缝隙连接传递伤害性信号促进小鼠肝缺血再灌注损伤及细胞凋亡

黄菲, 肖翠翠, 王敏学, 周少丽

新医学 ›› 2020, Vol. 51 ›› Issue (11) : 835-839.

PDF(3355 KB)
PDF(3355 KB)
新医学 ›› 2020, Vol. 51 ›› Issue (11) : 835-839. DOI: 10.3969/j.issn.0253-9802.2020.11.006
研究论著

缝隙连接传递伤害性信号促进小鼠肝缺血再灌注损伤及细胞凋亡

作者信息 +

Gap junction-mediated harmful signal transmission contributes to liver injury and cell apoptosis after hepatic ischemia-reperfusion

Author information +
文章历史 +

摘要

目的 探讨细胞缝隙连接(GJ)信号传递对肝缺血再灌注损伤(HIRI)及细胞凋亡的影响。方法 C57BL/6小鼠建立HIRI模型,随机分为假手术组(Sham组)、HIRI组、HIRI+2-氨基乙基联苯基硼酸酯(2APB)组、HIRI+维甲酸(RA)组。HIRI+2APB组及HIRI+RA组在造模前腹腔注射2APB或RA。采用HE染色观察肝组织病理损伤情况,生化检测血清ALT和AST评估肝功能情况,蛋白免疫印迹法检测凋亡相关蛋白Bim、Bax、Caspase-3的表达,TUNEL法检测肝组织细胞凋亡情况。结果 与HIRI组相比,HIRI+RA组肝损伤及凋亡明显增加,Bim介导的凋亡蛋白表达增强(P均< 0.05);相反,HIRI+2APB组可显著减轻肝损伤及细胞凋亡,并减少Bim介导的凋亡蛋白表达(P均< 0.05)。结论 GJ传递伤害性信号可能在HIRI进展中发挥重要作用。

Abstract

Objective To evaluate the effect of gap junction (GJ)-mediated signal transmission on liver injury and cell apoptosis after hepatic ischemia reperfusion (HIRI). Methods C57BL/6 mouse models of HIRI were established. All animals were randomly divided into the sham operation group (Sham group), HIRI group, HIRI+2-aminoethyldiphenyl borate (2APB) group and HIRI+retinoic acid (RA) group, respectively. In the HIRI+2APB and HIRI+RA groups, the animals were intraperitoneally injected with 2APB or RA before the establishment of HIRI models. The pathological injury of liver tissues was observed by HE staining. The liver function was evaluated by biochemical detection of serum ALT and AST levels. The expression levels of apoptosis-related proteins, such as Bim, Bax and Caspase-3 in liver tissues was quantitatively measured by Western blot. The cell apoptosis of liver tissues was detected by TUNEL assay. Results Compared with the HIRI group,the liver injury induced by HIRI and the cell apoptosis were aggravated and the expression level of Bim-mediated apoptosis-related protein was remarkably up-regulated in the HIRI+RA group (all P < 0.05). In the HIRI+2APB group, hepatic injury and cell apoptosis were significantly alleviated and the expression level of Bim-mediated apoptosis-related protein was remarkably down-regulated (all P < 0.05). Conclusion The harmful signal transmitted by GJ may play an important role in the development of HIRI-induced liver injury.

关键词

肝缺血再灌注损伤 / 凋亡 / 缝隙连接

Key words

Hepatic ischemia reperfusion injury / Apoptosis / Gap junction

引用本文

导出引用
黄菲 , 肖翠翠 , 王敏学 , 周少丽. 缝隙连接传递伤害性信号促进小鼠肝缺血再灌注损伤及细胞凋亡[J]. 新医学, 2020, 51(11): 835-839 https://doi.org/10.3969/j.issn.0253-9802.2020.11.006
Huang Fei , Xiao Cuicui , Wang Minxue , Zhou Shaoli. Gap junction-mediated harmful signal transmission contributes to liver injury and cell apoptosis after hepatic ischemia-reperfusion[J]. JOURNAL OF NEW MEDICINE, 2020, 51(11): 835-839 https://doi.org/10.3969/j.issn.0253-9802.2020.11.006
肝缺血再灌注损伤(HIRI)常见于肝移植、肝部分切除术、失血性休克等诸多过程中,可引起肝功能不全、肝衰竭甚至多器官功能衰竭。据报道,肝部分切除术后肝功能不全发生率约9.5%,是术后患者恢复延迟和死亡的主要原因[1]。HIRI机制复杂并具有多元化特点,目前尚未完全阐明。目前研究认为凋亡是肝缺血再灌注后早期肝损伤的主要形式[2]。Bcl-2家族在凋亡的调控中起着关键作用, 促凋亡蛋白Bim是近年来发现的Bcl-2家族中BH3-only亚家族的一员,也叫前凋亡蛋白,它是凋亡执行所必需的上游调节因子[3]
细胞缝隙连接(GJ)是细胞间直接联系通道,允许分子量小于1 KDa的物质在相邻细胞间双向性流动。GJ的信号传递在维持细胞内环境稳定和器官功能、调控细胞生长发育等过程中扮演重要角色[4]。GJ介导“旁细胞效应”被很多科学家形象地将其称为“死亡之吻”,并受到了科研界的广泛关注,因为这使对继发损伤程度的限制成为可能[5]。肝细胞间的GJ十分丰富,肝细胞主要表达Cx32和少量的Cx26,GJ发挥着重要的生理功能[6]。我们团队前期体外实验发现,缺氧复氧诱导BRL-3A肝细胞凋亡与Cx32组成的GJ通讯增强有关[7]。因此我们提出假设:Cx32组成的GJ传递伤害性信号可能加重HIRI。本研究旨在初步探讨Cx32组成的GJ在HIRI中的作用,为以GJ为靶点早期干预肝损伤进展提供实验依据。

