Role and mechanisms of mitochondria-associated endoplasmic reticulum membranes in the podocyte injury of diabetic kidney disease

Cao Yun, Feng Jun, Ding Guohua

PDF(19377 KB)
Chinese Journal of Nephrology ›› 2021, Vol. 37 ›› Issue (1) : 59-64. DOI: 10.3760/cma.j.cn441217-20200330-00012
Review

Role and mechanisms of mitochondria-associated endoplasmic reticulum membranes in the podocyte injury of diabetic kidney disease

Author information +
History +

Abstract

Cite this article

Download Citations
Cao Yun. , Feng Jun. , Ding Guohua. Role and mechanisms of mitochondria-associated endoplasmic reticulum membranes in the podocyte injury of diabetic kidney disease[J]. Chinese Journal of Nephrology, 2021, 37(1): 59-64. DOI: 10.3760/cma.j.cn441217-20200330-00012.
糖尿病肾病(diabetic kidney disease,DKD)已成为全球慢性肾脏病(chronic kidney disease,CKD)及终末期肾脏病(end-stage renal disease,ESRD)的首要病因[1]。据2017年流行病学统计,中国有1.14亿糖尿病(DM)患者,占全球DM患者数量(4.25亿)近四分之一[2]。DKD发病机制十分复杂,目前普遍认为与DKD发生发展相关的因素包括:遗传因素[微小RNA(miRNA)、DNA甲基化]、糖脂代谢异常、胰岛素抵抗(IR)、炎性反应、氧化应激 [活性氧类(ROS)]、内质网应激(endoplasmic reticulum stress,ERS)和线粒体功能障碍等[3]。上述致病因素大多与线粒体内质网耦联(mitochondria-associated endoplasmic reticulum membranes,MAMs)有关。因此,调控MAMs相关蛋白表达及信号传导通路或许可改善DKD肾脏损伤,起到肾脏保护作用。20世纪70年代在电子显微镜下观察到内质网(endoplasmic reticulum,ER)膜和线粒体膜间存在部分紧密接触,但受当时技术限制,难以证实两者之间确实存在连接[4]。直至1990年Vance[5]成功从大鼠肝脏中分离出ER与线粒体之间耦联的膜组分,并将这一膜组分命名为MAMs。我们系统地综述了MAMs在DKD足细胞损伤中的作用及其机制,为DKD的早期干预和延缓疾病进展,寻找新的治疗靶点提供思路。

一、 MAMs的结构与功能

MAMs是一系列蛋白质在ER和线粒体间形成的动态耦联平台,在该特殊部位ER膜和线粒体外膜维持着相对稳定的膜间距,相互之间不融合,通过高速数字成像显微镜和CT技术测量膜间距为10~80 nm[6]。富集在MAMs的蛋白大致可分为:(1)Ca2+通道及信号转导分子:如三磷酸肌醇受体(inositol 1,4,5-trisphosphate receptor,IP3R)[7]、三磷酸腺苷(5'-adenylate triphosphate,ATP)依赖性内质网钙泵(sarco/endoplasmic reticulum Ca2+ ATPase,SERCA)、电压依赖性阴离子通道1(voltage-dependent anion-selective channel 1,VDAC1)[7]、线粒体钙离子单向运载体(mitochondrial calcium uniporter,MCU)。(2)分子伴侣:如sigma-1受体(sigma-1 receptor,Sig-1R)[8]、葡萄糖调节蛋白78(glucose-regulated protein 78,GRP78)/免疫球蛋白重链结合蛋白(immunoglobulin heavy chain-binding protein,Bip)、GRP75。(3)脂质合成及转运酶:如类固醇合成快速调节蛋白(steroidogenic acute regulatory protein,StAR)、磷脂酰丝氨酸合成酶(phosphatidylserine synthase,PSS)。(4)线粒体动态调控蛋白:线粒体融合蛋白2(mitofusion 2,Mfn2)[9]、动力相关蛋白1(dynamin-related protein 1,Drp1)[10]。(5)凋亡相关蛋白:Bcl-2、Bcl-XL、Bax、Bak、细胞色素c(cytochrome c,Cyt c)。
近年来MAMs的神秘面纱正被逐步揭开,生理状态下MAMs协调ER和线粒体完成细胞的正常生命活动,病理状态下则相互影响,共同介导疾病的发生发展。MAMs是调节Ca2+稳态、线粒体功能、ERS、脂质代谢、炎性小体形成、自噬及凋亡等细胞生命活动的关键所在。MAMs结构和功能异常在肿瘤、代谢性疾病和神经退行性疾病等疾病模型中均有发现,提示MAMs结构域中的分子可能成为治疗相关疾病的新靶点[11]

