期刊文献+

热CO_2气腹对结肠癌细胞的增殖抑制作用及其机制 被引量:5

Anti-proliferative effect of hyperthermic CO_2 pneumoperitoneum on colonic cancer cells and its mechanism of action
下载PDF
导出
摘要 目的:探讨热CO2气腹对结肠癌细胞的增殖抑制作用及作用机制,明确其用于结直肠癌腹膜转移治疗的可行性。方法:建立热CO2气腹体外实验模型,热CO2气腹(43℃,2~4h)作用于结肠癌细胞株COLO205细胞。以WST-8法检测细胞增殖,Annexin-V/PI流式细胞术及透射电镜检测细胞死亡,流式细胞术检测细胞周期。结果:热CO2气腹(43℃,2~4h)对结肠癌细胞有显著的增殖抑制作用,热CO2显著诱导细胞凋亡及G1期细胞阻滞。结论:热CO2气腹通过诱导细胞凋亡及G1期细胞周期阻滞显著抑制结肠癌细胞的增殖,热CO2气腹具有应用于结直肠癌腹膜种植转移治疗的潜能。 Objective To evaluate the anti-proliferative effect of hyperthermic CO2 pneumoperitoneum (HT-CO2) on colonic cancer cells, to define its mechanism of action, and to assess the feasibility as a new therapeutic procedure for peritoneal carcinomatosis in colon cancer. Methods Based on an in vitro HT-CO2 study model, the cell proliferation was determined with WST-8 assay after treatment by HT-CO2 at 43℃ for 24 hours. Apoptosis was assessed by Annexin-V/PI flow cytometry and transmission electron microscopy. Cell cycle was detected by PI flow cytometry. Results HT-CO2 produced in significant anti-proliferative effect on colonic cancer cells. HT-CO2 sensibly induced apoptosis and G1 phrase cell cycle arrest. Conclusions HT-CO2 can significantly inhibit the proliferation of colonic cancer cells, and the anti- proliferation is attributable to induction of apoptosis and G1 phrase cell cycle arrest. HT-CO2 may potentially serve as a therapeutic procedure for the management of peritoneal carcinomatosis caused by colon cancer.
出处 《外科理论与实践》 2007年第5期455-459,共5页 Journal of Surgery Concepts & Practice
基金 上海市市级医院新兴前沿技术联合攻关项目(SHDC12006102) 上海交通大学医学院博士创新基金(BXJ0713)
关键词 结肠肿瘤 气腹 人工 二氧化碳 细胞周期 Colonic neoplasms Pneumoperitoneum, artificial Carbon dioxide Cell cycle
  • 相关文献

参考文献23

  • 1Knorr C, Reingruber B, Meyer T, et al. Peritoneal carcinomatosis of colorectal cancer: incidence, prognosis, and treatment modalities[J]. Int J Colorectal Dis, 2004, 19(3): 181-187.
  • 2Koppe M J, Boerman OC, Oyen WJ, et al. Peritoneal carcinomatosis of colorectal origin: incidence and current treatment strategies[J]. Ann Surg, 2006, 243(2):212-222.
  • 3Verwaal V J, van Ruth S, de Bree E, et al. Randomized trial of cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy and pallia tive surgery in patients with peritoneal carcinomatosis of colorectal cancer[J]. J Clin Oncol, 2003,21 (20):3737-3743.
  • 4Glehen O, Mohamed F, Gilly FN. Peritoneal carcinomatosis from digestive tract cancer: new management by cytoreductive surgery and intraperitoneal chemohyperthermia[J].Lancet Oncol, 2004,5(4):219-228.
  • 5Glehen O, Kwiatkowski F, Sugarbaker PH, et al. Cytoreductive surgery combined with perioperative intraperitoneal chemotherapy for the management of peritoneal carcinomatosis from colorectal cancer: a multi-institutional study[J]. J Clin Oncol,20(M, 22(16):3284-3292.
  • 6Hildebrandt B, Wust P, Ahlers O, et al. The cellular and molecular basis of hyperthermia [J]. Crit Rev Oncol Hematol, 2002, 43(1):33-56.
  • 7O' Neill KL, Fairbairn DW, Smith M J, et al. Critical parameters influencing hyperthermia-induced apoptosis in human lymphoid cell lines[J]. Apoptosis, 1998,3(5):369-375.
  • 8Vorotnikova E, Ivkov R, Foreman A, et al. The magnitude and time-dependence of the apoptotic response of normal and malignant cells subjected to ionizing radiation versus hyperthermia[J].Int J Radiat Biol,2006, 82(8):549-559.
  • 9Sturm I, Rau B, Schlag PM, et al. Genetic dissection of apoptosis and cell cycle control in response of colorectal cancer treated with preoperative radiochemotherapy [J]. BMC Cancer. 2006. 6:124.
  • 10Rong Y, Mack P. Apoptosis induced by hyperthermia in Dunn osteosarcoma cell line in vitro[J]. Int J Hyperthermia,2000,16(1):19-27.

同被引文献30

引证文献5

二级引证文献8

相关作者

内容加载中请稍等...

相关机构

内容加载中请稍等...

相关主题

内容加载中请稍等...

浏览历史

内容加载中请稍等...
;
使用帮助 返回顶部