中国科技论文统计源期刊 中文核心期刊  
美国《化学文摘》《国际药学文摘》
《乌利希期刊指南》
WHO《西太平洋地区医学索引》来源期刊  
日本科学技术振兴机构数据库(JST)
第七届湖北十大名刊提名奖  
医药导报, 2019, 38(1): 14-17
doi: 10.3870/j.issn.1004-0781.2019.01.003
Jab1在乳腺癌细胞中对曲妥珠单抗敏感性的影响
Effect of Jab1 on Trastuzumab Treatment Sensitivity in Breast Cancer Cell
沈倩1,, 刘勇2,, 于世英1,

摘要:

目的 探讨 Jab1在HER2过表达乳腺癌中对曲妥珠单抗治疗敏感性的影响。方法 收集使用曲妥珠单抗治疗的26例HER2过表达乳腺癌患者的标本,其中曲妥珠单药敏感组14例,耐药组12例,应用实时荧光定量聚合酶链反应(q-PCR)检测乳腺癌组织标本中Jab1的表达。运用Western blotting方法检测人乳腺癌细胞系BT474及曲妥珠单抗耐药乳腺癌细胞系C5和C6中Jab1的表达; 通过siRNA干扰技术敲除Jab1后,检测C5及C6细胞对曲妥珠单抗治疗敏感性的变化,以及对曲妥珠单抗诱导的凋亡作用的影响。结果 与曲妥珠单抗敏感的乳腺癌患者比较,Jab1在曲妥珠单抗耐药的乳腺癌患者中表达水平显著增高(P<0.05);在曲妥珠单抗耐药细胞株中C5和C6中,Jab1的蛋白表达水平显著高于药物敏感株;敲除Jab1后,耐药株可重获对曲妥珠单抗的敏感性,由曲妥珠单抗诱导的凋亡作用显著增强,在C5细胞系中,5 μg·mL-1曲妥珠单抗处理24 h后,Jab1敲低组与对照组凋亡率分别为(12.8±2.9)% 和(2.7±0.5)%(P<0.05);在C6细胞系中,Jab1敲低组与对照组的凋亡率分别为(18.8±3.6)% 和(4.1±1.1)%(P<0.05)。结论 Jab1表达上调导致HER2过表达乳腺癌细胞对曲妥珠单抗治疗敏感性下降,敲除Jab1后有望逆转耐药。

关键词: Jab1 ; 曲妥珠单抗 ; 耐药性 ; 乳腺癌 ; 人类表皮因子受体2

Abstract:

Objective To investigate the effect of Jab1 on trastuzumab (Ttzm) treatment sensitivity in HER2 positive breast cancer. Methods q-PCR was performed to detect the expression of Jab1 in 26 HER2 positive breast cancer patient with Ttzm treatment 14 patients were sensitive to Ttzm, while 12 patients were were Ttzm-resistant. The expression of Jab1 was examined by Western blotting in Ttzm-sensitive and resistant breast cancer cell lines. With the knockdown of Jab1 by siRNA, the sensitivity to the drug and the Ttzm-induced apoptosis were measured in related cell lines. Results The expression of Jab1 in Ttzm resistant breast cancer patient was significantly higher than these Ttzm sensitive patient(P<0.05). In Ttzm-sensitive breast cancer cell line C5 and C6, the expression of Jab1 was significantly lower than that in the Ttzm-resistant cells. After konckdown of Jab1 in Ttzm-resistant cells, C5 and C6 cells were re-sensitive to the Ttzm treatment, and the Ttzm-induced apoptosis was observed to be more obvious. In C5 cell line, after 24 hours of Ttzm treatment, the apoptotic rates in Jab1 knockdown group and the control group were (12.8±2.9)% and (2.7±0.5)%(P<0.05), while in the C6 cell line, the apoptosic rates were (18.8±3.6)% and (4.1±1.1)%(P<0.05), respectively. Conclusion Up-regulated expression of Jab1 is associated with the reduction of Ttzm treatment sensitivity in breast cancer cells. Knockdown of Jab1 might be a potential mechanism to antagonize the Ttzm-resistance.

