Expression of tumor necrosis factor-α induced protein 8 like-1 in cisplatin-induced acute kidney injury models

Liu Lichang, Suo Dongyang, Ding Yu, Wan Xiang, Zhang Yang, Yang Xiangdong

PDF(3005 KB)
Chinese Journal of Nephrology ›› 2020, Vol. 36 ›› Issue (7) : 543-549. DOI: 10.3760/cma.j.cn441217-20200223-00099
Basic Study

Expression of tumor necrosis factor-α induced protein 8 like-1 in cisplatin-induced acute kidney injury models

Author information +
History +

Abstract

Objective To investigate the expression and role of the tumor necrosis factor-α (TNF-α) induced protein 8 like-1 (TIPE1) in acute kidney injury (AKI) induced by cisplatin in animal model and cells. Methods Twelve male C57BL/6 mice aged 6-8 weeks were randomly divided into the control group and the model group. Mice in the model group received a single intraperitoneal injection of 20 mg/kg of cisplatin (20 mg/kg saline in the control group). All mice were euthanized after 5 days. Meanwhile, serum and kidney samples were collected. The levels of serum creatinine (Scr) and blood urea nitrogen (BUN) were detected by biochemical kits. Renal histopathological changes in mice were observed by HE staining. The expression of TIPE1 in kidney was examined using immunohistochemistry. qRT-PCR was used for testing the relative expression of TIPE1 mRNA in mice kidney. Western blotting was used for testing TIPE1 and NGAL protein relative expression in mice kidney. Human kidney proximal tubular cells (HK-2) were stimulated with 20 μmol/L cisplatin for 0, 6, 12 and 24 h to establish cisplatin-induced AKI cell model. The expressions of TIPE1 mRNA and protein were detected by qRT-PCR and Western blotting in HK-2 cells. The expression of TIPE1 gene in HK-2 cells was silenced by lentivirus containing TIPE1 siRNA sequence. Then, TIPE1 stable knockout HK-2 cell strains were treated with 20 μmol/L of cisplatin for 24 hours. The protein expression of tubular damage marker neutrophil gelatinase-associated lipocalin (NGAL), microtubule-associated protein 1 light chain 3 (LC3) and Beclin1 in HK-2 cells were detected by Western blotting. Results Compared with the control group, the expressions of TIPE1 mRNA and protein were up-regulated and NGAL protein expression was increased significantly in renal tissue of the model group (all P<0.05). The expressions of TIPE1 mRNA and protein were remarkably increased with the prolongation of cisplatin treatment in HK-2 cells (both P<0.05). Compared with the scramble siRNA group, the protein expressions of NGAL, LC3-Ⅱ and Beclin1 were increased significantly in the TIPE1 siRNA group after lentivirus interfered with the expression of TIPE1 gene in HK-2 cells (all P<0.05). Conclusions The mRNA and protein expressions of TIPE1 are increased in acute kidney injury models. Gene silencing of TIPE1 can promote the expressions of early renal tubular damage marker and autophagy-related proteins, which indicates the excessive autophagy aggravates renal tubular injury. It is suggested that TIPE1 may be involved in the pathogenesis of acute kidney injury.

Key words

Acute kidney injury / Autophagy / Cisplatin / TIPE1 / NGAL

Cite this article

Download Citations
Liu Lichang. , Suo Dongyang. , Ding Yu. , Wan Xiang. , Zhang Yang. , Yang Xiangdong. Expression of tumor necrosis factor-α induced protein 8 like-1 in cisplatin-induced acute kidney injury models[J]. Chinese Journal of Nephrology, 2020, 36(7): 543-549. DOI: 10.3760/cma.j.cn441217-20200223-00099.
急性肾损伤(AKI)是临床上常见且预后较差的严重并发症,其中药物相关性肾损伤约占所有AKI病例的25%[1]。肾毒性药物主要导致肾小管-间质损伤,其病理生理机制主要涉及近端肾小管上皮细胞凋亡和坏死[2-3]、氧化应激[4-5]和肾脏免疫炎性反应[6-7]等。肿瘤坏死因子α诱导蛋白8样分子1(tumor necrosis factor alpha-induced protein 8-like 1,TNFAIP8L1 or TIPE1)是肿瘤坏死因子α诱导蛋白8(TIPE)家族中的一员,该家族是新发现的一种可以调节免疫和肿瘤的蛋白家族。TIPE1也被称为Oxi-β,是一种没有信号肽和跨膜域的胞质蛋白。在大多数肿瘤疾病中,TIPE1具有促进肿瘤细胞凋亡和抑制肿瘤细胞增殖的作用[8]。Cui等[9]的研究显示,C57BL/6小鼠肾小管上皮细胞上的TIPE1表达较高。TIPE1是否参与AKI的肾小管损伤至今尚未见报道。我们采用顺铂诱导急性肾损伤动物和细胞模型,探讨TIPE1在药物性AKI中的作用,为药物性AKI的临床治疗提供新的策略。

