en
×

分享给微信好友或者朋友圈

使用微信“扫一扫”功能。
通讯作者:

蒋龙凤,E-mail:longfengjiang@njmu.edu.cn

中图分类号:R994.39

文献标识码:A

文章编号:1007-4368(2023)10-1350-07

DOI:10.7655/NYDXBNS20231003

参考文献 1
YANG T,WANG H,WANG X,et al.The dual role of innate immune response in acetaminophen ⁃induced liver injury[J].Biology(Basel),2022,11(7):1057
参考文献 2
SHEN T,LIU Y,SHANG J,et al.Incidence and etiology of drug ⁃induced liver injury in Mainland China[J].Gas⁃ troenterology,2019,156(8):2230-2241
参考文献 3
LEE W M.Acetaminophen(APAP)hepatotoxicity ⁃isn’t it time for APAP to go away?[J].J Hepatol,2017,67(6):1324-1331
参考文献 4
ORNING P,WENG D,STARHEIM K,et al.Pathogen blockade of TAK1 triggers caspase⁃8⁃dependent cleavage of gasdermin D and cell death[J].Science,2018,362(6418):1064-1069
参考文献 5
ZHENG Z Z,DENG W Y,BAI Y,et al.The lysosomal rag⁃ ragulator complex licenses RIPK1 and caspase⁃8⁃medi⁃ ated pyroptosis by Yersinia[J].Science,2021,372(6549):eabg0269
参考文献 6
WEINLICH R,OBERST A,BEERE H M,et al.Necropto⁃ sis in development,inflammation and disease[J].Nat Rev Mol Cell Biol,2017,18(2):127-136
参考文献 7
SHI J J,ZHAO Y,WANG K,et al.Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death [J].Nature,2015,526(7575):660-665
参考文献 8
NEWTON K,WICKLIFFE K E,MALTZMAN A,et al.Activity of caspase ⁃ 8 determines plasticity between cell death pathways[J].Nature,2019,575(7784):679-682
参考文献 9
JIANG L F,KE M,YUE S,et al.Blockade of Notch sig⁃ naling promotes acetaminophen ⁃induced liver injury[J].Immunol Res,2017,65(3):739-749
参考文献 10
YANG T,QU X,ZHAO J,et al.Macrophage PTEN con⁃ trols STING ⁃ induced inflammation and necroptosis through NICD/NRF2 signaling in APAP⁃induced liver in⁃ jury[J].Cell Commun Signal,2023,21(1):160
参考文献 11
WEI G,BERGQUIST A,BROOMÉ U,et al.Acute liver failure in Sweden:etiology and outcome[J].J Intern Med,2007,262(3):393-401
参考文献 12
KOLODZIEJCZYK A A,FEDERICI S,ZMORA N,et al.Acute liver failure is regulated by MYC⁃and microbiome⁃ dependent programs[J].Nat Med,2020,26(12):1899-1911
参考文献 13
SCHWARZER R,JIAO H P,WACHSMUTH L,et al.FADD and caspase⁃8 regulate gut homeostasis and inflam⁃ mation by controlling MLKL⁃and GSDMD⁃mediated death of intestinal epithelial cells[J].Immunity,2020,52(6):978-993
参考文献 14
MIFFLIN L,OFENGEIM D,YUAN J Y.Receptor ⁃inter⁃ acting protein kinase 1(RIPK1)as a therapeutic target [J].Nat Rev Drug Discov,2020,19(8):553-571
参考文献 15
XU P F,TAO C R,ZHU Y Y,et al.TAK1 mediates neu⁃ ronal pyroptosis in early brain injury after subarachnoid hemorrhage[J].J Neuroinflammation,2021,18(1):188
参考文献 16
FAJGENBAUM D C,JUNE C H.Cytokine storm[J].N Engl J Med,2021,384(16):2255-2273
参考文献 17
WANG W J,GAO W K,ZHU Q J,et al.TAK1:a molecu⁃ lar link between liver inflammation,fibrosis,steatosis,and carcinogenesis[J].Front Cell Dev Biol,2021,9:734749
参考文献 18
XU D C,JIN T J,ZHU H,et al.TBK1 suppresses RIPK1⁃ driven apoptosis and inflammation during development and in aging[J].Cell,2018,174(6):1477-1491
参考文献 19
MALIREDDI R K S,GURUNG P,KESAVARDHANA S,et al.Innate immune priming in the absence of TAK1 drives RIPK1 kinase activity ⁃ independent pyroptosis,apoptosis,necroptosis,and inflammatory disease[J].J Exp Med,2020,217(3):20191644
参考文献 20
TAN S X,ZHAO J,SUN Z Y,et al.Hepatocyte ⁃specific TAK1 deficiency drives RIPK1 kinase⁃dependent inflam⁃ mation to promote liver fibrosis and hepatocellular carci⁃ noma[J].PNAS,2020,117(25):14231-14242
目录contents

