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, 2017, 36(8): 875-879
doi: 10.3870/j.issn.1004-0781.2017.08.008
蟾毒灵对胃癌细胞侵袭转移的抑制作用*
Inhibition Effect of Bufalin on Invasion and Metastasis of Gastric Cancer Cells
王杰, 陈超, 张勇, 邢立凯, 许婕, 左青松, 蔡含, 蒋一鸣, 陈腾

摘要:

目的 探讨蟾毒灵对胃癌细胞侵袭转移能力的影响及其机制。方法 体外培养人胃癌AGS细胞,分为阴性对照组、蟾毒灵组和奥沙利铂组,采用噻唑蓝(MTT)法观察蟾毒灵对胃癌细胞增殖的影响,采用划痕实验和Transwell小室实验观察一定量蟾毒灵作用后胃癌细胞迁移和侵袭能力的变化,Western blotting 检测蟾毒灵对侵袭转移相关钙粘附蛋白E和基质金属蛋白酶(MMP)-2、MMP-9 的表达影响。结果 蟾毒灵可以抑制胃癌AGS细胞增殖,其抑制作用与剂量呈正相关;划痕实验和Transwell小室实验显示,与空白对照组比较蟾毒灵能够抑制胃癌细胞侵袭转移(P<0.05),与奥沙利铂组比较差异无统计学意义(P>0.05);Western-blotting结果显示,与空白对照组比较蟾毒灵组E-钙粘蛋白表达明显上调(P<0.05),而MMP-9和MMP-2表达明显下调(P<0.05),蟾毒灵组和奥沙利铂组比较差异无统计学意义。结论 蟾毒灵对胃癌细胞有显著的抗肿瘤作用,能够抑制胃癌细胞侵袭转移,调节侵袭转移相关基因的表达。

关键词: 蟾毒灵 ; ; ; 侵袭转移 ; 基质金属蛋白酶 ; E-钙粘蛋白

Abstract:

Objective To study effect of bufalin on invasion and metastasis of gastric cancer cells and related mechanism. Methods AGS human gastric cancer cell line was used for in vitro experiments.Cultured cells were treated by negative control group, bufalin group and oxaliplatin group.Cell proliferation was determined by MTT.Invasion and metastasis were observed by Wound-Healing Assay and Transwell Assay.Expression levels of E-cadherin,matrix metalloproteinases (MMP)-2,MMP-9 were detected by Western blotting. Results Bufalin was found to significantly inhibit the proliferation of AGS cells in a dose and time dependent manner.Wound-Healing Assay and Transwell Assay showed that as compared with the blank control group,bufalin inhibited invasion and metastasis of AGS cells (P<0.05),but there was no statistically significant difference between bufalin group and oxaliplatin group (P>0.05).Western blotting showed the expression of E-cadherin was increased in bufalin group as compared with the blank control group while the expression levels of MMP-9 and MMP-2 were down-regulated (P<0.05),but there was no statistically significant difference between bufalin group and oxaliplatin group (P>0.05). Conclusion Bufalin has anti-cancer activity on gastric cancer cells,and it has the ability of inhibiting cancer cell invasion and metastasis,and regulating the expression of some related gene.

Key words: Bufalin ; Cancer,gastric ; Invasion ; Metastasis ; Matrix metalloproteinases ; E-cadherin

胃癌是我国最常见的恶性肿瘤之一,位居恶性肿瘤的第2位,虽然最近几年胃癌的死亡率较前稍有下降,但其仍位居恶性肿瘤死亡率的第3位[1]。侵袭转移是恶性肿瘤的主要特征之一,胃癌一旦进入晚期或转移性阶段,手术对于提高患者的生存率起效甚微。因此更透彻地揭示胃癌的发生及发展的机制,寻找新的药物治疗靶点进行有针对性的治疗至关重要。蟾毒灵(bufalin)是从蟾酥中提取的多羟基甾体化合物,其分子式为C24H34O4,已有研究表明蟾毒灵能够在体外和体内选择性地对多种肿瘤具有杀伤及抑制作用,能够促进肿瘤细胞分化和诱导肿瘤细胞凋亡[2-4]。但蟾毒灵抗胃癌的作用及其机制研究较少,特别对胃癌侵袭转移能力的影响笔者还未见报道。因此本研究通过体外培养胃癌细胞,初步观察蟾毒灵对胃癌增殖及侵袭转移能力的影响,并探索其机制。

1 材料与方法
1.1 细胞培养和转染

人胃癌细胞株AGS 购于中国科学院上海细胞库,用含有10%胎牛血清(Gibco公司,批号:10099141),RPMI 1640培养液中(Gibco公司,批号:11875093),于37 ℃,5%二氧化碳(CO2)细胞培养箱中培养。蟾毒灵(Sigma公司,规格:每支10 mg,批号:B0261),奥沙利铂(赛诺菲制药有限公司,规格:每支50 mg,批号:J20100064);E-钙粘蛋白兔抗人单克隆抗体(美国CST公司,批号:5174)。基质金属蛋白酶(matrix metalloproteinase,MMP)-2(美国CST公司,批号:13132)、MMP-9兔抗人单克隆抗体(美国CST公司,批号:13667)。