材料与方法

一、 材料

1. 实验动物: SPF级健康雄性C57BL/6小鼠(6~8周龄)购自山东大学动物中心,在标准实验环境(光照12 h,黑暗12 h和室温21℃±2℃)下给予正常饮食和自由饮水。
2. 细胞培养: 人肾小管上皮细胞(HK-2)购于中国科学院细胞库,在37°C、5%CO2细胞孵箱中,用改良的Eagle培养基和营养混合物F-12按1∶1配置(DMEM/F-12)的培养基培养,培养液中同时加入10%胎牛血清和1%的青链霉素。
3. 主要试剂:顺铂(15663-27-1)购自美国TargetMol;兔抗TNFAIP8L1(BS60529)购自美国Bioworld公司;TNFAIP8L1 siRNA序列的慢病毒购自中国上海吉凯;RNA提取试剂(TRIzol Reagent,15596018)购自美国Invitrogen;反转录试剂盒 [SPARKscriptⅡ RT Plus Kit(with gDNA Eraser),AG0304]购自山东思科捷科学;实时荧光定量PCR试剂盒[SuperReal PreMix Plus(SYBR Green),FP205] 购自天根生化科技(北京);兔抗中性粒细胞明胶酶相关脂质运载蛋白(anti-lipocalin2/lcn2 antibody,NGAL,PB9609)购自武汉Boster;二抗试剂盒(SAP-9100)和DAB显色剂(ZLI-9018)购自北京中杉金桥生物;山羊抗兔(SA00001-2)和山羊抗鼠(SA00001-1)二抗购自中国Proteintech;肌酐检测试剂盒(C011-2-1)和尿素氮检测试剂盒(C013-2-1)购自南京建成生物工程研究所。

二、 实验方法

1. 动物模型的制备及分组: SPF级健康雄性C57BL/6小鼠(6~8周龄)适应性饲养1周后分成模型组(n=6)和对照组(n=6)。模型组小鼠给予单次20 mg/kg顺铂腹腔注射,注射完毕后恢复正常饮食饮水,5 d后处死小鼠。收集小鼠血液标本和肾组织,用于生化和组织病理学检查。对照组小鼠给予单次20 mg/kg生理盐水腹腔注射。实验动物的饲养严格遵循山东大学动物保护协会的原则。
2. 细胞模型的制备: 将HK-2接种到6孔板中,过夜后用20 μmol/L顺铂分别刺激0、6、12、24 h。
3. 血生化与肾组织病理检查: 摘小鼠眼球取血,血液静置后离心取上清,用肌酐和尿素氮检测试剂盒测定血清肌酐和尿素氮浓度。小鼠肾脏组织在4%多聚甲醛中固定,脱水、石蜡包埋后切片,用苏木素伊红(HE)染色分析肾组织形态结构的变化。
4. 免疫组织化学检查: 肾组织切片梯度乙醇脱蜡后,行抗原修复,用TIPE1(1∶100)一抗4℃孵育过夜。按照中杉金桥免疫组化试剂盒的方法进行DAB染色,并用苏木素将细胞核染色。
5. 蛋白免疫印迹: 用加入了蛋白酶抑制剂的RIPA裂解液(RIPA∶PMSF=100∶1)裂解细胞和组织。将等量的蛋白质样品在12%SDS-聚丙烯酰胺凝胶上进行电泳,转移至聚偏二氟乙烯膜,用5%BSA封闭60 min,分别加入一抗在4℃下孵育过夜。用Tris缓冲盐水/吐温(TBST)洗涤3次后,将膜与山羊抗鼠和山羊抗兔二抗在37℃下孵育1 h,洗涤曝光显影。以β-actin蛋白条带作为内参照,使用Image J软件对目的蛋白条带与β-actin蛋白条带的灰度比值进行半定量分析。
6. 实时荧光定量PCR: 用TRIzol试剂(美国Invitrogen)提取组织和细胞总RNA。用SPARKscriptⅡ反转录酶反转录1 μg的总RNA。实时荧光定量PCR使用SuperReal PreMix Plus (SYBR Green)和Bio-Rad实时PCR仪,根据制造商建议的步骤说明加样并上机。以β-actin作为内参照,用比较Ct法(ΔΔCt)计算目的基因半定量表达量,计算公式为2-ΔΔCt。引物序列如下:m-TIPE1正向:5'-CAGAACCCATGGACACCTTC-3',反向:5'-CTTCGTGGCCTGGTACAGTT-3';h-TIPE1正向:5'-TCTGCTTCGAGAGTAGGCCAT-3',反向:5'-GCCTTGGACGCCATCTTACT-3';m-actin正向:5'-GCGTGACATCAAAGAGAAG-3',反向:5'-TCC ATACCCAAGAAGGAAGG-3';h-actin正向:5'-AG TTGCGTTACACCCTTTC-3',反向:5'-CCTTCACCG TTCCAGTTT-3'。
7. 慢病毒沉默TIPE1基因的表达: 用包含TIPE1 siRNA序列的慢病毒靶向沉默TIPE1基因,TIPE1 RNAi的靶基因序列为:CCAGAAGATGCTC AAGAACCT,Scramble siRNA序列为:TTCTCCG AACGTGTCACGT。用制备的慢病毒感染HK-2细胞,72 h后用1 μg/ml嘌呤霉素筛选5 d。5 d后,将Scramble siRNA和TIPE1 siRNA细胞各分为对照组和顺铂处理组,分别用生理盐水和20 μmol/L顺铂处理24 h。