材料与方法

一、材料

SPF级健康雄性C57BL/6小鼠28只(8 ~ 10周,20 ~ 25 g)购于广东省动物实验中心;2-氨基乙基联苯基硼酸酯(2APB)及维甲酸(RA,Sigma,美国);Bim、Bax、Caspase-3单克隆抗体(Invitrogen,美国);HRP标记的兔抗小鼠IgG抗体(Sigma,美国);TUNEL检测试剂盒(Roche,美国)。

二、方法

1. 70%小鼠HIRI模型

本研究中所有动物实验均通过中山大学附属第三医院实验动物伦理委员会批准。根据我们既往经验,按照我们既往发表文献[8]的方法建立小鼠70%肝缺血再灌注模型。具体操作步骤如下:①术前常规禁食8 h,自由饮水;②氯胺酮(60 mg/kg)复合甲苯噻嗪(100 mg/kg)复合液腹腔注射进行麻醉;③手术取仰卧位,腹部备皮、消毒后腹部正中切口入腹;④进入腹腔后仔细游离进入肝左叶和肝中叶的血管;⑤使用无损伤的微血管夹阻断供应上述两个肝叶(缺血肝叶)的动脉和静脉,并保留供应肝脏尾状叶、右叶、乳头状叶及方叶的血流防止肠静脉充血;⑥缺血期间腹部伤口以湿润纱布覆盖,在进行60 min缺血后(观察缺血肝叶的颜色变化,颜色变灰白证明缺血成功),小心松开微血管夹,开始进行再灌注(再灌注后缺血肝叶颜色变化粗略判断再灌注成功)。仔细止血后,连续全层缝合腹壁切口关腹。实验过程中注意实验室的温度控制在30℃左右,并尽量减少出血。

2.实验动物分组

实验选取SPF级健康雄性野生型C57BL/6小鼠28只,随机分为假手术组(Sham组)、HIRI组、肝缺血再灌注+缝隙连接抑制剂(2APB)预处理组(HIRI+2APB组)、肝缺血再灌注+缝隙连接增强剂(RA)预处理组(HIRI+RA组)。假手术组在麻醉后只进行腹腔开腹和血管的分离,不进行肝脏的阻断和灌注。HIRI+2APB组及HIRI+RA组在造模前腹腔注射2APB(20 mg/kg,0.5 ml)或RA(10 mg/kg,0.5 ml)。

3. 标本的采集和处理

各组在肝脏再灌注后6 h采集标本。再次麻醉小鼠,并从下腔静脉取血0.5 ~ 1.0 ml,以1500转/分的速度离心后提取上清液,分装注入已编号标记好的EP管中,置入-20℃冰箱保存,用于后续的肝功能生化检测。收集缺血肝叶,部分肝组织用4%多聚甲醛溶液固定24 h,常规石蜡包埋、切片,HE染色,用于肝组织病理学检测;剩余肝组织放入冻存管后立即置入液氮中保存,并在实验完毕后迅速转运置入-80℃深低温冰箱进一步保存,用于后续的肝组织相关蛋白检测。

4. 肝损伤病理学评分标准

选取切片平整、组织细胞形态和结构清晰的HE染色组织切片进行光镜下观察和分析。参照Suzuki评分法对肝损伤程度进行病理学评分,具体评分标准如下:0分,无损伤, 肝实质与间质正常;1分,轻微病变,肝窦间隙轻微充血,很少空泡形成及单个肝细胞坏死;2分,肝窦间隙轻度充血,少数空泡形成,小于30%的肝细胞坏死;3 分,中度损伤,肝窦间隙中度度充血,中度空泡形成,约30% ~ 60%的肝细胞坏死;4分,严重损伤,肝窦间隙严重充血,重度空泡形成,超过60%的肝细胞坏死[9]

5. 蛋白免疫印迹法检测蛋白表达

取缺血肝叶肝组织冰浴中充分匀浆(加入裂解液),于4℃、12 000转/分离心30 min,二辛可宁酸(BCA)法蛋白定量,每份样品各取25 μg蛋白质。配胶、电泳,转膜后封闭2 h,加入β-actin(兔抗小鼠,1∶3000)、Cx32(兔抗小鼠,1∶1000)、Bim(兔抗小鼠,1∶1000)、Bcl-2(兔抗小鼠,1∶1000)、Caspase-3(兔抗小鼠,1∶1000)一抗,4℃摇床孵育过夜,磷酸盐缓冲液(PBS)洗膜3次,每次10 min。加入辣根过氧化物酶(HRP)标记的山羊抗兔IgG (1∶5000) 室温摇床孵育后PBS洗膜,暗室中曝光、显影,采用Image J软件检测β-actin及目的蛋白条带的灰度值,以目的蛋白条带灰度值/β-actin蛋白条带灰度值作为目的蛋白表达量。将3张以上条带灰度值平均值作为本次实验结果。