1.2 MTT实验

取对数生长期人胃癌AGS细胞,0.25%胰酶消化,用细胞培养液制成1×104·mL-1 单细胞悬液接种于96 孔板,每孔100 μL,于37 ℃、5%二氧化碳(CO2)及饱和湿度条件下培养12 h。实验设阴性对照组(正常细胞培养液);蟾毒灵组浓度分别为 0,50,100,200,500,1 000,2 000 nmol·L-1;奥沙利铂组(阳性对照组)浓度分别为0,1.25,2.5,5,10,20 μmol·L-1,每组6个复孔。12 h 后将阴性对照组换上新鲜培养液100 μL,蟾毒灵组和奥沙利铂组去除细胞培养液后换上相应浓度的药物的培养液100 μL。分别培养24,48,72 h 后,吸去培养液,各孔加入MTT(5 g·L-1)20 μL,继续培养4 h,吸去上层液体,加DMSO各120 μL,振荡10 min,在酶标仪上读取490 nm处吸光度(A)值。细胞生长抑制率=(A对照-A实验)/A对照。并用Bliss 法计算半数抑制浓度(IC50),作为后面细胞实验的药物浓度。

1.3 划痕实验

先用marker笔在6孔板背后,用直尺比着,均匀地划横线,每隔0.5~1 cm一道,横穿过孔,每孔至少穿过5条线。在孔中加入约5×105个细胞,接种原则为过夜后融合率达到100%。第2天用枪头比着直尺,尽量垂直于背后的横线划痕。PBS洗细胞3次,去除划下的细胞,加入无血清培养液,分为空白对照组、蟾毒灵(72 nmol·L-1)组、奥沙利铂(8.56 μmol·L-1)组。放入37 ℃,5%CO2培养箱继续培养0,24,48 h,时间点取样拍照。

1.4 Transwell 侵袭实验

取对数生长期的AGS细胞,分为空白对照组、蟾毒灵(72 nmol·L-1)组、奥沙利铂(8.56 μmol·L-1)组。用无血清培养基调整浓度为105·mL-1悬浮细胞;在下室加入600 μL含20%血清的培养基。上室加入150 μL细胞悬液,37 ℃、5%CO2、饱和湿度条件下继续培养48 h。用镊子小心取出chamber,吸干上室液体,移到预先加入约800 μL甲醇的孔中,室温固定30 min。取出chamber,吸干上室固定液,移到预先加入约800 μL结晶紫染液的孔中,室温染色30 min;轻轻用PBS冲洗浸泡数次,取出chamber,吸去上室液体,用湿棉棒小心擦去上室底部膜表面上的细胞。用小镊子小心揭下膜表面上的细胞,底面朝上晾干,移至载玻片上用中性树脂封片;显微镜下取5个随机视野计数,统计结果。

1.5 免疫印迹(Western blotting)实验

取对数生长期的胃癌AGS细胞,分为空白对照组、蟾毒灵(72 nmol·L-1)组、奥沙利铂(8.56 μmol·L-1)组。细胞培养箱中培养48 h,加入100 μL 预冷的细胞裂解液,冰上裂解30 min,提取总蛋白,采用BCA蛋白浓度测定试剂盒,取50 μg蛋白质100 ℃水溶变性后聚丙烯酰胺凝胶电泳(SDS-PAGE),半干法转移至聚偏二氟乙烯膜(PVDF)膜上,5%脱脂奶粉室温封闭1 h,一抗4 ℃摇床过夜,TBST洗膜后二抗室温孵育1 h,TBST洗膜3次,滴加电化学发光(ECL)液暗室中X线光片显影,图像扫描。以GAPDH蛋白作表达作为内参照。

1.6 统计学方法

采用SPSS 19.0版统计分析软件进行分析。计量资料以均数±标准差( x ¯ ±s)表示,采用单因素方差分析比较各组间差异,多重比较采用LSD-t检验,以P<0.05 为差异有统计学意义。

2 结果
2.1 蟾毒灵对胃癌细胞增殖的影响

MTT结果显示,蟾毒灵和奥沙利铂均呈剂量和时间依赖性抑制胃癌细胞增殖(图1),蟾毒灵和奥沙利铂在作用48 h对AGS细胞的半数抑制浓度分别为72 nmol·L-1和8.56 μmol·L-1,作为后面实验用药物浓度。

图1 不同浓度蟾毒灵(A)和奥沙利铂(B)分别对胃癌AGS细胞增殖的比较(x¯±s,n=3)
与空白对照组比较,*1P<0.05,*2P<0.01

Fig.1 Comparison of the inhibitory activity on AGS cells beween bufalin at different concentration(A) and oxaliplatin(B)(x¯±s,n=3)
Compared with blank control group,*1P<0.05,*2P<0.01

2.2 蟾毒灵对细胞侵袭转移的影响

划痕实验结果显示,蟾毒灵组和奥沙利铂组胃癌细胞的迁移速度与空白对照组比较明显减慢,且以奥沙利铂组最为明显(图2)。Transwell实验结果显示空白对照组穿膜细胞数为242.7±13.0;蟾毒灵组为120.0±5.5,奥沙利铂组为101.7±6.5,与空白对照组比较蟾毒灵组与奥沙利铂组肿瘤细胞穿膜数明显减少(图3,P<0.05),且奥沙利铂组与蟾毒灵组比较穿膜细胞数更少(图3,P<0.05)。

