用户名: 密码: 验证码:
转铁蛋白受体介导Tf-PEI-shRNA complex内吞靶向沉默缺氧诱导因子-1α抑制恶性黑素瘤生长的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
第一部分HIF-1α及其下游靶基因VEGF和GLUT-1在黑素瘤的表达
     目的研究缺氧诱导因子-1α(HIF-1α)、血管内皮生长因子(VEGF)和葡萄糖转运体-1(GLUT-1)在恶性黑素瘤组织和恶性黑素瘤细胞系A375,A875,KZ28中的表达,探讨HIF-1α作为黑素瘤基因治疗靶点的可能性。
     方法①应用免疫组织化学方法检测77例恶性黑素瘤组织HIF-1α、VEGF、GLUT-1的表达,分析其表达的相关性以及与临床病理关系,并且以20例色素痣组织作为对照研究。②应用免疫组织化学方法、Western blot方法、ELISA方法对缺氧条件下培养的A375,A875,KZ28中HIF-1α、VEGF、GLUT-1进行检测,并且以常氧条件下培养的上述细胞系作对照。
     结果①在77例恶性黑素瘤组织中,65例表达HIF-1α,55例表达VEGF,53例表达GLUT-1,而在20例色素痣组织2例表达HIF-1α、4例表达VEGF、3例表达GLUT-1;②在恶性黑素瘤组织中,HIF-1α与VEGF、GLUT-1的表达呈正相关;③在恶性黑素瘤组织中HIF-1α与VEGF、GLUT-1的表达与肿瘤的恶性程度相关。④HIF-1α、VEGF、GLUT-1在缺氧培养条件下的A375,A875,kZ28中表达明显高于上述三株细胞系在常氧培养条件下的表达。⑤分泌型的VEGF和膜型表达的GLUT-1随缺氧时间延长而增加,与HIF-1α表达正相关。
     结论①HIF-1α、VEGF、GLUT-1可作为区别良、恶性黑素细胞肿瘤的重要指标;②在黑素瘤中HIF-1α的表达影响其下游基因VEGF、GLUT-1的表达③HIF-1α是黑素瘤基因治疗的理想靶点。
     第二部分HIF-1α-shRNAs表达载体的构建及鉴定
     目的构建针对HIF-1α的发卡样shRNA真核表达载体HIF-1α-shRNA,并转染恶性黑素瘤细胞,证实其在体外培养恶性黑素瘤细胞中对HIF-1α的干扰作用及引起的效应。
     方法①人工合成三对互补并编码相应短发夹状HIF-1α-shRNAs的寡核苷酸链,将其插入到Pgenesil-1载体中,经酶切和测序鉴定所构建的重组体是否正确;②采用实时定量逆转录聚合酶链反应(real time RT-PCR)、Western blot分别检测转染细胞HIF-1αmRNA和蛋白的表达变化。对其下游基因分泌型VEGF和膜型GLUT-1蛋白表达的变化分别采用ELISA和流式细胞仪检测技术;③在人恶性黑素瘤细胞系A3758中,用G418筛选沉默效果最佳的HIF-1α-shRNAs的细胞,用FCM检测GFP的表达鉴定稳定转染的单克隆细胞系;④上述稳转细胞系在缺氧情况下培养,观察其凋亡和增殖变化。
     结果①经酶切和测序证明HIF-1α-shRNAs序列正确;②三株MM细胞系中转染HIF-1α-shRNAs载体后,shRNA1沉默效果最佳,HIF-1αmRNA和蛋白表达均较对照组明显下降(P<0.01),并在三株MM细胞系中具有平行作用,同时也可以下调VEGF和GLUT-1的表达。③荧光显微镜下观察到了G418筛选稳定表达shRNA1的A375细胞,FCM检测GFP表达均在98%以上;④稳定表达shRNA1的A375细胞在缺氧条件培养可以引起细胞凋亡和抑制细胞增殖。
     结论测序结果表明发卡样的HIF-1α-shRNA真核表达载体构建成功,转染A375、A875、KZ28细胞后获得稳定表达,并可特异性封闭HIF-1α的表达,下调VEGF和GLUT-1的表达,筛选和鉴定出稳定转染单克隆细胞系A375在缺氧条件下可以引起细胞凋亡和抑制细胞增殖,为进一步研究HIF-1α-shRNA载体在恶性黑素瘤治疗中的作用提供了实验基础。
     第三部分靶向分子TfR在恶性黒素瘤表达
     目的检测基因治疗靶向分子TfR在恶性黑素瘤组织和细胞系A375、A875、KZ28的表达,以明确TfR作为基因治疗的靶向分子的可能性。
     方法①应用免疫组织化学方法检测77例恶性黑素瘤组织TfR的表达,并且以20例色素痣组织作为对照研究。②应用免疫组织化学方法、western blot方法、流式细胞仪检测方法对常氧和缺氧条件下培养的A375,A875,KZ28中TfR表达进行检测,并且以已知低表达TfR的卵巢癌细胞系A2780进行对照。③分析在体外常氧和缺氧条件下培养的MM细胞株中TfR表达不同的可能机制。
     结果①在77例恶性黑素瘤组织中,70例表达TfR,而在色素痣组织只有5例表达;②在恶性黑素瘤组织中,TfR与肿瘤的病理分期相关;③在常氧培养的A375,A875,KZ28中TfR的表达分别为97%,89%,79%,A2780的表达为2%;④免疫组织化学方法、western blot方法检测恶性黑素瘤组织中TfR的表达与流式结果趋势相同。⑤缺氧在A375细胞株中对TfR的表达呈双相作用,早期表达减少,晚期表达增加。⑥缺氧对A375细胞株中对TfR的表达呈双相作用可能与缺氧早期促进凋亡,晚期引起细胞增殖有关。
     结论①TfR在恶性黑素瘤组织和细胞系A375,A875,KZ28明显高表达,是理想的黑素瘤基因治疗的靶向分子。②缺氧对黑素瘤细胞系TfR的表达呈双相作用,对我们个体化的利用TfR作为靶向分子提供了更加合理的理论依据。
