MicroRNAs (miRNAs) are short noncoding RNAs that regulate pathophysiological processes that suppress gene expression by binding to messenger RNAs. These biomolecules can be used to study gene regulation and protein expression, which will allow better understanding of many biological processes such as cell cycle progression and apoptosis that control the fate of cells. Several pathways have also been implicated to be involved in kidney diseases such as Transforming Growth Factor-β, Mitogen-Activated Protein Kinase signaling, and Wnt signaling pathways. The discovery of miRNAs has provided new insights into kidney pathologies and may provide new innovative and effective therapeutic strategies. Research has demonstrated the role of miRNAs in a variety of kidney diseases including renal cell carcinoma, diabetic nephropathy, nephritic syndrome, renal fibrosis, lupus nephritis and acute pyelonephritis. MiRNAs are implicated as playing a role in these diseases due to their role in apoptosis, cell proliferation, differentiation and development. As miRNAs have been detected in a stable condition in different biological fluids, they have the potential to be tools to study the pathogenesis of human diseases with a great potential to be used in disease prognosis and diagnosis. The purpose of this review is to examine the role of miRNA in kidney disease.
[52]
Grange C, Brossa A, Bussolati B. Extracellular vesicles and carried miRNAs in the progression of renal cell carcinoma[J]. Int J Mol Sci, 2019, 20(8): 1832. DOI: 10.3390/ijms20081832.
The formation and maintenance of renal cell carcinomas (RCC) involve many cell types, such as cancer stem and differentiated cells, endothelial cells, fibroblasts and immune cells. These all contribute to the creation of a favorable tumor microenvironment to promote tumor growth and metastasis. Extracellular vesicles (EVs) are considered to be efficient messengers that facilitate the exchange of information within the different tumor cell types. Indeed, tumor EVs display features of their originating cells and force recipient cells towards a pro-tumorigenic phenotype. This review summarizes the recent knowledge related to the biological role of EVs, shed by renal tumor cells and renal cancer stem cells in different aspects of RCC progression, such as angiogenesis, immune escape and tumor growth. Moreover, a specific role for renal cancer stem cell derived EVs is described in the formation of the pre-metastatic niche. We also highlight the tumor EV cargo, especially the oncogenic miRNAs, which are involved in these processes. Finally, the circulating miRNAs appear to be a promising source of biomarkers in RCC.
[53]
Dasgupta P, Kulkarni P, Majid S, et al. MicroRNA-203 inhibits long noncoding RNA HOTAIR and regulates tumorigenesis through epithelial-to-mesenchymal transition pathway in renal cell carcinoma[J]. Mol Cancer Ther, 2018, 17(5): 1061-1069. DOI: 10.1158/1535-7163.MCT-17-0925.
This study aims to investigate the role of miR-203-HOTAIR interaction in the suppression of renal cell carcinoma (RCC). We employed series of assays such as proliferation, invasion, migration, and colony formation along with tumor xenograft model. Profiling of miR-203 and HOTAIR expression revealed that miR-203 was significantly underexpressed, whereas HOTAIR was overexpressed in RCC cell lines and clinical specimens compared with normal cell line and tissue. Both miR-203 and HOTAIR expression significantly distinguished malignant from normal tissues and significantly correlated with clinicopathologic characteristics of patients. Overexpression of miR-203 significantly inhibited proliferation, migration, and invasion with an induction of apoptosis and cell-cycle arrest. However, HOTAIR suppression resulted in the similar functional effects in the same RCC cell lines., RNA-22 algorithm showed a binding site for miR-203 in HOTAIR. We observed a direct interaction between miR-203 and HOTAIR by RNA-immunoprecipitation (RIP) and luciferase reporter assays. We show that miR-203-HOTAIR interaction resulted in the inhibition of epithelial-to-mesenchymal transition (EMT) and metastatic genes as indicated by induction of key metastasis-suppressing proteins E-cadherin, claudin (epithelial markers), and PTEN along with induction of tumor suppressor genes p21 and p27. A significant decrease in vimentin (mesenchymal marker), KLF4, and Nanog (stemness markers) was also observed. This is the first report demonstrating miR-203-mediated regulation of HOTAIR induces tumor suppressor effects in RCC by regulating EMT and metastatic pathway genes. Thus, the study suggests that therapeutic regulation of HOTAIR by miR-203 overexpression may provide an opportunity to regulate RCC growth and metastasis..©2018 American Association for Cancer Research.