图2 3组胃癌AGS细胞迁移的病理切片图(×100)

Fig.2 Pathology slices for cell invasion of three groups of AGS cells(×100)

图3 3组胃癌AGS细胞侵袭能力的比较
A.切片图;B.矩形图;与空白对照组比较,*1P<0.05

Fig.3 Comparison of cell invasion among three groups of AGS cells
A.slice image;B.histogram;compared with blank control group,*1P<0.05

2.3 蟾毒灵对侵袭转移相关基因蛋白表达的影响

Western-blot实验结果显示,与空白对照组比较蟾毒灵组和奥沙利铂组E-钙粘蛋白表达明显升高(P<0.05),但蟾毒灵组与奥沙利铂组比较,差异无统计学意义(P>0.05);与空白对照组比较,蟾毒灵组和奥沙利铂组MMP9、MMP2蛋白表达明显下调(P<0.05),且蟾毒灵组与奥沙利铂组比较,差异无统计学意义(P>0.05)。见图4。

图4 3组胃癌AGS细胞E-cadherin、MMP-9、MMP-2蛋白表达的电泳图

Fig.4 Electrophoretogram for the protein expression of E-cadherin, MMP-9 and MMP-2 in three groups of AGS cells

3 讨论

蟾毒灵是从蟾酥中提取的有效生物活性成分,是目前已广泛用于临床抗肿瘤治疗药物华蟾素注射液的有效成分之一[5-6]。许多研究表明,蟾毒灵能够通过诱导人肝癌细胞自噬作用,抑制肝癌细胞增殖[7],通过调节EGFR1/VEGFR2/ EG-FR/c-Met-Akt/p44/42/p38-NF-κB 信号通路来抑制肺癌细胞分化[8],调节凋亡相关基因Survivin表达诱导急性早幼粒细胞白血病细胞凋亡[9]。已有研究发现蟾毒灵能够抑制胃癌细胞增殖,诱导细胞凋亡,其机制与调节凋亡通路PI3K/Akt及凋亡相关基因表达有关[4,10]。但是有关蟾毒灵对胃癌侵袭转移的抑制作用研究还比较少,在笔者的研究中发现蟾毒灵能够抑制胃癌AGS细胞增殖,蟾毒灵对胃癌细胞的IC50为72 nmol·L-1,因此选择72 nmol·L-1蟾毒灵作为后面侵袭转移实验的药物浓度。划痕实验和Transwell小室实验结果均显示蟾毒灵组胃癌细胞的迁移速度与空白对照组比较明显减慢(P<0.05),穿膜细胞数(P<0.05),另外笔者采用临床常用抗胃癌药物奥沙利铂作为阳性对照组,结果显示两组之间未见明显差异(P>0.05)。

MMPs是一种重要的蛋白水解酶家族,可以降解细胞外基质和基底膜成分以及一些非基质成分,从而改变肿瘤细胞的微环境,促进肿瘤的侵袭和转移[11]。研究表明MMP-2、MMP-9是作为Ⅳ型胶原酶的主要的细胞外基质降解蛋白酶。引起细胞外基质和基底膜降解,从而促进肿瘤细胞的迁徙和转移[12]。临床研究表明MMP-2和MMP-9与胃癌的进展相关[13-14]。姬瑞等[15]研究证实MMP-9表达的多少与胃癌组织的生成血管的情况相关,其参与了胃癌发生、发展、浸润和转移,且具有异质性,在转移灶中表达更高,是胃癌发生到晚期的主要作用原因。张金玲等[16]也研究显示,MMP-2在胃腺癌中表达的强弱与TNM分期、肿瘤浸润深度、淋巴转移与否呈正相关,其表达增高可增强低分化胃癌细胞对周围组织和器官的浸润能力,有利于淋巴转移。为进一步研究蟾毒灵抑制胃癌侵袭转移的机制,笔者采用免疫印迹方法检测了蟾毒灵作用后胃癌细胞中MMP-2和MMP-9蛋白的表达,研究发现蟾毒灵能够下调胃癌细胞MMP-2和MMP-9蛋白表达(P<0.05),蟾毒灵组与奥沙利铂组比较未见明显差异(P>0.05)。笔者推测蟾毒灵抑制胃癌细胞侵袭转移与下调MMP-2和MMP-9蛋白表达有关。

E-钙粘蛋白为一种钙粘蛋白分子家族中跨膜糖蛋白,由N端胞外区、C 端胞内区及高度疏水的跨膜区构成其分子结构。N端胞外区氨基酸末端与钙离子相结合,而胞内羧基末端则主要与细胞骨架蛋白的相互作用E-钙粘蛋白在粘着连接处集中表达,可有效维持上皮细胞的完整性、极性,对保持上皮细胞形态和组织结构起重要作用。大多数上皮组织来源的恶性肿瘤,随着肿瘤恶性程度的加深,E-钙粘蛋白介导的肿瘤细胞与正常细胞相比,其粘附能力逐渐下降[17]。E-钙粘蛋白在细胞分裂黏附和肿瘤早期浸润中有调控作用,其表达下调或缺失会导致肿瘤细胞失去接触抑制,增长和去分化不受控制,细胞之间的连接松散、容易脱落、排列紊乱、表现出较强的侵袭能力[18]。因此,在很多来源于上皮组织的肿瘤中,E-钙粘蛋白的表达下调或缺失是造成肿瘤细胞具有高侵袭的关键因素之一。在本研究中蟾毒灵作用后胃癌细胞中E-钙粘蛋白与空白对照组比较明显上调,且与奥沙利铂组达到了同样的效果。

综上所述,笔者初步研究显示蟾毒灵能够抑制胃癌细胞增殖,抑制肿瘤侵袭转移,其机制与抑制MMP-2、MMP-9表达并上调E-钙粘蛋白表达有关。但有关其对侵袭转移相关信号通路的影响及其涉及的信号靶点还有待于进一步研究。

The authors have declared that no competing interests exist.