Key words: Jab1 ; Trastuzumab ; Drug resistance ; Breast cancer ; Human epidermal growth factor receptor-2

乳腺癌是危害女性生命的最常见肿瘤,近年来发病率不断上升。 人类表皮因子受体2(human epidermal growth factor receptor-2,HER2)是一个重要的原癌基因,编码具有酪氨酸激酶活性的跨膜受体样蛋白[1]。HER2基因扩增导致肿瘤细胞表面HER2蛋白表达增加,从而使HER2受体活化[2]。在原发性乳腺癌患者中有25%~30%患者HER2过度表达[3]。目前研究表明,HER2是重要的乳腺癌预后判断因子,HER2过表达与乳腺癌侵袭转移密切相关,而且往往提示乳腺癌患者预后不佳[4]。曲妥珠单抗(trastuzumab)是一种人源化单克隆抗体,靶向作用于HER2受体,致使肿瘤细胞在G1阶段的生长终止,可以有效地抑制HER2过表达肿瘤细胞的增殖,是HER2过表达乳腺癌的最有效靶向治疗方式[5]。 尽管如此,对曲妥珠单抗的原发或继发耐药现象仍是临床上乳腺癌治疗的一大难题[6,7,8]。 目前研究显示, Jab1/CSN5(Jun activating binding protein) 作为COP9信号体的第5个亚单位,在乳腺癌、卵巢癌、肝癌、头颈部肿瘤等实体肿瘤中发挥重要作用。笔者检测正常组织和乳腺癌组织中Jab1表达水平,分析其与临床病理特征的相关性,利用小干扰RNA(siRNA)技术敲除乳腺癌细胞中Jab1,观察Jab1对于导致HER2过表达乳腺癌对曲妥珠单抗治疗敏感性的影响。

1 材料与方法
1.1 材料

人正常乳腺组织和乳腺癌组织标本来源于华中科技大学同济医学院附属同济医院病理科。人乳腺癌细胞(BT474、C5、C6)由华中科技大学同济医学院附属同济医院肿瘤中心实验室保存。达尔伯克改良伊格尔培养基(DMEM/G)及RPMI 1640培养基购自Hyclone ;胎牛血清、胰蛋白酶购自Gibco公司,曲妥珠单抗(Ttzm,赫赛汀,Roche公司),RIPA裂解液、LipofectamineTM3000脂质体(Invitmgen公司),抗体Jab1及二抗(Santa Cruz公司);Jab1 siRNA由Ambion公司合成。

1.2 细胞培养和Jab1 siRNA转染

BT474细胞培养于含有10%胎牛血清的DMEM/G培养基;人曲妥珠单抗耐药乳腺癌细胞系C5、C6培养于含有10%胎牛血清及2 μg·mL-1曲妥珠单抗的DMEM/G培养基常规培养。根据LipofectamineTM 2000说明书,将BT474和SKBR3以2×105·mL-1密度接种6孔板,每个细胞系分为两组:①转染空白siRNA的对照组;②转染Jab1 siRNA的实验组,每组分别使用100 pmol siRNA转染细胞6 h后,换新鲜培养基培养48 h,收集细胞进行相应实验。

1.3 3-(4,5-二甲基吡啶-2-基)-5-(3-羧基甲氨基苯基)-2-(4-磺苯基-2H-四唑)(MTS)实验检测曲妥珠单抗在乳腺癌细胞系中对细胞增殖的影响

MTS实验分为两部分,第一部分验证乳腺癌细胞系BT474及曲妥珠耐药株C5和C6的药物敏感性;第二部分检测Jab1敲低后,C5和C6耐药株对曲妥珠单抗敏感性的影响,每个细胞系分为两组:siNC空白组及Jab1 siRNA组,依据“1.2”项中实验步骤转染24 h后,用不同梯度浓度的曲妥珠单抗处理细胞。 两部分实验的药物处理时间均为72 h,药物梯度为12.5,25,50,100,200 μg·mL-1药物处理完毕后,用MTS法检测每孔中的细胞活性,计算并制作曲妥珠单抗的生长抑制曲线,并计算半数抑制浓度(IC50)。