三、 统计学方法

GraphPad Prism软件进行数据的统计学处理。正态分布的计量资料以x¯±s 表示,用t检验或方差分析比较组间差异。P<0.05被认为差异具有统计学意义。

结果

1. 急性肾损伤小鼠模型: 血生化结果显示,模型组小鼠Scr和血BUN较对照组显著升高(均P<0.05),提示顺铂诱导AKI模型制备成功。HE染色结果显示,与对照组相比,模型组小鼠肾小管上皮细胞上出现空泡样变性,细胞扁平、管腔扩张,刷状缘脱落,肾小管管腔内有脱落的上皮细胞碎片及细胞管型,可见肾小管内有压缩成三角形的凋亡的上皮细胞。Western印迹结果显示,模型组小鼠肾组织NGAL蛋白表达量较对照组显著升高(P<0.01)。提示模型组小鼠肾组织中存在明显的肾小管损伤。见图1
图1 顺铂诱导急性肾损伤小鼠的血生化、肾组织病理和NGAL蛋白表达的改变
注:A:血肌酐;B:尿素氮;C:肾组织HE染色(×400);D:NGAL蛋白表达(Western印迹);与对照组比较,aP<0.05,bP<0.01

Full size|PPT slide

2. 肾组织中TIPE1的表达: Western印迹和实时荧光定量PCR结果显示,与对照组相比,模型组小鼠肾组织TIPE1蛋白及mRNA表达量明显升高(均P<0.05)。免疫组化染色显示,TIPE1主要在肾小管上皮细胞中表达,肾小球中鲜见表达。与对照组相比,TIPE1在模型组小鼠近端肾小管上皮细胞中表达明显增加。提示TIPE1在急性肾损伤肾组织中的表达增加,且主要在近端肾小管上皮细胞中表达。见图2
图2 顺铂诱导AKI小鼠模型肾组织中TIPE1蛋白及mRNA的表达
注:A:TIPE1蛋白表达(Western印迹);B:TIPE1 mRNA表达 (实时荧光定量PCR);C:肾组织TIPE1表达(免疫组化 ×200、 ×400);与对照组比较,aP<0.05

Full size|PPT slide

3. 顺铂刺激对HK-2细胞TIPE1表达的影响: 分别用20 μmol/L顺铂刺激人肾小管上皮细胞6、12、24 h后,Western印迹和实时荧光定量PCR结果显示,顺铂刺激可增加HK-2细胞TIPE1蛋白及mRNA的表达,且呈时间依赖性(均P<0.05)。见图3
图3 顺铂刺激人肾小管上皮细胞6、12和24 h后TIPE1蛋白和mRNA表达的变化
注:A:TIPE1蛋白表达(Western印迹);B:TIPE1 mRNA表达 (实时荧光定量PCR);与0 h组比较,aP<0.05,bP<0.01

Full size|PPT slide

4. 敲除TIPE1基因对HK-2细胞自噬相关蛋白和肾小管早期损伤标志物表达的影响: 用慢病毒构建稳定敲除TIPE1基因的HK-2细胞株,Western印迹法验证TIPE1基因敲除的效果。见图4A。分别用20 μmol/L顺铂和生理盐水刺激Scramble siRNA和TIPE1 siRNA细胞24 h,检测自噬相关蛋白LC3和Beclin1及肾小管早期损伤标志物NGAL的表达。Western印迹结果显示,与Scramble siRNA组相比,TIPE1 siRNA组HK-2细胞在顺铂刺激下LC3-Ⅱ、Beclin1和NGAL蛋白表达明显增加(均P<0.05),见图4B图4C。提示敲除TIPE1基因后可引起HK-2细胞过度自噬,进一步加重肾小管损伤。
图4 敲除TIPE1基因对HK-2细胞自噬相关蛋白及肾小管早期损伤标志物表达的影响(Western印迹)
注:A:TIPE1基因敲除的验证;B:NGAL蛋白表达;C:LC3及Beclin1蛋白表达;1:Scramble siRNA组;2:TIPE1 siRNA组;与Scramble siRNA对照组比较,aP<0.05;与Scramble siRNA顺铂处理组比较,bP<0.05