参考文献

[1] 郑朝旭,郑荣寿,张思维,.中国2010 年胃癌发病与死亡分析[J].中国肿瘤,2014,23(10):795-800.
[本文引用:1]
[2] GAI Q,SHENG X,QIN J M,et al.The effect and mechani-sm of bufalin on regulating hepatocellular carcinoma cell invasion and metastasis via Wnt/β-catenin signaling pathway[J].Int J Oncol,2016,48(1):338-348.
Hepatocellular carcinoma (HCC) is a highly malignant tumor with an extremely poor prognosis. Our preliminary study indicated that bufalin could restrain the proliferation of human hepatoma BEL-7402 cells in a time- and dose-dependent manner. In the present study, the colony formation assay, the Transwell invasion assay, the western blot analysis and the immunofluorescence method were respectively used to investigate the effect and mechanism of bufalin against HCC cell invasion and metastasis. We found that: i) bufalin had significant inhibitory effect on the cell proliferation of BEL-7402 cells; ii) bufalin markedly inhibited the migration and invasion of BEL-7402 cells; iii) bufalin could suppress the phosphorylation of GSK-3β Ser9 site in BEL-7402 cells, decrease the expression of β-catenin, cyclin02D1, metalloproteinases-7 (MMP-7) and cyclooxygenase-2 (COX-2) in the cytoplasm, and increase the expression of E-cadherin and β-catenin on the cell membrane; and iv) the expression of α-fetoprotein significantly decreased and the expression of albumin increased in BEL-7402 cells after bufalin was used. Our results indicate that: i) bufalin can regulate the expression of associated factors in Wnt/β-catenin signaling pathway of BEL-7402 cells through suppressing the phosphorylation of GSK-3β Ser9 site; ii) bufalin can strengthen intercellular E-cadherin/β-catenin complex to control epithelial-mesenchymal transition; and iii) bufalin can reverse the malignant phenotype and promote the differentiation and maturation by regulating the AFP and ALB expression in BEL-7402 cells. These are very important mechanisms of bufalin on the inhibition of the invasion and metastasis of HCC cells.
DOI:10.3892/ijo.2015.3250      PMID:26648168      URL    
[本文引用:1]
[3] CHEN Y,LI M,LI Z,et al.Bufalin induces apoptosis in the U2OS human osteosarcoma cell line via triggering the mitochondrial pathway[J].Mol Med Rep,2016,13(1):817-822.
Bufalin has been shown to induce apoptosis in osteosarcoma cells; however, the underlying mechanism has not been elucidated. The purpose of the present study was to investigate whether mitochondriamediated signaling pathways trigger the process of apoptosis in the U2OS osteosarcoma cell line. Bufalin inhibited the proliferation and induced apoptosis in U2OS cells in a time- and dosedependent manner. Bufalininduced apoptosis was accompanied with a significant reduction of the mitochondrial membrane potential, release of mitochondrial cytochrome c into the cytosol, activation of caspase3, caspase9 and poly(adenosine diphosphate ribose) polymerase, as well as downregulation of B-cell lymphoma 2 (Bcl-2)/Bcl-2-associated X protein. Cyclosporin A, a specific inhibitor of the mitochondrial permeability transition pore, attenuated bufalin-induced apoptosis. In conclusion, the present study revealed that bufalin induced apoptosis in the U2OS human osteosarcoma cell line via triggering of the mitochondrial pathway.
DOI:10.3892/mmr.2015.4583      PMID:26648158      URL    
[本文引用:0]
[4] ZHAO H Y,ZHAO D L,TAN G,et al.Bufalin promotes ap-optosis of gastric cancer by down-regulation of miR-298 targeting bax[J].Int J Clin Exp Med,2015,8(3):3420-3428.
[本文引用:2]
[5] MENG Z,YANG P,SHEN Y,et al.Pilot study of huachansu in patients with hepatocellular carcinoma,nonsmall-cell lung cancer,or pancreatic cancer[J].Cancer,2009,115(22):5309-5318.
BACKGROUND:Huachansu, a Chinese medicine that comes from dried toad venom from the skin glands of Bufo gargarizans or B. melanostictus, has been used in the treatment of various cancers in China. The authors conducted a pilot study, using a phase 1 trial design, of huachansu in patients with advanced cancer.METHODS:Huachansu was administered intravenously for 14 days followed by 7 days off (1 cycle). Without significant adverse events or progressive disease, treatment continued beyond 2 cycles. The dose of huachansu was escalated as follows with 3 patients per cohort: 10 (level 1), 20 (level 2), 40 (level 3), 60 (level 4), and 90 (level 5) mL/m2.RESULTS:Fifteen patients (hepatocellular cancer, n = 11; nonsmall cell lung cancer, n = 2; pancreatic cancer, n = 2) were enrolled in the trial, and no dose-limiting toxicities (DLTs) were found. Eleven patients had no drug-related toxicity greater than grade 1. Six (40%) had stable disease (median duration, 6.0 months; range, 3.5-11.1 months). One of these patients (with hepatocellular cancer) had 20% regression (duration, 11 months) (dose level 1). Quality of life improved for patients with stable disease. Plasma bufalin concentration reached maximal levels at the end of the 2-hour infusion and was proportional to the amount of drug being administered (0.81-3.38 ng/mL).CONCLUSIONS:No DLT was observed with the use of huachansu at doses up to 8 higher than typically used in China. Six patients had prolonged stable disease or minor tumor shrinkage. Cancer 2009. 2009 American Cancer Society.