     第四部分Tf-PEI-HIF-1α-shRNA complex对恶性黑素瘤治疗靶向性实验研究
     目的研究Tf-PEI-HIF-1α-shRNA complex在TfR高表达的黑素瘤细胞系A375以及在A375荷瘤鼠的体内模型的靶向分布,以期为下一步系统使用Tf-PEI-HIF-1α-shRNA complex治疗A375荷瘤鼠提供理论依据和实验数据。
     方法①应用FCM方法检测转染不同实验组的GFP的表达,以TfR低表达的A2780细胞系做对照。②应用Real time RT-PCR、western blot方法检测Tf-PEI-HIF-1α- shRNA complex系统治疗的A375荷瘤鼠和A2780荷瘤鼠中肿瘤组织以及重要脏器GFP的表达。
     结果①在体外实验中,A375组中,Tf-PEI-HIF-1α-shRNA complex转染后GFP表达到达65%,与脂质体转染相当,在A2780组中,Tf-PEI-HIF-1α-shRNA complex转染后均在5%以下。②在体内实验中,Tf-PEI-HIF-1α-shRNA complex系统治疗的A375荷瘤鼠的肿瘤组织较重要脏器肝、脾、心、肾GFP的mRNA及蛋白质具有明显差异(P<0.01),在对照组A2780荷瘤鼠中肿瘤组织与重要脏器肝、脾、心、肾GFP的mRNA及蛋白质没有明显表达差异(P>0.05)。
     结论①Tf-PEI-HIF-1α-shRNA complex靶向治疗在TfR高表达的黑素瘤细胞系A375有特异性靶向分布,并且转染效率高。②在TfR高表达的黑素瘤细胞系A375荷瘤鼠中,Tf-PEI-HIF-1α-shRNA complex系统靶向治疗具有肿瘤靶向特异性,为黑素瘤基因治疗研究提供了实验依据。
     第五部分Tf-PEI-HIF-1α-shRNA complex对恶性黑素瘤荷瘤鼠生物学行为影响的实验研究
     目的观察Tf-PEI-HIF-1α-shRNA complex系统治疗对A375荷瘤鼠的生长抑制及可能的机制进行探讨。
     方法①分别对接受Tf-PEI-HIF-1α-shRNA complex系统治疗的A375、A2780荷瘤鼠肿瘤大小进行测量,按公式(L×W2)/2计算肿瘤体积大小。②治疗结束后处死荷瘤鼠,采用免疫组化和western blot方法检测HIF-1α、VEGF、GLUT1的表达情况。③治疗结束后处死荷瘤鼠,采用免疫组化方法检测MVD;酶显色法检测肿瘤组织糖酵解产物乳酸含量。末端脱氧核苷酸转移酶标记法(TUNEL)定量检测两组裸鼠模型的肿瘤组织凋亡;
     结果①体内实验证明,实验组肿瘤生长速度也明显减慢(P<0.01)②实验组HIF-1α、VEGF、GLUT1表达明显低于对照组(P<0.01)③实验组和对照组MVD无显著性差异(P>0.05)。④实验组肿瘤组织匀浆的乳酸含量明显低于对照组(P<0.01)。⑤实验组可见大量凋亡细胞,对照组仅见少许凋亡细胞,实验组凋亡指数与对照组相比有显著性差异(P<0.01)。
     结论①通过Tf-PEI-HIF-1α-shRNA complex靶向RNA干扰技术不但能阻断HIF-1α的表达,还能沉默其下游靶基因VEGF、GLUT1的表达,从而在体内可显著性抑制恶性黑素瘤生长,为恶性黑素瘤基因治疗研究提供了实验依据。②Tf-PEI-HIF-1α-shRNA complex抑制恶性黑素瘤生长,可能与抑制糖酵解,促进肿瘤组织凋亡有关,未发现与抑制肿瘤血管生成有关。
PartⅠExpressions of HIF-1αand its downstream genes VEGF and GLUT-1 in Malignant Melanoma
     Objectives To study expressions of HIF-1α,VEGF and GLUT-1 and their correlation to tumor clinical-pathology features in human malignant melanoma (MM) and MM cell lines. To analyze the possiblity of HIF-1αbeing an attractive therapeutic molecular target for MM
     Methods①Tissue sections from 77 MM patients were examined using immunohistochemical technique and morphological quantitative analysis for the protein expressions of HIF-1α,VEGF and GLUT-1 and their corelations to tumor clinical-pathology features in MM were analyzed. And tissue sections from 20 pigmented nevi patients were as controls.②MM cell lines under hypoxia condition were detected using immunocytochemistry ,western blot and ELISA for expressions of HIF-1α,VEGF and GLUT-1.And these cell lines under normoxia were as controls.