Full size|PPT slide

讨论

急性肾损伤是短时间内肾功能快速减退而出现的一系列临床综合征,占住院人数的10%~15%[10]。AKI的发病因素众多,其中药物相关性肾损伤已成为住院患者发生AKI的重要原因,约占所有AKI病例的25%。药物性肾损伤主要造成近端肾小管上皮细胞凋亡和坏死、氧化应激和免疫炎性反应等。顺铂是临床上治疗各种实体肿瘤的常用化疗药物,在近端肾小管上皮细胞中积累可诱发严重的肾毒性。在顺铂诱导的AKI小鼠模型中,HE染色结果显示肾小管上皮细胞出现空泡样变性和刷状缘脱落;小管管腔内出现脱落的上皮细胞碎片和细胞管型。
TNFAIP8家族是一类与细胞增殖、炎性反应和癌变有关的蛋白,由4个成员组成:TNFAIP8(TIPE)、TNFAIP8L1(TIPE1)、TNFAIP8L2(TIPE2)和TNFAIP8L3(TIPE3)[11]。4个成员之间虽然有高度序列同源性,但在组织、器官中的表达部位和程度不同,在不同疾病中的作用也存在较大的差别。目前对TIPE和TIPE2的研究较为详尽,而对TIPE1和TIPE3研究较少。在肿瘤疾病中,TIPE和TIPE3主要发挥促癌作用,而TIPE1和TIPE2则主要发挥抑癌作用[12]。研究者发现TIPE1在肝细胞癌[13]、肺癌[14]、胃癌[15]、食管癌[16]、骨肉瘤[17]和乳腺癌[18-19]中可通过调控其不同下游信号通路,如核转录因子(NF)-κB、c-jun氨基末端激酶(JNK)、细胞外信号调节激酶(ERK)、Wnt/β-catenin,或调控免疫细胞如巨噬细胞促进癌细胞凋亡、抑制肿瘤细胞生长和转移。然而,在子宫颈癌中,TIPE1可作为癌基因,并通过与p53结合而促进癌细胞的发生发展[20]。在对帕金森病的研究中,研究者发现TIPE1可与结节性硬化复合物2(tuberous sclerosis complex-2,TSC2)竞争性结合F-Box和WD40蛋白5(FBXW5),通过抑制FBXW5依赖的TSC2降解提高其稳定性,降低哺乳动物雷帕霉素靶蛋白(mTOR)磷酸化,并最终增强自噬,成为治疗帕金森病多巴胺神经元自噬失调的潜在靶标[21]。Shao等[22]在最新的研究中发现,TIPE1可通过氧化应激反应诱导血管内皮细胞功能障碍,促进动脉粥样硬化的形成。上述研究结果表明TIPE1在肿瘤疾病中既有促癌作用又有抑癌作用,同时TIPE1也与凋亡、自噬、氧化应激相关。提示TIPE1在肿瘤和炎性反应疾病中可能通过不同机制发挥重要作用。
本研究中,我们检测了TIPE1在顺铂诱导的AKI小鼠和细胞模型中的表达,发现肾损伤后的肾组织和顺铂刺激下的HK-2细胞中TIPE1的mRNA和蛋白的表达明显增加。免疫组化结果显示,TIPE1在AKI小鼠近端肾小管上皮细胞上阳性表达较对照组显著升高。沉默HK-2细胞TIPE1基因后,顺铂刺激下HK-2细胞的LC3-II、Beclin1和NGAL表达较对照组显著增加。我们的实验结果提示,TIPE1在顺铂诱导的AKI小鼠和HK-2细胞中表达增高,TIPE1可能通过减轻肾小管上皮细胞过度自噬,进而减轻肾损伤,发挥对肾脏的保护作用。
自噬是所有真核生物中的一种高度保守的代谢途径,对于维持细胞稳态起着重要作用。在正常生理状态下,细胞维持较低水平的自噬有利于细胞形态、结构和功能的稳定。当细胞受到过度不良刺激(如毒物、炎性反应、高糖环境等)时,过度自噬则造成细胞器损伤,诱导细胞自噬性死亡。本研究结果提示,干扰TIPE1表达后,HK-2细胞LC3-Ⅱ和Beclin1表达较对照组明显增加,提示自噬过度增强。
目前TIPE1在顺铂诱导的AKI中的发病机制仍未完全明确。本课题小组正在进一步研究,期待TIPE1可成为AKI的一个新治疗靶点。