DOI:10.1002/cncr.24602      PMID:19701908      URL    
[本文引用:1]
[6] QIN T J,ZHAO X H,YUN J,et al.Efficacy and safety of gemcitabine-oxaliplatin combined with huachansu in patients with advanced gallbladder carcinoma[J].World J Gastroenterol,2008,14(33):5210-5216.
AIM:To evaluate the efficacy and safety of gemcitabine-oxaliplatin(GEMOX)cornbined with huachansu(cinobufagin)injection treatment in patients with locally advanced or metastatic gallbladder carcinoma(GBC),and to assess the quality of life(QOL)of such patients.METHODS:Twenty-five patients with locally advanced or metastatic GBC were treated with intravenous gemcitabine(1000 mg/m2)over 30 min on days 1 and 8,2 h infusion of oxaliplatin(120 mg/m2)on day 1,and 2-3 h infusion of huachansu(20 mL/m2)on days-3-11,every 3-4 wk.Treatment was continued until occurrence of unacceptable toxicity or disease progression.QOL of patients was assessed by the EORTC QLQ-C30 at baseline,at the end of the first,third and sixth chemotherapy cycles,and 1 mo after the treatment.RESULTS:Among the 25 patients with a median age of 64 years(range 42-78 years),23 were evaluable in the study.A total of 137 cycles of therapy were performed and the median cycle was 5(range 1-8)per patient.Out of the 23 patients whose response could be evaluated,8 partial responses(PR)were observed(34.8%),while 7 patients(30.4%)demonstrated a stable disease(SD).The disease control rate was 65.2%.Progression of cancer was observed in 8(34.8%)patients.The median progression-free and overall survival time was 5.8 mo(95% CI:4.5-7.1 mo)and 10.5 mo,respectively.The therapy was well tolerated,with moderate myelosuppression as the main toxicity.Anemia grade 2 was seen in 16.0%,neutropenia grade 3 in 8.0% and thrombocytopenia grade 3 in 24.0% of patients,respectively.Non-hematologic toxicity ranged from mild to moderate.No death occurred due to toxicity.The QOL of patients was improved after chemotherapy,and the scores of QOL were increased by 10 t0 20 points.CONCLUSION:GEMOX combined with huachansu(cinobufagin)injection is well tolerated,effective,thus ireproving the QOL of patients with advanced GBC.(C)2008 The WJG Press.All rights reserved.
DOI:10.3748/wjg.14.5210      PMID:18777599      URL    
[本文引用:1]
[7] MIAO Q,BI L L,LI X,et al.Anticancer effects of bufalin on human hepatocellular carcinoma HepG2 cells:roles of apoptosis and autophagy[J].Int J Mol Sci,2013,14(1):1370-1382.
The traditional Chinese medicine bufalin, extracted from toadamp;#8217;s skin, has been demonstrated to exert anticancer activities in various kinds of human cancers. The mechanisms of action lie in its capacity to induce apoptosis, or termed type I programmed cell death (PCD). However, type II PCD, or autophagy, participates in cancer proliferation, progression, and relapse, as well. Recent studies on autophagy seem to be controversial because of the dual roles of autophagy in cancer survival and death. In good agreement with previous studies, we found that 100 nM bufalin induced extensive HepG2 cell apoptosis. However, we also noticed bufalin triggered autophagy and enhanced Beclin-1 expression, LC3-I to LC3-II conversion, as well as decreased p62 expression and mTOR signaling activation in HepG2 cells. Blockage of autophagy by selective inhibitor 3-MA decreased apoptotic ratio in bufalin-treated HepG2 cells, suggesting a proapoptotic role of bufalin-induced autophagy. Furthermore, we investigated the underlying mechanisms of bufalin-induced autophagy. Bufalin treatment dose-dependently promoted AMPK phosphorylation while AMPK inhibition by compound C significantly attenuated bufalin-induced autophagy. Taken together, we report for the first time that bufalin induces HepG2 cells PCD, especially for autophagy, and the mechanism of action is, at least in part, AMPK-mTOR dependent.
DOI:10.3390/ijms14011370      Magsci     URL    
[本文引用:1]
[8] JIANG Y Y,ZHANG Y,LUAN J L,et al.Effects of bufalin on the proliferation of human lung cancer cells and its molecular mechanisms of action[J].Cytotechnology,2010,62(6):573-583.