     Results①Positive HIF-1α,VEGF and GLUT-1 immunostaining were observed in 84.4% (65 cases),71.42% (55 cases) and 68.83%(53 cases) of 77 MM lesions respectively,whereas only one case of pigmented nevi expressed HIF-1αand two cases expressed VEGF and GLUT-1 ;②A positive correlation among HIF-1α,VEGF and GLUT-1 was observed in MM;③Expressions of HIF-1α,VEGF and GLUT-1 were positively correlated with malignant degree in MM;④Expressions of HIF-1α,VEGF and GLUT-1 in MM cell lines A375,A875 and KZ28 under hypoxia condition were higher than under normoxia⑤Expressions of secreted VEGF and membrane GLUT-1 in MM cell lines were steadily increased as hypoxia time was enlonged and were corelated to HIF-1αexpression. Conclusions①On the basis of the current data showing that malignant melanocytic tumors displayed strong HIF-1α,VEGF and GLUT-1 expressions, whereas benign melanocytic tumors showed almost no immunoreactivity for HIF-1α,VEGF and GLUT-1,they might be used as discriminating indicators to distinguish malignant melanocytic tumors from benign melanocytic tumors;②The strong upregulation of HIF-1αin MM found in this study suggested that HIF-1αmight be an important factor to MM growth and could be an attractive therapeutic molecular target for MM.
     PartⅡConstruction and Identification of HIF-1α-shRNAs
     Objectives Short interfering RNA (siRNA) eukaryotic expression vector for HIF-1αwas constructed and transfected into malignant melanoma cells. To explore its effect of interference for HIF-1αin malignant melanoma cells in vitro and observe its effect to cell apoptosis and proliferation.
     Methods①Three HIF-1α-shRNAs targeting human HIF-1αmRNA common sequence was synthesized and it was inserted into Bam HI-Hind III linearized Pgenesile-1 vector. The sequence of HIF-1α-shRNA plasmid was analyzed by DNA sequencer and restrict endonuclease cutting .②To screen the best silencing effect HIF-1αshRNA,the alteration of HIF-1αmRNA and protein was checked by real time Rt-PCR and western blot after HIF-1α-shRNAs were transfected in MM cell lines.And the level of secrected VEGF was checked by ELISA and membrance GLUT-1 was analyzed by FCM.③The monoclone A375 cells with stable expression of best effecting shRNA1 were obtained by G418 selection and were identified with checking GFP expression by FCM and observing under Fluorescence microscope④To observe cell apoptosis and proliferation ,shRNA1-A375 stable clone was cultured under hypoxia condition.
     Results①It was verified that the sequence of constructed recombinant plasmids were correct by DNA sequencing and restrict endonuclease cutting.②HIF-1αshRNAs showed variable efficacy in decreasing HIF-1αmRNA in the three cell lines, relative to the control scrambled shRNA and the mock group. ShRNA1 decreased HIF-1αmRNA by 74%, 69% and 70% in the three cell lines at time point of 24h after transfection, respectively. ShRNA3 decreased HIF-1αmRNA in the three cell lines by 32%, 24% and 22% at the same time point, respectively. Unexpectedly, shRNA2 resulted in a slightly decrease but no significant alteration of HIF-1αmRNA expression in the three cell lines. Among the HIF-1αshRNAs we tested , shRNA1 possessed the strongest inhibitory effect against HIF-1αand had paralleled effect on silencing HIF-1αin the three cell lines.And Silencing HIF-1αby HIF-1αshRNAs could down-regulated secreted VEGF and membrance GLUT-1 in MM cell lines.③We screened and obtained A375 cells with stable expressions of HIF-1α-siRNA as A375 stable clone could observed under Fluorescence microscope and 98% A375 stable clone expressed GFP detected by FCM;④The down-regulation of HIF-1αcould significantly prevent cell proliferation and enhance apoptosis in A375 exposed to hypoxic conditions.
     Conclusions It indicated that hairpin siRNA eukaryotic expression vector for HIF-1αwould be successfully established,and it played a specific inhibitory role in three kinds of tumor cell lines and had parellel silencing effect and also down-regulated VEGF and GLUT-1. At the same time we obtained cells with the stable expression of HIF-1α-shRNA which could cause cell apoptosis and inhibit cell proliferation under hypoxia condition in A375 stable clone. This study laid experimental foundation for further research of the therapy of HIF-1α-shRNA vector in MM.
