首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到18条相似文献,搜索用时 281 毫秒
1.
目的:利用HER2/neu胞外配体结合区2(RLD2)从噬菌体抗体库中筛选相应抗体,并进行初步检测。方法:设计合成引物,利用PCR方法克隆出RLD2基因后,将其连接到pET-24a( )载体中,在大肠杆菌中实现高效表达。对包涵体蛋白经纯化、透析复性后得到目的蛋白。以得到的目的蛋白为靶标,从人源性噬菌体抗体库中进行4轮筛选得到抗体,经ELISA法初步鉴定,并用MTT法检测阳性克隆。结果与结论:初步得到6株亲和力较高的抗HER2/neu抗体,选取其中2株进行了MTT法检测,表明对HER2高表达的乳腺癌细胞有较明显的抑制作用。  相似文献   

2.
HER2/neu 基因在肿瘤中的过度表达使其成为许多肿瘤的标志分子 . 为了增加过度表达 HER2/neu 的肿瘤细胞对肿瘤坏死因子 (TNF) 的敏感性和提高 HER2/neu 抗体的肿瘤杀伤效应,将抗 HER2/neu 单链抗体 C6.5 与人肿瘤坏死因子 hTNF-α融合,构建了 scFvC6.5-hTNF-α融合蛋白,完成了重组蛋白在大肠杆菌中的表达,产率为 400 μg/L 菌液 . 经过亲和层析和柱复性,融合蛋白的纯度达 95%以上 . ELISA 试验表明, scFvC6.5-hTNF-α能够特异结合 HER2/neu 阳性卵巢癌细胞 SKOV-3 和乳腺癌细胞 MCF-7 ,而不结合 HER2/neu 阴性的黑色素瘤细胞 A375. MTT 试验表明, scFvC6.5-hTNF-α能够选择性地杀伤 SKOV-3 和 MCF-7 细胞,而不影响 A375 细胞的生长 . 这种肿瘤细胞特异性杀伤作用提示该免疫毒素具有肿瘤靶向治疗的潜在应用价值 .  相似文献   

3.
应用RT PCR方法扩增了编码猪瘟病毒石门株 (CSFVshimenstrain)囊膜糖蛋白E2全基因 ,然后将其克隆到pMD 1 8T质粒中 ,获得重组质粒pMD E2。再以pMD E2为模板 ,另行设计两对引物 ,同时扩增其中一段适于在E .coli中表达且抗原反应性较好的基因片段 (E2蛋白A D抗原区基因序列 ) ,将扩增的两片段串联插入原核表达载体pET 32a中构建成重组质粒pET 2e。用酶切和序列分析鉴定插入目的基因的正确性。SDS PAGE和Western blot分析表明 ,经pET 2e转化、IPTG诱导的受体菌可表达目的蛋白 ,克隆在硫氧还蛋白 (thioredoxinprotein ,TrxA)基因下游的E2蛋白基因与TrxA基因获得了高效融合表达 ,并且具有免疫学反应活性 ,这为猪瘟的血清学诊断方法的建立打下了基础 。  相似文献   

4.
应用RT-PCR技术从人乳腺癌细胞系SK-BR-3中克隆出人表皮生长因子受体2(human epidermal growth factorreceptor 2,HER2)基因的胞外段,并插入到表达载体pET-30a中,得到重组表达载体pET30-HER2(Ex)。将该载体转化至大肠杆菌BL21(DE3)细胞中,加入IPTG进行诱导表达,成功获得HER2胞外段蛋白。分别提取培养液上清、大肠杆菌周质腔、细胞质可溶性及不可溶性组分蛋白进行SDS-PAGE电泳分析,确定目的蛋白定位于大肠杆菌细胞质包涵体中。通过改变诱导温度、诱导物浓度、诱导起始菌体密度和诱导时间,寻找最佳表达条件,使目的蛋白的表达量达到最高。结果表明,在37℃下,OD600达到1.0时,经终浓度为0.1 mmol/L的IPTG诱导4 h,目的蛋白的表达量最高。将重组表达菌进行超声破碎,分离出包涵体组分,经Ni2+亲和层析纯化后获得了纯度>90%的HER2胞外段蛋白,从而为抗HER2抗体的制备及肿瘤疫苗的研究奠定了基础。  相似文献   