    摘要

    目的:探讨Notch1信号通过转化生长因子β激活激酶1(transforming growth factor-β-activated kinase 1,TAK1)调控对乙酰氨基酚(acetyl-para-aminophenol,APAP)诱导的肝损伤(APAP induced liver injury,AILI)的作用机制。方法:髓系特异性 Notch1敲除(Notch1M-KO)和对照floxed Notch1(Notch1FL/FL)小鼠通过腹腔注射APAP构建AILI模型。留取小鼠血清标本,用全自动生化分析仪及酶联免疫反应分析法检测肝功能和细胞因子。留取小鼠肝组织标本,HE染色观察肝组织病理损伤情况,使用 Suzuki评分评估肝组织损伤程度,免疫印迹法检测TAK1、磷酸化TAK1(p-TAK1)、p65、磷酸化p65(p-p65)、Caspase-8(Casp-8)、受体相互作用蛋白激酶 1(receptor-interacting protein kinase 1,RIPK1)、磷酸化混合谱系激酶域样蛋白(mixed lineage kinase domain-like protein,p-MLKL)的表达,免疫荧光染色观察CD11b、p-TAK1的表达及活性氧(reactive oxygen species ROS)水平。结果:小鼠腹腔注射APAP后,肝脏病理提示肝细胞体积增大,窦道淤血,出现广泛的坏死。与Notch1FL/FL对照组相比,Notch1M-KO 小鼠血清丙氨酸氨基转移酶(alanine aminotransferase,ALT)、天冬氨酸氨基转移酶(aspartate aminotransferase,AST)明显升高,血清炎性因子水平上升,HE染色显示肝细胞体积增大更明显,伴大面积坏死及炎性细胞浸润,DCF探针检测显示原代肝细胞内ROS增加。肝组织p-TAK1表达增加,Casp-8的表达减少,RIPK1、p-MLKL表达增加。结论:在AILI中,髓系特异性Notch1敲除可活化TAK1,降低Casp-8水平,激活RIPK1-MLKL坏死性凋亡通路,加重肝损伤的发生。

    Abstract

    Objective:To investigate the mechanism by which Notch1 signaling regulates acetyl-para-aminophenol(APAP)-induced liver injury(AILI)via TAK1. Methods:AILI models were constructed on myeloid-specific Notch1 knockout(Notch1M - KO)and floxed Notch1(Notch1FL/FL)mice by intraperitoneal injection of APAP. Serum samples of mice were collected for detection of liver function and cytokines by fully automated biochemical analyser and enzyme - linked immunoassay(ELISA),respectively. The pathological damage of liver tissue was observed by HE staining,and the degree of liver tissue damage was evaluated by Suzuki score. Western blot analysis was performed to detect the expression levels of TAK1,phospho -TAK1,p65,phospho -p65,caspase -8,receptor -interacting protein kinase1(RIPK1),and phospho - mixed lineage kinase domain - like protein(MLKL)in the liver tissue. The expressions of CD11b,p - TAK1 and the level of reactive oxygen species(ROS)were observed by immunofluorescence staining. Results:After injected with APAP intraperitoneally in mice,liver histopathological examination suggested increased hepatocyte volume,sinus congestion,and extensive necrosis. Compared with Notch1FL/FL group,Notch1M- KO mice showed significantly elevated serum alanine aminotransferase(ALT) and aspartate aminotransferase(AST),increased inflammatory factor levels. HE staining showed more pronounced increase in hepatocyte volume,accompanied by extensive necrosis and increased infiltration of inflammatory cells. Additionally,the primary hepatocytes showed higher levels of ROS when assessed using the DCF probe. The expression of p-TAK1 in liver tissue elevated,and the expression of caspase-8 was down-regulated,while the expressions of RIPK1 and p-MLKL were up- regulated. Conclusion:In AILI,myeloid-specific Notch1 knockout activates TAK1 expression,which also decreases caspase-8 levels and promotes the RIPK1-MLKL necroptosis pathway,aggravating the liver injury.