     Part III Expression of TfR in MM
     Objectives To study expressions of TfR in human malignant melanoma (MM) and MM cell lines and to analyze the possiblity of TfR being an attractive target receptor for MM gene therapy.
     Methods①Tissue sections from 77 MM patients were examined using immunohistochemical technique and morphological quantitative analysis for the protein expressions of TfR and its corelations to tumor clinical-pathology features in MM were analyzed. And tissue sections from 20 pigmented nevi patients were as controls.②MM cell lines under normoxia and hypoxia condition were detected using immunocytochemistry ,western blot and FCM for expressions of TfR.And A2780 cell lines which TfR expression was low as controls.③To analyze the mechanism of the dual phase change of expression of TfR under hypoxia in different time course by checking cell apoptosis and proliferation under hypoxia.
     Results①Positive TfR immunostaining were observed in 90.9% (70 cases) of 77 MM lesions,whereas only five case of pigmented nevi expressed TfR ;②Expressions of TfR were positively correlated with tumor graduation in MM;③Expressions of TfR in MM cell lines A375,A875 , KZ28 and A2780 under normoxia condition were 97%,89%,79% and 2%,respectively.④hypoxia has a dual effect on the expression of TfR in human Melanoma A375 cell line during different hypoxic time course which hypoxia down-regulated TfR expression under hypoxia at time points 12 and 24h and up-regulated TfR expression under hypoxia at time point (36,48 and 60h).⑤hypoxia has multiple opposing effects on tumor growth, with a variable net outcome at various time periods and hypoxia might down-regulate TfR expression by inducing apoptosis which decreases the iron requirements in A375 cells in early hypoxic time course, and up-regulates TfR expression by promoting proliferation in A375 cells in late hypoxic time course.
     Conclusions①On the basis of the current data showing that malignant melanocytic tumors displayed strong TfR expressions,whereas benign melanocytic tumors showed almost no immunoreactivity for TfR,they might be used as target receptor for gene therapy in MM;②Hypoxia has dual effect on the expression of TfR in human Melanoma A375 cell line. These findings may have important implications for more rational, individualized gene-based therapy using TfR as target receptor in melanoma.
     Part IV To study Tumor-targeted distribution after administration of Tf-PEI-shRNA complex in vitro and in vivo
     Objectives By detecting the ability of Tf-PEI to deliver shRNA specifically to TfR-high-expressed cells in vitro and TfR-high-expressed cells bearing tumor tissue in vivo, we expect to provide the experimental data and basis of theory for the clinical treatment of malignant melanoma with Tf-PEI-shRNA complex.
     Methods①t ransfection efficiency for each group was estimated as the mean value of GFP per 10,000 cells by FCM in vitro.②tumors and major organs were excised for GFP mRNA and protein expression by real time RT-PCR and western blot analysis.
     Results①t ransfected shRNA by Lipofectamine2000 was comparably taken up by ~ 70% of both A2780 and A375. Neither A2780 and A375 appreciably took up shRNA by themselves nor when mixed either with Tf lacking PEI or with unmodified PEI.When mixed with Tf-PEI, did A2780 still not take up shRNA. In contrast, when mixed with Tf-PEI, shRNA was comparably taken up by ~ 52% in A375. Although lower than Lipofectamine2000 ~ 70% transfection in A375, Tf-PEI transfection specific delivery of shRNA into TfR-high-expressed MM cell was verified.
     ②the highest amounts of GFP mRNA and protein were detected in tumor tissues and in major organs such as liver, lungs, heart and kidneys only small amounts of GFP mRNA and protein were detected in the A375 bearing tumor mice at time point of 24h after the single injection. At the same time, the result revealed that the GFP mRNA and protein expression distribution in the tumor tissues and major organs of A2780 bearing tumor mice had no significant differences. The results demostrated that Tf-PEI could deliver shRNA with tumor-targeted specific distribution in the nude mice bearing A375 sc.xenograft tumors.
     Conclusions these results showed the tumor-targeted relatively specific distribution after administration of Tf-PEI-shRNA complex in vitro and in vivo.
     Part V Observation of the growth rate of MM xenograft tumor after injection with Tf-PEI- HIF-1αshRNA1 in vivo and exploration of potential mechanisms
     Objectives To observe growth inhibition of MM xenograft after receiving systemic admnistration of Tf-PEI- HIF-1αshRNA1 and to explore the potential mechanisms
     Methods①To evaluate therapeutic potential of Tf-PEI-shRNA1 complex in A375 tumor xenograft, the nude mice bearing A375 s.c.xenograft tumors and the nude mice bearing A2780 s.c.xenograft tumors were randomized and divided in three experimental groups, respectively: one group (n=12) was received direct injection of a total of 20μg of“therapeutic”plasmid Tf-PEI- shRNA1 complex conjugate in100μl volume through tail vein, whereas the second group (n=12) was injected with a total of 20μg of negative control Tf-PEI- scrambled shRNA conjugate in 100μl volume through the tail vein and the third group (n=12) was not received any treatment . Tumor diameter was measured at regular intervals with calipers, and tumor length (L) and width (W) were measured at the end of the experiment, and tumor volume was estimated by the formula (L×W2)/2.②At 25 days after injection, mice were sacrificed and tumors were dissected for further analysis.Expression of HIF-1α,VEGF and GLUT-1 and MVD in tumor tissues were detected by immunohistochemistry and western blot.③tumor glucolysis product Lactic Acid (LA) was detected by LA kit.④tumor cell apotosis was detected by TUNEL method.