6. TUNEL法检测肝组织细胞凋亡

厚度3 μm的组织切片37℃温箱烘烤过夜→脱蜡及水化→3%的双氧水甲醇溶液37℃恒温箱孵育10 min(TBS洗3次,每次5 min)→蛋白酶K渗透,37℃恒温箱孵育10 min(TBS洗3次,每次5 min)→滴加标记缓冲液,37℃恒温箱孵育2 h(TBS 洗3次,每次5 min)→封闭液封闭,室温孵育30 min→滴加TUNEL反应液到组织上,37℃恒温箱孵育30 min(TBS洗3次,每次5 min)→滴加POD转化剂到组织上,37℃恒温箱孵育30 min(TBS洗3次,每次5 min)→DAB显色→苏木素复染细胞核90 s,充分水洗、脱水、透明及中性树脂封片→数据分析。

三、统计学处理

采用Sigmaplot 10.0分析数据。实验数据以x¯±s表示,组间比较采用单因素方差分析,两两比较采用LSD-t检验,P < 0.05为差异有统计学意义。

结果

一、肝损伤及评分

我们在光镜下观察肝组织HE染色切片,结果显示,假手术Sham组肝组织形态和结构基本正常,未见明显肝窦充血和肝细胞坏死;肝缺血6 h后(HIRI组)肝组织病理损伤严重,可见肝窦明显充血,肝细胞肿胀、空泡形成,炎性细胞浸润,并出现片状坏死,见图1A。各组Suzuki评分在统计学上存在差异(F = 54.33,P < 0.05)。与HIRI组相比,HIRI+RA组小鼠肝组织病理学损伤进一步加重,坏死区域面积扩大,Suzuki评分升高(P < 0.05)。相反,与HIRI组相比,HIRI+2APB组小鼠肝组织病理学损伤明显减轻,肝窦充血和肝细胞肿胀减少,坏死区域面积缩小,Suzuki评分有所降低(P < 0.05),表现出肝损伤的好转,见图1B
图1 各组小鼠肝组织病理损伤情况及肝功能情况
A:各组小鼠肝组织HE染色(×100);B:各组小鼠肝组织病理损伤Suzuki评分;C:各组小鼠血清ALT水平;D:各组小鼠血清AST水平;与Sham组比较,*P < 0.05;与HIRI组比较,#P < 0.05

Full size|PPT slide

二、肝功能情况

各组小鼠血清ALT和AST水平存在差异(F值分别为147.9和86.05,P 均< 0.05)。与Sham组相比,HIRI组小鼠血清ALT水平升高(P < 0.05),提示存在明显的肝细胞损伤;与HIRI组相比,HIRI+RA组小鼠血清ALT水平进一步升高(P < 0.05),提示肝细胞损伤进一步加重;相反,HIRI+2APB组ALT水平下降(P < 0.05),表现出肝功能的保护作用,见图1C。各组小鼠血清AST水平变化与ALT呈现相同的趋势,见图1D

三、肝组织中凋亡相关蛋白Bim、Bax、Caspase-3表达

各组小鼠肝组织Bim、Bax和Caspase-3蛋白表达存在差异(F值分别为63.15、36.61和35.65,P 均< 0.05)。与Sham组相比,HIRI组小鼠肝组织Bim、Bax、Caspase-3蛋白表达上调(P 均< 0.05),与HIRI组相比,HIRI+RA组Bim、Bax和Caspase-3蛋白表达进一步上调(P均< 0.05),相反,HIRI+2APB组上述蛋白表达有所下降(P均< 0.05),见图2
图2 各组小鼠肝组织凋亡相关蛋白表达情况
A:蛋白免疫印迹法检测各组小鼠肝组织中凋亡相关蛋白的表达;B:Bim蛋白表达灰度分析;C:Bax蛋白表达灰度分析;D:Caspase-3蛋白表达灰度分析;与Sham组比较,*P < 0.05;与HIRI组比较,#P < 0.05

Full size|PPT slide

四、肝组织中细胞凋亡情况

与Sham组相比,HIRI组小鼠肝组织细胞凋亡明显增加;与HIRI组相比,HIRI+RA组细胞凋亡进一步增加;相反,HIRI+2APB组细胞凋亡明显减少,见图3
图3 TUNEL法检测各组小鼠肝组织细胞凋亡情况(×100)
蓝色荧光为正常细胞,红色箭头所指的绿色荧光为凋亡细胞