二、 MAMs在DKD足细胞损伤中的作用及机制

足细胞是一种位于肾小球基底膜外侧的终末分化细胞,其相邻足突紧密交织形成裂孔,裂孔表面覆盖一层裂孔隔膜(slit diaphragm,SD),形成阻止蛋白质等大分子滤过的最后屏障[12]。DKD动物模型中观察到受损的足细胞足突增宽或消失、肥大、上皮间充质转分化(epithelial mesenchymal transformation,EMT)、剥脱和凋亡等一系列病理改变[13]。MAMs可通过调控细胞内Ca2+稳态、ERS及线粒体功能,诱导自噬或凋亡等调节DKD中足细胞的生命活动。
1. Ca2+稳态: ER是细胞内最大的Ca2+库,主要通过SERCA摄取和IP3R通路释放Ca2+来发挥生理功能。线粒体外膜上的VDAC1和线粒体内膜上的MCU则是将ER释放到MAMs间隙的Ca2+摄取入线粒体基质的重要蛋白。Rizzuto等[14]在荧光显微镜下观察发现,ER和线粒体间物理连接处存在利于两者进行Ca2+信号转导的高浓度Ca2+微区。人为地缩小或增大MAMs膜间距时,线粒体基质内Ca2+浓度也随之增高或降低。
MAMs中多种蛋白与Ca2+信号转导相关,胞质的Ca2+主要通过ER外膜上的SERCA进入ER,SERCA对维持ER中Ca2+稳态及调节细胞凋亡至关重要,其中SERCA2b的Ca2+亲和力最高,是MAMs调控Ca2+信号流起始的基础。SERCA2b在db/db小鼠及链脲佐菌素(streptozocin,STZ)诱导的DKD小鼠肾皮质中均出现表达和活性降低。在条件永生化人足细胞中,敲除SERCA2b可抵消黄芪甲苷对ERS和细胞凋亡的改善作用,而过表达SERCA2b具有抗凋亡作用[15]。IP3R-GRP75-VDAC1复合体负责传导ER和线粒体间的Ca2+信号,ER外膜上的IP3R激活后释放ER腔内的Ca2+,在GRP75的辅助下,被线粒体外膜上的VDAC1摄入,经线粒体内膜的MCU进入线粒体基质,形成MAMs中的Ca2+信号流,发挥生物功能[7,16]
肾脏是胰岛素的靶器官,足细胞是胰岛素高度敏感的效应细胞,胰岛素刺激下足细胞可快速将葡萄糖转运至细胞质,为维持其肌动蛋白骨架和正常滤过功能提供能量。丝氨酸-苏氨酸激酶(serine-threonine kinase,Akt)、蛋白磷酸酶2A(protein phosphatase 2A,PP2A)和哺乳动物雷帕霉素靶蛋白(mammalian target of rapamycin,mTOR)复合体2(mTOR complex 2,mTORC2)均是定位于MAMs的胰岛素信号通路蛋白。胰岛素受体底物(insulin receptor substrate 1,IRS1)/磷脂酰肌醇3激酶(phosphatidylinositol-3-kinase,PI3K)/Akt2信号通路活性低下被广泛认为是足细胞发生IR的重要因素[17]。胰岛素累积上调Akt的磷酸化水平,Akt活化后可抑制IP3R通路,减少Ca2+释放和细胞凋亡[18-19]。mTORC2和PP2A可分别通过磷酸化和去磷酸化Akt调节细胞内Ca2+信号流。在肝脏和骨骼肌组织,特异性敲除Akt或mTORC2可破坏MAMs完整性并导致糖代谢紊乱,提示胰岛素信号传递与MAMs结构完整性及功能密切相关[20]。由此推测MAMs可通过调控胰岛素信号影响足细胞Ca2+稳态和糖代谢,介导DKD足细胞损伤。
2. 内质网应激: ERS可分为未折叠蛋白质反应(unfolded protein response,UPR)、内质网超负荷反应(endoplasmic reticulum overload response,EOR)和固醇调节级联反应[21],其中UPR是目前被认为与DM和DKD密切相关的信号转导通路,主要通过肌醇必需蛋白1(inositol requiring protein 1,IRE1)、蛋白激酶RNA样内质网激酶 (protein kinase RNA-like ER kinase,PERK)以及激活转录因子6(activating transcription factor 6,ATF6)3种ERS传感蛋白介导后续信号通路的活化,减少细胞内蛋白翻译、促进未折叠蛋白质的降解或正确折叠,在应激早期维持细胞内稳态。首次证实MAMs和UPR之间存在联系是研究者发现磷酸弗林蛋白酶酸性氨基酸簇选蛋白2(phosphofurin acidic cluster sortingprotein 2,PACS2)缺失可导致MAMs结构及功能紊乱并诱导UPR[22]
足细胞含有丰富的ER系统,适度的ERS是细胞应对外界刺激的保护机制,在UPR早期可增加ER-线粒体连接,上调线粒体呼吸速率和能量合成,但持续或过强的应激反应将导致细胞功能受损甚至凋亡。在6周到12周的DKD小鼠足细胞中均检测到ERS激活而未发现明显的凋亡现象,提示DKD足细胞中ERS先于凋亡发生。Dong等[23]发现,相比心肌梗死的对照大鼠,有心肌梗死的DKD大鼠中GRP78蛋白、CCAAT/增强子结合蛋白同源蛋白(CCAAT/enhancer-binding protein homologous protein,CHOP)、半胱天冬酶(caspases)的表达升高,足细胞损伤、肾小球结构功能紊乱程度更重,提示高糖可上调足细胞ERS标志蛋白表达,激活ERS介导的凋亡通路。
既往的研究发现,生理状态下Mfn2与UPR传感蛋白PERK均定位于MAMs,两者结合使PERK处于稳定非活化状态,敲除或抑制Mfn2蛋白表达可活化PERK,从而磷酸化下游的真核翻译起始因子2α(eukaryotic translation initiation factor 2α,eIF2α),启动UPR[24-25]。白杨黄素(5,7-dihydroxyflavone)可抑制PERK磷酸化,从而阻断PERK/eIF2α/ATF4/CHOP通路,减少db/db小鼠足细胞损伤[26]。Sig-1R广泛表达于神经系统、肾脏、肝脏、肺和眼等组织,非应激条件下以无活性状态与GRP78结合,两者解离后Sig-1R活化[27]。ERS状态下,Sig-1R与活化的IRE1结合强化其作用,敲除Sig-1R可抑制IRE1-X盒子结合蛋白1(x-box binding protein 1,XBP1)通路活化,并上调ERS诱导的凋亡相关蛋白CHOP的表达[8]。过表达Sig-1R可抑制PERK和ATF6磷酸化,发挥抗凋亡作用。苏维维等[28]发现,抑制条件永生化小鼠足细胞系(MPC5)中的Sig-1R基因表达后,nephrin表达减少;Hochest核染色显示染色质边集凝聚、核固缩及核碎裂等细胞凋亡改变明显增加;活化的caspase-12(定位于ER外膜,是介导ER凋亡通路的关键分子)、caspase-3蛋白表达量明显增加,提示Sig-1R对足细胞有保护作用,抑制Sig-1R基因表达可通过加重ERS介导足细胞凋亡。
3. 线粒体功能障碍: 电镜下可见,足细胞除了胞体,狭窄的足突部位同样有线粒体分布。线粒体的核心作用是产生能量,其电子传递复合物(electron transfer complex,ETC)和ATP合成系统产生大量ATP来维持足细胞以肌动蛋白为主的细胞骨架结构[29]。随着DKD病程发展,足细胞可发生不同程度的线粒体形态异常和功能障碍,包括线粒体动力学紊乱、ROS 产生增加、进行性呼吸链功能异常等[30]
线粒体是一种高度动态的细胞器,通过持续的分裂和融合来调控自身的形态、数量、分布和功能等。线粒体动力学相关的主要蛋白均为具有GTP酶功能的动力蛋白家族成员,其中Drp1与Mfn2定位于MAMs,Drp1介导线粒体分裂,Mfn2与Mfn1共同介导线粒体外膜融合。当细胞受到内外部刺激时,线粒体动力学由分裂融合平衡转为趋向发生线粒体分裂。大量研究表明,线粒体动力学紊乱在DKD的发生发展中发挥重要作用。
在哺乳动物细胞内,线粒体裂变区高度定位于ER-线粒体膜接触区域,ER小管缠绕线粒体裂变区并募集Drp1形成螺旋体,启动线粒体分裂[9]。融合增多或分裂减少都会促进线粒体形成长管状结构, 当融合减少或分裂增多时, 线粒体则多呈现为小球状结构。Ma等[31]发现,体外高糖刺激的足细胞和DKD大鼠模型足细胞内长管状线粒体减少而小球状线粒体增多,且DKD患者肾活检组织中的足细胞也有同样改变。DKD肾组织中线粒体碎片增多伴随Drp1表达及磷酸化水平的明显上调,Mfn2缺乏同样易导致线粒体碎片形成。Chen等[32]发现,在STZ诱导的DKD大鼠肾脏足细胞及高糖刺激的人永生化足细胞中,PKA/A型激酶锚定蛋白1(PKA/A kinase anchoring protein 1,AKAP1)表达上调,活化的AKAP1促使Drp1在第637位丝氨酸残基(Ser637)处磷酸化,p-Drp1随后募集到线粒体外膜介导线粒体分裂。非活性状态下,Bax蛋白主要存在于细胞质而Bak蛋白锚定在线粒体外膜上,线粒体分裂前,ER-线粒体膜接触增加,募集到线粒体外膜的Drp1在此诱导Bax和Bak蛋白激活,发生构象变化,并分别或相互寡聚化形成成孔蛋白低聚物,通过打开环形孔的方式破坏脂质双分子层的连续性,从而介导线粒体外膜通透(mitochondrial outer membrane permeabilization,MOMP),释放线粒体凋亡途径相关蛋白如Cyt c到胞质,引发caspase级联反应,最终导致细胞凋亡[33];敲除Drp1可阻断该信号通路,减少线粒体分裂及其诱导的细胞凋亡。
除线粒体ETC产生ROS外,ER中蛋白质折叠时二硫键的形成或断裂过程也伴随着胞内约1/4的ROS生成[34]。高糖诱导ROS过量产生是DKD的主要初始机制,也是促进DM肾损伤的关键因素。Coughlan等[35]发现,DKD大鼠肾组织内线粒体ATP合成明显下降,过氧化氢产物生成增加,抗氧化物活性降低等线粒体功能异常均早于蛋白尿出现。线粒体是ROS的产生中心和主要攻击靶点,过量ROS有直接损伤足细胞、激活足细胞还原型烟酰胺腺嘌呤二核苷酸磷酸(reduced nicotinamide adenine dinucleotide phosphate,NADPH)氧化酶,通过p38MAPK/caspase-3途径诱导足细胞凋亡或影响胞内信号传递致足细胞凋亡等多种作用[36]
高糖刺激足细胞24 h可上调ROS激活自噬,其可能机制是ROS活化PERK,磷酸化eIF2α并氧化自噬相关蛋白4(autophagy protein 4,Atg4),促进微管相关蛋白1轻链3(microtubule-associated protein 1 light chain 3,LC3)生成的同时抑制mTOR活化,从而激活自噬[37]。长期暴露于高糖环境下则会抑制自噬,引起溶酶体功能障碍和足细胞凋亡。
4. 自噬: 哺乳动物细胞内自噬分为巨自噬(macroautophagy)、微自噬(microautophagy)和分子伴侣介导的自噬(chaperone-mediated autophagy),巨自噬即通常所称自噬。自噬更多地被认为是发生于亚细胞层面的促细胞存活机制,是真核生物对细胞内物质进行周转的重要过程。生理条件下,自噬可降解有毒的蛋白质聚合体以及老化受损的细胞器,在营养物质缺乏时则降解一些非必需物质, 产生能量和氨基酸等以维持细胞的基本功能[38]。在肾脏中,自噬主要发生于足细胞及近端肾小管细胞,足细胞是终末分化细胞,再生能力差,需要不断更新胞内细胞器来维持细胞的存活和正常功能,因此足细胞相较于其他肾脏固有细胞基础自噬水平更高[39]。自噬过程中胞质型LC3Ⅰ转化为膜结合型LC3Ⅱ标志着自噬体的形成,体外高糖刺激的永生鼠足细胞自噬相关蛋白LC3Ⅱ/LC3Ⅰ比值降低,足细胞标志蛋白synaptopodin显著减少,提示高糖可介导足细胞自噬缺失及损伤[40]。DM环境下,自噬失衡会改变足细胞结构和功能,自噬抑制可导致足细胞自我更新不足、发生空泡化及足突融合,加重足细胞损伤;上调STZ诱导的DKD小鼠细胞内自噬水平可缓解肾小球损伤及减少大量蛋白尿,起到足细胞保护作用[41]
真核细胞的巨自噬过程分为3个阶段:(1)吞噬泡(phagophore)阶段:自噬前体包绕待降解蛋白质、细胞器和部分胞质,形成杯状吞噬泡;(2)自噬体(autophagosome)阶段:吞噬泡延伸至形成封闭自噬体,完全包绕待降解蛋白质和细胞器;(3)自噬溶酶体(autolysosome)阶段:自噬体与溶酶体融合形成自噬溶酶体,其内容物被水解酶降解[42]。饥饿条件下,自噬体特异地在MAMs处形成,在包括DM等多种疾病中发现,敲除MAMs相关蛋白如Mfn2可导致自噬紊乱、自噬体蓄积甚至细胞凋亡[43]。Hamasaki等[44]发现,自噬前体标志物Atg14早期便募集到MAMs,启动自噬体的构建。
生理状态下,足细胞通过维持低水平mTOR活性和高水平自噬活性以促进细胞更新。在哺乳动物中mTOR信号通路负向调节自噬,涉及2种mTOR复合物,mTORC1及mTORC2。其中mTORC1被认为是自噬的直接调节因子,DM患者以及DM动物模型中均表现为mTORC1表达上调,而敲除或下调mTORC1的活性可增强自噬,有效缓解DM肾损伤。DM中主要通过活化PI3K/Akt途径和抑制单磷酸腺苷酸活化蛋白激酶(adenosine 5'-monophosphate-activated proteinkinase,AMPK)途径激活mTOR信号通路,从而抑制自噬,介导足细胞损伤和DKD进展:能量、营养状况等上游调控因子通过活化PI3K,激活Akt并活化mTORC1;AMPK通过磷酸化Unc-51样自噬活化激酶1(unc-51-like kinase 1,ULK1)直接调控自噬,这种相互作用可被mTORC1拮抗。调控IP3R通道Ca2+释放水平也可通过AMPK通路及mTOR通路调节自噬:ER外膜上的IP3R释放Ca2+分别进入胞质和线粒体,胞质内Ca2+可以活化钙调蛋白依赖性蛋白激酶β(calmodulin-dependent protein kinase β,CaMKKβ),激活AMPK,抑制mTOR从而间接启动自噬[45];线粒体基质内Ca2+浓度过低时,AMP/ATP比值升高,激活AMPK并活化ULK1,直接启动自噬[45-46]
5. 凋亡: 细胞凋亡是细胞死亡的最主要方式,生物体通过细胞凋亡的方式清除体内多余、受损或有害的细胞。细胞凋亡主要分为线粒体通路、受体通路和ER通路等3条途径,其中大部分的信号转导通路都与Ca2+相关,Ca2+通过激活凋亡执行者caspase各种亚型和Bcl-2家族蛋白介导细胞凋亡。MAMs蛋白可通过介导Ca2+的释放及摄取调控细胞凋亡,其反应的强度及结果主要受Bcl-2家族蛋白调控。Bcl-2家族中至少有18个不同的成员,根据它们在细胞凋亡中的作用、是否包含BH结构域及所包含BH结构域的数量可分为三类:BH3-only蛋白、多域抗凋亡蛋白和多域促凋亡蛋白。其中Bax和Bak通过上调Ca2+转运促凋亡,Bax和Bak过表达可加速凋亡相关的Ca2+进程[47]
足细胞损伤及凋亡增加现象在db/db小鼠和STZ诱导的DKD大鼠等多种动物模型、2型糖尿病患者肾组织、AngⅡ或高糖刺激的永生化足细胞中均可观察到[17,48-49]。多种机制参与调控高糖介导的内环境紊乱,导致肾脏足细胞凋亡。Ca2+稳态失调如IP3R的表达和磷酸化水平可调控Ca2+从ER转移到胞质或线粒体,调节细胞对凋亡的敏感性;线粒体数量减少、形态异常及功能障碍如ROS产生增加可诱导肾脏足细胞凋亡[48];在大部分细胞的凋亡过程中发现,Drp1、Mfn2与Bax、Bak高度共定位,上调Mfn2表达可引起Bax亚家族蛋白表达升高;严重或持续的ERS如UPR主要通过PERK/eIF2α/ATF4/CHOP通路、IRE1/c-jun氨基末端激酶(c-jun N-terminal kinases,JNK)通路和caspase-12通路介导足细胞凋亡。应用ERS抑制剂牛磺熊去氧胆酸(ursodeoxycholic acid,UDCA)或4-苯基丁酸(4-phenylbutyrate,4-PBA)可下调db/db小鼠肾脏组织中GRP78、p-PERK、CHOP、p-IRE1及caspase-12等蛋白表达,提示抑制ERS可减少DKD的足细胞凋亡[50]