     Results①Tumor growth delay was statistically significant (P=0.0014) from day 5 after the beginning of therapy until the day the mice were sacrificed. During day 10-15 after treatment we observed the strongest effect of Tf-PEI-shRNA1 complex; A2780 group tumors in the three groups had a volume of 710±145mm3 , 755±90mm3 and 695±120mm3, which were 7.5-fold larger than the starting volume. Tumor growth delay was not found in the“therapeutic”group.②the immunohistochemistry results shown that in the nude mice bearing A375 s.c.xenograft tumors, HIF-1α,VEGF and GLUT-1 expressions in Tf-PEI-shRNA1 complex group were significantly lower than in Tf-PEI-scrambled shRNA and A375 groups, and no significant difference in HIF-1αexpression was found between Tf-PEI-scrambled shRNA and A375 groups.③But expression of MVD in the three groups has no significant diference (P>0.05)④tumor glucolysis product Lactic Acid (LA) in Tf-PEI-shRNA1 complex group were significantly lower than in Tf-PEI-scrambled shRNA and A375 groups, and no significant difference in HIF-1αexpression was found between Tf-PEI-scrambled shRNA and A375 groups.⑤the number of apotosis tumor cell was larger in Tf-PEI-shRNA1 complex group that other two groups.
     Conclusions①Tf-PEI was able to deliver shRNAs specifically to TfR-expressed MM cells to suppress MM growth even when administered systemically.②systemic Tf-PEI-HIF-1α-shRNA complex administration in the nude mice bearing A375 sc.xenograft tumors could knock down HIF-1αwhich slowed the growth rate of MM xenograft tumor.③Inhibiting tumor glucolysis ,not tumor angiogenesis maybe involve to enhance tumor cell apotosis and to slow the growth rate of MM xenograft tumor.
引文
1. Bataille V. [Genetic and epidemiological aspects of melanoma]. Ann Dermatol Venereol 2006; 133: 56-62.
    2. Dunbar R, Findlay M, Stevens G. Melanoma control: few answers, many questions. N Z Med J 2006; 119: U2172.
    3. Haase VH. The VHL/HIF oxygen-sensing pathway and its relevance to kidney disease. Kidney Int 2006; 69: 1302-7.
    4. Becker JC, Kirkwood JM, Agarwala SS et al. Molecularly targeted therapy for melanoma: current reality and future options. Cancer 2006; 107: 2317-27.
    5. Acker T, Plate KH. Role of hypoxia in tumor angiogenesis-molecular and cellular angiogenic crosstalk. Cell Tissue Res 2003; 314: 145-55.
    6. Acker T, Plate KH. Hypoxia and hypoxia inducible factors (HIF) as important regulators of tumor physiology. Cancer Treat Res 2004; 117: 219-48.
    7. Brahimi-Horn C, Berra E, Pouyssegur J. Hypoxia: the tumor's gateway to progression along the angiogenic pathway. Trends Cell Biol 2001; 11: S32-6.
    8. Belozerov VE, Van Meir EG. Hypoxia inducible factor-1: a novel target for cancer therapy. Anticancer Drugs 2005; 16: 901-9.
    9. Powis G, Kirkpatrick L. Hypoxia inducible factor-1alpha as a cancer drug target. Mol Cancer Ther 2004; 3: 647-54.
    10. Airley R, Loncaster J, Davidson S et al. Glucose transporter glut-1 expression correlates with tumor hypoxia and predicts metastasis-free survival in advanced carcinoma of the cervix. Clin Cancer Res 2001; 7: 928-34.
    11. Airley RE, Loncaster J, Raleigh JA et al. GLUT-1 and CAIX as intrinsic markers of hypoxia in carcinoma of the cervix: relationship to pimonidazole binding. Int J Cancer 2003; 104: 85-91.
    12. Palit V, Phillips RM, Puri R et al. Expression of HIF-1alpha and Glut-1 in human bladder cancer. Oncol Rep 2005; 14: 909-13.
    13. Urano N, Fujiwara Y, Doki Y et al. Overexpression of hypoxia-inducible factor-1 alpha in gastric adenocarcinoma. Gastric Cancer 2006; 9: 44-9.
    14. Tan C, de Noronha RG, Roecker AJ et al. Identification of a novel small-molecule inhibitor of the hypoxia-inducible factor 1 pathway. Cancer Res 2005; 65: 605-12.