5.
目的为了构建人分泌型磷脂酶A2(secretaryphospholipaseA2,sPLA2-IIA)的有效表达系统,从胎脾中提取总RNA。方法采用RT-PCR方法扩增出编码sPLA2-IIA的基因定向地克隆于硫氧环蛋白基因融合表达载体pET32a的TrxA基因3′末端,构建符合读码框的融合表达载体pET32a-sPLA2-IIA。37℃下经IPTG诱导,hsPLA2-IIA融合蛋白在大肠杆菌BL21(DE3)中获得高效表达,表达产物以包涵体的形式存在。包涵体经8mol/L尿素溶解、复性后检测结果显示具有较高的催化活性并呈现剂量依赖关系。结论以大肠杆菌为宿主,成功表达了hsPLA2-IIA蛋白,为进一步进行hsPLA2-IIA的大量生产和功能研究奠定了基础。  相似文献   

6.
以复制子为p15A的质粒pACU184为基础 ,构建了 3种表达硫氧还蛋白 (TrxA)或 和二硫键异构酶 (DsbC)的表达质粒 .经IPTG诱导 ,克隆的DsbC和TrxA都以可溶的形式高表达 .分别将构建的 3种表达质粒与复制子为colE1并克隆有人源化鼠抗人纤维蛋白单链抗体 低分子量尿激酶融合基因 (C6 UK)的表达质粒共转化大肠杆菌XL1 blue ,在 30℃用IPTG诱导表达 .SDS PAGE显示 ,共表达TrxA或DsbC都能导致C6 UK融合蛋白的部分可溶性表达 ,而且同时共表达TrxA和DsbC 2种分子时 ,C6 UK完全以可溶形式表达 ,但表达量降低 .分别用溶圈法和ELISA检测了各种共表达时可溶表达产物的生物活性 .结果显示 ,只有共表达DsbC时才能检测到明显的C6 UK融合蛋白的双功能  相似文献   

7.
HER-2/neu癌基因在许多肿瘤,如乳腺癌、卵巢癌、非小细胞肺癌等肿瘤中高表达,在肿瘤的发生与发展中起重要作用,与肿瘤的转化、转移、复发、预后差、患者生存期缩短有关。HER-2/neu在乳腺癌过度表达率约为20%~30%,编码蛋白P185HER2属生长因子受体家族,抗P185HER2单克隆抗体(Herceptin)作为靶向药物已临床应用治疗HER2/neu高表达乳腺癌。  相似文献   

8.
目的:构建人角质细胞生长因子2(hKGF2)基因的高效原核表达载体pET-30a( )-hKGF2,并在大肠杆菌BL21(DE3)中获得表达。方法:从培养的人胚胎肺成纤维细胞中提取总RNA,采用RT-PCR技术扩增出去除了信号肽部分的hKGF2基因,克隆到pMD18-T载体,经DNA序列分析后与pET-30a( )表达载体连接,在大肠杆菌BL21(DE3)诱导表达6×His-hKGF2,用SDS-PAGE及Western印迹鉴定表达蛋白。结果:构建了表达载体pET-30a( )-hKGF2,经IPTG诱导后,表达的重组蛋白理论相对分子质量约为23000,约占菌体总蛋白的20%。结论:6×His-hKGF2蛋白在大肠杆菌BL21(DE3)中为可溶性高效表达,为获得高纯度、高活性的产物,以及进一步的大规模生产和应用研究奠定了基础。  相似文献   