  • 药物性肝损伤(drug⁃induced liver injury,DILI) 是临床上最常见的肝脏疾病,由某种特定的药物或/ 和其代谢产物直接或间接作用于肝脏导致,其临床表现多样[1]。一项来自308个医学中心25 927例患者的回顾性研究显示,DILI 发生率为 0.023 8%[2]。对乙酰氨基酚(acetyl⁃para⁃aminophenol,APAP)是临床上最常用的解热镇痛药物,过量的APAP 摄入是导致严重肝损伤最常见的病因[3]。然而APAP导致 DILI的发病机制还有待进一步研究。

  • 在APAP诱导的肝损伤(APAP induced liverinjury, AILI)中,肝脏固有免疫反应激活和肝细胞死亡是其发病机制中的重要环节。既往研究显示,转化生长因子β激活激酶1(transforming growth factor⁃β⁃activated kinase1,TAK1)是核转录因子(nuclear factor,NF)⁃κB (p⁃65)信号通路激活过程中的关键激酶,在机体固有免疫和适应性免疫中发挥重要作用,如在巨噬细胞中,TAK1可调控gasdermin D(GSDMD)的水解,从而调控细胞死亡[4]。而在巨噬细胞中,caspase ⁃8 (Casp⁃8)已被证明是 NLRP3 炎症小体形成、耶尔森菌诱导的促炎细胞因子表达及焦亡过程中必需的[5]。进一步研究显示,抑制Casp⁃8的表达或敲除 Casp⁃8后,受体相互作用蛋白激酶1(receptor⁃inter⁃ acting protein kinase1,RIPK1)⁃RIPK3 ⁃ 混合谱系激酶域样蛋白(mixed lineage kinase domain⁃like protein, MLKL)途径被激活,诱导肿瘤坏死因子(tumor necro⁃ sis factor,TNF)释放,加剧细胞损伤,甚至死亡[6-8]; 说明 Casp⁃8 可能是控制细胞死亡并防止组织损伤的关键。在AILI中,肝巨噬细胞(如Kupffer细胞)通过释放大量的促炎因子介导炎症反应,从而加重肝细胞死亡,参与肝损伤的进展[1]。本课题组既往研究发现阻断巨噬细胞Notch1信号可以加重AILI[9]; 但Notch1信号调控APAP诱导肝细胞死亡的机制仍不清楚,有待进一步研究。

  • 本研究构建腹腔注射APAP诱导小鼠肝损伤模型,探索髓系特异性 Notch1 敲除(myeloid ⁃ specific Notch1 knockout,Notch1M ⁃ KO)诱导肝细胞死亡的机制。研究发现,髓系特异性 Notch1 敲除通过上调 TAK1 的表达,从而抑制 Casp⁃8,调控 RIPK1⁃MLKL 坏死性凋亡信号,加重肝组织损伤。

  • 1 材料和方法

  • 1.1 材料

  • SPF级雄性C57BL/6J背景的髓系特异性Notch1 敲除(Notch1M ⁃ KO)和对照 floxed Notch1(Notch1FL/FL) 小鼠,6~8 周龄,来自美国 Jackson 实验室。ELISA 试剂盒(江苏菲亚生物科技有限公司),APAP(Sigma⁃ Aldrich 公司,美国)。Notch1 抗体、TAK1 抗体、p65 抗体、p⁃p65抗体、Casp⁃8抗体、RIPK1抗体、p⁃MLKL 抗体(CST公司,美国),p⁃TAK1抗体(赛默飞公司,美国),荧光标记二抗(Jackson Immunoresearch,美国),CD11b 抗体(Abcam 公司,美国),H2DCFDA 活性氧荧光探针(ThermoFisher 公司,美国)。荧光显微镜(Keyence 公司,日本),全自动生化分析仪 (Olympus公司,日本)。

  • 1.2 方法

  • 1.2.1 小鼠肝细胞和肝巨噬细胞分离

  • 小鼠饲养于南京医科大学实验动物中心,温度 (25±1)°C,湿度(55±5)%,光照时间维持 12 h 光照和12 h 黑暗周期,并给予充足的食物和无菌蒸馏水供其自由采食,所有实验动物符合南京医科大学实验动物福利伦理(批准编号:IACUC⁃2206039)。