[54]
Hensley CT, Faubert B, Yuan Q, et al. Metabolic heterogeneity in human lung tumors[J]. Cell, 2016, 164(4): 681-694. DOI: 10.1016/j.cell.2015.12.034.
Non-small cell lung cancer (NSCLC) is heterogeneous in the genetic and environmental parameters that influence cell metabolism in culture. Here, we assessed the impact of these factors on human NSCLC metabolism in vivo using intraoperative (13)C-glucose infusions in nine NSCLC patients to compare metabolism between tumors and benign lung. While enhanced glycolysis and glucose oxidation were common among these tumors, we observed evidence for oxidation of multiple nutrients in each of them, including lactate as a potential carbon source. Moreover, metabolically heterogeneous regions were identified within and between tumors, and surprisingly, our data suggested potential contributions of non-glucose nutrients in well-perfused tumor areas. Our findings not only demonstrate the heterogeneity in tumor metabolism in vivo but also highlight the strong influence of the microenvironment on this feature.Copyright © 2016 Elsevier Inc. All rights reserved.
[55]
Pierini S, Fang C, Rafail S, et al. A tumor mitochondria vaccine protects against experimental renal cell carcinoma[J]. J Immunol, 2015, 195(8): 4020-4027. DOI: 10.4049/jimmunol.1500281.
Mitochondria provide energy for cells via oxidative phosphorylation. Reactive oxygen species, a byproduct of this mitochondrial respiration, can damage mitochondrial DNA (mtDNA), and somatic mtDNA mutations have been found in all colorectal, ovarian, breast, urinary bladder, kidney, lung, and pancreatic tumors studied. The resulting altered mitochondrial proteins or tumor-associated mitochondrial Ags (TAMAs) are potentially immunogenic, suggesting that they may be targetable Ags for cancer immunotherapy. In this article, we show that the RENCA tumor cell line harbors TAMAs that can drive an antitumor immune response. We generated a cellular tumor vaccine by pulsing dendritic cells with enriched mitochondrial proteins from RENCA cells. Our dendritic cell-based RENCA mitochondrial lysate vaccine elicited a cytotoxic T cell response in vivo and conferred durable protection against challenge with RENCA cells when used in a prophylactic or therapeutic setting. By sequencing mtDNA from RENCA cells, we identified two mutated molecules: COX1 and ND5. Peptide vaccines generated from mitochondrial-encoded COX1 but not from ND5 had therapeutic properties similar to RENCA mitochondrial protein preparation. Thus, TAMAs can elicit effective antitumor immune responses, potentially providing a new immunotherapeutic strategy to treat cancer. Copyright © 2015 by The American Association of Immunologists, Inc.
[56]
Dutta P, Haller E, Sharp A, et al. MIR494 reduces renal cancer cell survival coinciding with increased lipid droplets and mitochondrial changes[J]. BMC Cancer, 2016, 16: 33. DOI: 10.1186/s12885-016-2053-3.
Background: miRNAs can regulate cellular survival in various cancer cell types. Recent evidence implicates the formation of lipid droplets as a hallmark event during apoptotic cell death response. It is presently unknown whether MIR494, located at 14q32 which is deleted in renal cancers, reduces cell survival in renal cancer cells and if this process is accompanied by changes in the number of lipid droplets. Methods: 769-P renal carcinoma cells were utilized for this study. Control or MIR494 mimic was expressed in these cells following which cell viability (via crystal violet) and apoptotic cell numbers (via Annexin V/PI staining) were assessed. By western blotting, MIR494 cellular responses were validated using MIR494 