当前位置: X-MOL 学术J. Bone Oncol. › 论文详情
Our official English website, www.x-mol.net, welcomes your feedback! (Note: you will need to create a separate account there.)
The P2RX7B splice variant modulates osteosarcoma cell behaviour and metastatic properties
Journal of Bone Oncology ( IF 3.1 ) Pub Date : 2021-10-20 , DOI: 10.1016/j.jbo.2021.100398
Luke Tattersall 1 , Karan M Shah 1 , Darren L Lath 1 , Archana Singh 2 , Jennifer M Down 1 , Elena De Marchi 3 , Alex Williamson 1 , Francesco Di Virgilio 3 , Dominique Heymann 4 , Elena Adinolfi 3 , William D Fraser 5, 6 , Darrell Green 5 , Michelle A Lawson 1 , Alison Gartland 1
Affiliation  

Background

Osteosarcoma (OS) is the most common type of primary bone cancer affecting children and adolescents. OS has a high propensity to spread meaning the disease is often incurable and fatal. There have been no improvements in survival rates for decades. This highlights an urgent need for the development of novel therapeutic strategies. Here, we report in vitro and in vivo data that demonstrates the role of purinergic signalling, specifically, the B isoform of the purinergic receptor P2RX7 (P2RX7B), in OS progression and metastasis.

Methods

TE85 and MNNG-HOS OS cells were transfected with P2RX7B. These cell lines were then characterised and assessed for proliferation, cell adhesion, migration and invasion in vitro. We used these cells to perform both paratibial and tail vein injected mouse studies where the primary tumour, bone and lungs were analysed. We used RNA-seq to identify responsive pathways relating to P2RX7B.

Results

Our data shows that P2RX7B expression confers a survival advantage in TE85 + P2RX7B and MNNG-HOS + P2RX7B human OS cell lines in vitro that is minimised following treatment with A740003, a specific P2RX7 antagonist. P2RX7B expression reduced cell adhesion and P2RX7B activation promoted invasion and migration in vitro, demonstrating a metastatic phenotype. Using an in vivo OS xenograft model, MNNG-HOS + P2RX7B tumours exhibited cancer-associated ectopic bone formation that was abrogated with A740003 treatment. A pro-metastatic phenotype was further demonstrated in vivo as expression of P2RX7B in primary tumour cells increased the propensity of tumour cells to metastasise to the lungs. RNA-seq identified a novel gene axis, FN1/LOX/PDGFB/IGFBP3/BMP4, downregulated in response to A740003 treatment.

Conclusion

Our data illustrates a role for P2RX7B in OS tumour growth, progression and metastasis. We show that P2RX7B is a future therapeutic target in human OS.



中文翻译:

P2RX7B 剪接变体调节骨肉瘤细胞行为和转移特性

背景

骨肉瘤 (OS) 是影响儿童和青少年的最常见的原发性骨癌类型。OS 具有很高的传播倾向,这意味着该疾病通常是无法治愈和致命的。几十年来,存活率一直没有提高。这突显出迫切需要开发新的治疗策略。在这里,我们报告了证明嘌呤能信号传导的作用的体外体内数据,特别是嘌呤能受体 P2RX7 (P2RX7B) 的 B 亚型在 OS 进展和转移中的作用。

方法

用 P2RX7B 转染 TE85 和 MNNG-HOS OS 细胞。然后对这些细胞系进行表征并在体外评估增殖、细胞粘附、迁移和侵袭。我们使用这些细胞进行胫骨旁和尾静脉注射的小鼠研究,其中分析了原发性肿瘤、骨骼和肺。我们使用 RNA-seq 来识别与 P2RX7B 相关的响应途径。

结果

我们的数据显示,P2RX7B 表达在体外的 TE85 + P2RX7B 和 MNNG-HOS + P2RX7B 人 OS 细胞系中赋予生存优势,这种优势在用特异性 P2RX7 拮抗剂 A740003 治疗后最小化。P2RX7B 表达降低了细胞粘附,P2RX7B 激活促进了体外侵袭和迁移,证明了转移表型。使用体内OS 异种移植模型,MNNG-HOS + P2RX7B 肿瘤表现出与癌症相关的异位骨形成,而 A740003 治疗消除了这种异位骨形成。体内进一步证实了促转移表型,因为原发性肿瘤细胞中 P2RX7B 的表达增加了肿瘤细胞转移到肺部的倾向。RNA-seq 确定了一个新的基因轴,FN1 / LOX / PDGFB / IGFBP3 / BMP4,响应 A740003 处理而下调。

结论

我们的数据说明了 P2RX7B 在 OS 肿瘤生长、进展和转移中的作用。我们表明 P2RX7B 是人类 OS 的未来治疗靶点。

更新日期:2021-10-27
down
wechat
bug