Full size|PPT slide

讨论

本研究所使用的模型为70%HIRI模型,这是目前非常成熟的非致命性肝脏热缺血再灌注损伤模型,国内外大量研究均以此模型来模拟肝脏缺血再灌注过程而进行HIRI方面的研究[10]。我们团队前期研究观察了C57BL/6小鼠肝缺血1 h后不同时间点肝损伤情况的动态变化,发现再灌注6 h肝损伤最重[11]。据此,我们在本次研究中,采用肝缺血1 h再灌注6 h作为模型。
肝组织细胞中的GJ十分丰富,其中Cx32是肝脏中表达最多的Cx蛋白,占肝组织Cx蛋白总量的90%以上[12]。本研究结果显示,使用GJ增强剂可加重肝损伤及细胞凋亡,相反,使用GJ抑制剂可明显减轻肝损伤和细胞凋亡,提示在HIRI过程中,GJ可能传递伤害性信号放大损伤和凋亡。那么,究竟是哪一种Cx蛋白组成的GJ发挥的作用呢?目前认为,2APB是Cx32特异性的GJ抑制剂,只对Cx32组成的GJ功能有抑制作用,对其他Cx蛋白组成的GJ无影响,那么,我们的结果提示Cx32组成的GJ在此过程中可能发挥了重要作用[13]
在细胞凋亡的进程中, 有两个家族的蛋白质起到非常重要的作用:Caspase家族是凋亡的执行者,而Bcl-2家族则为凋亡的决策者。Bcl-2家族又分为抗凋亡的Bcl-2亚家族及促凋亡的Bax和BH3-only亚家族。BH3-only亚家族被认为是Caspase级联反应的启动者[14]。在BH3-only亚家族成员中,Bim是目前已知最重要的促凋亡因子,因为它不仅可激活多种Bcl-2家族的促凋亡蛋白,还可与该家族中几乎所有抗凋亡蛋白结合抑制其作用[15]。有学者报道,Bim在内质网应激诱导的细胞凋亡中扮演不可或缺的哨兵角色,是上游死亡信号级联反应和下游Bax/Bak依赖的线粒体凋亡程序的连接者[16]。在造血细胞、肾脏上皮细胞、黑素细胞、神经细胞、干细胞以及自身免疫性胸腺细胞中的研究证明,Bim是凋亡最关键的始动因子,在其凋亡过程中必不可少[17,18]。本研究结果显示,通过GJ工具药改变GJ功能可以明显影响Bim及下游凋亡蛋白Bax的表达,提示GJ参与Bim介导的凋亡,但GJ究竟传递何种信号,通过何种途径促进Bim表达,还需要进一步的研究。
本实验证实了改变GJ功能可以明显影响HIRI程度和细胞凋亡数量,并改变Bim介导的凋亡相关蛋白的表达,其机制可能与GJ传递伤害性信号有关,但此伤害性信号的性质尚不清楚,值得深入研究,为以GJ为靶点,减轻HIRI提供实验依据。