三、 展望

综上所述,ER和线粒体是真核细胞中两大重要的功能单位,两者间建立了复杂的动态细胞内信号转导平台——MAMs。实际上MAMs处于一个不断形成和解离的过程,不同状态下通过改变表达于MAMs上的蛋白和信号传导通路影响细胞命运。为了更系统和定量地描述其动态过程尚需要更多的研究。尽管MAMs结构广泛存在于不同类型的细胞中,但特定细胞内MAMs是否由相同蛋白组成及其功能是否完全相同尚未可知。
传统DKD的临床治疗手段主要是控制血糖、血压和血脂等,但多数DKD患者仍发生进行性肾损伤[3]。DKD足细胞损伤因素如氧化应激、代谢紊乱等均受到MAMs调节,提示MAMs在维持足细胞内环境稳态上扮演着重要角色。本文探讨了DKD中Ca2+稳态失调、内质网应激和线粒体功能障碍诱导足细胞发生自噬和凋亡可能的作用机制。足细胞作为肾脏中的重要细胞屏障在DKD的发生发展中至关重要,通过调控MAMs相关蛋白的表达和信号转导通路可以改善DKD足细胞和其他肾脏固有细胞损伤,起到保护肾脏的作用。将MAMs作为足细胞损伤的干预靶点可为DKD的治疗带来新思路和转机。

