...
首页> 外文期刊>American Journal of Clinical and Experimental Urology >Platelet TSP-1 controls prostate cancer-induced osteoclast differentiation and bone marrow-derived cell mobilization through TGFβ-1
【24h】

Platelet TSP-1 controls prostate cancer-induced osteoclast differentiation and bone marrow-derived cell mobilization through TGFβ-1

机译:血小板TSP-1通过TGFβ-1控制前列腺癌诱导的骨髓细胞分化和骨髓衍生细胞培养

获取原文

摘要

The development of distant metastasis is the leading cause of prostate cancer (CaP)-related death, with the skeleton being the primary site of metastasis. While the progression of primary tumors and the growth of bone metastatic tumors are well described, the mechanisms controlling pre-metastatic niche formation and homing of CaP to bone remain unclear. Through prior studies, we demonstrated that platelet secretion was required for ongoing tumor growth and pre-metastatic tumor-induced bone formation. Platelets stimulated bone marrow-derived cell (BMDC) mobilization to tumors supporting angiogenesis. We hypothesized that proteins released by the platelet α granules were responsible for inducing changes in the pre-metastatic bone niche. We found that the classically anti-angiogenic protein thrombospondin (TSP)-1 was significantly increased in the platelets of mice with RM1 murine CaP tumors. To determine the role of increased TSP-1, we implanted tumors in TSP-1 null animals and assessed changes in tumor growth and pre-metastatic niche. TSP-1 loss resulted in increased tumor size and enhanced angiogenesis by immunohistochemistry. Conversely, TSP-1 deletion reduced BMDC mobilization and enhanced osteoclast formation resulting in decreased tumor-induced bone formation as measured by microcomputed tomography. We hypothesized that changes in the pre-metastatic niche were due to the retention of TGF-β1 in the platelets of mice after TSP-1 deletion. To assess the importance of platelet-derived TGF-β1, we implanted RM1 CaP tumors in mice with platelet factor 4-driven deletion of TGF-β1 in platelets and megakaryocytes. Like TSP-1 deletion, loss of platelet TGF-β1 resulted in increased angiogenesis with a milder effect on tumor size and BMDC release. Within the bone microenvironment, platelet TGF-β1 deletion prevented tumor-induced bone formation due to increased osteoclastogenesis. Thus, we demonstrate that the TSP-1/TGF-β1 axis regulates pre-metastatic niche formation and tumor-induced bone turnover. Targeting the platelet release of TSP-1 or TGF-β1 represents a potential method to interfere with the process of CaP metastasis to bone.
机译:远处转移的发展是前列腺癌(帽子)的致命原因 - 相关死亡,骨骼是转移的主要部位。虽然原发性肿瘤的进展和骨转移肿瘤的生长良好描述,但控制覆盖帽的前转移性Niche形成和归巢的机制仍不清楚。通过先前的研究,我们证明了持续肿瘤生长和预转移性肿瘤诱导的骨形成所需的血小板分泌。血小板刺激骨髓衍生的细胞(BMDC)调动支持血管生成的肿瘤。我们假设血小板α颗粒释放的蛋白质负责诱导转移性骨骼的变化。我们发现,具有RM1鼠帽肿瘤的小鼠血小板中,典型的抗血管生成蛋白血栓样蛋白(TSP)-1显着增加。为了确定增加的TSP-1的作用,我们植入TSP-1零毒动动物中的肿瘤,并评估肿瘤生长和转移性乳蛋白的变化。 TSP-1损失导致免疫组织化学增加肿瘤大小和增强的血管生成。相反,TSP-1缺失降低了BMDC动员和增强的破骨细胞形成,导致肿瘤诱导的骨形成减少,如微仿性断层扫描测量的。我们假设预转移性地基的变化是由于TSP-1缺失后小鼠血小板中的TGF-β1。为了评估血小板衍生的TGF-β1的重要性,我们在小鼠中植入小鼠的RM1帽肿瘤,血小板和巨粒细胞中的TGF-β1缺失。与TSP-1删除一样,血小板TGF-β1的损失导致血管生成增加,对肿瘤大小和BMDC释放的影响较高。在骨髓微环境中,血小板TGF-β1缺失防止肿瘤诱导的骨形成由于骨细胞发生增加。因此,我们证明了TSP-1 / TGF-β1轴调节转移性的乳头形成和肿瘤诱导的骨质周转。靶向TSP-1或TGF-β1的血小板释放代表潜在的方法,以干扰帽转移到骨的过程。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号