参考文献

[1]
彭娜, 王彦峰, 何维阳, 李明霞, 胡晓燕, 叶啟发. 肝移植供肝缺血再灌注损伤的研究进展. 中华肝胆外科杂志, 2016,22(5):349-351.
[2]
王冰, 林颖, 刘慧玲, 金海, 汪根树, 吴斌, 陈规划. 己酮可可碱对大鼠肝脏缺血再灌注损伤的影响. 新医学, 2011,42(8):601-604.
[3]
Sionov RV, Vlahopoulos SA, Granot Z. Regulation of bim in health and disease. Oncotarget, 2015,6(27):23058-23134.
The BH3-only Bim protein is a major determinant for initiating the intrinsic apoptotic pathway under both physiological and pathophysiological conditions. Tight regulation of its expression and activity at the transcriptional, translational and post-translational levels together with the induction of alternatively spliced isoforms with different pro-apoptotic potential, ensure timely activation of Bim. Under physiological conditions, Bim is essential for shaping immune responses where its absence promotes autoimmunity, while too early Bim induction eliminates cytotoxic T cells prematurely, resulting in chronic inflammation and tumor progression. Enhanced Bim induction in neurons causes neurodegenerative disorders including Alzheimer's, Parkinson's and Huntington's diseases. Moreover, type I diabetes is promoted by genetically predisposed elevation of Bim in beta-cells. On the contrary, cancer cells have developed mechanisms that suppress Bim expression necessary for tumor progression and metastasis. This review focuses on the intricate network regulating Bim activity and its involvement in physiological and pathophysiological processes.
[4]
Maes M, Crespo Yanguas S, Willebrords J, Cogliati B, Vinken M. Connexin and pannexin signaling in gastrointestinal and liver disease. Transl Res, 2015,166(4):332-343.
Gap junctions, which mediate intercellular communication, are key players in digestive homeostasis. They are also frequently involved in gastrointestinal and liver pathology. This equally holds true for connexin (Cx) hemichannels, the structural precursors of gap junctions, and pannexin (Panx) channels, Cx-like proteins assembled in a hemichannel configuration. Both Cx hemichannels and Panx channels facilitate extracellular communication and drive a number of deteriorative processes, such as cell death and inflammation. Cxs, Panxs, and their channels underlie a wide spectrum of gastrointestinal and liver diseases, including gastritis and peptic ulcer disease, inflammatory intestinal conditions, acute liver failure, cholestasis, hepatitis and steatosis, liver fibrosis and cirrhosis, infectious gastrointestinal pathologies, and gastrointestinal and liver cancer. This could open promising perspectives for the characterization of new targets and biomarkers for therapeutic and diagnostic clinical purposes in the area of gastroenterology and hepatology.
[5]
Suzuki M. Significance of radiation-induced bystander effects in radiation therapy. Igaku Butsuri, 2014,34(2):70-78.
Since 1994, a Phase I/II clinical study and radiotherapy have carried out using carbon-ion beams produced with the Heavy Ion Medical Accelerator in Chiba (HIMAC) at National Institute of Radiological Sciences. Now we constructed the new treatment facility for the advanced carbon-ion therapy at HIMAC applying a 3D fast spot scanning system with pencil beams. In the field of fundamental biological studies for high-LET heavy ions, there are some reports regarding bystander effects after exposure to alpha particles derived from 238Pu or He-ion microbeams. However, only limited sets of studies have examined bystander effects after exposure to different ion species heavier than helium, such as carbon ions. We have been investigating bystander cellular responses in both normal human and human tumor cells irradiated with the HIMAC carbon ions. Bystander cell-killing effect was observed in the cells harboring wild-type P53 gene, but not in the P53-mutated cells. Moreover, observed bystander effect was suppressed by treating with a specific inhibitor of gap-junction mediated cell-cell communication. There is clear evidence that the carbon-ion irradiation enables the enhanced cell killing in cells with wild-type P53 gene via gap-junction mediated bystander effect.
[6]
Patel SJ, Milwid JM, King KR, Bohr S, Iracheta-Vellve A, Li M, Vitalo A, Parekkadan B, Jindal R, Yarmush ML. Gap junction inhibition prevents drug-induced liver toxicity and fulminant hepatic failure. Nat Biotechnol, 2012,30(2):179-183.
Drug-induced liver injury (DILI) limits the development and application of many therapeutic compounds and presents major challenges to the pharmaceutical industry and clinical medicine. Acetaminophen-containing compounds are among the most frequently prescribed drugs and are also the most common cause of DILI. Here we describe a pharmacological strategy that targets gap junction communication to prevent amplification of fulminant hepatic failure and acetaminophen-induced hepatotoxicity. We demonstrate that connexin 32 (Cx32), a key hepatic gap junction protein, is an essential mediator of DILI by showing that mice deficient in Cx32 are protected against liver damage, acute inflammation and death caused by liver-toxic drugs. We identify a small-molecule inhibitor of Cx32 that protects against liver failure and death in wild-type mice when co-administered with known hepatotoxic drugs. These findings indicate that gap junction inhibition could provide a pharmaceutical strategy to limit DILI and improve drug safety.
[7]
Wang R, Huang F, Chen Z, Li S. Downregulation of connexin 32 attenuates hypoxia/reoxygenation injury in liver cells. J Biochem Mol Toxicol, 2015,29(4):189-197.