References

[1]
Umanath K, Lewis JB. Update on diabetic nephropathy: core curriculum 2018[J]. Am J Kidney Dis, 2018, 71(6): 884-895. DOI: 10.1053/j.ajkd.2017.10.026.
Diabetic kidney disease and diabetic nephropathy are the leading cause of end-stage kidney disease in the United States and most developed countries. Diabetes accounts for 30% to 50% of the incident cases of end-stage kidney disease in the United States. Although this represents a significant public health concern, it is important to note that only 30% to 40% of patients with diabetes develop diabetic nephropathy. Specific treatment of patients with diabetic nephropathy can be divided into 4 major arenas: cardiovascular risk reduction, glycemic control, blood pressure control, and inhibition of the renin-angiotensin system (RAS). Recommendations for therapy include targeting a hemoglobin A concentration < 7% and blood pressure < 140/90mmHg with therapy anchored around the use of a RAS-blocking agent. The single best evidence-based therapy for diabetic nephropathy is therapy with a RAS-blocking medication. This Core Curriculum outlines and discusses in detail the epidemiology, pathophysiology, diagnosis, and management of diabetic nephropathy.Copyright © 2017 National Kidney Foundation, Inc. Published by Elsevier Inc. All rights reserved.
[2]
中华医学会糖尿病学分会. 中国2型糖尿病防治指南(2017年版)[J]. 中华糖尿病杂志, 2018, 10(1): 4-67. DOI: 10.3760/cma.j.issn.1674-5809.2018.01.003.
[3]
Lin YC, Chang YH, Yang SY, et al. Update of pathophysiology and management of diabetic kidney disease[J]. J Formos Med Assoc, 2018, 117(8): 662-675. DOI: 10.1016/j.jfma.2018.02.007.
[4]
Morré DJ, Merritt W, Lembi C. Connections between mitochondria and endoplasmic reticulum in rat liver and onion stem[J]. Protoplasma, 1971, 73(1): 43-49. DOI: 10.1007/bf01286410.
[5]
Vance JE. Phospholipid synthesis in a membrane fraction associated with mitochondria[J]. J Biol Chem, 1990, 265(13): 7248-7256.
A crude rat liver mitochondrial fraction that was capable of the rapid, linked synthesis of phosphatidylserine (PtdSer), phosphatidylethanolamine (PtdEtn), and phosphatidylcholine (PtdCho) labeled from [3-3H] serine has been fractionated. PtdSer synthase, PtdEtn methyltransferase, and CDP-choline:diacylglycerol cholinephosphotransferase activities were present in the crude mitochondrial preparation but were absent from highly purified mitochondria and could be attributed to the presence of a membrane fraction, X. Thus, previous claims of the mitochondrial location of some of these enzymes might be explained by the presence of fraction X in the mitochondrial preparation. Fraction X had many similarities to microsomes except that it sedimented with mitochondria (at 10,000 x g). However, the specific activities of PtdSer synthase and glucose-6-phosphate phosphatase in fraction X were almost twice that of microsomes, and the specific activities of CTP:phosphocholine cytidylyltransferase and NADPH:cytochrome c reductase in fraction X were much lower than in microsomes. The marker enzymes for mitochondria, Golgi apparatus, plasma membrane, lysosomes, and peroxisomes all had low activities in fraction X. Polyacrylamide gel electrophoresis revealed distinct differences, as well as similarities, among the proteins of fraction X, microsomes, and rough and smooth endoplasmic reticulum. The combined mitochondria-fraction X membranes can synthesize PtdSer, PtdEtn, and PtdCho from serine. Thus, fraction X in combination with mitochondria might be responsible for the observed compartmentalization of a serine-labeled pool of phospholipids previously identified (Vance, J. E., and Vance, D. E. (1986) J. Biol. Chem. 261, 4486-4491) and might be involved in the transfer of lipids between the endoplasmic reticulum and mitochondria.
[6]
Filadi R, Theurey P, Pizzo P. The endoplasmic reticulum-mitochondria coupling in health and disease: molecules, functions and significance[J]. Cell Calcium, 2017, 62: 1-15. DOI: 10.1016/j.ceca.2017.01.003.
The close apposition between endoplasmic reticulum (ER) and mitochondria represents a key platform, capable to regulate different fundamental cellular pathways. Among these, Ca signaling and lipid homeostasis have been demonstrated over the last years to be deeply modulated by ER-mitochondria cross-talk. Given its importance in cell life/death decisions, increasing evidence suggests that alterations of the ER-mitochondria axis could be responsible for the onset and progression of several diseases, including neurodegeneration, cancer and obesity. However, the molecular identity of the proteins controlling this inter-organelle apposition is still debated. In this review, we summarize the main cellular pathways controlled by ER-mitochondria appositions, focusing on the principal molecules reported to be involved in this interplay and on those diseases for which alterations in organelles communication have been reported.Copyright © 2017 Elsevier Ltd. All rights reserved.
[7]
Yuan L, Liu Q, Wang Z, et al. EI24 tethers endoplasmic reticulum and mitochondria to regulate autophagy flux[J]. Cell Mol Life Sci, 2020, 77(8): 1591-1606. DOI: 10.1007/s00018-019-03236-9.
Etoposide-induced protein 2.4 (EI24), located on the endoplasmic reticulum (ER) membrane, has been proposed to be an essential autophagy protein. Specific ablation of EI24 in neuronal and liver tissues causes deficiency of autophagy flux. However, the molecular mechanism of the EI24-mediated autophagy process is still poorly understood. Like neurons and hepatic cells, pancreatic β cells are also secretory cells. Pancreatic β cells contain large amounts of ER and continuously synthesize and secrete insulin to maintain blood glucose homeostasis. Yet, the effect of EI24 on autophagy of pancreatic β cells has not been reported. Here, we show that the autophagy process is inhibited in EI24-deficient primary pancreatic β cells. Further mechanistic studies demonstrate that EI24 is enriched at the ER-mitochondria interface and that the C-terminal domain of EI24 is important for the integrity of the mitochondria-associated membrane (MAM) and autophagy flux. Overexpression of EI24, but not the EI24-ΔC mutant, can rescue MAM integrity and decrease the aggregation of p62 and LC3II in the EI24-deficient group. By mass spectrometry-based proteomics following immunoprecipitation, EI24 was found to interact with voltage-dependent anion channel 1 (VDAC1), inositol 1,4,5-trisphosphate receptor (IP3R), and the outer mitochondrial membrane chaperone GRP75. Knockout of EI24 impairs the interaction of IP3R with VDAC1, indicating that these proteins may form a quaternary complex to regulate MAM integrity and the autophagy process.
[8]
Alam S, Abdullah CS, Aishwarya R, et al. Sigmar1 regulates endoplasmic reticulum stress-induced C/EBP-homologous protein expression in cardiomyocytes[J]. Biosci Rep, 2017, 37(4): BSR20170898. DOI: 10.1042/BSR20170898.
C/EBP-homologous protein (CHOP) is a ubiquitously expressed stress-inducible transcription factor robustly induced by maladaptive endoplasmic reticulum (ER) stresses in a wide variety of cells. Here, we examined a novel function of Sigma 1 receptor (Sigmar1) in regulating CHOP expression under ER stress in cardiomyocytes. We also defined Sigmar1-dependent activation of the adaptive ER-stress pathway in regulating CHOP expression. We used adenovirus-mediated Sigmar1 overexpression as well as Sigmar1 knockdown by siRNA in neonatal rat ventricular cardiomyocytes (NRCs); to induce ER stress, cardiomyocytes were treated with tunicamycin. Sigmar1-siRNA knockdown significantly increased the expression of CHOP and significantly induced cellular toxicity by sustained activation of ER stress in cardiomyocytes. Sigmar1 overexpression decreased the expression of CHOP and significantly decreased cellular toxicity in cells. Using biochemical and immunocytochemical experiments, we also defined the specific ER-stress pathway associated with Sigmar1-dependent regulation of CHOP expression and cellular toxicity. We found that Sigmar1 overexpression significantly increased inositol requiring kinase 1α (IRE1α) phosphorylation and increased spliced X-box-binding proteins (XBP1s) expression as well as nuclear localization. In contrast, Sigmar1 knockdown significantly decreased IRE1α phosphorylation and decreased XBP1s expression as well as nuclear transport. Taken together, these results indicate that Sigmar1-dependent activation of IRE1α-XBP1s ER-stress response pathways are associated with inhibition of CHOP expression and suppression of cellular toxicity. Hence, Sigmar1 is an essential component of the adaptive ER-stress response pathways eliciting cellular protection in cardiomyocytes.
[9]
Bernard-Marissal N, van Hameren G, Juneja M, et al. Altered interplay between endoplasmic reticulum and mitochondria in Charcot - Marie - Tooth type 2A neuropathy[J]. Proc Natl Acad Sci U S A, 2019, 116(6): 2328-2337. DOI: 10.1073/pnas.1810932116.
Interactions between mitochondria and the endoplasmic reticulum (ER) at the level of mitochondria-associated membranes (MAM) constitute a key signaling hub, emerging as a shared target altered in multiple neurodegenerative diseases. We use both in vivo and in vitro models of Charcot–Marie–Tooth type 2A, an axonal form of neuropathy, to demonstrate that the presence of mutated Mitofusin 2 leads to altered MAM. In neurons, these modifications occur concomitantly with activation of ER stress response, dysregulated calcium handling, and alterations in mitochondrial morphology and transport, collectively contributing to axonopathy. Importantly, the reported results indicate that the pathological consequences of mutated Mitofusin 2 may be targeted with drugs reinforcing the ER–mitochondria cross-talk and/or reducing ER stress.
[10]
Friedman JR, Lackner LL, West M, et al. ER tubules mark sites of mitochondrial division[J]. Science, 2011, 334(6054): 358-362. DOI: 10.1126/science.1207385.
Mitochondrial structure and distribution are regulated by division and fusion events. Mitochondrial division is regulated by Dnm1/Drp1, a dynamin-related protein that forms helices around mitochondria to mediate fission. Little is known about what determines sites of mitochondrial fission within the mitochondrial network. The endoplasmic reticulum (ER) and mitochondria exhibit tightly coupled dynamics and have extensive contacts. We tested whether ER plays a role in mitochondrial division. We found that mitochondrial division occurred at positions where ER tubules contacted mitochondria and mediated constriction before Drp1 recruitment. Thus, ER tubules may play an active role in defining the position of mitochondrial division sites.
[11]
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.
[12]
Wang X, Gao L, Lin H, et al. Mangiferin prevents diabetic nephropathy progression and protects podocyte function via autophagy in diabetic rat glomeruli[J]. Eur J Pharmacol, 2018, 824: 170-178. DOI: 10.1016/j.ejphar.2018.02.009.
Diabetic nephropathy (DN) is one of the most severe microangiopathies of diabetes mellitus and is a leading cause of end stage renal disease. Numerous studies suggest that podocyte injury contributes to progressive proteinuria. Podocytes are highly specialized, terminally differentiated cells that are unable to proliferate, autophagy plays a key role in maintaining the structure and function of podocytes. Autophagy impairment is involved in the pathogenesis of podocyte loss, which leads to massive proteinuria in DN. In the present study, we investigated the effects of mangiferin on nephropathy in streptozotocin (STZ)-induced diabetic rats; we focused on pathological factors related to autophagy in podocytes and the AMPK-mTOR-ULK1 pathway. The results showed that chronic treatment with mangiferin significantly decreased albuminuria, inhibited glomerular extracellular matrix expansion and restored the expression of nephrin, a podocyte marker, in diabetic rats; these results suggest that mangiferin delayed the process of DN and protected the podocytes. In addition, mangiferin induced autophagy, as shown by the up-regulation of LC3 II and the down-regulation of p62 in both DN rats and podocytes. Transmission electron microscope analyses showed that mangiferin increased the number of autophagosomes in the podocytes of DN rats. This underlying mechanism was associated with the up-regulation of AMPK phosphorylation, the down-regulation of mTOR phosphorylation and the up-regulation of p-ULK1. Taken together, mangiferin delayed the progression of DN and protected the podocytes by enhancing autophagy under diabetic conditions via the AMPK-mTOR-ULK1 pathway. These findings provide new insights into the molecular mechanisms underlying the renoprotective effects of mangiferin in DN.Copyright © 2018 Elsevier B.V. All rights reserved.