1.4 Annexin V/PI 双染法检测细胞凋亡

取对数生长期细胞,消化计数后,将耐药株C5和C6细胞以细胞密度为2.5×105·mL-1接种于直径6 cm的细胞培养皿中,每个细胞系的实验分组如下:①空载体siRNA+二甲亚砜(DMSO)组;②空载体siRNA+曲妥珠单抗组;③Jab1 siRNA+DMSO组;④Jab1 siRNA+曲妥珠单抗组。第2天细胞贴壁后,依照“1.2”项所述,行细胞转染,24 h后,依分组加入5 μg·mL-1曲妥珠单抗或对应体积的DMSO,处理24 h后,收集每组中含有上清液里细胞的全部细胞,1000 r·min-1离心3 min,吸弃上清液,加入磷酸盐缓冲液(PBS)重悬漂洗细胞,再次以1000 r·min-1离心3 min,吸弃上清液PBS,加入凋亡抗体反应缓冲液100 μL,然后加入Annexin V 5 μL,于室温下避光反应5~10 min,于上机前加入碘化丙啶(propidium iodide,PI)400 μL,用流式细胞仪检测每组中的凋亡细胞比例,其中Annexin V阳性/PI阴性的细胞为早期凋亡细胞,Annexin V阳性/PI阳性的细胞则为晚期凋亡细胞,统计这两部分细胞在整体细胞中的比例作为该组的凋亡率。

1.5 定量聚合酶链反应(q-PCR)实验检测乳腺癌组织标本中Jab1表达

按流程提取RNA,并逆转成cDNA,在PCR仪上完成q-PCR,实验条件:变性温度为95 ℃,退火温度为55 ℃,Jab1引物序列如下:F:5'-GCAATCGGGTGGTATCATAGC-3';R:5'-TGCGGATA-TTGTTCTTGTTGGA-3',完成PCR取得每个标本CT值后,取曲妥珠单抗敏感中1例乳腺癌组织当做对照组,其Jab1表达记为1,依照公式:其他组织表达量倍数=2-△Ct ,其中△Ct =其他组织CT值-对照组CT值,计算完毕后,使用Graphpad Prism 5.0制图,并计算两组间Jab1表达量的差异。

1.6 Western blotting 检测Jab1水平

RIPA裂解液提取蛋白,BCA法测定蛋白浓度。每个样本取30 μg蛋白进行十二烷基硫酸钠-聚丙烯酰胺凝胶电泳(SDS-PAGE)电泳,转印至聚偏氟乙烯(PVDF)膜,用5%脱脂乳封闭1 h,4 ℃一抗孵育过夜(Jab1 1:1000、β-actin 1:1000)。磷酸盐吐温缓冲液(PBST)洗涤3×10 min,室温二抗孵育1 h(1:3000)。PBST洗涤3×10 min,增强型化学发光试剂(ECL)法显色后显影成像,以β-actin作为内参。

1.7 统计学方法

计量资料用均数±标准差( x ¯ ±s)表示,所有实验均重复3次,组间差异通过student-t检验统计两组间差异,以 P<0.05为差异有统计学意义。

2 结果
2.1 Jab1在曲妥珠单抗耐药及敏感乳腺癌组织中的表达

26例使用曲妥珠单抗治疗的乳腺癌患者组织q-PCR结果显示,在曲妥珠单抗耐药的乳腺癌组织中Jab1的表达水平显著高于敏感的乳腺癌组织(图1),耐药组中Jab1表达的平均值为敏感组的2.1倍(t=3.564,P<0.01)。