Gap junction intercellular communication is involved in ischemia-reperfusion (IR) injury of organs. Connexins are proteins that are critical to the function of gap junctions. To clarify the role of gap junctions in IR injury in liver cells, the function of gap junctions was modulated in an in vitro hypoxia/reoxygenation (H/R) model. BRL-3A rat liver cells, endogenously expressing connexins Cx32 and Cx43, were used to model the process of hepatic IR injury. Suppression of gap junction activity was achieved genetically, using Cx32-specific small interfering RNA (siRNA), or chemically, with pharmacological inhibitors, oleamide, and 18-alpha-GA. BRL-3A cells subjected to H/R exhibited reduced cell survival and pathologies indicative of IR injury. Cx32-specific siRNA, oleamide, and 18-alpha-GA, respectively, decreased gap junction permeability, as assessed by the parachute assay. Pretreatment with Cx32-specific siRNA increased cell survival. Pretreatment with oleamide or 18-alpha-GA did not improve cell survival. Modulating gap junction by Cx32 gene silencing protected BRL-3A liver cells from H/R.
[8]
Ge M, Yao W, Wang Y, Yuan D, Chi X, Luo G, Hei Z. Propofol alleviates liver oxidative stress via activating Nrf2 pat-hway. J Surg Res, 2015,196(2):373-381.
BACKGROUND: Nuclear factor-E2-related factor 2 (Nrf2)-mediated antioxidant response is the main protective system of graft-liver against ischemia-reperfusion injury after liver transplantation. Propofol is considered to confer protective effects on different organs; thus, we explored the possibility that whether propofol could attenuate graft-liver injury in a rat autologous orthotopic liver transplantation (AOLT) model and mechanisms were associated with activation of Nrf2 pathway. METHODS: Sprague-Dawley rats were randomly divided into four groups: sham-operated group, saline-treated AOLT group, low-dose propofol intervention group, and high-dose propofol intervention group. Liver injury was determined, and concentration of hydroxyl free radical (*OH), superoxide anion (O2(*-)), and malondialdehyde in the liver tissue were detected. The expression of Keap1, Nrf2, HO-1, and NQO1 were explored by Western blotting, and also the change of Nrf2 and keap1 was assessed by immunofluorescence. RESULTS: Compared with sham group, pathologic damage of graft-livers was in a time-dependent manner, accompanied with the increased level of oxidative stress in the AOLT group, and nuclear Nrf2 expression and its downstream antioxidant enzyme, HO-1 and NQO1, were also increased in this group. However, in propofol pretreatment groups especially in the high-dose group, the pathologic score was significantly decreased, accompanied with a lower level of *OH, O2(*-), and malondialdehyde than that of the AOLT group. The change of oxidative stress might be related to the Nrf2 pathway, evidenced as the elevation of protein expression level of NQO1, HO-1, and nuclear Nrf2. CONCLUSIONS: Protective effects of propofol against liver transplantation-induced graft-liver injury may be related with Keap1-Nrf2 signal pathway activation.
[9]
Zhang Y, Yuan D, Yao W, Zhu Q, Liu Y, Huang F, Feng J, Chen X, Huang Y, Chi X, Hei Z. Hyperglycemia aggravates hepatic ischemia reperfusion injury by inducing chronic oxidative stress and inflammation. Oxid Med Cell Longev, 2016,2016:3919627.
Aim. To investigate whether hyperglycemia will aggravate hepatic ischemia reperfusion injury (HIRI) and the underlying mechanisms. Methods. Control and streptozotocin-induced diabetic Sprague-Dawley rats were subjected to partial hepatic ischemia reperfusion. Liver histology, transferase, inflammatory cytokines, and oxidative stress were assessed accordingly. Similarly, BRL-3A hepatocytes were subjected to hypoxia/reoxygenation (H/R) after high (25 mM) or low (5.5 mM) glucose culture. Cell viability, reactive oxygen species (ROS), and activation of nuclear factor-erythroid 2-related factor 2 (Nrf2) and nuclear factor of kappa light polypeptide gene enhancer in B-cells (NF-kappaB) were determined. Results. Compared with control, diabetic rats presented more severe hepatic injury and increased hepatic inflammatory cytokines and oxidative stress. HIRI in diabetic rats could be ameliorated by pretreatment of N-acetyl-L-cysteine (NAC) or apocynin. Excessive ROS generation and consequent Nrf2 and NF-kappaB translocation were determined after high glucose exposure. NF-kappaB translocation and its downstream cytokines were further increased in high glucose cultured group after H/R. While proper regulation of Nrf2 to its downstream antioxidases was observed in low glucose cultured group, no further induction of Nrf2 pathway by H/R after high glucose culture was identified. Conclusion. Hyperglycemia aggravates HIRI, which might be attributed to chronic oxidative stress and inflammation and potential malfunction of antioxidative system.
[10]
黑子清. 肝移植围术期器官损伤机制及器官保护策略研究进展. 中山大学学报(医学版), 2019,40(4):488-492.
[11]
Wu S, Yao W, Chen C, Chen H, Huang F, Liu Y, Cai J, Yuan D, Hei Z. Connexin 32 deficiency protects the liver against ischemia/reperfusion injury. Eur J Pharmacol, 2020,876:173056.
Hepatic ischemia/reperfusion (I/R) injury is a common complication in the clinical setting. Our previous study has shown that connexin 32 (Cx32) plays a major role in renal I/R injury; however, the role of Cx32 in hepatic I/R injury remains unknown. Liver tissue and serum samples from patients undergoing orthotopic liver transplantation (OLT) were used to evaluate the function of Cx32 in OLT post-reperfusion injury. Then, partial hepatic ischemia was established in global Cx32 knockout mice and wild-type mice followed by reperfusion. Hepatic injury markers were examined. Cx32 small interfering RNA and the p53 inhibitor, pifithrin-alpha, tenovin-1 were used to examine the relationship between Cx32 and the p53/puma pathways in the BRL-3A and murine primary hepatocytes hypoxia/reoxygenation (H/R) model. Corresponding to liver damage, Cx32 was significantly induced both during OLT in human patients and partial hepatic I/R in mice. Cx32 KO mice exhibited less liver injury than controls. Cx32 deficiency significantly suppressed the p53/puma pathways and hepatocyte apoptosis. Similar results were observed in the BRL-3A and murine primary hepatocytes H/R model. Propofol protected against OLT post-reperfusion injury and hepatocyte apoptosis by inhibiting Cx32. In conclusion Cx32 is a novel regulator of hepatic I/R injury through the modulation of hepatocyte apoptosis and damage, largely via the p53/puma signaling pathway.