    15. 15 Das B, Yeger H, Tsuchida R et al. A hypoxia-driven vascular endothelial growth factor/Flt1 autocrine loop interacts with hypoxia-inducible factor-1alpha through mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 pathway in neuroblastoma. Cancer Res 2005; 65: 7267-75.
    16. Harris AL. von Hippel-Lindau syndrome: target for anti-vascular endothelial growth factor (VEGF) receptor therapy. Oncologist 2000; 5 Suppl 1: 32-6.
    17. Bagnato A, Spinella F. Emerging role of endothelin-1 in tumor angiogenesis. Trends Endocrinol Metab 2003; 14: 44-50.
    18. Choi KS, Bae MK, Jeong JW et al. Hypoxia-induced angiogenesis during carcinogenesis. J Biochem Mol Biol 2003; 36: 120-7.
    19. Mizuno T, Nagao M, Yamada Y et al. Small interfering RNA expression vector targeting hypoxia-inducible factor 1 alpha inhibits tumor growth in hepatobiliary and pancreatic cancers. Cancer Gene Ther 2006; 13: 131-40.
    20. HIF-1alpha, HIF-2alpha and VHL mRNA expression in different cell lines under hypoxia]. Ukr Biokhim Zh 2006; 78: 62-72.
    21. Chung J, Roberts AM, Chow J et al. Homotypic association between tumour-associated VHL proteins leads to the restoration of HIF pathway. Oncogene 2006; 25: 3079-83.
    22. Brahimi-Horn C, Pouyssegur J. When hypoxia signalling meets the ubiquitin-proteasomal pathway, new targets for cancer therapy. Crit RevOncol Hematol 2005; 53: 115-23.
    23. Kim DH, Rossi JJ. Strategies for silencing human disease using RNA interference. Nat Rev Genet 2007; 8: 173-84.
    24. Aharinejad S, Sioud M, Lucas T et al. Target validation using RNA interference in solid tumors. Methods Mol Biol 2007; 361: 227-38.
    25. Kawakami S, Hashida M. Targeted delivery systems of small interfering RNA by systemic administration. Drug Metab Pharmacokinet 2007; 22: 142-51.
    26. Liu G, Wong-Staal F, Li QX. Development of new RNAi therapeutics. Histol Histopathol 2007; 22: 211-7.
    27. Chakraborty C. Potentiality of small interfering RNAs (siRNA) as recent therapeutic targets for gene-silencing. Curr Drug Targets 2007; 8: 469-82.
    28. Daniels TR, Delgado T, Rodriguez JA et al. The transferrin receptor part I: Biology and targeting with cytotoxic antibodies for the treatment of cancer. Clin Immunol 2006; 121: 144-58.
    29. Daniels TR, Delgado T, Helguera G et al. The transferrin receptor part II: targeted delivery of therapeutic agents into cancer cells. Clin Immunol 2006; 121: 159-76.
    1. Acker T, Plate KH. Hypoxia and hypoxia inducible factors (HIF) as important regulators of tumor physiology. Cancer Treat Res 2004; 117: 219-48.
    2. Belozerov VE, Van Meir EG. Hypoxia inducible factor-1: a novel target for cancer therapy. Anticancer Drugs 2005; 16: 901-9.
    3. Blagosklonny MV. Hypoxia-inducible factor: Achilles' heel of antiangiogenic cancer therapy (review). Int J Oncol 2001; 19: 257-62.
    4. Brahimi-Horn C, Pouyssegur J. The role of the hypoxia-inducible factor in tumor metabolism growth and invasion. Bull Cancer 2006; 93: E73-80.
    5. Evans A, Bates V, Troy H et al. Glut-1 as a therapeutic target: increased chemoresistance and HIF-1-independent link with cell turnover is revealed through COMPARE analysis and metabolomic studies. Cancer Chemother Pharmacol 2008; 61: 377-93.
    6. Richardson SM, Knowles R, Tyler J et al. Expression of glucose transporters GLUT-1, GLUT-3, GLUT-9 and HIF-1alpha in normal and degenerate human intervertebral disc. Histochem Cell Biol 2008.
    7. Yasuda M, Miyazawa M, Fujita M et al. Expression of hypoxia inducible factor-1alpha (HIF-1alpha) and glucose transporter-1 (GLUT-1) in ovarian adenocarcinomas: difference in hypoxic status depending on histological character. Oncol Rep 2008; 19: 111-6.
    8. HIF-1 blockade inhibits gastric cancer growth in mice. Cancer Biol Ther 2004; 3: 587.
    9. Yu RM, Chen EX, Kong RY et al. Hypoxia induces telomerase reverse transcriptase (TERT) gene expression in non-tumor fish tissues in vivo: the marine medaka (Oryzias melastigma) model. BMC Mol Biol 2006; 7: 27.
    10. Chin L, Garraway LA, Fisher DE. Malignant melanoma: genetics and therapeutics in the genomic era. Genes Dev 2006; 20: 2149-82.