9.
目的 为了构建人分泌型磷脂酶A2(secretary phospholipase A2, sPLA2-IIA) 的有效表达系统,本文从胎脾中提取总RNA,采用RT-PCR方法扩增出编码sPLA2-IIA的基因定向地克隆于硫氧环蛋白基因融合表达载体pET32a的TrxA基因3’末端,构建符合读码框的融合表达载体pET32a-sPLA2-IIA。37℃下经IPTG诱导,hsPLA2-IIA融合蛋白在大肠杆菌BL21(DE3)中获得高效表达,表达产物以包涵体的形式存在。包涵体经8M尿素溶解、复性后检测结果显示具有较高的催化活性并呈现剂量依赖关系。结论:以大肠杆菌为宿主,成功表达了hsPLA2-IIA蛋白,为进一步进行hsPLA2-IIA的大量生产和功能研究奠定了基础。  相似文献   

10.
目的:研究胃癌组织中HER2基因及P53蛋白表达情况,分析其对临床诊疗的意义与价值。方法:选取2013年7月到2015年7月我院确诊的胃癌患者110例,检测患者胃癌组织中的HER2蛋白表达与基因扩增,及P53蛋白的表达情况,分析HER2基因扩增与P53蛋白表达与病理关系间的关系。结果:HER2基因扩增率为17.3%(19/110),HER2蛋白表达率为42.7%(47/110),P53蛋白表达率为58.2%(64/110),其中HER2蛋白表达3+、2+者HER2基因的扩增比例分别为3/4、6/9与1+表达者的3/16比较具有统计学意义(P0.05);HER2基因扩增和P53蛋白表达与胃癌淋巴结转移以及浸润程度有关(P0.05);相关性分析显示:HER2基因扩增与P53蛋白表达具有正相关关系(P0.05)。结论:胃癌组织中HER2基因扩增和P53蛋白协同表达,促进胃癌浸润和淋巴结转移,对胃癌早期诊断具有重要参考价值。  相似文献   

11.
HER2胞外区基因的克隆及其在大肠杆菌中的可溶性表达   总被引:1,自引:0,他引:1  
采用反转录PCR和PCR方法分别克隆P185^HER2/neu胞外区基因和噬菌体M13K07g3p—N1结构域基因,然后将二偶联入pET-22b( )载体中,在大肠杆菌中进行融合表达。可溶性目的蛋白表达量占细菌可溶性表达产物总量的30%72右.并通过镍亲和层析纯化出目的蛋白。以上结果为从噬菌体抗体库中筛选抗P185^HER2/neu的抗体奠定了基础。  相似文献   

12.
为验证真核表达的携带绿色荧光的抗HER2单链抗体应用于临床诊断HER2阳性肿瘤细胞和病理组织的可靠性,构建融合基因Anti HER2 ScFv-GFP,重组入pFAST Bac HT A载体,在昆虫细胞Sf9中表达,以Ni2+-NTA亲和层析法纯化Anti HER2 ScFv-GFP融合蛋白,测定其浓度与纯度,将同浓度的纯化蛋白分别与3种乳腺癌细胞BT474、SKBR3和MCF7各混合12 h、24 h和48 h,分析其在不同时间段结合HER2阳性肿瘤细胞的稳定性。用纯化蛋白直接检测经抗原修复的乳腺癌病理组织,与免疫组织化学法检测结果对比。结果在昆虫细胞Sf9中可观察到明显绿色荧光,纯化的融合蛋白相对分子量约60 kDa,浓度为115.5μg/mL,纯度约97%,SKBR3和BT474鉴定为HER2阳性。结合12 h、24 h、48 h后其细胞表面均有明显绿色荧光,而HER2阴性MCF7被洗脱后无荧光,该抗体滴度为1:64,48 h内该荧光抗体仍具有稳定性。携带绿色荧光的融合抗体检测病理组织与IHC法的结果完全一致。表明成功表达的携带绿色荧光的抗HER2单链抗体可特异性检测HER2阳性乳腺癌细胞BT474和SKBR3,在HER2阳性肿瘤细胞和临床病理组织检测上具有应用前景。  相似文献   

