...
首页> 外文期刊>Cancer Management and Research >Mechanical ventilation promotes lung metastasis in experimental 4T1 breast cancer lung-metastasized models
【24h】

Mechanical ventilation promotes lung metastasis in experimental 4T1 breast cancer lung-metastasized models

机译:机械通风在实验4T1乳腺癌肺转移模型中促进肺转移

获取原文

摘要

Background/purpose: The aim of this study was to test the hypothesis that mechanical ventilation (MV) during cancer surgery induces lung stroma/tissue milieu changes, creating a favorable microenvironment for postoperative lung metastatic tumor establishment. Materials and methods: In Protocol A, female BALB/c mice were divided into an MV group and a control (no MV) group, both of which were anesthetized and subjected to intravenous injection of green fluorescent protein (GFP)-labeled mouse mammary carcinoma cell line (4T1) cells. After 24 h, the lung tissue was removed and the number of GFP-labeled 4T1 cells was calculated. In Protocol B, the clinically relevant mouse model of spontaneous breast cancer lung metastasis was used with surgical resection of the primary tumor to investigate the MV event that dictates postoperative lung metastasis. Female BALB/c mice were inoculated in the mammary fat pad with 4T1 cells. After 14-d growth, mice were anesthetized and divided into an MV group and a control (no MV) group during surgical procedures (mastectomy). Metastatic tumor burden was assessed two weeks after mastectomy by both macroscopic metastatic nodule count, hematoxylin–eosin histology, immunohistochemistry for the macrophage marker (CD68), and epithelial cell adhesion molecule (EpCAM). Results: MV was associated with a significant increase in the number of circulating breast tumor cells (GFP-labeled 4T1 cells) remaining in the microvasculature of the lung ( P <0.01). Immunohistochemical results showed increased infiltration of CD68-positive macrophages within injured lung parenchyma and metastatic tumor as well as increased expression of EpCAM in metastatic nodules. Postoperative metastases were more prevalent in the mechanically ventilated mice group compared to the non-ventilated group ( P <0.05). Conclusion: MV-induced lung metastasis occurs by attracting circulating tumor cells to the site of the lung injury and by accelerating the proliferation of preexisting micro-metastases in the lung. These observations indicate that the metastasis-enhancing effect of MV should be considered in general anesthesia during cancer surgery.
机译:背景/目的:本研究的目的是测试癌症手术期间机械通气(MV)的假设诱导肺基质/组织Milieu的变化,为术后肺转移性肿瘤建立产生良好的微环境。材料和方法:在方案A中,将雌性BALB / C小鼠分为MV组和对照(NO MV)组,两者被麻醉并进行静脉内注射绿色荧光蛋白(GFP) - 标记的小鼠乳腺癌细胞系(4T1)细胞。 24小时后,除去肺组织,计算GFP标记的4T1细胞的数量。在协议B中,使用临床相关的乳腺癌肺转移模型与原发性肿瘤的手术切除使用,以研究术后肺转移的MV事件。雌性BALB / C小鼠接种在乳腺脂肪垫中,含有4T1细胞。在14-D生长后,在手术程序(乳房切除术)期间麻醉并分成MV组和对照(NO MV)组的小鼠。通过宏观转移性结节计数,血毒性标记(CD68)的免疫组织化学和上皮细胞粘附分子(EPCAM),通过宏观转移性结节计数,血毒转移性结节计数,血毒性转移性结节计数,免疫组织化学评估转移性肿瘤负担。结果:MV与肺部微血管结构中剩余的循环乳腺肿瘤细胞(GFP标记的4T1细胞)的数量显着增加(P <0.01)。免疫组织化学结果表明,受损肺实质和转移性肿瘤内CD68阳性巨噬细胞的渗透增加,以及EPCAM在转移结节中的表达增加。与非通风组相比,机械通风小鼠在机械通风小鼠中更普遍(P <0.05)。结论:通过将循环肿瘤细胞吸引到肺损伤的部位,并加速肺中预先存在的微转移的增殖,发生MV诱导的肺转移。这些观察结果表明,在癌症手术期间,在全身麻醉中应考虑mV的转移增强作用。

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