欢迎来到《四川大学学报(医学版)》
王齐, 王一涵, 黎鹰, 等. 基于黑猩猩腺病毒6型的新型溶瘤病毒抑瘤研究[J]. 四川大学学报(医学版), 2022, 53(1): 71-76. DOI: 10.12182/20220160504
引用本文: 王齐, 王一涵, 黎鹰, 等. 基于黑猩猩腺病毒6型的新型溶瘤病毒抑瘤研究[J]. 四川大学学报(医学版), 2022, 53(1): 71-76. DOI: 10.12182/20220160504
WANG Qi, WANG Yi-han, LI Ying, et al. Anti-Tumor Effect of a Novel Oncolytic Virus Based on Chimpanzee Adenovirus Type 6[J]. Journal of Sichuan University (Medical Sciences), 2022, 53(1): 71-76. DOI: 10.12182/20220160504
Citation: WANG Qi, WANG Yi-han, LI Ying, et al. Anti-Tumor Effect of a Novel Oncolytic Virus Based on Chimpanzee Adenovirus Type 6[J]. Journal of Sichuan University (Medical Sciences), 2022, 53(1): 71-76. DOI: 10.12182/20220160504

基于黑猩猩腺病毒6型的新型溶瘤病毒抑瘤研究

Anti-Tumor Effect of a Novel Oncolytic Virus Based on Chimpanzee Adenovirus Type 6

  • 摘要:
      目的   以6型黑猩猩腺病毒(AdC6)为载体,构建一组新型溶瘤腺病毒,使之获得瘤内特异性复制能力。在体内外检测其抑瘤效果,并探讨其溶瘤机制。
      方法  以AdC6载体为基础,以人端粒酶逆转录酶(hTERT)启动子驱动该腺病毒的复制相关基因E1A表达,获得重组溶瘤病毒AdC6-htertE1A-ΔE3,同源构建表达粒细胞-巨噬细胞集落刺激因子(GM-CSF/CSF2)的AdC6-htertE1A-ΔE3(CSF2)、复制缺陷型的腺病毒AdC6-ΔE1-ΔE3。重组腺病毒在HEK293细胞中包装,纯化后进行酶切鉴定。以上述3种腺病毒感染不同种类的肿瘤细胞(RD、SW-620、HeLa、Huh7、RM-1与MC-38),24 h后Western blot检测CSF2的表达,72 h后CCK8实验检测肿瘤细胞的存活率。以上述3种腺病毒感染HeLa细胞,12 h、24 h、48 h后以Western blot检测凋亡信号通路蛋白表达水平。在C57BL/6小鼠背部皮下注射含有1×106个小鼠结肠癌MC38细胞或小鼠前列腺癌RM-1细胞的细胞悬液,建立2种荷瘤小鼠模型实验,分为4组,每组分别瘤内注射50 μL的PBS、1×108 PFU的AdC6-ΔE1-ΔE3、AdC6-htertE1A-ΔE3、AdC6-htertE1A-ΔE3(CSF2)。当PBS组荷瘤小鼠的肿瘤达到2 500 mm3以上时统一处死小鼠,取肿瘤组织进行TUNEL染色,在显微镜下观察凋亡阳性细胞并计数。
      结果  酶切鉴定发现,成功构建了溶瘤病毒AdC6-htertE1A-ΔE3、AdC6-htertE1A-ΔE3(CSF2)及AdC6-ΔE1-ΔE3。Western blot检测发现,AdC6-htertE1A-ΔE3(CSF2)能够感染不同的肿瘤细胞,并稳定表达外源基因CSF2。CCK8实验结果表明AdC6-htertE1A-ΔE3和AdC6-htertE1A-ΔE3(CSF2)对RD、SW-620、HeLa、Huh7、RM-1与MC-38等多种肿瘤细胞具有明显的杀伤效果;相较于复制缺陷的腺病毒AdC6-ΔE1-ΔE3,感染复数为100 MOI时的AdC6-htertE1A-ΔE3、AdC6-htertE1A-ΔE3(CSF2)对肿瘤细胞的杀伤效果极为明显(P<0.05)。Western blot实验证明AdC6-htertE1A-ΔE3和AdC6-htertE1A-ΔE3(CSF2)通过激活P53依赖的通路,诱导肿瘤细胞凋亡。在前列腺癌及结直肠癌荷瘤小鼠模型中注射溶瘤病毒可显著抑制肿瘤生长,甚至清除肿瘤。
      结论  以AdC6为载体的溶瘤病毒通过促凋亡机制在体内外杀伤肿瘤,具有治疗肿瘤的巨大潜力。