图1 Jab1在乳腺癌患者中表达情况

Fig.1 Jab1 expression in breast cancer patients

2.2 Jab1在乳腺癌细胞株中的表达

Western blotting检测结果显示Jab1在曲妥珠单抗耐药株乳腺癌细胞C5、C6中的表达水平显著高于曲妥珠单抗敏感乳腺癌细胞系BT474(图2),灰度值分析显示,C5和C6细胞系中Jab1在蛋白水平的表达分别是BT474细胞的18.3和15.1倍。

图2 Jab1在乳腺癌细胞系中的表达

Fig.2 Jab1 expression in breast cancer cell lines

2.3 Jab1敲除对曲妥珠单抗耐药性的影响

Western blotting检测结果显示,Jab1 siRNA 能够显著降低C5及C6细胞系中Jab1的表达(图3),灰度值分析显示C5细胞系中,Jab1 siRNA 组中Jab1的表达为对照组的23.5%,而C6细胞系中,Jab1 siRNA组为对照组的45.6%。MTS结果显示,Jab1敲除后对曲妥珠单抗耐药的乳腺癌细胞株系C5 和C6重新对曲妥珠单抗敏感(P<0.05)(图4)。

图3 敲除后Jab1在C5和C6中的表达

Fig.3 Jab1 expression in C5 cells and C6 cells after Jab1 knockdown

图4 Jab1敲除后对C5和C6细胞存活率的影响

Fig.4 Viability of C5 cells and C6 cells after Jab1 knockdown

2.4 Jab1敲除对曲妥珠单抗治疗诱导的凋亡的影响

Annexin V/PI 染色结果显示,在Jab1敲除后的细胞系C5和C6,曲妥珠单抗导致的凋亡作用显著增强,在C5细胞系中,100 μg·mL-1曲妥珠单抗处理24 h后,Jab1敲除组与对照组凋亡率为(12.8±2.9)% 和(2.7±0.5)%(P<0.05)(图5)。而在C6细胞系中,Jab1敲除组与对照组的凋亡率分别为(18.8±3.6)% 和(4.1±1.1)%(P<0.05)(图6)。

图5 Jab1敲除后对C5细胞凋亡的影响

Fig.5 Apoptosis rate of C5 cells after Jab1 knockdown

图6 Jab1敲除后对C6细胞凋亡的影响

Fig.6 Apoptosis rate of C6 cells after Jab1 knockdown

3 讨论

HER2是乳腺癌重要的预后判断因子,目前以HER2为靶点药物曲妥珠单抗在大部分HER2阳性的乳腺癌患者中取得了良好的疗效,改善了预后,但是在治疗过程中大部分患者发生了曲妥珠单抗耐药的情况,这也是目前乳腺癌治疗研究中的热点与难点。在乳腺癌中,HER2的过表达能够激活一些重要的信号通路,如MAPK及PI3K/AKT信号通路等,这些重要信号通路的激活能够促进肿瘤的增殖及侵袭。基于此现象,通过抑制这些重要通路来逆转曲妥珠单抗耐药取得了进展,动物实验及前期临床试验表明,以这些信号通路为靶点的药物联合曲妥珠单抗在HER2乳腺癌治疗中取得了一定疗效,但对HER2过表达乳腺癌曲妥珠单抗耐药的进一步研究仍然是必要的。Jab1/CSN5是COP9信号体的第5个亚单位,在许多恶性肿瘤组织中过表达的现象,包括乳腺癌、胃癌、结直肠癌、胶质瘤等,实验表明,Jab1对这些肿瘤的增殖及侵袭性有显著影响,因此Jab1在很多肿瘤中认为是一个潜在的治疗靶点,此外,HER2能够在转录水平激活Jab1从而在诸多肿瘤发生发展中发挥重要作用[9]。在乳腺中,HER2是重要的治疗靶点,HER2和Jab1的联系使得笔者做了大胆的假设,并研究Jab1在曲妥珠单抗耐药中是否发挥一定的作用。本研究结果显示,Jab1在曲妥珠单抗耐药乳腺癌患者组织中表达显著高于曲妥珠单抗敏感乳腺癌患者,同样的,在曲妥珠单抗耐药的乳腺癌细胞株C5和C6中,Jab1蛋白表达水平显著高于敏感组细胞株。这提示Jab1可能作为预测HER2过表达乳腺癌对于曲妥珠单抗治疗敏感性的有效预测指标。进一步利用RNA干扰技术敲除耐药细胞株中Jab1的表达,可见细胞株IC50明显下降。凋亡实验显示,Jab1敲除后增强了曲妥珠单抗诱导的凋亡作用。本研究结果提示,在乳腺癌中,Jab1过表达可能诱导其发生对曲妥珠单抗治疗耐药,虽然这一现象的具体机制需要进一步的研究,但是本研究对临床上逆转乳腺癌治疗中的曲妥珠单药耐药提供了新的思路。

