低氧诱导因子HIF-1α调控CX3CR1在胰腺癌细胞中的表达

刘俊秀 赵天锁 贾玲玲 张圣杰 赵潇 孙军伟 张欢 任贺 郝继辉

刘俊秀, 赵天锁, 贾玲玲, 张圣杰, 赵潇, 孙军伟, 张欢, 任贺, 郝继辉. 低氧诱导因子HIF-1α调控CX3CR1在胰腺癌细胞中的表达[J]. 中国肿瘤临床, 2013, 40(4): 187-189, 194. doi: 10.3969/j.issn.1000-8179.2013.04.002
引用本文: 刘俊秀, 赵天锁, 贾玲玲, 张圣杰, 赵潇, 孙军伟, 张欢, 任贺, 郝继辉. 低氧诱导因子HIF-1α调控CX3CR1在胰腺癌细胞中的表达[J]. 中国肿瘤临床, 2013, 40(4): 187-189, 194. doi: 10.3969/j.issn.1000-8179.2013.04.002
Junxiu LIU, Tiansuo ZHAO, Lingling JIA, Shengjie ZHANG, Xiao ZHAO, Junwei SUN, Huan ZHANG, He REN, Jihui HAO. Regulation of CX3CR1 expression in pancreatic cancer cells by hypoxia inducible factor 1α[J]. CHINESE JOURNAL OF CLINICAL ONCOLOGY, 2013, 40(4): 187-189, 194. doi: 10.3969/j.issn.1000-8179.2013.04.002
Citation: Junxiu LIU, Tiansuo ZHAO, Lingling JIA, Shengjie ZHANG, Xiao ZHAO, Junwei SUN, Huan ZHANG, He REN, Jihui HAO. Regulation of CX3CR1 expression in pancreatic cancer cells by hypoxia inducible factor 1α[J]. CHINESE JOURNAL OF CLINICAL ONCOLOGY, 2013, 40(4): 187-189, 194. doi: 10.3969/j.issn.1000-8179.2013.04.002

低氧诱导因子HIF-1α调控CX3CR1在胰腺癌细胞中的表达

doi: 10.3969/j.issn.1000-8179.2013.04.002
基金项目: 

天津市自然科学基金 10JCZDJC 20200

详细信息
    通讯作者:

    郝继辉  jihuihao@yahoo.com

Regulation of CX3CR1 expression in pancreatic cancer cells by hypoxia inducible factor 1α

Funds: 

the Municipal Natural Science Foundation of Tianjin 10JCZDJC 20200

More Information
  • 摘要:   目的  以胰腺癌细胞株Patu8988为研究对象, 通过过表达和干扰低氧诱导因子(HIF-1α), 观察CX3CR1表达水平的变化, 并探讨CX3CR1在胰腺癌中的调控机制。  方法  分别构建pcDNA3.1-HIF-1α过表达质粒和HIF-1α-siRNA, 转染胰腺癌细胞株Patu8988, 经Western blot、半定量PCR检测CX3CR1的表达情况。采用染色质免疫沉淀(chromatin immunoprecipitation, ChIP)、荧光素酶技术探查HIF-1α与CX3CR1启动子区的结合情况。  结果  Patu8988转染pcDNA3.1-HIF-1α后CX3CR1的表达增加, 敲除HIF-1α后CX3CR1的表达减少。HIF-1α与CX3CR1启动子区的低氧反应元件直接结合, 并上调CX3CR1启动子的活性(P < 0.01)。  结论  HIF-1α调控CX3CR1在胰腺癌细胞中的表达。

     

  • 图  1  过表达和干扰HIF-1α前后CX3CR1的表达变化

    Figure  1.  Changes in CX3CR1 before and after pcDNA3.1 HIF-1α or siHIF-1α was overexpressed and transfected

    A: Patu8988 cells transfected with pcDNA3.1 HIF-1α plasmids (1 μg, 2 μg) and assessed by Western blot analysis after 48 h; B: Patu8988 cells transfected with pcDNA3.1 HIF-1 α plasmids (1 μ g, 2 μ g) and assessed by semi-quantitative PCR after 48 h; C: Patu8988 cells transfected with siHIF-1 α (50 nM) and assessed by Western blot analysis after 48 h; D: Patu8988 cells transfected with siHIF-1α (50 nM) and assessed by semi-quantitative PCR after 48 h

    图  2  HIF-1α直接结合CX3CR1启动子的HRE区并上调其活性

    Figure  2.  HIF-1αcould directly interact and upregulate CX3CR1 promoter activity