13.
14.
Apigenin is a low toxicity and non-mutagenic phytopolyphenol and protein kinase inhibitor. It exhibits anti-proliferating effects on human breast cancer cells. Here we examined several human breast cancer cell lines having different levels of HER2/neu expression and found that apigenin exhibited potent growth-inhibitory activity in HER2/neu-overexpressing breast cancer cells but was much less effective for those cells expressing basal levels of HER2/neu. Induction of apoptosis was also observed in HER2/neu-overexpressing breast cancer cells in a dose- and time-dependent manner. However, the one or more molecular mechanisms of apigenin-induced apoptosis in HER2/neu-overexpressing breast cancer cells remained to be elucidated. A cell survival pathway involving phosphatidylinositol 3-kinase (PI3K), and Akt is known to play an important role in inhibiting apoptosis in response to HER2/neu-overexpressing breast cancer cells, which prompted us to investigate whether this pathway plays a role in apigenin-induced apoptosis in HER2/neu-overexpressing breast cancer cells. Our results showed that apigenin inhibits Akt function in tumor cells in a complex manner. First, apigenin directly inhibited the PI3K activity while indirectly inhibiting the Akt kinase activity. Second, inhibition of HER2/neu autophosphorylation and transphosphorylation resulting from depleting HER2/neu protein in vivo was also observed. In addition, apigenin inhibited Akt kinase activity by preventing the docking of PI3K to HER2/HER3 heterodimers. Therefore, we proposed that apigenin-induced cellular effects result from loss of HER2/neu and HER3 expression with subsequent inactivation of PI3K and AKT in cells that are dependent on this pathway for cell proliferation and inhibition of apoptosis. This implies that the inhibition of the HER2/HER3 heterodimer function provided an especially effective strategy for blocking the HER2/neu-mediated transformation of breast cancer cells. Our results also demonstrated that apigenin dissociated the complex of HER2/neu and GRP94 that preceded the depletion of HER2/neu. Apigenin-induced degradation of mature HER2/neu involves polyubiquitination of HER2/neu and subsequent hydrolysis by the proteasome.  相似文献   

15.
The human epidermal growth factor receptor 2/neuregulin (HER2/neu) receptor is overexpressed in highly malignant mammary and ovarian tumors and correlates with a poor prognosis. It is a target for therapy; humanized monoclonal antibodies to HER2 have led to increased survival of patients with HER2/neu-positive breast cancer. As a first step in the design of an oncolytic herpes simplex virus able to selectively infect HER2/neu-positive cells, we constructed two recombinants, R-LM11 and R-LM11L, that carry a single-chain antibody (scFv) against HER2 inserted at residue 24 of gD. The inserts were 247 or 256 amino acids long, and the size of the gD ectodomain was almost doubled by the insertion. We report the following. R-LM11 and R-LM11L infected derivatives of receptor-negative J or CHO cells that expressed HER2/neu as the sole receptor. Entry was dependent on HER2/neu, since it was inhibited in a dose-dependent manner by monoclonal antibodies to HER2/neu and by a soluble form of the receptor. The scFv insertion in gD disrupted the ability of the virus to enter cells through HVEM but maintained the ability to enter through nectin1. This report provides proof of principle that gD can tolerate fusion to a heterologous protein almost as large as the gD ectodomain itself without loss of profusion activity. Because the number of scFv's to a variety of receptors is continually increasing, this report makes possible the specific targeting of herpes simplex virus to a large collection of cell surface molecules for both oncolytic activity and visualization of tumor cells.  相似文献   