The authors have declared that no competing interests exist.

参考文献

[1] DI COSIMO S,BASELGA J.Management of breast cancer with target agents:importance of heterogeneity[J].Nat Rev Clin Oncol,2010,7(3):139-147.
Breast cancer is a heterogeneous disease with different molecular drivers regulating its growth, survival and response to therapy. Breast cancer is divided in three major subtypes based on the pattern of expression of hormone receptors and HER2: luminal tumors (or HR positive), HER2 amplified tumors, and the remaining subtypes being collectively referred to as triple-negative breast cancer. While tumors within these subtypes have similar gene-expression patterns, clinical outcomes, and response to therapy, this division is far from perfect and subgroups within these groups are beginning to be identified. In terms of therapy, an increasingly rational drug development effort has resulted in agents against new molecular targets that are active against only those tumors with the targeted molecular alteration or phenotype. These agents include receptor and non-receptor tyrosine kinase inhibitors (HER1, HER2, HER3, insulin-like growth factor receptor, c-met, fibroblast growth factor receptor and HSP 90 inhibitors), intracellular signaling pathways (PI3K, AKT, mTOR), angiogenesis inhibitors and agents that interfere with DNA repair (PARP inhibitors). Thus, the overall management of breast cancer will increasingly require an understanding of breast cancer heterogeneicity, the biological nature of any given tumor as well the existence of increased personalized treatment options.
DOI:10.1038/nrclinonc.2009.234      PMID:20125090      URL    
[本文引用:1]
[2] YARDEN Y,PINES G.The ERBB network:at last,cancer therapy meets systems biology[J].Nat Rev Caner,2012,12(4):553-563.
Although it is broadly agreed that the improved treatment of patients with will depend on a deeper molecular understanding of the underlying , only a few examples are already available. This Timeline article focuses on the (also known as HER) network of receptor tyrosine kinases (RTKs), which exemplifies how a constant dialogue between basic research and medical oncology can translate into both a sustained pipeline of novel drugs and ways to overcome acquired treatment resistance in patients. We track the key early discoveries that linked this RTK family to , the course of pioneering clinical research and their merger into a systems-biology framework that is likely to inspire further generations of effective therapeutic strategies.
DOI:10.1038/nrc3309      PMID:22785351      URL    
[本文引用:1]
[3] LI C I,URIBE D J,DALING J R.Clinical characteristics of different histologic types of breast cancer[J].Br J Cancer,2005,93(8):1046-1052.
Breast cancer is a heterogeneous disease, though little is known about some of its rarer forms, including certain histologic types. Using Surveillance, Epidemiology, and End Results Program data on 13565157 invasive breast cancer cases diagnosed from 1992 to 2001, relationships between nine histologic types of breast cancer and various tumour characteristics were assessed. Among women aged 50–89 years at diagnosis, lobular and ductal/lobular carcinoma cases were more likely to be diagnosed with stage III/IV, 825.065cm, and node-positive tumours compared to ductal carcinoma cases. Mucinous, comedo, tubular, and medullary carcinomas were less likely to present at an advanced stage. Lobular, ductal/lobular, mucinous, tubular, and papillary carcinomas were less likely, and comedo, medullary, and inflammatory carcinomas were more likely to be oestrogen receptor (ER) negative/progesterone receptor (PR) negative and high grade (notably, 68.2% of medullary carcinomas were ER61/PR61 vs 19.3% of ductal carcinomas). In general, similar differences were observed among women diagnosed at age 30–49 years. Inflammatory carcinomas are associated with more aggressive tumour phenotypes, and mucinous, tubular, and papillary tumours are associated with less aggressive phenotypes. The histologic types of breast cancer studied here differ greatly in their clinical presentations, and the differences in their hormone receptor profiles and grades point to their likely different aetiologies.