antagomir and Argonaute 2 siRNA. Transmission electron microscopy (TEM) was performed in MIR494-transfected 769-P cells to identify ultrastructural changes. LipidTOX green neutral lipid staining and cholesterol measurements were conducted to assess accumulation of lipids droplets and total cholesterol levels, respectively, in MIR494 expressing 769-P cells. Indirect immunofluorescence and western analyses were also performed to examine changes in mitochondria organization. Co-transfection of MIR494 mimic with siRNA targeting LC3B and ATG7 was conducted to assess their contribution to formation of lipid droplets in MIR494-expressing cells. Results: MIR494 expression reduces viability of 769-P renal cancer cells; this was accompanied by increased cleaved PARP (an apoptotic marker) and LC3B protein. Further, expression of MIR494 increased LC3B mRNA levels and LC3B promoter activity (2.01-fold; 50 % increase). Interestingly, expression of MIR494 markedly increased multilamellar bodies and lipid droplets (by TEM and validated by LipidTOX immunostaining) while reducing total cholesterol levels. Via immunocytochemistry, we observed increased LC3B-associated endogenous punctae upon MIR494 expression. In contrast to ATG7 siRNA, knockdown of LC3B reduced the numbers of lipid droplets in MIR494-expressing cells. Our results also identified that MIR494 expression altered the organization of mitochondria which was accompanied by co-localization with LC3B punctae, decreased PINK1 protein, and altered Drp1 intracellular distribution. Conclusion: Collectively, our findings indicate that MIR494 reduces cell survival in 769-P renal cancer cells which is accompanied by increased lipid droplet formation (which occurs in a LC3B-dependent manner) and mitochondrial changes.
[57]
Nakada C, Hijiya N, Tsukamoto Y, et al. A transgenic mouse expressing miR-210 in proximal tubule cells shows mitochondrial alteration: possible association of miR-210 with a shift in energy metabolism[J]. J Pathol, 2020, 251(1): 12-25. DOI: 10.1002/path.5394.
Previously we reported that the microRNA miR‐210 is aberrantly upregulated in clear cell renal cell carcinoma (ccRCC) via deregulation of the VHL–HIF pathway. In the present study, to investigate the biological impact of miR‐210 in ccRCC tumorigenesis, we developed a transgenic mouse line expressing miR‐210 in proximal tubule cells under control of the mouse SGLT2/Slc5a2 promoter. Light microscopy revealed desquamation of the tubule cells and regeneration of the proximal tubule, suggesting that miR‐210 expression led to damage of the proximal tubule cells. Electron microscopy revealed alterations to the mitochondria in proximal tubule cells, with marked reduction of the mitochondrial inner membrane, which is the main site of ATP production via oxidative phosphorylation (OxPhos). An additional in vitro study revealed that this loss of the inner membrane was associated with downregulation of Iscu and Ndufa4, the target genes of miR‐210, suggesting that the miR‐210–ISCU/NDUFA4 axis may affect mitochondrial energy metabolism. Furthermore, metabolome analysis revealed activation of anaerobic glycolysis in miR‐210‐transfected cells, and consistent with this the secretion of lactate, the final metabolite of anaerobic glycolysis, was significantly increased. Lactate concentration was higher in the kidney cortex of transgenic mice relative to wild‐type mice, although the difference was not significant (p = 0.070). On the basis of these findings, we propose that miR‐210 may induce a shift of energy metabolism from OxPhos to glycolysis by acting on the mitochondrial inner membrane. In addition to activation of glycolysis, we observed activation of the pentose phosphate pathway (PPP) and an increase in the total amount of amino acids in miR‐210‐transfected cells. This may help cells synthesize nucleotides and proteins for building new cells. These results suggest that miR‐210 may be involved in the metabolic changes in the early stage of ccRCC development, helping the cancer cells to acquire growth and survival advantages. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley &amp; Sons, Ltd.
PDF(766 KB)

2265

Accesses

0

Citation

Detail

Sections
Recommended

/