16.
Breast and ovarian cancer are two of the leading causes of cancer deaths among women in the United States. Overexpression of the HER2/neu oncoprotein has been reported in patients affected with breast and ovarian cancers, and is associated with poor prognosis. To develop a novel targeted therapy for HER2/neu expressing tumors, we have constructed a fully human IgE with the variable regions of the scFv C6MH3-B1 specific for HER2/neu. This antibody was expressed in murine myeloma cells and was properly assembled and secreted. The Fc region of this antibody triggers in vitro degranulation of rat basophilic cells expressing human FcεRI (RBL SX-38) in the presence of murine mammary carcinoma cells that express human HER2/neu (D2F2/E2), but not the shed (soluble) antigen (ECD(HER2)) alone. This IgE is also capable of inducing passive cutaneous anaphylaxis in a human FcεRIα transgenic mouse model, in the presence of a cross-linking antibody, but not in the presence of soluble ECD(HER2). Additionally, IgE enhances antigen presentation in human dendritic cells and facilitates cross-priming, suggesting that the antibody is able to stimulate a secondary T-cell anti-tumor response. Furthermore, we show that this IgE significantly prolongs survival of human FcεRIα transgenic mice bearing D2F2/E2 tumors. We also report that the anti-HER2/neu IgE is well tolerated in a preliminary study conducted in Macaca fascicularis (cynomolgus) monkeys. In summary, our results suggest that this IgE should be further explored as a potential therapeutic against HER2/neu overexpressing tumors, such as breast and ovarian cancers.  相似文献   

17.
The HER2/neu proto-oncogene encodes a 185-kDa trans-membrane glycoprotein kinase with extensive homology to the epidermal growth factor receptor and plays a key role in the transformation and growth of malignant tumors. To date, two antibody drugs targeting HER2/neu have been developed successfully. In order to reduce the cost and the time of clinical treatment, we produced a fusion protein composed of human beta defensin 2 (hBD2) and anti-HER2/neu single-chain variable fragment (scFv 4D5), which is capable of specifically targeting, significantly inhibiting, and promptly killing HER2/neu-positive cancer cells. The recombinant protein was expressed in Escherichia coli using the small ubiquitin-related modifier (SUMO) as the molecular chaperone, and the optimal expression level reached to 40.2 % of the total supernatant protein. After purifying by Ni-NTA affinity chromatography, the fusion protein was cleaved with a SUMO-specific protease to obtain hBD2–4D5, which was further purified by Ni-NTA affinity chromatography. The purity of hBD2–4D5 was higher than 95 %, and the yield was 19?±?2 mg/L in flask fermentation. The cell number count and flow cytometry results showed that hBD2–4D5 exerted cytotoxic and anti-proliferative effects on HER2/neu-positive breast cancer cell line, SKBR-3. The results of scanning electron microscope and transmission electron microscope observation indicated that hBD2–4D5 could induce intracellular ultrastructure changes and cell necrosis by disrupting the cell membrane. Immunofluorescence analysis showed that hBD2–4D5 could bind to SKBR-3 cells and further be internalized into the cytoplasm. Moreover, hBD2–4D5 could also mediate apoptosis of SKBR-3 cells by up-regulating the ratio of Bax to Bcl-2.  相似文献   

18.
BACKGROUND AND PURPOSE: Expression of the HER2/neu proto-oncogene, a receptor-like transmembrane protein expressed at low levels on some normal cells, is markedly increased in a subset of human breast, colon, lung, and ovarian cancers. A humanized HER2/neu antibody has been tested as a therapeutic agent in several clinical trials, with promising results. We have developed a family of anti-HER2/neu fusion proteins. To evaluate the immunologic efficacy of these proteins, it is critical that tumors expressing the target antigen can grow in immunologically intact mice. METHOD: To produce murine tumors expressing human HER2/neu on the surface, CT26, MC38, and EL4 murine cell lines were transduced by use of a retroviral construct containing the cDNA encoding the human HER2/neu gene. RESULTS: Histologic features and kinetics of tumor growth in subcutaneous space of the human HER2/neu-expressing cells were similar to those of the respective parental cell lines. Intravenous inoculation with these cells induced disseminated malignant disease. Flow cytometric and immmunohistochemical analyses of freshly isolated tumors revealed in vivo expression of human HER2/neu. Secretion of antigen was not detected by use of an ELISA. CONCLUSION: Although an antibody response against the human HER2/neu antigen was observed, this response does not affect the growth rate of the HER2/neu-expressing cells. These murine models may be useful tools for evaluation of anti-cancer therapeutic approaches that target human HER2/neu.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号