References

[1]
Bentley ML, Corwin HL, Dasta J. Drug-induced acute kidney injury in the critically ill adult: recognition and prevention strategies[J]. Crit Care Med, 2010, 38(< W>6 Suppl): S169-S174. DOI:10.1097/CCM.0b013e3181de0c60.
[2]
Perazella MA. Drug-induced acute kidney injury: diverse mechanisms of tubular injury[J]. Curr Opin Crit Care, 2019, 25(6): 550-557. DOI: 10.1097/mcc.0000000000000653.
Medications are a relatively common cause of acute kidney injury (AKI), especially in hospitalized patients who are exposed to numerous agents. Drug-related acute tubular/tubulointerstitial injury is the most common cause of AKI associated with these agents. Toxic effects of drugs and their renal handling often lead to various forms of AKI.The inherent nephrotoxicity of drugs and their transport and metabolism by the kidneys play an important role in the occurrence of acute tubular injury. Apical transport of the aminoglycosides by endocytosis and apical pinocytosis of filtered hydroxyethyl starch into cells lead to acute tubular dysfunction. Transport of tenofovir and cisplatin by organic anion and cation transporters in the basolateral surface of the proximal tubule, respectively, are associated with intracellular drug accumulation and injury. Intratubular deposition of drug crystals with associated AKI occurs with several drugs, in particular the anticancer agent methotrexate. A potentially new mechanism of drug-induced AKI was described with vancomycin - acute vancomycin-related cast nephropathy. Immune-mediated acute tubulointerstitial injury is another cause of drug-induced AKI, as seen with immune checkpoint inhibitors.Drugs lead to AKI through mechanisms that involve their inherent toxicity as well as their transport and handling by the kidneys.
[3]
Volarevic V, Djokovic B, Jankovic MG, et al. Molecular mechanisms of cisplatin-induced nephrotoxicity: a balance on the knife edge between renoprotection and tumor toxicity[J]. J Biomed Sci, 2019, 26(1): 25. DOI: 10.1186/s12929-019-0518-9.
Cisplatin (cis-diamminedichloroplatinum II, CDDP) is one of the most effective chemotherapeutic agents. However, its clinical use is limited due to the severe side effects, including nephrotoxicity and acute kidney injury (AKI) which develop due to renal accumulation and biotransformation of CDDP. The alleviation or prevention of CDDP-caused nephrotoxicity is currently accomplished by hydration, magnesium supplementation or mannitol-induced forced diuresis which is considered for high-dose CDDP-treated patients. However, mannitol treatment causes over-diuresis and consequent dehydration in CDDP-treated patients, indicating an urgent need for the clinical use of safe and efficacious renoprotective drug as an additive therapy for high dose CDDP-treated patients.In this review article we describe in detail signaling pathways involved in CDDP-induced apoptosis of renal tubular cells, oxidative stress and inflammatory response in injured kidneys in order to pave the way for the design of new therapeutic approaches that can minimize CDDP-induced nephrotoxicity. Most of these molecular pathways are, at the same time, crucially involved in cytotoxic activity of CDDP against tumor cells and potential alterations in their function might mitigate CDDP-induced anti-tumor effects.Despite the fact that many molecules were designated as potential therapeutic targets for renoprotection against CDDP, modulation of CDDP-induced nephrotoxicity still represents a balance on the knife edge between renoprotection and tumor toxicity.
[4]
Aydinoz S, Uzun G, Cermik H, et al. Effects of different doses of hyperbaric oxygen on cisplatin-induced nephrotoxicity[J]. Ren Fail, 2007, 29(3): 257-263. DOI: 10.1080/0886022060 1166487.
[5]
Hosohata K. Role of oxidative stress in drug-induced kidney injury[J]. Int J Mol Sci, 2016, 17(11): 1826. DOI: 10.3390/ijms17111826.
[6]
Ramesh G, Reeves WB. TNF-alpha mediates chemokine and cytokine expression and renal injury in cisplatin nephrotoxicity[J]. J Clin Invest, 2002, 110(6): 835-842. DOI: 10.1172/jci15606.
The purpose of these studies was to examine the role of cytokines in the pathogenesis of cisplatin nephrotoxicity. Injection of mice with cisplatin (20 mg/kg) led to severe renal failure. The expression of cytokines, chemokines, and ICAM-1 in kidney was measured by ribonuclease protection assays and RT-PCR. We found significant upregulation of TNF-alpha, TGF-beta, RANTES, MIP-2, MCP-1, TCA3, IL-1beta, and ICAM-1 in kidneys from cisplatin-treated animals. In addition, serum, kidney, and urine levels of TNF-alpha measured by ELISA were increased by cisplatin. Inhibitors of TNF-alpha production (GM6001, pentoxifylline) and TNF-alpha Ab's reduced serum and kidney TNF-alpha protein levels and also blunted the cisplatin-induced increases in TNF-alpha, TGF-beta, RANTES, MIP-2, MCP-1, and IL-1beta, but not ICAM-1, mRNA. In addition, the TNF-alpha inhibitors also ameliorated cisplatin-induced renal dysfunction and reduced cisplatin-induced structural damage. Likewise, TNF-alpha-deficient mice were resistant to cisplatin nephrotoxicity. These results indicate cisplatin nephrotoxicity is characterized by activation of proinflammatory cytokines and chemokines. TNF-alpha appears to play a central role in the activation of this cytokine response and also in the pathogenesis of cisplatin renal injury.
[7]
Faubel S, Lewis EC, Reznikov L, et al. Cisplatin-induced acute renal failure is associated with an increase in the cytokines interleukin (IL)-1beta, IL-18, IL-6, and neutrophil infiltration in the kidney[J]. J Pharmacol Exp Ther, 2007, 322(1): 8-15. DOI: 10.1124/jpet.107.119792.