  • 采用“两步灌注法”分离Notch1MKO和Notch1FL/FL 小鼠的原代肝细胞及肝脏巨噬细胞[10]。每组取3只小鼠,打开小鼠腹腔,暴露门静脉,门静脉穿刺成功后固定,10 mL/min流量经过门静脉灌注含1%双抗的 EGTA 溶液(9.5 mg EGTA 粉末加至 50 mL HBSS 中,37℃预热30 min),总量为40 mL,再用含1%双抗的Ⅱ型胶原酶(0.01 g胶原酶加至40 mL GBSS 溶液中,37℃预热30 min)灌注肝脏,灌注完成后可见肝脏呈土黄色。灌注完毕后用组织剪将肝脏从腹腔分离后置入含有胶原酶维持液的培养皿中,用镊子钝性分离肝脏直至无明显块状组织,制成细胞悬液。70目过滤网过滤后离心,GBSS重悬离心(50 g, 5 min)清洗2次,Percoll密度梯度离心纯化,分别获得原代肝细胞和肝巨噬细胞,0.4%台酚蓝测定细胞状态并计数,DMEM+10%胎牛血清(FBS)+1%青链霉素培养,接种至6孔板中,细胞数为1.0×106~1.5× 106 个/孔,培养箱(37℃、5% CO2)中培养3 h后,37℃预热的 PBS 清洗细胞 3 次,去除非贴壁细胞。24 h 换液,并收集上清液,-80℃储存。按照每1×106 个细胞加入 100 μL RIPA,混匀后冰浴静置 30 min 充分裂解细胞。BCA定量蛋白,用于Western blot检测。

  • 1.2.2 小鼠AILI模型的制备及生存率分析

  • 取 Notch1M ⁃ KO和 Notch1FL/FL小鼠各 6 只,腹腔注射 APAP 以构建 AILI 模型,APAP 溶液在实验使用前1 h配制,将APAP溶解在PBS中,浓度为10 mg/mL,并在水浴箱中加热到40℃。雄性小鼠禁食16 h,然后用 400 mg/kg APAP 腹腔注射。在注射后的 72 h 内每12 h观察小鼠存活情况并记录。

  • 1.2.3 小鼠分组及处理

  • Notch1M ⁃KO和 Notch1FL/FL小鼠各 12 只,分别随机取 6 只,通过腹腔注射 APAP 构建 AILI 模型 (Notch1FL/FL+APAP组、Notch1FL/FL+APAP组),具体方法同前。其余6只腹腔注射PBS作为对照(Notch1FL/FL+ PBS 组、Notch1M ⁃KO+PBS 组)。24 h 后通过乙醚吸入对小鼠进行安乐死,收集血液和肝脏组织。

  • 1.2.4 肝功能及细胞因子检测

  • 小鼠的血液样本室温静置 30 min,3 000 r/min 离心12 min,收集上清。采用全自动生化分析仪检测丙氨酸氨基转移酶(alanine aminotransferase,ALT)、天冬氨酸氨基转移酶(aspartate aminotransferase, AST)。TNF⁃α、白介素(interleukin,IL)⁃1β、IL⁃6、转化生长因子(transforming growth factor,TGF)⁃β1 使用 ELISA检测。

  • 1.2.5 肝脏组织切片染色

  • 收集小鼠肝组织,部分肝组织石蜡包埋后切片 (5 μm)行 HE 染色观察肝脏病理情况,Suzuki 评分评估肝组织损伤程度。部分肝组织OCT包埋后,冰冻切片(10 μm)行免疫荧光染色观察肝组织CD11b 和p⁃TAK1的表达情况;冰冻切片室温复温30 min, 4%福尔马林固定 10 min,0.1% Triton X ⁃100 破膜 (2.5% BSA),CD11b抗体、p⁃TAK1抗体(1∶100)4℃ 孵育过夜,荧光标记二抗(1∶250)室温避光孵育1 h, DAPI封片,在荧光显微镜下观察并采集图像。

  • 1.2.6 Western blot检测

  • 50 mg 肝组织加入 RIPA 蛋白裂解液超声碾磨后提取总蛋白,BCA 试剂盒定量,确保蛋白上样量为30 μg/孔。经过电泳,转膜,封闭,Notch1抗体、TAK1抗体、p⁃TAK1抗体、p65抗体、p⁃p65抗体、 Casp⁃8 抗体、RIPK1 抗体、p ⁃MLKL 抗体(1∶1 000) 4℃孵育过夜,根据一抗种属进行二抗(1∶3 000) 室温孵育 1 h,曝光得到条带后经 Image J 分析灰度值。

  • 1.2.7 原代肝细胞ROS检测

  • 分离 Notch1FL/FL +APAP 组、Notch1M ⁃ KO+APAP 组小鼠(每组4只)原代肝细胞进行ROS检测,DCF探针(1∶500)加入不含血清培养基的原代肝细胞, 37℃孵育20~30 min。在荧光显微镜下观察并采集图像。

  • 1.3 统计学方法

  • 采用Prism8软件进行数据统计学分析,Kaplan⁃ Meier法进行小鼠生存分析;计量数据采用均值±标准差(x-±s)表示,组间比较采用t检验。P <0.05为差异有统计学意义。