    11. Taylor CT. Mitochondria and cellular oxygen sensing in the HIF pathway. Biochem J 2008; 409: 19-26.
    12. Bardos JI, Ashcroft M. Hypoxia-inducible factor-1 and oncogenic signalling. Bioessays2004; 26: 262-9.
    13. Acker T, Plate KH. Role of hypoxia in tumor angiogenesis-molecular and cellular angiogenic crosstalk. Cell Tissue Res 2003; 314: 145-55.
    14. Blancher C, Harris AL. The molecular basis of the hypoxia response pathway: tumour hypoxia as a therapy target. Cancer Metastasis Rev 1998; 17: 187-94.
    15. Blagosklonny MV. Antiangiogenic therapy and tumor progression. Cancer Cell 2004; 5: 13-7.
    1. Bedogni B, Welford SM, Cassarino DS et al. The hypoxic microenvironment of the skin contributes to Akt-mediated melanocyte transformation. Cancer Cell 2005; 8: 443-54.
    2. Busca R, Berra E, Pouyssegur J et al. [Hypoxia inducible factor 1a is a new target of microphthalmia-associated transcription factor (MITF) in melanoma cells]. Med Sci (Paris) 2006; 22: 10-3.
    3. Aharinejad S, Sioud M, Lucas T et al. Target validation using RNA interference in solid tumors. Methods Mol Biol 2007; 361: 227-38.
    4. Kim DH, Rossi JJ. Strategies for silencing human disease using RNA interference. Nat Rev Genet 2007; 8: 173-84.
    5. Chakraborty C. Potentiality of small interfering RNAs (siRNA) as recent therapeutic targets for gene-silencing. Curr Drug Targets 2007; 8: 469-82.
    6. Chen M, Du Q, Zhang HY et al. High-throughput screening using siRNA (RNAi) libraries. Expert Rev Mol Diagn 2007; 7: 281-91.
    7. Fuchs U, Borkhardt A. The application of siRNA technology to cancer biology discovery. Adv Cancer Res 2007; 96: 75-102.
    8. Yang H, He S, Quan Z et al. Small interfering RNA-mediated caveolin-1 knockout on plasminogen activator inhibitor-1 expression in insulin-stimulated human vascular endothelial cells. Acta Biochim Biophys Sin (Shanghai) 2007; 39: 224-33.
    9. Aviles JA, Lazaro P. [Genetic predisposition in cutaneous melanoma]. Actas Dermosifiliogr 2006; 97: 229-40.
    10. Giblin AV, Thomas JM. Incidence, mortality and survival in cutaneous melanoma. J Plast Reconstr Aesthet Surg 2007; 60: 32-40.
    11. Belozerov VE, Van Meir EG. Hypoxia inducible factor-1: a novel target for cancer therapy. Anticancer Drugs 2005; 16: 901-9.
    12. Lee SH, Sinko PJ. siRNA--getting the message out. Eur J Pharm Sci 2006; 27: 401-10.
    13. Dallas A, Vlassov AV. RNAi: a novel antisense technology and its therapeutic potential. Med Sci Monit 2006; 12: RA67-74.
    14. Tong AW, Zhang YA, Nemunaitis J. Small interfering RNA for experimental cancertherapy. Curr Opin Mol Ther 2005; 7: 114-24.
    15. Paddison PJ, Caudy AA, Hannon GJ. Stable suppression of gene expression by RNAi in mammalian cells. Proc Natl Acad Sci U S A 2002; 99: 1443-8.
    16. Tan FL, Yin JQ. Application of RNAi to cancer research and therapy. Front Biosci 2005; 10: 1946-60.
    17. Ameyar-Zazoua M, Guasconi V, Ait-Si-Ali S. siRNA as a route to new cancer therapies. Expert Opin Biol Ther 2005; 5: 221-4.
    18. Cheng JC, Moore TB, Sakamoto KM. RNA interference and human disease. Mol Genet Metab 2003; 80: 121-8.
    19. Ryther RC, Flynt AS, Phillips JA, 3rd et al. siRNA therapeutics: big potential from small RNAs. Gene Ther 2005; 12: 5-11.
    20. Campbell TN, Choy FY. RNA interference: past, present and future. Curr Issues Mol Biol 2005; 7: 1-6.
    21. Ait-Si-Ali S, Guasconi V, Harel-Bellan A. [RNA interference and its possible use in cancer therapy]. Bull Cancer 2004; 91: 15-8.
    22. Gartel AL, Kandel ES. RNA interference in cancer. Biomol Eng 2006; 23: 17-34.
    23. Woessmann W, Damm-Welk C, Fuchs U et al. RNA interference: new mechanisms for targeted treatment? Rev Clin Exp Hematol 2003; 7: 270-91.
    24. Aigner A. Gene silencing through RNA interference (RNAi) in vivo: strategies based on the direct application of siRNAs. J Biotechnol 2006; 124: 12-25.