[13]
杨倩, 梁伟, 丁国华. 足细胞损伤与糖尿病肾病[J]. 中国医学前沿杂志(电子版), 2012, (9): 10-14. DOI: 10.3969/j.issn.1674-7372.2012.09.005.
[14]
Rizzuto R, Pinton P, Carrington W, et al. Close contacts with the endoplasmic reticulum as determinants of mitochondrial Ca2+ responses[J]. Science, 1998, 280(5370): 1763-1766. DOI: 10.1126/science.280.5370.1763.
The spatial relation between mitochondria and endoplasmic reticulum (ER) in living HeLa cells was analyzed at high resolution in three dimensions with two differently colored, specifically targeted green fluorescent proteins. Numerous close contacts were observed between these organelles, and mitochondria in situ formed a largely interconnected, dynamic network. A Ca2+-sensitive photoprotein targeted to the outer face of the inner mitochondrial membrane showed that, upon opening of the inositol 1,4,5-triphosphate (IP3)-gated channels of the ER, the mitochondrial surface was exposed to a higher concentration of Ca2+ than was the bulk cytosol. These results emphasize the importance of cell architecture and the distribution of organelles in regulation of Ca2+ signaling.
[15]
Guo H, Wang Y, Zhang X, et al. Astragaloside IV protects against podocyte injury via SERCA2-dependent ER stress reduction and AMPKα-regulated autophagy induction in streptozotocin-induced diabetic nephropathy[J]. Sci Rep, 2017, 7(1): 6852. DOI: 10.1038/s41598-017-07061-7.
Aberrant endoplasmic reticulum (ER) stress and autophagy are associated with diabetic nephropathy. Here we investigated the effect of astragaloside IV (AS-IV) on the progression of diabetic nephropathy (DN) and the underlying mechanism involving ER stress and autophagy in streptozotocin (STZ)-induced diabetic mice and high glucose (HG)-incubated podocytes. The diabetic mice developed progressive albuminuria and glomerulosclerosis within 8 weeks, which were significantly ameliorated by AS-IV treatment in a dose-dependent manner. Moreover, diabetes or HG-induced podocyte apoptosis was markedly attenuated by AS-IV, paralleled by a marked remission in ER stress and a remarkable restoration in impaired autophagy, which were associated with a significant improvement in the expression of sarcoendoplasmic reticulum Ca2+ ATPase 2b (SERCA2b) and AMP-activated protein kinase alpha (AMPK alpha) phosphorylation, respectively. Knockdown of SERCA2 in podocytes induced ER stress and largely abolished the protective effect of AS-IV, but had no obvious effect on the expression of autophagy-associated proteins. On the other hand, blockade of either autophagy induction or AMPK alpha activation could also significantly mitigate AS-IV-induced beneficial effect. Collectively, these results suggest that AS-IV prevented the progression of DN, which is mediated at least in part by SERCA2-dependent ER stress attenuation and AMPK alpha-promoted autophagy induction.
[16]
Xing Y, Wang M, Wang J, et al. Dimerization of MICU proteins controls Ca2+ influx through the mitochondrial Ca2+ uniporter[J]. Cell Rep, 2019, 26(5): 1203-1212.e4. DOI: 10.1016/j.celrep.2019.01.022.
[17]
Gao Y, Zhang M, Zhang R, et al. Whole grain brown rice extrudate ameliorates the symptoms of diabetes by activating the IRS1/PI3K/AKT insulin pathway in db/db Mice[J]. J Agric Food Chem, 2019, 67(42): 11657-11664. DOI: 10.1021/acs.jafc.9b04684.
[18]
Zhang Y, Zhang Y, Yu Y. Global phosphoproteomic analysis of insulin/Akt/mTORC1/S6K signaling in rat hepatocytes[J]. J Proteome Res, 2017, 16(8): 2825-2835. DOI: 10.1021/acs.jproteome.7b00140.
Insulin resistance is a hallmark of type 2 diabetes. Although multiple genetic and physiological factors interact to cause insulin resistance, deregulated signaling by phosphorylation is a common underlying mechanism. In particular, the specific phosphorylation-dependent regulatory mechanisms and signaling outputs of insulin are poorly understood in hepatocytes, which represents one of the most important insulin-responsive cell types. Using primary rat hepatocytes as a model system, we performed reductive dimethylation (ReDi)-based quantitative mass spectrometric analysis and characterized the phosphoproteome that is regulated by insulin as well as its key downstream kinases including Akt, mTORC1, and S6K. We identified a total of 12 294 unique, confidently localized phosphorylation sites and 3805 phosphorylated proteins in this single cell type. Detailed bioinformatic analysis on each individual data set identified both known and previously unrecognized targets of this key insulin downstream effector pathway. Furthermore, integrated analysis of the hepatic Akt/mTORC1/S6K signaling axis allowed the delineation of the substrate specificity of several close-related kinases within the insulin signaling pathway. We expect that the data sets will serve as an invaluable resource, providing the foundation for future hypothesis-driven research that helps delineate the molecular mechanisms that underlie the pathogenesis of type 2 diabetes and related metabolic syndrome.
[19]
Shinjo S, Jiang S, Nameta M, et al. Disruption of the mitochondria-associated ER membrane (MAM) plays a central role in palmitic acid-induced insulin resistance[J]. Exp Cell Res, 2017, 359(1): 86-93. DOI: 10.1016/j.yexcr.2017.08.006.
The mitochondria-associated ER membrane (MAM) is a specialized subdomain of ER that physically connects with mitochondria. Although disruption of inter-organellar crosstalk via the MAM impairs cellular homeostasis, its pathological significance in insulin resistance in type 2 diabetes mellitus remains unclear. Here, we reveal the importance of reduced MAM formation in the induction of fatty acid-evoked insulin resistance in hepatocytes. Palmitic acid (PA) repressed insulin-stimulated Akt phosphorylation in HepG2 cells within 12h. Treatment with an inhibitor of the ER stress response failed to restore PA-mediated suppression of Akt activation. Mitochondrial reactive oxygen species (ROS) production did not increase in PA-treated cells. Even short-term exposure (3h) to PA reduced the calcium flux from ER to mitochondria, followed by a significant decrease in MAM contact area, suggesting that PA suppressed the functional interaction between ER and mitochondria. Forced expression of mitofusin-2, a critical component of the MAM, partially restored MAM contact area and ameliorated the PA-elicited suppression of insulin sensitivity with Ser473 phosphorylation of Akt selectively improved. These results suggest that loss of proximity between ER and mitochondria, but not perturbation of homeostasis in the two organelles individually, plays crucial roles in PA-evoked Akt inactivation in hepatic insulin resistance.Copyright © 2017 Elsevier Inc. All rights reserved.
[20]
Betz C, Stracka D, Prescianotto-Baschong C, et al. Feature article: mTOR complex 2-Akt signaling at mitochondria-associated endoplasmic reticulum membranes (MAM) regulates mitochondrial physiology[J]. Proc Natl Acad Sci U S A, 2013, 110(31): 12526-12534. DOI: 10.1073/pnas.1302455110.
\n The target of rapamycin (TOR) is a highly conserved protein kinase and a central controller of growth. Mammalian TOR complex 2 (mTORC2) regulates AGC kinase family members and is implicated in various disorders, including cancer and diabetes. Here we report that mTORC2 is localized to the endoplasmic reticulum (ER) subcompartment termed mitochondria-associated ER membrane (MAM). mTORC2 localization to MAM was growth factor-stimulated, and mTORC2 at MAM interacted with the IP\n 3\n receptor (IP3R)-Grp75–voltage-dependent anion-selective channel 1 ER-mitochondrial tethering complex. mTORC2 deficiency disrupted MAM, causing mitochondrial defects including increases in mitochondrial membrane potential, ATP production, and calcium uptake. mTORC2 controlled MAM integrity and mitochondrial function via Akt mediated phosphorylation of the MAM associated proteins IP3R, Hexokinase 2, and phosphofurin acidic cluster sorting protein 2. Thus, mTORC2 is at the core of a MAM signaling hub that controls growth and metabolism.\n
[21]
Chaudhari N, Talwar P, Parimisetty A, et al. A molecular web: endoplasmic reticulum stress, inflammation, and oxidative stress[J]. Front Cell Neurosci, 2014, 8: 213. DOI: 10.3389/fncel.2014.00213.
Execution of fundamental cellular functions demands regulated protein folding homeostasis. Endoplasmic reticulum (ER) is an active organelle existing to implement this function by folding and modifying secretory and membrane proteins. Loss of protein folding homeostasis is central to various diseases and budding evidences suggest ER stress as being a major contributor in the development or pathology of a diseased state besides other cellular stresses. The trigger for diseases may be diverse but, inflammation and/or ER stress may be basic mechanisms increasing the severity or complicating the condition of the disease. Chronic ER stress and activation of the unfolded-protein response (UPR) through endogenous or exogenous insults may result in impaired calcium and redox homeostasis, oxidative stress via protein overload thereby also influencing vital mitochondrial functions. Calcium released from the ER augments the production of mitochondrial Reactive Oxygen Species (ROS).Toxic accumulation of ROS within ER and mitochondria disturbs fundamental organelle functions. Sustained ER stress is known to potentially elicit inflammatory responses via UPR pathways. Additionally, ROS generated through inflammation or mitochondrial dysfunction could accelerate ER malfunction. Dysfunctional UPR pathways have been associated with a wide range of diseases including several neurodegenerative diseases, stroke, metabolic disorders, cancer, inflammatory disease, diabetes mellitus, cardiovascular disease, and others. In this review, we have discussed the UPR signaling pathways, and networking between ER stress-induced inflammatory pathways, oxidative stress, and mitochondrial signaling events, which further induce or exacerbate ER stress.
[22]
Simmen T, Aslan JE, Blagoveshchenskaya AD, et al. PACS-2 controls endoplasmic reticulum-mitochondria communication and bid-mediated apoptosis[J]. EMBO J, 2005, 24(4): 717-729. DOI: 10.1038/sj.emboj.7600559.
The endoplasmic reticulum (ER) and mitochondria form contacts that support communication between these two organelles, including synthesis and transfer of lipids, and the exchange of calcium, which regulates ER chaperones, mitochondrial ATP production, and apoptosis. Despite the fundamental roles for ER-mitochondria contacts, little is known about the molecules that regulate them. Here we report the identification of a multifunctional sorting protein, PACS-2, that integrates ER-mitochondria communication, ER homeostasis, and apoptosis. PACS-2 controls the apposition of mitochondria with the ER, as depletion of PACS-2 causes BAP31-dependent mitochondria fragmentation and uncoupling from the ER. PACS-2 also controls formation of ER lipid-synthesizing centers found on mitochondria-associated membranes and ER homeostasis. However, in response to apoptotic inducers, PACS-2 translocates Bid to mitochondria, which initiates a sequence of events including the formation of mitochondrial truncated Bid, the release of cytochrome c, and the activation of caspase-3, thereby causing cell death. Together, our results identify PACS-2 as a novel sorting protein that links the ER-mitochondria axis to ER homeostasis and the control of cell fate, and provide new insights into Bid action.
[23]
Dong Z, Wu P, Li Y, et al. Myocardial infarction worsens glomerular injury and microalbuminuria in rats with pre-existing renal impairment accompanied by the activation of ER stress and inflammation[J]. Mol Biol Rep, 2014, 41(12): 7911-7921. DOI: 10.1007/s11033-014-3685-5.