Bufalin, a naturally occurring small-molecule compound from Traditional Chinese Medicine (TCM) Chansu showed inhibitory effects against human prostate, hepatocellular, endometrial and ovarian cancer cells, and leukemia cells. However, whether or not bufalin has inhibitory activity against the proliferation of human non-small cell lung cancer (NSCLC) cells is unclear. The aim of this study is to study the effects of bufalin on the proliferation of NSCLC and its molecular mechanisms of action. The cancer cell proliferation was measured by MTT assay. The apoptosis and cell cycle distribution were analyzed by flow cytometry. The protein expressions and phosphorylation in the cancer cells were detected by Western blot analysis. In the present study, we have demonstrated that bufalin suppressed the proliferation of human NSCLC A549 cell line in time- and dose-dependent manners. Bufalin induced the apoptosis and cell cycle arrest by affecting the protein expressions of Bcl-2/Bax, cytochrome c, caspase-3, PARP, p53, p21WAF1, cyclinD1, and COX-2 in A549 cells. In addition, bufalin reduced the protein levels of receptor expressions and/or phosphorylation of VEGFR1, VEGFR2, EGFR and/or c-Met in A549 cells. Furthermore, bufalin inhibited the protein expressions and phosphorylation of Akt, NF- B, p44/42 MAPK (ERK1/2) and p38 MAPK in A549 cells. Our results suggest that bufalin inhibits the human lung cancer cell proliferation via VEGFR1/VEGFR2/EGFR/c-Met-Akt/p44/42/p38-NF- B signaling pathways; bufalin may have a wide therapeutic and/or adjuvant therapeutic application in the treatment of human NSCLC.
DOI:10.1007/s10616-010-9310-0      PMID:20963488      Magsci     URL    
[本文引用:1]
[9] ZHU Z,LI E,LIU Y,et al.Bufalin induces the apoptosis of acute promyelocytic leukemia cells via the downregulation of survivin expression[J].Acta Haematologic,2012,128(3):144-150.
Abstract BACKGROUND AND AIMS: Bufalin is a cardiotonic steroid isolated from the Chinese toad venom preparation Chan'su and has been shown to induce leukemia cell differentiation and apoptosis under certain experimental conditions. However, the detailed mechanism by which bufalin induces the apoptosis of acute promyelocytic leukemia cells is largely unexplored. METHODS: The acute promyelocytic leukemia cell line NB4 was treated with bufalin, then the proliferation was evaluated by cell viability assay and apoptosis was detected by flow cytometry analysis. In addition, NB4 cells were treated by MEK inhibitor PD98059 in combination with bufalin, and the expression of survivin and activation of caspase-3 were detected by Western blot analysis. RESULTS: Bufalin inhibited the proliferation and induced the apoptosis of NB4 cells in a dose- and time-dependent manner. Moreover, bufalin synergized with PD98059 to inhibit the proliferation and induce the apoptosis of NB4 cells, which was associated with the downregulation of survivin expression and the upregulation of caspase-3 activation. CONCLUSIONS: Bufalin is a potential regimen to be used in combination with conventional chemotherapeutic drugs to improve acute promyelocytic leukemia therapy. Copyright 2012 S. Karger AG, Basel.
DOI:10.1159/000339424      PMID:22890165      URL    
[本文引用:1]
[10] LI D,QU X J,HOU K Z,et al.PI3K/Akt is involved in bufalin-induced apoptosis in gastric cancer cells[J].Anti-Cancer Drugs,2009,20(1):59-64.
Bufalin is the active ingredient of the Chinese medicine Chan Su, and it has been reported that bufalin induces apoptosis in some human leukemia and solid cancer cell lines. The exact mechanism of bufalin-induced apoptosis is, however, still not clear. In this study, we demonstrated that bufalin inhibited the proliferation of gastric cancer MGC803 cells in a dose-dependent and time-dependent manner. At a low concentration (20 nmol/l), bufalin induced M-phase cell cycle arrest, whereas at a high concentration (80 nmol/l) it induced apoptosis in MGC803 cells. Bufalin increased the Bax/Bcl-2 ratio and activated caspase-3 during the apoptotic process of MGC803 cells. It should be noted that bufalin transiently activated the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway and then inhibited it completely, and upregulated the Casitas B-lineage lymphoma (Cbl) family of ubiquitin ligases, upstream modulators of PI3K. A combination of bufalin and LY294002, a PI3K-specific inhibitor, enhanced apoptosis, but PD98059, an extracellular-regulated protein kinase-specific inhibitor, had no significant effect on bufalin-induced apoptosis. These results suggested that the PI3K/Akt pathway might play a key role in bufalin-induced apoptosis in gastric cancer MGC803 cells.
DOI:10.1002/eej.10318      PMID:19343001      URL    
[本文引用:1]
[11] PATTERSON M L,ATJUBSIB S J,KNAUPER V,et al.Specific collagenolysis by gelatinasc A,MMP-2,is determined by the hemopexin domain and not the fibronectin-like domain[J].Febs Letters,2001,503(2-3):158-162.
In view of the essential role of the hemopexin domain of the traditional interstitial collagenases, MMP-1, -8, -13 and MT1-MMP (MMP-14), in determining specific collagen cleavage we have studied the function of this domain in MMP-2, relative to that of the fibronectin-like domain that promotes gelatinolysis. Although the fibronectin-like domain promotes avid binding to collagen, our data demonstrate that the catalytic and hemopexin domains of MMP-2 are sufficient to effect the critical step in cleavage of rat type I collagen into 3/4 and 1/4 fragments. The mechanism of MMP-2 cleavage of collagen proceeds in two phases, the first resembling that of the interstitial collagenases, followed by gelatinolysis, promoted by the fibronectin-like domain.
DOI:10.1016/S0014-5793(01)02723-5      PMID:11513874      URL    
[本文引用:1]