     

    Abstract:
      Objective  To construct, with chimpanzee adenovirus serotype 6 (AdC6) as the vector, a novel oncolytic adenovirus, enabling it to selectively replicate intratumorally, to test its tumor suppressive effect in vitro and in vivo, and to study its oncolytic mechanism.
      Methods  Based on the AdC6 vector, the human telomerase reverse transcriptase (hTERT) promoter was used to drive the expression of E1A, the adenovirus replication-related gene, and the recombinant oncolytic virus AdC6-htertΔE1A-ΔE3 was thus obtained. The oncolytic virus AdC6-htertE1A-ΔE3 (CSF2) expressing granulocyte macrophage colony-stimulating factor (GM-CSF/CSF2) and replication-deficient adenovirus AdC6-ΔE1-ΔE3 were constructed by homologous recombination, respectively. The recombinant adenovirus was packaged in HEK293 cells, purified and then identified with restriction enzyme digestion. Different types of tumor cells, including RD, SW-620, HeLa, Huh7, RM-1 and MC-38 were infected with the three adenoviruses. Twenty-four hours after infection, Western blot was used to determine the expression of CSF2 24 hours after infection. CCK8 assay was used to determine the survival rate of tumor cells 72 hours after infection. HeLa cells were infected with the three adenoviruses, and the expression levels of apoptosis signaling pathway proteins were examined with Western blot at 12 h, 24 h, and 48 h. C57BL/6 mice were subcutaneously injected with cell suspension containing 1×106 MC38 murine colon cancer cells and RM-1 murine prostate cancer cells to construct two tumor-bearing mice models. The tumor-bearing mice were divided into 4 groups, receiving intratumoral injection of 50 μL of PBS, AdC6-ΔE1-ΔE3 (1×108 PFU), AdC6-htertE1A-ΔE3 (1×108 PFU), and AdC6-htertE1A-ΔE3 (CSF2) (1×108 PFU), respectively. When the tumor size of PBS group reached 2 500 mm3, all the mice were sacrificed and the tumor tissue was collected for TUNEL staining. Then, apoptosis-positive cells were observed and counted under a microscope.
      Results  Restriction digestion revealed that the oncolytic viruses AdC6-htertE1A-ΔE3, AdC6-htertE1A-ΔE3 (CSF2) and AdC6-ΔE1-ΔE3 were successfully constructed. Western blot confirmed that AdC6-htertE1A-ΔE3 (CSF2) could infect different tumor cells and stably express CSF2, the exogenous gene. CCK8 results showed that AdC6-htertE1A-ΔE3 and AdC6-htertE1A-ΔE3 (CSF2) had obvious killing effects on RD, SW-620, HeLa, Huh7, RM-1and MC-38. Compared with the replication-deficient adenovirus AdC6-ΔE1-ΔE3, AdC6-htertE1A-ΔE3 and AdC6-htertE1A-ΔE3 (CSF2) at a multiplicity of infection of 100 MOI had extremely obvious killing effects on tumor cells (P<0.05). Western blot showed that AdC6-htertE1A-ΔE3 and AdC6-htertE1A-ΔE3 (CSF2) induced tumor cell apoptosis by activating the P53-dependent pathway. Injection of oncolytic virus in tumor-bearing mouse models of prostate cancer and colorectal cancer could significantly inhibit the tumor growth and even clear the tumor.
      Conclusion  Oncolytic virus based on AdC6 could eliminate tumor in vivo and in vitro through mechanisms that induced apoptosis, showing great potential for the treatment of tumors.

     

© 2022 《四川大学学报(医学版)》编辑部 版权所有 cc

开放获取 本文遵循知识共享署名—非商业性使用4.0国际许可协议(CC BY-NC 4.0),允许第三方对本刊发表的论文自由共享(即在任何媒介以任何形式复制、发行原文)、演绎(即修改、转换或以原文为基础进行创作),必须给出适当的署名,提供指向本文许可协议的链接,同时标明是否对原文作了修改;不得将本文用于商业目的。CC BY-NC 4.0许可协议详情请访问 https://creativecommons.org/licenses/by-nc/4.0

/

返回文章
返回