We have demonstrated that caspase-1-deficient (caspase-1(-/-)) mice are functionally and histologically protected against cisplatin-induced acute renal failure (ARF). Caspase-1 exerts proinflammatory effects via the cytokines interleukin (IL)-1beta, IL-18, IL-6, and neutrophil recruitment. We sought to determine the role of the cytokines IL-1beta, IL-18, and IL-6 and neutrophil recruitment in cisplatin-induced ARF. We first examined IL-1beta; renal IL-1beta increased nearly 2-fold in cisplatin-induced ARF and was reduced in the caspase-1(-/-) mice. However, inhibition with IL-1 receptor antagonist (IL-1Ra) did not attenuate cisplatin-induced ARF. Renal IL-18 increased 2.5-fold; however, methods to inhibit IL-18 using IL-18 antiserum and transgenic mice that overproduce IL-18-binding protein (a natural inhibitor of IL-18) did not protect. Renal IL-6 increased 3-fold; however, IL-6-deficient (IL-6(-/-)) mice still developed cisplatin-induced ARF. We next examined neutrophils; blood neutrophils increased dramatically after cisplatin injection; however, prevention of peripheral neutrophilia and renal neutrophil infiltration with the neutrophil-depleting antibody RB6-8C5 did not protect against cisplatin-induced ARF. In summary, our data demonstrated that cisplatin-induced ARF is associated with increases in the cytokines IL-1beta, IL-18, and IL-6 and neutrophil infiltration in the kidney. However, inhibition of IL-1beta, IL-18, and IL-6 or neutrophil infiltration in the kidney is not sufficient to prevent cisplatin-induced ARF.
[8]
Niture S, Moore J, Kumar D. TNFAIP8: inflammation, immunity and human diseases[J]. J Cell Immunol, 2019, 1(2): 29-34.
Tumor necrosis factor (TNF)-alpha-induced protein 8 (TNFAIP8 /TIPE) family proteins are known to be involved in maintaining immune homeostasis. The TIPE family contains four members: tumor necrosis factor-α-induced protein 8 (TNFAIP8), TNFAIP8 like 1 (TIPE1), TNFAIP8 like 2 (TIPE2), and TNFAIP8 like 3 (TIPE3). Here we review the latest roles and associations of a founding member of TIPE family protein - TNFAIP8 in cellular function/signaling, inflammation, and immunity related human diseases.
[9]
Cui J, Zhang G, Hao C, et al. The expression of TIPE1 in murine tissues and human cell lines[J]. Mol Immunol, 2011, 48(12-13): 1548-1555. DOI: 10.1016/j.molimm.2011.04.023.
Members of the tumor necrosis factor-alpha-induced protein-8 (TNFAIP8 or TIPE) family play important roles in immune homeostasis and cancer. TIPE1 (TNFAIP8-like 1) is a new member of the TIPE family that may regulate cell death. However, due to the lack of a suitable antibody, the nature of cells and tissues that express TIPE1 protein has not been determined. In this study, we generated a highly specific antibody to TIPE1 and examined TIPE1 expression in various murine tissues and human cell lines by immunohistochemistry, reverse transcription real-time PCR, and Western blot. We found that TIPE1 protein was detected in a wide variety of tissues in C57BL/6 mice, such as neurons in brain, hepatocytes, germ cells of female and male reproductive organs, muscular tissues, and a variety of cells of the epithelial origin, particularly those with secretory functions. TIPE1 protein was not expressed in mature T or B lymphocytes, but detectable in human B lymphoblast cell line HMy2.CIR and murine T cell line EL4. Furthermore, high levels of TIPE1 mRNA were detected in most human carcinoma cell lines, especially in cells transformed with viral genomes. These results indicate that TIPE1 may perform functions in cell secretion and carcinogenesis, but not in immunity.Copyright © 2011 Elsevier Ltd. All rights reserved.
[10]
Ronco C, Bellomo R, Kellum JA. Acute kidney injury[J]. Lancet, 2019, 394(10212): 1949-1964. DOI: 10.1016/s0140-6736(19)32563-2.
Acute kidney injury (AKI) is defined by a rapid increase in serum creatinine, decrease in urine output, or both. AKI occurs in approximately 10-15% of patients admitted to hospital, while its incidence in intensive care has been reported in more than 50% of patients. Kidney dysfunction or damage can occur over a longer period or follow AKI in a continuum with acute and chronic kidney disease. Biomarkers of kidney injury or stress are new tools for risk assessment and could possibly guide therapy. AKI is not a single disease but rather a loose collection of syndromes as diverse as sepsis, cardiorenal syndrome, and urinary tract obstruction. The approach to a patient with AKI depends on the clinical context and can also vary by resource availability. Although the effectiveness of several widely applied treatments is still controversial, evidence for several interventions, especially when used together, has increased over the past decade.Copyright © 2019 Elsevier Ltd. All rights reserved.
[11]
Bordoloi D, Banik K, Shabnam B, et al. TIPE family of proteins and its implications in different chronic diseases[J]. Int J Mol Sci, 2018, 19(10): 2974. DOI: 10.3390/ijms19102974.
The tumor necrosis factor-α-induced protein 8-like (TIPE/TNFAIP8) family is a recently identified family of proteins that is strongly associated with the regulation of immunity and tumorigenesis. This family is comprised of four members, namely, tumor necrosis factor-α-induced protein 8 (TIPE/TNFAIP8), tumor necrosis factor-α-induced protein 8-like 1 (TIPE1/TNFAIP8L1), tumor necrosis factor-α-induced protein 8-like 2 (TIPE2/TNFAIP8L2), and tumor necrosis factor-α-induced protein 8-like 3 (TIPE3/TNFAIP8L3). Although the proteins of this family were initially described as regulators of tumorigenesis, inflammation, and cell death, they are also found to be involved in the regulation of autophagy and the transfer of lipid secondary messengers, besides contributing to immune function and homeostasis. Interestingly, despite the existence of a significant sequence homology among the four members of this family, they are involved in different biological activities and also exhibit remarkable variability of expression. Furthermore, this family of proteins is highly deregulated in different human cancers and various chronic diseases. This review summarizes the vivid role of the TIPE family of proteins and its association with various signaling cascades in diverse chronic diseases.