[12]
Masia R, Diagacomo E, Adams D, King KR, Piaker S, Reinecker HC, Yarmush ML, Argemi J, Bataller R, Dienstag JL, Chung RT, Patel SJ. Hepatic gap junctions amplify alcohol liver injury by propagating cGAS-mediated IRF3 activation. Proc Natl Acad Sci USA, 2020,117(21):11667-11673.
Alcohol-related liver disease (ALD) accounts for the majority of cirrhosis and liver-related deaths worldwide. Activation of IFN-regulatory factor (IRF3) initiates alcohol-induced hepatocyte apoptosis, which fuels a robust secondary inflammatory response that drives ALD. The dominant molecular mechanism by which alcohol activates IRF3 and the pathways that amplify inflammatory signals in ALD remains unknown. Here we show that cytoplasmic sensor cyclic guanosine monophosphate-adenosine monophosphate (AMP) synthase (cGAS) drives IRF3 activation in both alcohol-injured hepatocytes and the neighboring parenchyma via a gap junction intercellular communication pathway. Hepatic RNA-seq analysis of patients with a wide spectrum of ALD revealed that expression of the cGAS-IRF3 pathway correlated positively with disease severity. Alcohol-fed mice demonstrated increased hepatic expression of the cGAS-IRF3 pathway. Mice genetically deficient in cGAS and IRF3 were protected against ALD. Ablation of cGAS in hepatocytes only phenocopied this hepatoprotection, highlighting the critical role of hepatocytes in fueling the cGAS-IRF3 response to alcohol. We identified connexin 32 (Cx32), the predominant hepatic gap junction, as a critical regulator of spreading cGAS-driven IRF3 activation through the liver parenchyma. Disruption of Cx32 in ALD impaired IRF3-stimulated gene expression, resulting in decreased hepatic injury despite an increase in hepatic steatosis. Taken together, these results identify cGAS and Cx32 as key factors in ALD pathogenesis and as potential therapeutic targets for hepatoprotection.
[13]
Bai D, del Corsso C, Srinivas M, Spray DC. Block of specific gap junction channel subtypes by 2-aminoethoxydiphenyl borate (2-APB). J Pharmacol Exp Ther, 2006,319(3):1452-1458.
2-Aminoethoxydiphenyl borate (2-APB), an inositol 1,4,5-triphosphate receptor modulator, inhibits capacitive current transients measured in normal rat kidney and human embryonic kidney 293 cells, an indication of blocking gap junction channels between these cells. Here, we used the dual whole-cell patch-clamp method to study the actions of 2-APB on gap junction channels formed by selected connexins expressed in a communication-deficient neuroblastoma cell line (N2A). 2-APB dose-dependently and reversibly blocked junctional currents of connexin (Cx) 50 gap junction channels. The concentration-inhibition curve of 2-APB on the junctional current indicated an IC(50) of 3.7 microM, lower than that of most gap junction inhibitors. At a concentration of 20 microM, 2-APB also significantly blocked junctional conductance in cell pairs coupled by Cx26, Cx30, Cx36, Cx40, and Cx45 but did not appreciably affect coupling in cell pairs expressing Cx32, Cx43, and Cx46. Although concentration inhibition curves of 2-APB on Cx36 channels were similar to Cx50 (Cx36; IC(50), 3.0 microM), IC(50) values were higher for Cx43 (51.6 microM), Cx45 (18.1 microM), and Cx46 (29.4 microM). The blocking action of 2-APB did not substantially alter transjunctional voltage-dependent gating of Cx50 gap junction channels, and recordings from poorly coupled pairs of Cx50-transfected N2A cells indicated that 2-APB reduced gap junction channel open probability without changing the main state single-channel conductance. The differential efficacy of block by 2-APB of gap junction channels formed by different connexins may provide a useful tool that could be exploited in gap junction research to selectively block certain gap junction channel subtypes.
[14]
Huang K, O'Neill KL, Li J, Zhou W, Han N, Pang X, Wu W, Struble L, Borgstahl G, Liu Z, Zhang L, Luo X. BH3-only proteins target BCL-xL/MCL-1, not BAX/BAK, to initiate apoptosis. Cell Res, 2019,29(11):942-952.
It has been widely accepted that mitochondria-dependent apoptosis initiates when select BH3-only proteins (BID, BIM, etc.) directly engage and allosterically activate effector proteins BAX/BAK. Here, through reconstitution of cells lacking all eight pro-apoptotic BH3-only proteins, we demonstrate that all BH3-only proteins primarily target the anti-apoptotic BCL-2 proteins BCL-xL/MCL-1, whose simultaneous suppression enables membrane-mediated spontaneous activation of BAX/BAK. BH3-only proteins' apoptotic activities correlate with affinities for BCL-xL/MCL-1 instead of abilities to directly activate BAX/BAK. Further, BID and BIM do not distinguish BAX from BAK or accelerate BAX/BAK activation following inactivation of BCL-xL/MCL-1. Remarkably, death ligand-induced apoptosis in cells lacking BH3-only proteins and MCL-1 is fully restored by BID mutants capable of neutralizing BCL-xL, but not direct activation of BAX/BAK. Taken together, our findings provide a
[15]