Deterioration of renal function occurs after chronic heart failure in approximately one-third of patients, particularly in those with pre-existing renal impairment such as diabetic nephropathy. Impaired renal function in these patients is always associated with a worse prognosis. However, the mechanisms underlying such deterioration of renal function are still largely unknown. In three separate protocols, we compared 1) sham operation (Ctr, n = 10) with surgically induced myocardial infarction (MI, n = 10); 2) unilateral nephrectomy (UNX, n = 10) with UNX + MI (n = 10); and 3) STZ-induced type 1 diabetes (DB, n = 10) with DB + MI (n = 10). The differences between combined injury models (UNX + MI, DB + MI) and simple MI were also examined. Renal remodeling, function, ER stress (CHOP and GRP78) and inflammation (infiltration of inflammatory cells, NF-κB p65) were evaluated 12 weeks after MI. In common SD rats, MI activated less glomerular ER stress and inflammation, resulting in a minor change of glomerular remodeling and microalbuminuria. However, MI significantly increased the glomerular expression of GRP78 and CHOP in UNX and DB rats. In addition, it also promoted the infiltration of CD4+ T cells, particularly inflammatory cytokine (IFN-γ, IL-17, IL-4)-producing CD4+ T cells, and the expression of NF-κB p65 in the glomeruli. By contrast, significant glomerular fibrosis, glomerulosclerosis, podocyte injury and microalbuminuria were found in rats with UNX + MI and DB + MI. MI significantly increased chronic glomerular injury and microalbuminuria at 12 weeks in rats with pre-existing renal impairment, i.e., UNX and DB, but not common SD rats. These changes were accompanied by increased glomerular ER stress and immune-associated inflammation.
[24]
Muñoz JP, Ivanova S, Sánchez-Wandelmer J, et al. Mfn2 modulates the UPR and mitochondrial function via repression of PERK[J]. EMBO J, 2013, 32(17): 2348-2361. DOI: 10.1038/emboj.2013.168.
Mitofusin 2 (Mfn2) is a key protein in mitochondrial fusion and it participates in the bridging of mitochondria to the endoplasmic reticulum (ER). Recent data indicate that Mfn2 ablation leads to ER stress. Here we report on the mechanisms by which Mfn2 modulates cellular responses to ER stress. Induction of ER stress in Mfn2-deficient cells caused massive ER expansion and excessive activation of all three Unfolded Protein Response (UPR) branches (PERK, XBP-1, and ATF6). In spite of an enhanced UPR, these cells showed reduced activation of apoptosis and autophagy during ER stress. Silencing of PERK increased the apoptosis of Mfn2-ablated cells in response to ER stress. XBP-1 loss-of-function ameliorated autophagic activity of these cells upon ER stress. Mfn2 physically interacts with PERK, and Mfn2-ablated cells showed sustained activation of this protein kinase under basal conditions. Unexpectedly, PERK silencing in these cells reduced ROS production, normalized mitochondrial calcium, and improved mitochondrial morphology. In summary, our data indicate that Mfn2 is an upstream modulator of PERK. Furthermore, Mfn2 loss-of-function reveals that PERK is a key regulator of mitochondrial morphology and function.
[25]
Carreras - Sureda A, Pihán P, Hetz C. The unfolded protein response: at the intersection between endoplasmic reticulum function and mitochondrial bioenergetics[J]. Front Oncol, 2017, 7: 55. DOI: 10.3389/fonc.2017.00055.
Endoplasmic reticulum (ER) to mitochondria communication has emerged in recent years as a signaling hub regulating cellular physiology with a relevant contribution to diseases including cancer and neurodegeneration. This functional integration is exerted through discrete interorganelle structures known as mitochondria-associated membranes (MAMs). At these domains, ER/mitochondria physically associate to dynamically adjust metabolic demands and the response to stress stimuli. Here, we provide a focused overview of how the ER shapes the function of the mitochondria, giving a special emphasis to the significance of local signaling of the unfolded protein response at MAMs. The implications to cell fate control and the progression of cancer are also discussed.
[26]
Kang MK, Park SH, Kim YH, et al. Chrysin ameliorates podocyte injury and slit diaphragm protein loss via inhibition of the PERK-eIF2α-ATF-CHOP pathway in diabetic mice[J]. Acta Pharmacol Sin, 2017, 38(8): 1129-1140. DOI: 10.1038/aps.2017.30.
[27]
Yano H, Bonifazi A, Xu M, et al. Pharmacological profiling of sigma 1 receptor ligands by novel receptor homomer assays[J]. Neuropharmacology, 2018, 133: 264-275. DOI: 10.1016/j.neuropharm.2018.01.042.
The sigma 1 receptor (σR) is a structurally unique transmembrane protein that functions as a molecular chaperone in the endoplasmic reticulum (ER), and has been implicated in cancer, neuropathic pain, and psychostimulant abuse. Despite physiological and pharmacological significance, mechanistic underpinnings of structure-function relationships of σR are poorly understood, and molecular interactions of selective ligands with σR have not been elucidated. The recent crystallographic determination of σR as a homo-trimer provides the foundation for mechanistic elucidation at the molecular level. Here we report novel bioluminescence resonance energy transfer (BRET) assays that enable analyses of ligand-induced multimerization of σR and its interaction with BiP. Haloperidol, PD144418, and 4-PPBP enhanced σR homomer BRET signals in a dose dependent manner, suggesting their significant effects in stabilizing σR multimerization, whereas (+)-pentazocine and several other ligands do not. In non-denaturing gels, (+)-pentazocine significantly decreased whereas haloperidol increased the fraction of σR multimers, consistent with the results from the homomer BRET assay. Further, BRET assays examining heteromeric σR-BiP interaction revealed that (+)-pentazocine and haloperidol induced opposite trends of signals. From molecular modeling and simulations of σR in complex with the tested ligands, we identified initial clues that may lead to the differed responses of σR upon binding of structurally diverse ligands. By combining multiple in vitro pharmacological and in silico molecular biophysical methods, we propose a novel integrative approach to analyze σR-ligand binding and its impact on interaction of σR with client proteins.Published by Elsevier Ltd.
[28]
苏维维, 丁桂霞, 朱春华, 等. Sig-1R基因表达抑制对内质网应激和足细胞损伤的影响[J]. 南京医科大学学报(自然科学版), 2011, 31(5): 667-672.
[29]
Reiter RJ, Tan DX, Rosales-Corral S, et al. Melatonin mitigates mitochondrial meltdown: interactions with SIRT3[J]. Int J Mol Sci, 2018, 19(8): 2439. DOI: 10.3390/ijms19082439.
Melatonin exhibits extraordinary diversity in terms of its functions and distribution. When discovered, it was thought to be uniquely of pineal gland origin. Subsequently, melatonin synthesis was identified in a variety of organs and recently it was shown to be produced in the mitochondria. Since mitochondria exist in every cell, with a few exceptions, it means that every vertebrate, invertebrate, and plant cell produces melatonin. The mitochondrial synthesis of melatonin is not photoperiod-dependent, but it may be inducible under conditions of stress. Mitochondria-produced melatonin is not released into the systemic circulation, but rather is used primarily in its cell of origin. Melatonin’s functions in the mitochondria are highly diverse, not unlike those of sirtuin 3 (SIRT3). SIRT3 is an NAD+-dependent deacetylase which regulates, among many functions, the redox state of the mitochondria. Recent data proves that melatonin and SIRT3 post-translationally collaborate in regulating free radical generation and removal from mitochondria. Since melatonin and SIRT3 have cohabitated in the mitochondria for many eons, we predict that these molecules interact in many other ways to control mitochondrial physiology. It is predicted that these mutual functions will be intensely investigated in the next decade and importantly, we assume that the findings will have significant applications for preventing/delaying some age-related diseases and aging itself.
[30]
Yang S, Han Y, Liu J, et al. Mitochondria: a novel therapeutic target in diabetic nephropathy[J]. Curr Med Chem, 2017, 24(29): 3185-3202. DOI: 10.2174/0929867324666170509121003.
Diabetic nephropathy (DN) is an important diabetic microvascular complication, and it is becoming the leading cause of end-stage renal disease worldwide. Unfortunately, there are no effective therapies to treat established DN. Therefore, new therapeutic targets are urgently required. Accumulating studies indicate that mitochondrial dysfunction is central to the pathogenesis of DN, and therapies targeted mitochondria might effectively delay the progression of DN.A structured search of previously research literature about mitochondrial structure and function, mitochondrial DNA, mitochondrial biogenesis, mitochondrial dynamics change, mitophagy, mitochondrial ROS, mitochondrial apoptosis and therapies targeted mitochondria has been performed in several databases.176 papers were included in this review, the results from these papers indicated that mitochondrial dysfunction is a pivotal issue for the development of DN, such as elevated oxidative stress induced by disorders of the mitochondrial respiratory chain complex and mitochondrial dynamic disorders, mutation of mitochondrial DNA, mitochondrial abnormal biogenesis, mitochondrial excessive fission, mitochondrial ROS overproduction. In addition, several therapeutic agents targeting the mitochondria (e.g mitochondrial ROS modulators, mitochondrial fragmentation inhibitors and mitochondrial biogenesis activators) have shown perfect therapeutic effect and security for DN.The finding of this review has further confirmed the vital role of mitochondrial dysfunction in the progression of DN, management strategies for recovering the normal mitochondrial function will offer potential novel therapeutic targets for DN.Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.org.
[31]
Ma Y, Chen Z, Tao Y, et al. Increased mitochondrial fission of glomerular podocytes in diabetic nephropathy[J]. Endocr Connect, 2019, 8(8): 1206-1212. DOI: 10.1530/EC-19-0234.
Previous studies showed that abnormal mitochondrial structure and function were involved in the pathological process of diabetic nephropathy (DN). The dynamic mitochondrial processes, including fusion and fission, maintain the mass and quantity of mitochondria. Podocyte injury is a critical factor in the development and progression of DN. The present study evaluated the mitochondrial fission of podocytes in patients with DN.We recruited 31 patients with biopsy-confirmed DN. A quantitative analysis of the mitochondrial morphology was conducted with electron microscopy using a computer-assisted morphometric analysis application to calculate the aspect ratio values. Immunofluorescence assays were used to evaluate protein colocalization in the glomeruli of patients.The urine protein level was significantly increased in DN patients compared to non-DN patients (P < 0.001), and the mitochondria in the podocytes from DN patients were more fragmentated than those from patients without DN. The mitochondrial aspect ratio values were negatively correlated with the proteinuria levels (r = -0.574, P = 0.01), and multiple regression analysis verified that the mitochondrial aspect ratio was significantly and independently associated with the urine protein level (β = -0.519, P = 0.007). In addition, Drp1, a mitochondrial fission factor, preferentially combines with AKAP1, which is located in the mitochondrial membrane.In the podocytes of DN patients, mitochondrial fragmentation was increased, and mitochondrial aspect ratio values were correlated with the proteinuria levels. The AKAP1-Drp1 pathway may contribute to mitochondrial fission in the pathogenesis of DN.
[32]
Chen Z, Ma Y, Yang Q, et al. AKAP1 mediates high glucose-induced mitochondrial fission through the phosphorylation of Drp1 in podocytes[J]. J Cell Physiol, 2020, 235(10): 7433-7448. DOI: 10.1002/jcp.29646.
Increasing evidence suggests that mitochondrial dysfunction plays a critical role in the development of diabetic kidney disease (DKD), however, its specific pathomechanism remains unclear. A-kinase anchoring protein (AKAP) 1 is a scaffold protein in the AKAP family that is involved in mitochondrial fission and fusion. Here, we show that rats with streptozotocin (STZ)-induced diabetes developed podocyte damage accompanied by AKAP1 overexpression and that AKAP1 closely interacted with the mitochondrial fission enzyme dynamin-related protein 1 (Drp1). At the molecular level, high glucose (HG) promoted podocyte injury and Drp1 phosphorylation at Ser637 as proven by decreased mitochondrial membrane potential, elevated reactive oxygen species generation, reduced adenosine triphosphate synthesis, and increased podocyte apoptosis. Furthermore, the AKAP1 knockdown protected HG-induced podocyte injury and suppressed HG-induced Drp1 phosphorylation at Ser637. AKAP1 overexpression aggravated HG-induced mitochondrial fragmentation and podocyte apoptosis. The coimmunoprecipitation assay showed that HG-induced Drp1 interacted with AKAP1, revealing that AKAP1 could recruit Drp1 from the cytoplasm under HG stimulation. Subsequently, we detected the effect of drp1 phosphorylation on Ser637 by transferring several different Drp1 mutants. We demonstrated that activated AKAP1 promoted Drp1 phosphorylation at Ser637, which promoted the transposition of Drp1 to the surface of the mitochondria and accounts for mitochondrial dysfunction events. These findings indicate that AKAP1 is the main pathogenic factor in the development and progression of HG-induced podocyte injury through the destruction of mitochondrial dynamic homeostasis by regulating Drp1 phosphorylation in human podocytes.© 2020 Wiley Periodicals, Inc.