  • 2 结果

  • 2.1 髓系特异性Notch1敲除验证

  • 分别分离Notch1FL/FL、Notch1MKO小鼠的原代肝细胞和肝巨噬细胞,提取蛋白,Western blot 检测 Notch1的表达。结果显示,Notch1MKO小鼠肝巨噬细胞不表达Notch1,而Notch1FL/FL小鼠的原代肝细胞和肝巨噬细胞中Notch1正常表达(图1A)。

  • 2.2 Notch1MKO加重APAP诱导的肝损伤

  • APAP 腹腔注射构建小鼠 AILI 模型,观察小鼠死亡率。结果显示,与 Notch1FL/FL +APAP 组相比, Notch1MKO+APAP组小鼠死亡率有所增加,但差异无统计学意义(图1B)。

  • HE染色结果显示,PBS处理的小鼠肝脏病理未见明显异常。小鼠腹腔注射APAP 后,肝脏病理提示肝细胞体积增大,窦道淤血,并出现广泛的坏死。与Notch1FL/FL+APAP组相比,经APAP腹腔注射的Notch1MKO小鼠肝窦淤血,肝细胞体积增大及空泡样变性,细胞边界形态模糊等更为严重,出现广泛细胞的坏死和炎性细胞浸润;两组Suzuki评分差异有统计学意义(P <0.001,图1C)。血清转氨酶是评价肝功能损伤的一个重要标志,PBS处理的小鼠血清ALT及AST水平正常,而APAP处理组表达明显升高。与 Notch1FL/FL +APAP 组相比,Notch1M ⁃ KO+APAP 组小鼠血清ALT和AST水平升高更明显,差异具有统计学意义(P <0.001,图1D)。进一步通过免疫荧光染色观察肝脏切片中 CD11b 的表达情况,APAP 处理后,肝组织切片中 CD11b 表达明显升高,说明肝损伤后肝内巨噬细胞浸润增多;而与Notch1FL/FL+ APAP 组相比,Notch1M ⁃KO+APAP 组肝脏巨噬细胞浸润增加更明显,差异有统计学意义(P <0.001,图1E)。

  • 图1 Notch1M-KO加重APAP诱导的肝损伤及炎性细胞浸润

  • Figure1 Myeloid⁃specific Notch1 knockout exacerbates APAP⁃induced liver injury and immune cell accumulation

  • 2.3 Notch1MKO通过激活 TAK1 信号抑制细胞坏死性凋亡

  • Notch1 敲除后,APAP 处理组小鼠肝组织中 p⁃TAK1 表达水平明显升高(P <0.001),同时伴有 p⁃p65 表达上调(图2A),但 Casp⁃8 表达受到抑制, RIPK1 和 p ⁃MLKL 的表达上调(P <0.001,图2A)。而 PBS 处理的 Notch1FL/F和 Notch1MKO小鼠肝组织中 p⁃TAK1、p⁃p65、Casp⁃8、RIPK1和p⁃MLKL蛋白的表达未见明显差异。说明APAP可激活坏死性凋亡通路,导致肝细胞坏死增加,符合组织学结果。进一步通过免疫荧光染色检测 p⁃TAK1 在肝组织中表达情况,经APAP腹腔注射的Notch1MKO小鼠肝组织中p⁃TAK1阳性的细胞明显增多(P <0.001,图2B)。结合Western blot和免疫荧光染色结果,说明Notch1 敲除后,激活 p⁃TAK1 的表达促进肝脏细胞凋亡坏死,加重APAP诱导肝损伤的发生。

  • 2.4 Notch1M ⁃ KO加重全身炎症反应及原代肝细胞 ROS产生

  • 与Notch1FL/FL+APAP组小鼠相比,Notch1MKO+APAP 组小鼠血清中TGF⁃β1、IL⁃1β、IL⁃6、TNF⁃α的表达明显升高,差异具有统计学意义(图3A~D)。进一步通过DCF探针检测原代肝细胞ROS情况,结果显示 Notch1MKO小鼠肝细胞的ROS水平明显升高(P <0.01,图3E)。

  • 3 讨论

  • AILI可能不是DILI首位病因,却是导致急性肝损伤的主要病因之一[3]。在美国,46%的急性肝损伤是服用过量APAP导致的[11]。急性肝损伤随后引起各种并发症甚至是多器官衰竭,进一步危及患者生命,最终需要行肝移植手术,而肝源缺乏导致只有少数患者才能得到救治。因此,进一步研究AILI 发病机制,为临床提供潜在治疗靶点和预防策略显得非常重要。