Frank DO, Dengjel J, Wilfling F, Kozjak-Pavlovic V, Häcker G, Weber A. The pro-apoptotic BH3-only protein Bim interacts with components of the translocase of the outer mitochondrial membrane (TOM). PLoS One, 2015,10(4):e0123341.
The pro-apoptotic Bcl-2-family protein Bim belongs to the BH3-only proteins known as initiators of apoptosis. Recent data show that Bim is constitutively inserted in the outer mitochondrial membrane via a C-terminal transmembrane anchor from where it can activate the effector of cytochrome c-release, Bax. To identify regulators of Bim-activity, we conducted a search for proteins interacting with Bim at mitochondria. We found an interaction of Bim with Tom70, Tom20 and more weakly with Tom40, all components of the Translocase of the Outer Membrane (TOM). In vitro import assays performed on tryptically digested yeast mitochondria showed reduced Bim insertion into the outer mitochondrial membrane (OMM) indicating that protein receptors may be involved in the import process. However, RNAi against components of TOM (Tom40, Tom70, Tom22 or Tom20) by siRNA, individually or in combination, did not consistently change the amount of Bim on HeLa mitochondria, either at steady state or upon de novo-induction. In support of this, the individual or combined knock-downs of TOM receptors also failed to alter the susceptibility of HeLa cells to Bim-induced apoptosis. In isolated yeast mitochondria, lack of Tom70 or the TOM-components Tom20 or Tom22 alone did not affect the import of Bim into the outer mitochondrial membrane. In yeast, expression of Bim can sensitize the cells to Bax-dependent killing. This sensitization was unaffected by the absence of Tom70 or by an experimental reduction in Tom40. Although thus the physiological role of the Bim-TOM-interaction remains unclear, TOM complex components do not seem to be essential for Bim insertion into the OMM. Nevertheless, this association should be noted and considered when the regulation of Bim in other cells and situations is investigated.
[16]
Huang DC, Strasser A. BH3-Only proteins-essential initiators of apoptotic cell death. Cell, 2000,103(6):839-842.
[17]
Shukla S, Saxena S, Singh BK, Kakkar P. BH3-only protein BIM: an emerging target in chemotherapy. Eur J Cell Biol, 2017,96(8):728-738.
BH3-only proteins constitute major proportion of pro-apoptotic members of B-cell lymphoma 2 (Bcl-2) family of apoptotic regulatory proteins and participate in embryonic development, tissue homeostasis and immunity. Absence of BH3-only proteins contributes to autoimmune disorders and tumorigenesis. Bim (Bcl-2 Interacting Mediator of cell death), most important member of BH3-only proteins, shares a BH3-only domain (9-16 aa) among 4 domains (BH1-BH4) of Bcl-2 family proteins and highly pro-apoptotic in nature. Bim initiates the intrinsic apoptotic pathway under both physiological and patho-physiological conditions. Reduction in Bim expression was found to be associated with tumor promotion and autoimmunity, while overexpression inhibited tumor growth and drug resistance as cancer cells suppress Bim expression and stability. Apart from its role in normal homeostasis, Bim has emerged as a central player in regulation of tumorigenesis, therefore gaining attention as a plausible target for chemotherapy. Regulation of Bim expression and stability is complicated and regulated at multiple levels viz. transcriptional, post-transcriptional, post-translational (preferably by phosphorylation and ubiquitination), epigenetic (by promoter acetylation or methylation) including miRNAs. Furthermore, control over Bim expression and stability may be exploited to enhance chemotherapeutic efficacy, overcome drug resistance and select anticancer drug regimen as various chemotherapeutic agents exploit Bim as an executioner of cell death. Owing to its potent anti-tumorigenic activity many BH3 mimetics e.g. ABT-737, ABT-263, obatoclax, AT-101and A-1210477 have been developed and entered in clinical trials. It is more likely that in near future strategies commanding Bim expression and stability ultimately lead to Bim based therapeutic regimen for cancer treatment.
[18]
Qian G, Yao W, Zhang S, Bajpai R, Hall WD, Shanmugam M, Lonial S, Sun SY. Co-inhibition of BET and proteasome enhances ER stress and Bim-dependent apoptosis with augmented cancer therapeutic efficacy. Cancer Lett, 2018,435:44-54.
Agents that inhibit bromodomain and extra-terminal domain (BET) protein have been actively tested in the clinic as potential anticancer drugs. Proteasome inhibitors such as carfilzomib (CFZ) are FDA-approved for the treatment of patients with advanced multiple myeloma and have been tested against other cancers. The current study focuses on the combination of a BET inhibitor (e.g., JQ1) and a proteasome inhibitor (e.g., CFZ) as a novel cancer therapeutic strategy and the underlying mechanisms. The tested combination (JQ1 with CFZ) synergistically decreased cell survival and enhanced apoptosis in vitro and inhibited tumor growth in vivo. The dramatic induction of apoptosis was accompanied by enhanced elevation of Bim and ER stress. Bim knockout significantly attenuated apoptosis induced by the combination, suggesting a critical role of Bim induction in mediating the enhanced induction of apoptosis by BET and proteasome co-inhibition. The combination significantly increased Bim mRNA levels with limited effect on Bim protein stability, suggesting a primary transcriptional regulation of enhanced Bim expression. Our findings warrant further investigation of this combinatorial strategy as an effective regimen against cancer in the clinic.

基金

广东省自然科学基金自由申请项目(2017A030313492)
广东省医学科研基金(A2017042)

版权

版权所有,未经授权,不得转载、摘编本刊文章,不得使用本刊的版式设计。
PDF(3355 KB)

921

Accesses

0

Citation

Detail

段落导航
相关文章

/