    ▶A: DNA sequence of the CX3CR1 promoter. Hypoxia-response elements were located at different sites; B: Chromatin immunoprecipitation analysis of Patu8988 cells. PCR products of the vascular endothelial growth factor promoter were used as the positive control sample. H, N, and NC indicate hypoxia, normoxia, and negative control, respectively; C: Luciferase analysis of Patu8988 cells. The cells were cotransfected with pGL3-CX3CR1 (1μg) and pcDNA3.1 HIF-1 α plasmids (1μg). Luciferase analysis was performed with a dual-luciferase reporter assay system. Y axis: pGL3-CX3CR1-related luciferase activity. **P < 0.01 versus the control group

  • [1] Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012[J]. CA Cancer J Clin, 2012, 62(1): 10-29. doi: 10.3322/caac.20138
    [2] Gao YJ, Ji RR. Chemokines, neuronal-glial interactions, and central processing of neuropathic pain[J]. Pharmacol Ther, 2010, 126 (1): 56-68. doi: 10.1016/j.pharmthera.2010.01.002
    [3] Zhao T, Gao S, Wang X, et al. Hypoxia-inducible factor-1α regulates chemotactic migration of pancreatic ductal adenocarcinoma cells through directly transactivating the CX3CR1 gene[J]. PLoS One, 2012, 7(8): e43399. doi: 10.1371/journal.pone.0043399
    [4] Marchesi F, Piemonti L, Fedele G, et al. The chemokine receptor CX3CR1 is involved in the neural tropism and malignant behavior of pancreatic ductal adenocarcinoma[J]. Cancer Res, 2008, 68(21): 9060-9069. doi: 10.1158/0008-5472.CAN-08-1810
    [5] Imai T, Hieshima K, Haskell C, et al. Identification and molecular characterization of fractalkine receptor CX3CR1, which mediates both leukocyte migration and adhesion[J]. Cell, 1997, 91(4): 521-530. doi: 10.1016/S0092-8674(00)80438-9
    [6] Liebig C, Ayala G, Wilks JA, et al. Perineural invasion in cancer: a review of the literature[J]. Cancer, 2009, 115(15): 3379-3391. doi: 10.1002/cncr.24396
    [7] Jamieson-Gladney WL, Zhang Y, Fong AM, et al. The chemokine receptor CX?CR1 is directly involved in the arrest of breast cancer cells to the skeleton[J]. Breast Cancer Res, 2011, 13(5): R91. doi: 10.1186/bcr3016
    [8] Bazan JF, Bacon KB, Hardiman G, et al. A new class of membrane-bound chemokine with a CX3C motif[J]. Nature, 1997, 385 (6617): 640-644. doi: 10.1038/385640a0
    [9] Pan Y, Lloyd C, Zhou H, et al. Neurotactin, a membrane-anchored chemokine upregulated in brain inflammation[J]. Nature, 1997, 387(6633): 611-617. doi: 10.1038/42491
    [10] Haskell CA, Cleary MD, Charo IF. Unique role of the chemokine domain of fractalkine in cell capture. Kinetics of receptor dissociation correlate with cell adhesion[J]. J Biol Chem, 2000, 275(44): 34183-34189. doi: 10.1074/jbc.M005731200
    [11] Marchesi F, Locatelli M, Solinas G, et al. Role of CX3CR1/ CX3CL1 axis in primary and secondary involvement of the nervous system by cancer[J]. J Neuroimmunol, 2010, 224(1-2): 39-44. doi: 10.1016/j.jneuroim.2010.05.007
    [12] Cardona AE, Pioro EP, Sasse ME, et al. Control of microglial neurotoxicity by the fractalkine receptor[J]. Nat Neurosci, 2006, 9(7): 917-924. doi: 10.1038/nn1715
    [13] Marchesi F, Locatelli M, Solinas G, et al. Role of CX3CR1/ CX3CL1 axis in primary and secondary involvement of the nervous system by cancer[J]. J Neuroimmunol, 2010, 224(1-2): 39-44. doi: 10.1016/j.jneuroim.2010.05.007
    [14] Mimura I, Tanaka T, Wada Y, et al. Pathophysiological response to hypoxia - from the molecular mechanisms of malady to drug discovery: epigenetic regulation of the hypoxic response via hypoxia-inducible factor and histone modifying enzymes[J]. J Pharmacol Sci, 2011, 115(4): 453-458. doi: 10.1254/jphs.10R19FM
    [15] Semenza GL. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics[J]. Oncogene, 2010, 29(5): 625-634. doi: 10.1038/onc.2009.441
  • 加载中
图(2)
计量
  • 文章访问数:  46
  • HTML全文浏览量:  2
  • PDF下载量:  1
  • 被引次数: 0
出版历程
  • 收稿日期:  2012-10-08
  • 修回日期:  2012-12-20

目录

    /

    返回文章
    返回