  • 本研究通过对 Notch1 特异性敲除小鼠腹腔注射APAP 构建AILI 动物模型。研究发现,与PBS 处理组相比,APAP 处理组小鼠肝脏病理提示肝细胞水肿及肝细胞坏死,伴炎性细胞浸润,说明 AILI 造模成功。与Notch1FL/FL+APAP组相比,Notch1M ⁃KO+ APAP 组小鼠肝组织损伤更严重,使用 Suzuki 量化肝组织损伤程度,Suzuki评分明显高于对照组,且血清ALT、AST表达明显升高,说明髓系特异性Notch1 敲除后加重了APAP诱导的肝损伤。

  • 图2 Notch1MKO通过激活TAK1信号抑制细胞坏死性凋亡

  • Figure2 Myeloid⁃specific Notch1 knockout inhibits necroptosis through activation of TAK1 signaling

  • 图3 Notch1MKO加重全身炎症反应及原代肝细胞ROS产生

  • Figure3 Myeloid⁃specific Notch1 knockout exacerbates systemic inflammatory response and ROS generation in primary hepatocytes

  • 肝脏巨噬细胞在肝脏遭遇损伤应激后扮演重要角色,研究显示,在AILI中,有51.5%的Kupffer细胞被激活[12]。本研究采用免疫荧光染色观察CD11b 的表达情况,结果显示,经APAP处理后,Notch1MKO 组CD11b+ 细胞比例明显升高,说明肝内巨噬细胞浸润增加。浸润的巨噬细胞随后释放各种细胞因子,如TNF⁃α等,与靶细胞膜表面的TNF受体相结合,激活RIPK1⁃MLKL信号通路放大炎症信号[13],甚至启动细胞程序性死亡[14]。程序性死亡是细胞应对外界应激损伤的一个重要方式。既往研究表明可通过 NF ⁃κB 和丝裂原活化蛋白激酶(mitogen ⁃activated protein kinase,MAPK)信号调控炎症反应[15]。APAP 诱导肝损伤后,坏死的肝细胞释放损伤相关的分子模式(damage ⁃associated molecular patterns,DAMP) 招募炎性细胞。浸润的炎性细胞随后释放多种促炎因子,导致“炎性因子风暴”或炎症因子释放综合征(cytokine release syndrome,CRS),进一步危及机体的生命[16]。本研究结果显示,APAP 处理后小鼠RIPK1、p⁃MLKL表达增加,且Notch1MKO组表达增加更明显,说明巨噬细胞 Notch1 缺乏导致炎性因子释放增加,从而促进肝细胞坏死性凋亡通路激活,进一步加重肝损伤的发生。

  • 既往研究发现,TAK1是NF⁃κB和c⁃Jun N端激酶的上游细胞内蛋白激酶,它被众多细胞因子、生长因子和微生物产物激活[17]。而TAK1和RIPK1的相互作用在多种模型中被证实[18-20]。本研究通过 Western blot 和免疫荧光染色,发现在 Notch1M ⁃KO组 AILI 模型中 p⁃TAK1 表达明显升高,说明 Notch1 缺乏可促进肝脏 TAK1 的磷酸化。进一步研究显示, p⁃p65的表达上调,而Casp⁃8的表达下调,最终激活 RIPK1⁃MLKL 信号通路,导致肝细胞坏死性凋亡增加。ROS是反映细胞氧化应激的一个重要标志,过量的APAP在体内耗尽谷胱甘肽后形成APAP蛋白结合物,进而导致肝细胞内线粒体膜通透性转换孔开放,ROS释放,从而进一步加重氧化应激,最终使细胞裂解死亡[1]。通过 DCF 探针检测肝细胞中 ROS,进一步证实Notch1MKO组ROS水平明显升高。

  • 综上所述,本研究成功构建小鼠 AILI 模型,并通过髓系特异性敲除进一步探讨固有免疫 Notch1 信号缺陷对 AILI 肝细胞死亡机制的作用。结果显示,髓系特异性 Notch1 敲除加重肝细胞损伤,其可能机制是通过上调肝内TAK1信号,抑制Casp⁃8的表达,从而激活RIPK1⁃MLKL信号通路启动肝细胞坏死性凋亡,最终导致损伤加重。本研究有可能为 DILI 提供潜在的治疗靶点,但也存在一定的缺陷。如没有通过 Notch1 过表达进一步验证下游信号通路表达情况,Notch1信号在巨噬细胞中如何调节炎性细胞因子的释放,以及细胞间接触相互影响如何,TAK1 如何调控 Casp8 和 RIPK1 的表达等,均有待进一步研究。