[12] HAGEDOM H G,BACHMEIER B E,NERLICH A G.Synthesis and degradation of basement membranes and extracellular matrix and their regulation by TGF-beta in invasive carcinomas[J].Int J Oncol,2001,18(4):669-681.
The proper structure of the extracellular matrix, in particular of the basement membrane and the adjacent interstitial matrix, are essential prerequisits for a proper function of tissues. Invasive growth in malignant tumors is associated with a destruction of various matrix structures. Due to extensive recent analyses significant advances have been made in the knowledge of the structure of the extracellular matrix, the composition of its most important constituents, their metabolism and that of matrix degrading enzymes. This information provides insight into the pathophysiology of malignant growth. Thereby, it has been shown that malignant tumor growth is associated with a loss of basement membrane (BM) material which, however, disappears not homogeneously, but affects various BM components to different degree. The loss of an intact BM as the first barrier is therefore the initial step of tumor invasion. Despite this loss there is evidence that the de novo synthesis of BM constituents in tumor and adjacent stromal cells is enhanced. Thus, it is obvious that BM material is degraded during the invasion process to significant degree. In addition, since there is a positive correlation between the amount of retained peritumoral BM and a higher degree of tumor cell differentiation the amount of retained BM material seems to represent a marker for the biological behaviour of the tumor cells. The loss of BM material is well explained by a significant expression of major matrix degrading enzymes, the matrix metalloproteinases (MMPs) both on the mRNA and protein level. Here again, there is considerable data indicating that both tumor and stroma cells are involved in the MMP synthesis. In addition to the loss of BM substances, the interstitial extracellular matrix (ECM) is disarranged. This disarrangement may comprise enhanced de novo synthesis (
DOI:10.3892/ijo.18.4.669      PMID:11251160      URL    
[本文引用:1]
[13] ANATOLY P B,RINA I G,MARAT R G,et al.Stomach cancer:interconnection between the redox state,activity of MMP-2,MMP-9 and stage of tumor growth[J].Cancer Microenviron,2016,9(1):27-32.
Abstract High levels of reactive oxygen (ROS) and nitrogen (RNS) species can lead to the destruction of extracellular matrix facilitating tumor progression. ROS can activate matrix metalloproteinases (MMP), damage DNA and RNA. Therefore, the levels of MMP, ROS and RNS can serve as additional prognostic markers and for the estimation of the effectiveness of tumor therapy. Concerning gastric cancer, the prognostic role of MMP, its connection with the cancer staging remains controversial and correlations between the activity of MMP with the ROS and RNS levels are insufficiently confirmed. Superoxide generation rates, nitric oxide (NO) levels, concentrations of active forms of matrix metalloproteinases MMP-2 and MMP-9 in tumor and adjacent tissues of patients with stomach cancer at different disease stages were measured by electron spin resonance (ESR) including spin-trapping and polyacrylamide gel zymography. It is shown that the activity of MMP-2 and MMP-9 in tumor tissue correlate with the superoxide radicals generation rate and NO levels (r = 0.48 0.67, p < 0.05). The activity of MMP-2 and MMP-9 in tumor tissues and superoxide radical generation rates correlate positively with the stage of regional dissemination (r = 0.45 and 0.37, correspondingly, p < 0.05), but MMP-2 and MMP-9 activity inversely depends on distant metastatic degree of stomach cancer (r = 0.58; p < 0.05). Additionally, the feasibility of ESR to locally determine oxidative stress is demonstrated.
DOI:10.1007/s12307-016-0182-5      PMID:26905073      URL    
[本文引用:1]
[14] GIALELI C,THEOCHARIS A D,KARAMANOS N K.Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting[J].Febs J,2011,278(1):16-27.
Matrix metalloproteinases (MMPs) consist of a multigene family of zinc-dependent extracellular matrix (ECM) remodeling endopeptidases implicated in pathological processes, such as carcinogenesis. In this regard, their activity plays a pivotal role in tumor growth and the multistep processes of invasion and metastasis, including proteolytic degradation of ECM, alteration of the cell-cell and cell-ECM interactions, migration and angiogenesis. The underlying premise of the current minireview is that MMPs are able to proteolytically process substrates in the extracellular milieu and, in so doing, promote tumor progression. However, certain members of the MMP family exert contradicting roles at different stages during cancer progression, depending among other factors on the tumor stage, tumor site, enzyme localization and substrate profile. MMPs are therefore amenable to therapeutic intervention by synthetic and natural inhibitors, providing perspectives for future studies. Multiple therapeutic agents, called matrix metalloproteinase inhibitors (MMPIs) have been developed to target MMPs, attempting to control their enzymatic activity. Even though clinical trials with these compounds do not show the expected results in most cases, the field of MMPIs is ongoing. This minireview critically evaluates the role of MMPs in relation to cancer progression, and highlights the challenges, as well as future prospects, for the design, development and efficacy of MMPIs.