[12]
Lou Y, Liu S. The TIPE (TNFAIP8) family in inflammation, immunity, and cancer[J]. Mol Immunol, 2011, 49(1-2): 4-7. DOI: 10.1016/j.molimm.2011.08.006.
[13]
Zhang Z, Liang X, Gao L, et al. TIPE1 induces apoptosis by negatively regulating Rac1 activation in hepatocellular carcinoma cells[J]. Oncogene, 2015, 34(20): 2566-2574. DOI:10.1038/onc.2014.208.
TIPE1 (tumor necrosis factor-α-induced protein 8-like 1 or TNFAIP8L1) is a newly identified member of the TIPE (TNFAIP8) family, which play roles in regulating cell death. However, the biologic functions of TIPE1 in physiologic and pathologic conditions are largely unknown. Here, we report the roles of TIPE1 in hepatocellular carcinoma (HCC). Evaluated by immunohistochemical staining, HCC tissues showed significantly downregulated TIPE1 expression compared with adjacent non-tumor tissues, which positively correlated with tumor pathologic grades and patient survival. Using a homograft tumor model in Balb/c mice, we discovered that TIPE1 significantly diminished the growth and tumor weight of murine liver cancer homografts. Consistently, TIPE1 inhibited both cell growth and colony formation ability of cultured HCC cell lines, which was further identified to be due to TIPE1-inducing apoptosis in a caspase-independent, necrostatin-1 (Nec-1)-insensitive manner. Furthermore, mechanistic investigations revealed that TIPE1 interacted with Rac1, and inhibited the activation of Rac1 and its downstream p65 and c-Jun N-terminal kinase pathway. Moreover, overexpression of constitutively active Rac1 partially rescued the apoptosis induced by TIPE1, and Rac1 knockdown significantly restored the deregulated cell growth induced by TIPE1 small interfering RNA. Our findings revealed that TIPE1 induced apoptosis in HCC cells by negatively regulating Rac1 pathway, and loss of TIPE1 might be a new prognostic indicator for HCC patients.
[14]
Wu X, Ma Y, Cheng J, et al. TIPE1 function as a prognosis predictor and negative regulator of lung cancer[J]. Oncotarget, 2017, 8(45): 78496-78506. DOI: 10.18632/oncotarget.19655.
TIPE1 (tumor necrosis factor-α-induced protein 8-like 1 or TNFAIP8L1) belongs to the TIPE (TNFAIP8) family, which act as a regulator of cell death. However, the expression and biologic functions of TIPE1 in lung cancer are largely unknown. Here, we investigated the roles of TIPE1 in lung cancer. Evaluated by qRT-PCR and immunohistochemical staining, lower TIPE1 mRNA and protein expression was found in the lung tumor tissue, compared with adjacent non-tumor tissues, which positively correlated with tumor patient survival. Overexpression of TIPE1 by lentivirus system in TIPE1-downregulated lung cancer cells significantly diminished cell growth and colony formation, companied with proliferation inhibition, apoptosis induction and invasion inhibition. It was identified to be due to TIPE1-regulated Cyclin D1, Cyclin B1, caspase 8, Caspase3, MM2 and MMP9 expression. Consistently, using a homograft tumor model in Balb/c mice, we discovered that TIPE1 prevented the growth and tumor weight of murine lung cancer homografts. Our findings revealed the anti-tumor role of TIPE1 in lung cancer cells and TIPE1 might be a novel prognostic indicator for lung cancer patients.
[15]
Liu W, Chen Y, Xie H, et al. TIPE1 suppresses invasion and migration through down-regulating Wnt/beta-catenin pathway in gastric cancer[J]. J Cell Mol Med, 2018, 22(2): 1103-1117. DOI:10.1111/jcmm.13362.
Epithelial–mesenchymal transition (EMT) plays an important role in the invasiveness and metastasis of gastric cancer. Therefore, identifying key molecules involved in EMT will provide new therapeutic strategy for treating patients with gastric cancer. TIPE1 is a newly identified member of the TIPE (TNFAIP8) family, and its contributions to progression and metastasis have not been evaluated. In this study, we found that the levels of TIPE1 were significantly reduced and inversely correlated with differentiation status and distant metastasis in primary gastric cancer tissues. We further observed overexpression of TIPE1 in aggressive gastric cancer cell lines decreased their metastatic properties both in vitro and in vivo as demonstrated by markedly inhibiting EMT and metastasis of gastric cancer cells in nude mice. Consistently, gene silencing of TIPE1 in well‐differentiated gastric cancer cell line (AGS) inhibited these processes. Mechanistically, we found that TIPE1‐medicated Wnt/β‐catenin signalling was one of the critical signal transduction pathways that link TIPE1 to EMT inhibition. Importantly, TIPE1 dramatically restrained the expression and activities of MMP2 and MMP9 which are demonstrated to promote tumour progression and are implicated in EMT. Collectively, these findings provide new evidence for a better understanding of the biological activities of TIPE1 in progression and metastasis of gastric cancer and suggest that TIPE1 may be an innovative diagnostic and therapeutic target of gastric cancer.
[16]
王道军, 赵山虎, 夏平, 等. 肿瘤坏死因子α诱导蛋白8样分子1在食管癌中的表达及其作用机制研究[J]. 疑难病杂志, 2019, 18(12): 1258-1262. DOI: 10.3969/j.issn.1671-6450.2019.12.016.
[17]
Chen P, Zhou J, Li J, et al. TIPE1 suppresses osteosarcoma tumor growth by regulating macrophage infiltration[J]. Clin Transl Oncol, 2019, 21(3): 334-341. DOI: 10.1007/s12094-018-1927-z.