  • 参考文献

    • [1] YANG T,WANG H,WANG X,et al.The dual role of innate immune response in acetaminophen ⁃induced liver injury[J].Biology(Basel),2022,11(7):1057

    • [2] SHEN T,LIU Y,SHANG J,et al.Incidence and etiology of drug ⁃induced liver injury in Mainland China[J].Gas⁃ troenterology,2019,156(8):2230-2241

    • [3] LEE W M.Acetaminophen(APAP)hepatotoxicity ⁃isn’t it time for APAP to go away?[J].J Hepatol,2017,67(6):1324-1331

    • [4] ORNING P,WENG D,STARHEIM K,et al.Pathogen blockade of TAK1 triggers caspase⁃8⁃dependent cleavage of gasdermin D and cell death[J].Science,2018,362(6418):1064-1069

    • [5] ZHENG Z Z,DENG W Y,BAI Y,et al.The lysosomal rag⁃ ragulator complex licenses RIPK1 and caspase⁃8⁃medi⁃ ated pyroptosis by Yersinia[J].Science,2021,372(6549):eabg0269

    • [6] WEINLICH R,OBERST A,BEERE H M,et al.Necropto⁃ sis in development,inflammation and disease[J].Nat Rev Mol Cell Biol,2017,18(2):127-136

    • [7] SHI J J,ZHAO Y,WANG K,et al.Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death [J].Nature,2015,526(7575):660-665

    • [8] NEWTON K,WICKLIFFE K E,MALTZMAN A,et al.Activity of caspase ⁃ 8 determines plasticity between cell death pathways[J].Nature,2019,575(7784):679-682

    • [9] JIANG L F,KE M,YUE S,et al.Blockade of Notch sig⁃ naling promotes acetaminophen ⁃induced liver injury[J].Immunol Res,2017,65(3):739-749

    • [10] YANG T,QU X,ZHAO J,et al.Macrophage PTEN con⁃ trols STING ⁃ induced inflammation and necroptosis through NICD/NRF2 signaling in APAP⁃induced liver in⁃ jury[J].Cell Commun Signal,2023,21(1):160

    • [11] WEI G,BERGQUIST A,BROOMÉ U,et al.Acute liver failure in Sweden:etiology and outcome[J].J Intern Med,2007,262(3):393-401

    • [12] KOLODZIEJCZYK A A,FEDERICI S,ZMORA N,et al.Acute liver failure is regulated by MYC⁃and microbiome⁃ dependent programs[J].Nat Med,2020,26(12):1899-1911

    • [13] SCHWARZER R,JIAO H P,WACHSMUTH L,et al.FADD and caspase⁃8 regulate gut homeostasis and inflam⁃ mation by controlling MLKL⁃and GSDMD⁃mediated death of intestinal epithelial cells[J].Immunity,2020,52(6):978-993

    • [14] MIFFLIN L,OFENGEIM D,YUAN J Y.Receptor ⁃inter⁃ acting protein kinase 1(RIPK1)as a therapeutic target [J].Nat Rev Drug Discov,2020,19(8):553-571

    • [15] XU P F,TAO C R,ZHU Y Y,et al.TAK1 mediates neu⁃ ronal pyroptosis in early brain injury after subarachnoid hemorrhage[J].J Neuroinflammation,2021,18(1):188

    • [16] FAJGENBAUM D C,JUNE C H.Cytokine storm[J].N Engl J Med,2021,384(16):2255-2273

    • [17] WANG W J,GAO W K,ZHU Q J,et al.TAK1:a molecu⁃ lar link between liver inflammation,fibrosis,steatosis,and carcinogenesis[J].Front Cell Dev Biol,2021,9:734749

    • [18] XU D C,JIN T J,ZHU H,et al.TBK1 suppresses RIPK1⁃ driven apoptosis and inflammation during development and in aging[J].Cell,2018,174(6):1477-1491

    • [19] MALIREDDI R K S,GURUNG P,KESAVARDHANA S,et al.Innate immune priming in the absence of TAK1 drives RIPK1 kinase activity ⁃ independent pyroptosis,apoptosis,necroptosis,and inflammatory disease[J].J Exp Med,2020,217(3):20191644

    • [20] TAN S X,ZHAO J,SUN Z Y,et al.Hepatocyte ⁃specific TAK1 deficiency drives RIPK1 kinase⁃dependent inflam⁃ mation to promote liver fibrosis and hepatocellular carci⁃ noma[J].PNAS,2020,117(25):14231-14242