DOI:10.1111/j.1742-4658.2010.07919.x      PMID:21087457      Magsci     URL    
[本文引用:1]
[15] 姬瑞,周永宁,任涛文,.MMP-9在胃癌及其淋巴结组织中表达的临床意义[J].中国肿瘤临床,2010,37(7):377-380.
目的:研究MMP-9在胃癌组织中的表达状况,分析其与临床病理特征和生存之间的关系.方法:应用免疫组化SP法,采用组织芯片技术,分析189例胃癌、54例癌旁组织、41例淋巴结转移灶及32例正常对照组织中c-Jun和MMP-9蛋白表达.结果:胃癌中MMP-9蛋白的阳性表达率为78.3%.MMP-9的表达与浸润深度、淋巴结转移、分化程度及Lauren分型有关(P0.05).结论:MMP-9在胃癌组织中均有高表达.MMP-9阳性表达与肿瘤的浸润、转移密切相关,可作为判断胃癌恶性行为的重要生物学标记物.MMP-9在促进血管生成中起重要作用.MMP-9蛋白阳性表达对胃癌患者来说是一个不利的预后因素,但尚不能作为一个独立的预后指标.
DOI:10.3969/j.issn.1000-8179.2010.07.005      Magsci     URL    
[本文引用:1]
[16] 张金玲,费雁,陈伟,.MMP-2、MMP-14、TIMP-2在胃癌组织中的表达及意义[J].华中科技大学学报(医学版),2013,42(2):227-230.
[本文引用:1]
[17] EIDELMAN S,DAMSKY C H,WHEELOCK M J,et al.Expression of the cell-cell adhesion glycoprotein cell-CAM120 /80 in normal human tissues and tumors[J].Am J Pathol,1989,135(1):101-110.
Polyclonal and monoclonal antibodies raised to the 80 kd glycoprotein component of the cell to cell adhesion molecule cell-CAM 120/80 were used to map its distribution immunohistochemically in normal human tissues and in benign and malignant tumors. Cell-CAM 120/80 was found in all normal epithelial tissues, but was not expressed on neural, lymphoid, smooth, striated and cardiac muscle, connective tissue, or the germ cells in either sex. The expression of this adhesion molecule was polarized in ductal and glandular epithelia and evenly circumferential in squamous and transitional epithelia. Some organs, such as the kidney, liver and endocrine glands, showed unique organ to tissue specific patterns. Maturation-dependent loss of cell-CAM 120/80 was noticed in superficial layers of squamous epithelium and the placenta. Benign epithelial tumors expressed cell-CAM 120/80 in a manner comparable with their tissue of origin. Malignant tumors expressed cell-CAM 120/80 either in a manner similar to the tissue of their origin or assumed a less polarized phenotype. Overall, the immunoreactivity in many malignant tumors appeared weaker and the polarization was less pronounced. Thus, cell-CAM 120/80 is a universal marker of human epithelial cells, but its mode of expression differs in various anatomic sites, and may be influenced by maturation or malignant transformation of cells.
DOI:10.1007/BF00724904      PMID:2774055      URL    
[本文引用:1]
[18] HERMISTON M L,WONG M H,GORDON J I.Forced expression of E-cadherin in the mouse intestinal epithelium slows cell migration and provides evidence for nonauto-nomous regulation of cell fate in a self-renewing system[J].Genes Dev,1996,10(8):985-996.
The adult mouse small intestinal epithelium is self-renewing. Its crypt-villus unit provides a model for studying many of the processes that occur during tissue morphogenesis such as control of proliferative status, specification of cell fate, regulation of differentiation, and induction of death. To assess the contributions of cell-cell and cell-substratum interactions to the coordinated control of these processes, 129/Sv embryonic stem (ES) cells, transfected with a recombinant DNA consisting of a fatty acid-binding protein gene (Fabp1) promoter that functions along the entire length of the crypt-villus axis linked to mouse E-cadherin, were introduced into normal C57Bl/6 (B6) blastocysts. Analyses of adult B6 129/Sv mice indicated that forced expression of E-cadherin suppresses proliferation and induces apoptosis in the crypt, and slows cell movement up the villus. The slowed migration is not accompanied by a change in distribution of terminal differentiation markers along the crypt-villus axis suggesting that differentiation is largely cell nonautonomous. To determine whether the slowed migration was a direct effect of forced expression of E-cadherin or a secondary effect of reduced crypt cell production, another Fabp promoter was used to restrict overproduction of E-cadherin to the villus epithelium of transgenic mice. Enterocytic migration was slowed, although proliferation and apoptosis were not perturbed in crypts. Augmentation of cellular E-cadherin pools was accompanied by an increase in beta-catenin levels. These findings establish that cadherins and their associated proteins modulate cellular migration, proliferation, and death programs in an adult vertebrate organ.
DOI:10.1101/gad.10.8.985      PMID:8608945      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)
蟾毒灵
侵袭转移
基质金属蛋白酶
E-钙粘蛋白

Bufalin
Cancer,gastric
Invasion
Metastasis
Matrix metalloproteinases
E-cadherin

作者
王杰
陈超
张勇
邢立凯
许婕
左青松
蔡含
蒋一鸣
陈腾

WANG Jie
CHEN Chao
ZHANG Yong
XING Likai
XU Jie
ZUO Qingshong
CAI Han
JIANG Yiming
CHEN Teng