Osteosarcoma is the most common primary malignancy of the bone, and macrophages play a promotional role during osteosarcoma development and progression. TIPE1 is known to function as a tumor suppressor in diverse cancers by inducing cell arrest and apoptosis. However, the biological function of TIPE1 in osteosarcoma is still unclear.The purpose of this study was to investigate the expression and function of TIPE1 in osteosarcoma.In the present study, TIPE1 expression in osteosarcoma cancer cells was determined by qPCR and western blotting. A subcutaneous tumor model was established to investigate the potential anti-tumor activity of TIPE1 in osteosarcoma. Further, flow cytometry, western blotting, immunofluorescence staining, and ELISA were performed to clarify the underlying mechanism by which TIPE1 regulates growth of osteosarcoma.Our results suggest that TIPE1 is downregulated in osteosarcoma cancer cells, and ectopic expression TIPE1 significantly inhibited osteosarcoma tumor growth in vivo. Furthermore, TIPE1 inhibits the infiltration of macrophages in osteosarcoma tumor by suppressing MCP-1 expression in osteosarcoma cells. Further in vivo study revealed that inhibition of MCP-1/CCR2 axis by Bindarit blocked the inhibitory effect of TIPE1 on osteosarcoma growth.Collectively, our results demonstrate the anti-tumor role of TIPE1 in osteosarcoma and reveal a novel therapy target for osteosarcoma.
[18]
Hu W, Feng CM, Liu LY, et al. TIPE1 inhibits breast cancer proliferation by downregulating ERK phosphorylation and predicts a favorable prognosis[J]. Front Oncol, 2019, 9: 400. DOI: 10.3389/fonc.2019.00400.
TIPE1, which acts as a cell death regulator, has emerged as a tumor suppressor in the process of carcinogenesis. However, our recent research demonstrated that it serves as an oncogene in the pathogenesis of cervical cancer, indicating that the role of TIPE1 in carcinogenesis needs to be further evaluated. In this study, we show that TIPE1 is able to inhibit breast cancer cell growth both and. Functionally, TIPE1 inhibits cancer cell proliferation preferentially by downregulating ERK phosphorylation. Furthermore, the expression of TIPE1 is decreased in breast cancer tissues compared to matched adjacent tissues, and its expression is positively correlated with patients' lifespan. These data indicate that TIPE1 suppresses breast cancer proliferation by inhibiting the ERK signaling pathway. This study also suggests that TIPE1 could serve as a potential therapeutic target and a diagnostic biomarker for breast cancer.
[19]
Qiu S, Hu W, Ma Q, et al. TIPE1 suppresses the invasion and migration of breast cancer cells and inhibits epithelial-to-mesenchymal transition primarily via the ERK signaling pathway[J]. Acta Biochim Biophys Sin (Shanghai), 2019, 51(10): 1008-1015. DOI: 10.1093/abbs/gmz099.
[20]
Zhao P, Pang X, Jiang J, et al. TIPE1 promotes cervical cancer progression by repression of p53 acetylation and is associated with poor cervical cancer outcome[J]. Carcinogenesis, 2019, 40(4): 592-599. DOI: 10.1093/carcin/bgy163.
Previous studies have shown that TIPE1 inhibits tumor proliferation and metastasis in certain cancers; however, increased expression of TIPE1 is observed in cervical cancer cell lines and tissues, indicating it might exert a distinctive role in cervical cancer. Cell and xenograft tumorigenicity assays showed that TIPE1 facilitates cervical cancer progression in this study. Further investigation demonstrated that TIPE1 binds to p53 and impairs its activity via inhibition of its acetylation. In addition, TIPE1 promoted cell proliferation and suppressed cisplatin susceptibility in a p53-dependent manner, indicating that TIPE1 facilitates cervical cancer progression primarily through the p53 pathway. TIPE1 expression in clinical samples also demonstrated that its upregulation predicts poor prognosis in patients with cervical cancer. Taken together, the results of this study showed that TIPE1 serves as an oncogene by restricting p53 activity in the development of cervical cancer, suggesting that TIPE1 will provide a new potential target for cervical cancer therapy and can be used as a biomarker to predict patient prognosis.© The Author(s) 2018. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.
[21]
Ha JY, Kim JS, Kang YH, et al. Tnfaip8 l1/Oxi-beta binds to FBXW5, increasing autophagy through activation of TSC2 in a Parkinson's disease model[J]. J Neurochem, 2014, 129(3): 527-538. DOI: 10.1111/jnc.12643.
Abnormal autophagy may contribute to neurodegeneration in Parkinson's disease (PD). However, it is largely unknown how autophagy is dysregulated by oxidative stress (OS), one of major pathogenic causes of PD. We recently discovered the potential autophagy regulator gene family including Tnfaip8/Oxi‐α, which is a mammalian target of rapamycin (mTOR) activator down‐regulated by OS in dopaminergic neurons (J. Neurochem., 112, 2010, 366). Here, we demonstrate that the OS‐induced Tnfaip8 l1/Oxi‐β could increase autophagy by a unique mechanism that increases the stability of tuberous sclerosis complex 2 (TSC2), a critical negative regulator of mTOR. Tnfaip8 l1/Oxi‐β and Tnfaip8/Oxi‐α are the novel regulators of mTOR acting in opposition in dopaminergic (DA) neurons. Specifically, 6‐hydroxydopamine (6‐OHDA) treatment up‐regulated Tnfaip8 l1/Oxi‐β in DA neurons, thus inducing autophagy, while knockdown of Tnfaip8 l1/Oxi‐β prevented significantly activation of autophagic markers by 6‐OHDA. FBXW5 was identified as a novel binding protein for Tnfaip8 l1/Oxi‐β. FBXW5 is a TSC2 binding receptor within CUL4 E3 ligase complex, and it promotes proteasomal degradation of TSC2. Thus, Tnfaip8 l1/Oxi‐β competes with TSC2 to bind FBXW5, increasing TSC2 stability by preventing its ubiquitination. Our data show that the OS‐induced Tnfaip8 l1/Oxi‐β stabilizes TSC2 protein, decreases mTOR phosphorylation, and enhances autophagy. Therefore, altered regulation of Tnfaip8 l1/Oxi‐β may contribute significantly to dysregulated autophagy observed in dopaminergic neurons under pathogenic OS condition.\nimage\n
[22]
Shao J, Li Y, Zhou C, et al. TIPE1 accelerates atherogenesis by inducing endothelial dysfunction in response to oxidative stress[J]. Biochim Biophys Acta Mol Basis Dis, 2020, 1866(1): 165578. DOI: 10.1016/j.bbadis.2019.165578.

Funding

National Natural Science Foundation of China(81670660)
Key R&D Project of Shandong Province(GG201809250293)
PDF(3005 KB)

963

Accesses

0

Citation

Detail

Sections
Recommended

/