  • 参考文献

    • [1] YANG T,WANG H,WANG X,et al.The dual role of innate immune response in acetaminophen ⁃induced liver injury[J].Biology(Basel),2022,11(7):1057

    • [2] SHEN T,LIU Y,SHANG J,et al.Incidence and etiology of drug ⁃induced liver injury in Mainland China[J].Gas⁃ troenterology,2019,156(8):2230-2241

    • [3] LEE W M.Acetaminophen(APAP)hepatotoxicity ⁃isn’t it time for APAP to go away?[J].J Hepatol,2017,67(6):1324-1331

    • [4] ORNING P,WENG D,STARHEIM K,et al.Pathogen blockade of TAK1 triggers caspase⁃8⁃dependent cleavage of gasdermin D and cell death[J].Science,2018,362(6418):1064-1069

    • [5] ZHENG Z Z,DENG W Y,BAI Y,et al.The lysosomal rag⁃ ragulator complex licenses RIPK1 and caspase⁃8⁃medi⁃ ated pyroptosis by Yersinia[J].Science,2021,372(6549):eabg0269

    • [6] WEINLICH R,OBERST A,BEERE H M,et al.Necropto⁃ sis in development,inflammation and disease[J].Nat Rev Mol Cell Biol,2017,18(2):127-136

    • [7] SHI J J,ZHAO Y,WANG K,et al.Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death [J].Nature,2015,526(7575):660-665

    • [8] NEWTON K,WICKLIFFE K E,MALTZMAN A,et al.Activity of caspase ⁃ 8 determines plasticity between cell death pathways[J].Nature,2019,575(7784):679-682

    • [9] JIANG L F,KE M,YUE S,et al.Blockade of Notch sig⁃ naling promotes acetaminophen ⁃induced liver injury[J].Immunol Res,2017,65(3):739-749

    • [10] YANG T,QU X,ZHAO J,et al.Macrophage PTEN con⁃ trols STING ⁃ induced inflammation and necroptosis through NICD/NRF2 signaling in APAP⁃induced liver in⁃ jury[J].Cell Commun Signal,2023,21(1):160

    • [11] WEI G,BERGQUIST A,BROOMÉ U,et al.Acute liver failure in Sweden:etiology and outcome[J].J Intern Med,2007,262(3):393-401

    • [12] KOLODZIEJCZYK A A,FEDERICI S,ZMORA N,et al.Acute liver failure is regulated by MYC⁃and microbiome⁃ dependent programs[J].Nat Med,2020,26(12):1899-1911

    • [13] SCHWARZER R,JIAO H P,WACHSMUTH L,et al.FADD and caspase⁃8 regulate gut homeostasis and inflam⁃ mation by controlling MLKL⁃and GSDMD⁃mediated death of intestinal epithelial cells[J].Immunity,2020,52(6):978-993

    • [14] MIFFLIN L,OFENGEIM D,YUAN J Y.Receptor ⁃inter⁃ acting protein kinase 1(RIPK1)as a therapeutic target [J].Nat Rev Drug Discov,2020,19(8):553-571

    • [15] XU P F,TAO C R,ZHU Y Y,et al.TAK1 mediates neu⁃ ronal pyroptosis in early brain injury after subarachnoid hemorrhage[J].J Neuroinflammation,2021,18(1):188

    • [16] FAJGENBAUM D C,JUNE C H.Cytokine storm[J].N Engl J Med,2021,384(16):2255-2273

    • [17] WANG W J,GAO W K,ZHU Q J,et al.TAK1:a molecu⁃ lar link between liver inflammation,fibrosis,steatosis,and carcinogenesis[J].Front Cell Dev Biol,2021,9:734749

    • [18] XU D C,JIN T J,ZHU H,et al.TBK1 suppresses RIPK1⁃ driven apoptosis and inflammation during development and in aging[J].Cell,2018,174(6):1477-1491

    • [19] MALIREDDI R K S,GURUNG P,KESAVARDHANA S,et al.Innate immune priming in the absence of TAK1 drives RIPK1 kinase activity ⁃ independent pyroptosis,apoptosis,necroptosis,and inflammatory disease[J].J Exp Med,2020,217(3):20191644

    • [20] TAN S X,ZHAO J,SUN Z Y,et al.Hepatocyte ⁃specific TAK1 deficiency drives RIPK1 kinase⁃dependent inflam⁃ mation to promote liver fibrosis and hepatocellular carci⁃ noma[J].PNAS,2020,117(25):14231-14242

  • 通知关闭
    郑重声明