[33]
Uren RT, O'Hely M, Iyer S, et al. Disordered clusters of Bak dimers rupture mitochondria during apoptosis[J]. Elife, 2017, 6: e19944. DOI: 10.7554/eLife.19944.
During apoptosis, Bak and Bax undergo major conformational change and form symmetric dimers that coalesce to perforate the mitochondrial outer membrane via an unknown mechanism. We have employed cysteine labelling and linkage analysis to the full length of Bak in mitochondria. This comprehensive survey showed that in each Bak dimer the N-termini are fully solvent-exposed and mobile, the core is highly structured, and the C-termini are flexible but restrained by their contact with the membrane. Dimer-dimer interactions were more labile than the BH3:groove interaction within dimers, suggesting there is no extensive protein interface between dimers. In addition, linkage in the mobile Bak N-terminus (V61C) specifically quantified association between dimers, allowing mathematical simulations of dimer arrangement. Together, our data show that Bak dimers form disordered clusters to generate lipidic pores. These findings provide a molecular explanation for the observed structural heterogeneity of the apoptotic pore.
[34]
Amodio G, Moltedo O, Faraonio R, et al. Targeting the endoplasmic reticulum unfolded protein response to counteract the oxidative stress-induced endothelial dysfunction[J]. Oxid Med Cell Longev, 2018, 2018: 4946289. DOI: 10.1155/2018/4946289.
[35]
Coughlan MT, Nguyen TV, Penfold SA, et al. Mapping time-course mitochondrial adaptations in the kidney in experimental diabetes[J]. Clin Sci (Lond), 2016, 130(9): 711-720. DOI: 10.1042/CS20150838.
Oxidative phosphorylation (OXPHOS) drives ATP production by mitochondria, which are dynamic organelles, constantly fusing and dividing to maintain kidney homoeostasis. In diabetic kidney disease (DKD), mitochondria appear dysfunctional, but the temporal development of diabetes-induced adaptations in mitochondrial structure and bioenergetics have not been previously documented. In the present study, we map the changes in mitochondrial dynamics and function in rat kidney mitochondria at 4, 8, 16 and 32 weeks of diabetes. Our data reveal that changes in mitochondrial bioenergetics and dynamics precede the development of albuminuria and renal histological changes. Specifically, in early diabetes (4 weeks), a decrease in ATP content and mitochondrial fragmentation within proximal tubule epithelial cells (PTECs) of diabetic kidneys were clearly apparent, but no changes in urinary albumin excretion or glomerular morphology were evident at this time. By 8 weeks of diabetes, there was increased capacity for mitochondrial permeability transition (mPT) by pore opening, which persisted over time and correlated with mitochondrial hydrogen peroxide (H2O2) generation and glomerular damage. Late in diabetes, by week 16, tubular damage was evident with increased urinary kidney injury molecule-1 (KIM-1) excretion, where an increase in the Complex I-linked oxygen consumption rate (OCR), in the context of a decrease in kidney ATP, indicated mitochondrial uncoupling. Taken together, these data show that changes in mitochondrial bioenergetics and dynamics may precede the development of the renal lesion in diabetes, and this supports the hypothesis that mitochondrial dysfunction is a primary cause of DKD.© 2016 Authors; published by Portland Press Limited.
[36]
Lee SH, Moon SJ, Paeng J, et al. Podocyte hypertrophy precedes apoptosis under experimental diabetic conditions[J]. Apoptosis, 2015, 20(8): 1056-1071. DOI: 10.1007/s10495-015-1134-0.
Podocyte hypertrophy and apoptosis are two hallmarks of diabetic glomeruli, but the sequence in which these processes occur remains a matter of debate. Here we investigated the effects of inhibiting hypertrophy on apoptosis, and vice versa, in both podocytes and glomeruli, under diabetic conditions. Hypertrophy and apoptosis were inhibited using an epidermal growth factor receptor inhibitor (PKI 166) and a pan-caspase inhibitor (zAsp-DCB), respectively. We observed significant increases in the protein expression of p27, p21, phospho-eukaryotic elongation factor 4E-binding protein 1, and phospho-p70 S6 ribosomal protein kinase, in both cultured podocytes exposed to high-glucose (HG) medium, and streptozotocin-induced diabetes mellitus (DM) rat glomeruli. These increases were significantly inhibited by PKI 166, but not by zAsp-DCB. In addition, the amount of protein per cell, the relative cell size, and the glomerular volume were all significantly increased under diabetic conditions, and these changes were also blocked by treatment with PKI 166, but not zAsp-DCB. Increased protein expression of cleaved caspase-3 and cleaved poly (ADP-ribose) polymerase, together with increased Bax/Bcl-2 ratios, were also observed in HG-stimulated podocytes and DM glomeruli. Treatment with either zAsp-DCB or PKI 166 resulted in a significant attenuation of these effects. Both PKI 166 and zAsp-DCB also inhibited the increase in number of apoptotic cells, as assessed by Hoechst 33342 staining and TUNEL assay. Under diabetic conditions, inhibition of podocyte hypertrophy results in attenuated apoptosis, whereas blocking apoptosis has no effect on podocyte hypertrophy, suggesting that podocyte hypertrophy precedes apoptosis.
[37]
Verfaillie T, Rubio N, Garg AD, et al. PERK is required at the ER-mitochondrial contact sites to convey apoptosis after ROS-based ER stress[J]. Cell Death Differ, 2012, 19(11): 1880-1891. DOI: 10.1038/cdd.2012.74.
Endoplasmic reticulum stress is emerging as an important modulator of different pathologies and as a mechanism contributing to cancer cell death in response to therapeutic agents. In several instances, oxidative stress and the onset of endoplasmic reticulum (ER) stress occur together; yet, the molecular events linking reactive oxygen species (ROS) to ER stress-mediated apoptosis are currently unknown. Here, we show that PERK (RNA-dependent protein kinase (PKR)-like ER kinase), a key ER stress sensor of the unfolded protein response, is uniquely enriched at the mitochondria-associated ER membranes (MAMs). PERK(-/-) cells display disturbed ER morphology and Ca(2+) signaling as well as significantly weaker ER-mitochondria contact sites. Re-expression of a kinase-dead PERK mutant but not the cytoplasmic deletion mutant of PERK in PERK(-/-) cells re-establishes ER-mitochondria juxtapositions and mitochondrial sensitization to ROS-mediated stress. In contrast to the canonical ER stressor thapsigargin, during ROS-mediated ER stress, PERK contributes to apoptosis twofold by sustaining the levels of pro-apoptotic C/EBP homologous protein (CHOP) and by facilitating the propagation of ROS signals between the ER and mitochondria through its tethering function. Hence, this study reveals an unprecedented role of PERK as a MAMs component required to maintain the ER-mitochondria juxtapositions and propel ROS-mediated mitochondrial apoptosis. Furthermore, it suggests that loss of PERK may cause defects in cell death sensitivity in pathological conditions linked to ROS-mediated ER stress.
[38]
Choi ME. Autophagy in kidney disease[J]. Annu Rev Physiol, 2020, 82: 297-322. DOI: 10.1146/annurev-physiol-021119-034658.
Autophagy is a cellular homeostatic program for the turnover of cellular organelles and proteins, in which double-membraned vesicles (autophagosomes) sequester cytoplasmic cargos, which are subsequently delivered to the lysosome for degradation. Emerging evidence implicates autophagy as an important modulator of human disease. Macroautophagy and selective autophagy (e.g., mitophagy, aggrephagy) can influence cellular processes, including cell death, inflammation, and immune responses, and thereby exert both adaptive and maladaptive roles in disease pathogenesis. Autophagy has been implicated in acute kidney injury, which can arise in response to nephrotoxins, sepsis, and ischemia/reperfusion, and in chronic kidney diseases. The latter includes comorbidities of diabetes and recent evidence for chronic obstructive pulmonary disease-associated kidney injury. Roles of autophagy in polycystic kidney disease and kidney cancer have also been described. Targeting the autophagy pathway may have therapeutic benefit in the treatment of kidney disorders.
[39]
Lenoir O, Jasiek M, Hénique C, et al. Endothelial cell and podocyte autophagy synergistically protect from diabetes-induced glomerulosclerosis[J]. Autophagy, 2015, 11(7): 1130-1145. DOI: 10.1080/15548627.2015.1049799.
The glomerulus is a highly specialized capillary tuft, which under pressure filters large amounts of water and small solutes into the urinary space, while retaining albumin and large proteins. The glomerular filtration barrier (GFB) is a highly specialized filtration interface between blood and urine that is highly permeable to small and midsized solutes in plasma but relatively impermeable to macromolecules such as albumin. The integrity of the GFB is maintained by molecular interplay between its 3 layers: the glomerular endothelium, the glomerular basement membrane and podocytes, which are highly specialized postmitotic pericytes forming the outer part of the GFB. Abnormalities of glomerular ultrafiltration lead to the loss of proteins in urine and progressive renal insufficiency, underlining the importance of the GFB. Indeed, albuminuria is strongly predictive of the course of chronic nephropathies especially that of diabetic nephropathy (DN), a leading cause of renal insufficiency. We found that high glucose concentrations promote autophagy flux in podocyte cultures and that the abundance of LC3B II in podocytes is high in diabetic mice. Deletion of Atg5 specifically in podocytes resulted in accelerated diabetes-induced podocytopathy with a leaky GFB and glomerulosclerosis. Strikingly, genetic alteration of autophagy on the other side of the GFB involving the endothelial-specific deletion of Atg5 also resulted in capillary rarefaction and accelerated DN. Thus autophagy is a key protective mechanism on both cellular layers of the GFB suggesting autophagy as a promising new therapeutic strategy for DN.
[40]
He Y, Zhang M, Wu Y, et al. Aberrant activation of Notch-1 signaling inhibits podocyte restoration after islet transplantation in a rat model of diabetic nephropathy[J]. Cell Death Dis, 2018, 9(10): 950. DOI: 10.1038/s41419-018-0985-z.
Signaling abnormalities play important roles during podocyte injury and have been indicated as crucial events for triggering many glomerular diseases. There is emerging evidence demonstrating significant improvements in preventing renal injury and restoring podocytes after islet transplantation. However, whether signaling abnormalities affect the therapeutic efficacy of islet transplantation remain unclear. This study was established to investigate the impact of Notch-1 signaling activation on renal injury and podocyte restoration after islet transplantation. Experiments were performed in vivo and in vitro under conditions of diabetic nephropathy and high-glucose medium, respectively. Podocyte injury in vitro was induced by high-glucose concentration, and expression levels of genes associated with the Notch-1 pathway were also regulated by Jagged-1/FC and NJN-(3,5-Difluorophenacetyl)-L-alanylj- S-phenylglycine t-butyl ester (DAPT). Podocytes were co-cultured with islets to investigate the protective effect of islets in high-glucose conditions. Histopathological staining and transmission electron microscopy were performed to assess pathological changes in podocytes in glomeruli. The results from this study showed that Notch-1 signaling in podocytes was significantly decreased by functional islet cells in vivo and in vitro. Compared with the co-cultured group and transplanted group, highly activated Notch-1 signaling significantly moderated the effect of islets in affecting podocyte restoration and renal injury. Renal damage and podocyte injury were alleviated after DAPT treatment. Furthermore, the balance between apoptosis and autophagy was diverse under different treatments. All the data in this study showed that highly activated Notch-1 signaling could affect the therapeutic efficacy of islet transplantation on renal injury and podocyte restoration in high-glucose conditions. The balance between apoptosis and autophagy was also closely associated with the degree of podocyte restoration. This finding may suggest that the in vivo microenvironment plays a critical role in podocyte restoration after islet transplantation, which provides a promising and individual assessment and targeting treatment for different diabetic nephropathy patients after islet transplantation into the future.