DOI:10.1038/sj.bjc.6602787      PMID:2361680      URL    
[本文引用:1]
[4] SLAMON D J,LEYL B,JONES S,et al.Use of chemothera-py plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2[J].N Engl J Med,2001,344(11):783-792.
DOI:10.1056/NEJM200103153441101      URL    
[本文引用:1]
[5] YEON C H,PEGRAM M D.Anti-erbB-2 antibody trastuzu-mab in the treatment of HER2-amplified breast cancer[J].Inv New Drugs,2005,23(3):391-409.
Human epidermal growth factor receptor-2 (HER2/erbB-2) is a member of a family of four transmembrane receptor tyrosine kinases that regulate cell growth, survival and differentiation via multiple signal transduction pathways. Amplification of the HER2 gene occurs in 20 25% of human breast cancers. This amplification event is an independent adverse prognostic factor as well as a predictive factor for increased response to doxorubicin-based combination chemotherapy, response to trastuzumab and decreased response to hormonal therapy. Methods for detecting protein overexpression or gene amplification in clinical tumor specimens include immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) techniques, with the latter considered by some to be more accurate. Trastuzumab (Herceptin) is a recombinant humanized monoclonal antibody which targets an epitope in the extracellular domain of the HER2 protein. Preclinical models demonstrated that this antibody has significant anti-tumor activity as a single agent and has synergy with certain chemotherapeutic drugs. Phase II and III clinical trials performed in women with metastatic breast cancer that overexpress HER2 have shown that trastuzumab has clinical activity when used as first-, second- or third-line monotherapy, and improves survival when used as first-line therapy in combination with chemotherapy. Newer combinations with numerous chemotherapeutic drugs have also shown significant clinical activity in phase II studies. In all of these trials, trastuzumab was generally well-tolerated, but cardiac toxicity (particularly when the antibody was combined with anthracyclines) was an unexpected adverse effect. Although trastuzumab is currently usually administered on a weekly intravenous schedule, evidence suggests that a triple dose of the drug given once every three weeks has a pharmacokinetic profile expected to be equally efficacious. Neither the optimal schedule nor the optimal duration of trastuzumab therapy has yet been clearly defined in controlled clinical trials. Current clinical investigations of trastuzumab include its use in both the adjuvant and neoadjuvant settings as well as in combination with other chemotherapy drugs or new biologic targeted agents.
DOI:10.1007/s10637-005-2899-8      PMID:16133791      URL    
[本文引用:1]
[6] ARTEAGA C L,SLIWKOWSKI M X,OSBORNE C K,et al.Treatment of HER2-positive breast cancer:current status and future perspectives[J].Nat Rev Clin Oncol,2012,9(1):16-32.
The advent of HER2-directed therapies has significantly improved the outlook for patients with HER2-positive early stage breast cancer. However, a significant proportion of these patients still relapse and die of breast cancer. Trials to define, refine and optimize the use of the two approved HER2-targeted agents (trastuzumab and lapatinib) in patients with HER2-positive early stage breast cancer are ongoing. In addition, promising new approaches are being developed including monoclonal antibodies and small-molecule tyrosine kinase inhibitors targeting HER2 or other HER family members, antibodies linked to cytotoxic moieties or