[41]
Li X, Zhu Q, Zheng R, et al. Puerarin attenuates diabetic nephropathy by promoting autophagy in podocytes[J]. Front Physiol, 2020, 11: 73. DOI: 10.3389/fphys.2020.00073.
Puerarin, an active compound of radix puerariae, is a major compound used in Chinese herbal medicines to treat patients with diabetic nephropathy (DN). In the previous studies, we showed that puerarin exerts renoprotective effects in Streptozocin (STZ)-induced diabetic mice through activation of Sirt1 and anti-oxidative effects. Here, we further investigated the underlying mechanism mediating the renal protective effects of puerarin in DN. We studied the effects and mechanism of puerarin in STZ-induced diabetic mice and in cultured immortalized mouse podocytes treated with high glucose. We confirmed that puerarin ameliorated urinary albumin creatinine ratio and kidney injury in STZ-induced DN mice. We found that expression of heme oxygenase 1 (HMOX-1) and Sirt1 was suppressed in diabetic glomeruli but restored by puerarin treatment at both mRNA and protein levels. Additionally, we found that puerarin induced autophagy in the kidney of DN mice. In conditionally immortalized mouse podocytes, puerarin inhibited HG-induced apoptosis and restored the mRNA and protein levels of HMOX-1 and Sirt1. Interestingly, we showed that puerarin decreased liver kinase B1 (LKB1) acetylation, thereby promoting adenosine 5'-monophosphate-activated protein kinase-dependent autophagy. Knockdown of HMOX-1 and Sirt1 expression or treatment with the autophagy inhibitor 3-methyladenine abolished the protective effects of puerarin in HG-treated podocytes. Taken together, these results suggest that puerarin protects podocytes from diabetes-induced injury through HMOX1 and Sirt1-mediated upregulation of autophagy, a novel mechanism explaining its renal protective effects in DN.Copyright © 2020 Li, Zhu, Zheng, Yan, Wei, Fan, Deng and Zhong.
[42]
Galluzzi L, Green DR. Autophagy-independent functions of the autophagy machinery[J]. Cell, 2019, 177(7): 1682-1699. DOI: 10.1016/j.cell.2019.05.026.
Macroautophagy (herein referred to as autophagy) is an evolutionary ancient mechanism that culminates with the lysosomal degradation of superfluous or potentially dangerous cytosolic entities. Over the past 2 decades, the molecular mechanisms underlying several variants of autophagy have been characterized in detail. Accumulating evidence suggests that most, if not all, components of the molecular machinery for autophagy also mediate autophagy-independent functions. Here, we discuss emerging data on the non-autophagic functions of autophagy-relevant proteins.Copyright © 2019 Elsevier Inc. All rights reserved.
[43]
Filadi R, Pendin D, Pizzo P. Mitofusin 2: from functions to disease[J]. Cell Death Dis, 2018, 9(3): 330. DOI: 10.1038/s41419-017-0023-6.
Mitochondria are highly dynamic organelles whose functions are essential for cell viability. Within the cell, the mitochondrial network is continuously remodeled through the balance between fusion and fission events. Moreover, it dynamically contacts other organelles, particularly the endoplasmic reticulum, with which it enterprises an important functional relationship able to modulate several cellular pathways. Being mitochondria key bioenergetics organelles, they have to be transported to all the specific high-energy demanding sites within the cell and, when damaged, they have to be efficiently removed. Among other proteins, Mitofusin 2 represents a key player in all these mitochondrial activities (fusion, trafficking, turnover, contacts with other organelles), the balance of which results in the appropriate mitochondrial shape, function, and distribution within the cell. Here we review the structural and functional properties of Mitofusin 2, highlighting its crucial role in several cell pathways, as well as in the pathogenesis of neurodegenerative diseases, metabolic disorders, cardiomyopathies, and cancer.
[44]
Hamasaki M, Furuta N, Matsuda A, et al. Autophagosomes form at ER - mitochondria contact sites[J]. Nature, 2013, 495(7441): 389-393. DOI: 10.1038/nature11910.
[45]
Zhou D, Zhou M, Wang Z, et al. Progranulin alleviates podocyte injury via regulating CAMKK/AMPK-mediated autophagy under diabetic conditions[J]. J Mol Med (Berl), 2019, 97(11): 1507-1520. DOI: 10.1007/s00109-019-01828-3.
Podocyte injury is considered a major contributor to the development of diabetic nephropathy (DN). Therefore, identification of potential therapeutic targets for preventing podocyte injury has clinical importance. Recent studies have indicated that autophagy is a key homeostatic mechanism to maintaining podocyte integrity and function. This study was to elucidate the role of progranulin (PGRN), a secreted glycoprotein, in the modulation of podocyte autophagic process and podocyte injury under a diabetic condition. PGRN was downregulated in the kidney from diabetic mice and podocytes under a high-glucose (HG) condition. PGRN deficiency exacerbated the renal dysfunction and glomerular structural alterations. In vitro, treatment with recombinant human PGRN (rPGRN) attenuated HG-induced podocyte injury accompanied by enhanced autophagy. Inhibition of autophagy disturbed the protective effects of PGRN in HG-induced podocytotoxicity. Furthermore, PGRN induced autophagy via the PGRN-CAMKK-AMPK pathway. Collectively, our data identified the protective role of PGRN in podocyte injury via restoring autophagy and activating the CAMKK-AMPK pathway, which may pave the road to new therapeutic modalities for the treatment of diabetic nephropathy. KEY MESSAGES: • PGRN level is reduced in kidney of diabetic mice and high-glucose-treated podocytes. • PGRN deficiency exacerbates renal injury in diabetic mice. • PGRN protects against high-glucose-induced podocyte injury. • PGRN restores high-glucose-inhibited autophagy in podocytes. • CAMKK-AMPK pathway is required for the protective role of PGRN in podocyte injury.
[46]
Yang Y, Li N, Chen T, et al. Trimetazidine ameliorates sunitinib-induced cardiotoxicity in mice via the AMPK/mTOR/autophagy pathway[J]. Pharm Biol, 2019, 57(1): 625-631.
Sunitinib (SU) is a multi-targeted tyrosine kinase inhibitor anticancer agent whose clinical use is often limited by cardiovascular complications. Trimetazidine (TMZ) is an anti-angina agent that has been demonstrated cardioprotective effects in numerous cardiovascular conditions, but its potential effects in SU-induced cardiotoxicity have not been investigated. This study investigates the effect of TMZ in sunitinib-induced cardiotoxicity and and molecular mechanisms. Male 129S1/SvImJ mice were treated with vehicle, SU (40 mg/kg/d) or SU and TMZ (20 mg/kg/d) via oral gavage for 28 days, and cardiovascular functions and cardiac protein expressions were examined. H9c2 cardiomyocytes were treated with vehicle, SU (2-10 μM) or SU and TMZ (40-120 μM) for 48 h, and cell viability, apoptosis, autophagy, and protein expression was tested. SU induces hypertension (systolic blood pressure [SBP] + 28.33 ± 5.00 mmHg) and left ventricular dysfunction (left ventricular ejection fraction [LVEF] - 11.16 ± 2.53%) in mice. In H9c2 cardiomyocytes, SU reduces cell viability (IC 4.07 μM) and inhibits the AMPK/mTOR/autophagy pathway (< 0.05). TMZ co-administration with SU reverses SU-induced cardiotoxicity in mice (SBP - 23.75 ± 4.69 mmHg, LVEF + 10.95 ± 3.317%), alleviates cell viability loss in H9c2 cardiomyocytes (< 0.01) and activates the AMPK/mTOR/autophagy pathway (< 0.001) and (< 0.05). Our results suggest TMZ as a potential cardioprotective approach for cardiovascular complications during SU regimen, and potentially for cardiotoxicity of other anticancer chemotherapies associated with cardiomyocyte autophagic pathways.
[47]
Jeng PS, Inoue-Yamauchi A, Hsieh JJ, et al. BH3-dependent and independent activation of BAX and BAK in mitochondrial apoptosis[J]. Curr Opin Physiol, 2018, 3: 71-81. DOI: 10.1016/j.cophys.2018.03.005.
Mitochondria play key roles in mammalian apoptosis, a highly regulated genetic program of cell suicide. Multiple apoptotic signals culminate in mitochondrial outer membrane permeabilization (MOMP), which not only couples the mitochondria to the activation of caspases but also initiates caspase-independent mitochondrial dysfunction. The BCL-2 family proteins are central regulators of MOMP. Multidomain pro-apoptotic BAX and BAK are essential effectors responsible for MOMP, whereas anti-apoptotic BCL-2, BCL-X, and MCL-1 preserve mitochondrial integrity. The third BCL-2 subfamily of proteins, BH3-only molecules, promotes apoptosis by either activating BAX and BAK or inactivating BCL-2, BCL-X, and MCL-1. Through an interconnected hierarchical network of interactions, the BCL-2 family proteins integrate developmental and environmental cues to dictate the survival versus death decision of cells by regulating the integrity of the mitochondrial outer membrane. Over the past 30 years, research on the BCL-2-regulated apoptotic pathway has not only revealed its importance in both normal physiological and disease processes, but has also resulted in the first anti-cancer drug targeting protein-protein interactions.
[48]
Fan Y, Yang Q, Yang Y, et al. Sirt6 suppresses high glucose-induced mitochondrial dysfunction and apoptosis in podocytes through AMPK activation[J]. Int J Biol Sci, 2019, 15(3): 701-713. DOI: 10.7150/ijbs.29323.
Previous studies have shown that mitochondrial dysfunction plays an important role in high- glucose(HG)-induced podocyte injury and thus contributes to the progression of diabetic nephropathy(DN). The histone deacetylase Sirtuin6 (Sirt6) has been revealed to have an essential role in the regulation of mitochondrial function in skeletal muscle and cardiomyocytes. However, its specific role in mitochondrial homeostasis in podocytes is undetermined. Here, we aimeds to explore the physiological function of Sirt6 in podocyte mitochondria and apoptosis under HG conditions and explore the possible mechanism. Herein, we observed that Sirt6-WT-1 colocalization was suppressed in the glomeruli of patients with DN. In addition, diabetic mice exhibited reduced Sirt6 expression and AMP kinase (AMPK) dephosphorylation accompanied by mitochondrial morphological abnormalities., podocytes exposed to HG presented with mitochondrial morphological alterations and podocyte apoptosis accompanied by Sirt6 and p-AMPK downregulation. In addition, HG promoted a decrease in mitochondrial number and an increase in mitochondrial superoxide production as well as a decreased mitochondrial membrane potential. ROS production was also increased in HG-treated podocytes. Conversely, all these mitochondrial defects induced by HG were significantly alleviated by Sirt6 plasmid transfection. Sirt6 overexpression simultaneously alleviated HG-induced podocyte apoptosis and oxidative stress, as well as increased AMPK phosphorylation. Increased levels of H3K9ac and H3K56ac induced by HG were attenuated in podocytes transfected with Sirt6 plasmids. Therefore, these results elucidated that Sirt6 protects mitochondria of podocytes and exerts anti-apoptotic effects via activating AMPK pathway. The present findings provide key insights into the pivotal role of mitochondria regulation by SIRT6 in its protective effects on podocytes.
[49]
Fang L, Zhou Y, Cao H, et al. Autophagy attenuates diabetic glomerular damage through protection of hyperglycemia-induced podocyte injury[J]. PLoS One, 2013, 8(4): e60546. DOI: 10.1371/journal.pone.0060546.
[50]
Cao AL, Wang L, Chen X, et al. Ursodeoxycholic acid and 4-phenylbutyrate prevent endoplasmic reticulum stress-induced podocyte apoptosis in diabetic nephropathy[J]. Lab Invest, 2016, 96(6): 610-622. DOI: 10.1038/labinvest.2016.44.

所有作者均声明不存在利益冲突

PDF(19377 KB)

2001

Accesses

0

Citation

Detail

Sections
Recommended

/