modified to improve their immunological function, immunostimulatory peptides, and targeting the PI3K and IGF-1R pathways. Improved understanding of the HER2 signaling pathway, its relationship with other signaling pathways and mechanisms of resistance has also led to the development of rational combination therapies and to a greater insight into treatment response in patients with HER2-positive breast cancer. Based on promising results with new agents in HER2-positive advanced-stage disease, a series of large trials in the adjuvant and neoadjuvant settings are planned or ongoing. This Review focuses on current treatment for patients with HER2-positive breast cancer and aims to update practicing clinicians on likely future developments in the treatment for this disease according to ongoing clinical trials and translational research.
DOI:10.1038/nrclinonc.2011.177      PMID:22124364      URL    
[本文引用:1]
[7] THUY V U,SLIWKOWSKI M X,CLARET F X.Personali-zed drug combinations to overcome trastuzumab resistance in HER2-positive breast cancer[J].Biochim Biophysica Acta,2014,10(3):353-365.
[本文引用:1]
[8] MARIA L C,PAULA G T,YOLANDA F P,et al.Mechan-isms behind the resistance to trastuzumab in HER2-amplied breast cancer and strategies to overcome it[J].Clin Med Ins:Oncol,2016,10(Suppl 1):21-30.
Purpose of review: This review discusses the mechanisms of anti-human epidermal growth factor receptor 2 (HER2) resistance in breast cancer patients, detailing possible predictive biomarkers of therapy benefit that could implement novel therapeutic strategies. Recent findings: Despite a remarkable improvement in survival over the past two decades, up to 30% of early-stage HER2+ breast... [Show full abstract]
DOI:10.4137/CMO.S34537      PMID:4811269      URL    
[本文引用:1]
[9] HSU M C,CHANG H C,HUNG W C.HER-2/neu transcrip-tionally activates Jab1 expression via the AKT/beta-catenin pathway in breast cancer cells[J].Endocr Relat Cancer,2007,14(3):655-667.
Jab1 is a co-activator of activating protein-1 (AP-1) transcription factor and the fifth subunit of the constitutive photomorphogenesis 9 (COP9) signalosome, which has been shown to mediate nuclear exportation and ubiquitin-dependent degradation of the tumor suppressor p27(Kip1). Jab1 is overexpressed in several types of human cancer. However, de-regulation of Jab1 gene expression in cancer cells is largely unclear. In this study, we reported that expression of Jab1 was stimulated by HER-2/neu oncogene via transcriptional activation. Promoter deletion and mutation analysis indicated that HER-2/neu stimulated Jab1 via the T cell factor (TCF) binding site located at the -380/-368 region of the human Jab1 promoter. DNA affinity precipitation assay and chromatin immunoprecipitation assay verified that binding of beta-catenin and TCF-4 to this consensus site was increased by HER-2/neu. In addition, dominant-negative mutant of TCF significantly attenuated the stimulatory effect of HER-2/neu. We also demonstrated that HER-2/neu increased beta-catenin/TCF-mediated Jab1 expression via the AKT signaling pathway because chemical inhibitor or dominant-negative mutant of AKT effectively attenuated the stimulatory action of HER-2/neu. IGF-I, which is a well-known AKT activator, also up-regulated the expression of Jab1 in NIH/3T3 and MCF-7 cells. Knockdown of Jab1 by small interfering RNA (siRNA) preferentially inhibited proliferation of HER-2/neu-overexpressing breast cancer cells. Taken together, our results suggest that HER-2/neu transcriptionally activates Jab1 expression to promote proliferation of breast cancer cells.
DOI:10.1677/ERC-07-0077      PMID:17914096      URL    
[本文引用:1]
资源
PDF下载数    
RichHTML 浏览数    
摘要点击数    

分享
导出

相关文章:
关键词(key words)
Jab1
曲妥珠单抗
耐药性
乳腺癌
人类表皮因子受体2

Jab1
Trastuzumab
Drug resistance
Breast cancer
Human epidermal growth fa...

作者
沈倩
刘勇
于世英

SHEN Qian
LIU Yong
YU Shiying