    25. Bi F, Liu N, Fan D. Small interfering RNA: a new tool for gene therapy. Curr Gene Ther 2003; 3: 411-7.
    26. Sui G, Soohoo C, Affar el B et al. A DNA vector-based RNAi technology to suppress gene expression in mammalian cells. Proc Natl Acad Sci U S A 2002; 99: 5515-20.
    27. Harborth J, Elbashir SM, Vandenburgh K et al. Sequence, chemical, and structural variation of small interfering RNAs and short hairpin RNAs and the effect on mammalian gene silencing. Antisense Nucleic Acid Drug Dev 2003; 13: 83-105.
    28. Aoki Y, Cioca DP, Oidaira H et al. RNA interference may be more potent than antisense RNA in human cancer cell lines. Clin Exp Pharmacol Physiol 2003; 30: 96-102.
    29. Kittler R, Buchholz F. RNA interference: gene silencing in the fast lane. Semin CancerBiol 2003; 13: 259-65.
    30. Izquierdo M. Short interfering RNAs as a tool for cancer gene therapy. Cancer Gene Ther 2005; 12: 217-27.
    31. Liu F, He CW, Zhang YF et al. RNA interference by expression of short hairpin RNAs suppresses bcl-xL gene expression in nasopharyngeal carcinoma cells. Acta Pharmacol Sin 2005; 26: 228-34.
    32. Czauderna F, Santel A, Hinz M et al. Inducible shRNA expression for application in a prostate cancer mouse model. Nucleic Acids Res 2003; 31: e127.
    33. Karagiannis TC, El-Osta A. RNA interference and potential therapeutic applications of short interfering RNAs. Cancer Gene Ther 2005; 12: 787-95.
    34. Dang CV, Lewis BC, Dolde C et al. Oncogenes in tumor metabolism, tumorigenesis, and apoptosis. J Bioenerg Biomembr 1997; 29: 345-54.
    35. Goda N, Dozier SJ, Johnson RS. HIF-1 in cell cycle regulation, apoptosis, and tumor progression. Antioxid Redox Signal 2003; 5: 467-73.
    36. Greijer AE, van der Wall E. The role of hypoxia inducible factor 1 (HIF-1) in hypoxia induced apoptosis. J Clin Pathol 2004; 57: 1009-14.
    37. Fan LF, Diao LM, Chen DJ et al. [Expression of HIF-1 alpha and its relationship to apoptosis and proliferation in lung cancer]. Ai Zheng 2002; 21: 254-8.
    38. Ghafar MA, Anastasiadis AG, Chen MW et al. Acute hypoxia increases the aggressive characteristics and survival properties of prostate cancer cells. Prostate 2003; 54: 58-67.
    39. Carmeliet P, Dor Y, Herbert JM et al. Role of HIF-1alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 1998; 394: 485-90.
    40. Bacon AL, Harris AL. Hypoxia-inducible factors and hypoxic cell death in tumour physiology. Ann Med 2004; 36: 530-9.
    41. Acker T, Plate KH. Role of hypoxia in tumor angiogenesis-molecular and cellular angiogenic crosstalk. Cell Tissue Res 2003; 314: 145-55.
    42. Brahimi-Horn MC, Pouyssegur J. Harnessing the hypoxia-inducible factor in cancer and ischemic disease. Biochem Pharmacol 2006.
    1. Ameyar-Zazoua M, Guasconi V, Ait-Si-Ali S. siRNA as a route to new cancer therapies. Expert Opin Biol Ther 2005; 5: 221-4.
    2. Campbell TN, Choy FY. RNA interference: past, present and future. Curr Issues Mol Biol 2005; 7: 1-6.
    3. Qian ZM, Li H, Sun H et al. Targeted drug delivery via the transferrin receptor-mediated endocytosis pathway. Pharmacol Rev 2002; 54: 561-87.
    4. Tao J, Liu YQ, Li Y et al. Hypoxia: dual effect on the expression of transferrin receptor in human melanoma A375 cell line. Exp Dermatol 2007; 16: 899-904.
    5. Kasibhatla S, Jessen KA, Maliartchouk S et al. A role for transferrin receptor in triggering apoptosis when targeted with gambogic acid. Proc Natl Acad Sci U S A 2005; 102: 12095-100.
    6. Daniels TR, Delgado T, Helguera G et al. The transferrin receptor part II: targeted delivery of therapeutic agents into cancer cells. Clin Immunol 2006; 121: 159-76.
    7. Testa U, Pelosi E, Peschle C. The transferrin receptor. Crit Rev Oncog 1993; 4: 241-76.
    8. Hogrefe RI, Lebedev AV, Zon G et al. Chemically modified short interfering hybrids (siHYBRIDS): nanoimmunoliposome delivery in vitro and in vivo for RNAi of HER-2. Nucleosides Nucleotides Nucleic Acids 2006; 25: 889-907.
    9. Masiero M, Nardo G, Indraccolo S et al. RNA interference: implications for cancer treatment. Mol Aspects Med 2007; 28: 143-66.
    10. Chakraborty C. Potentiality of small interfering RNAs (siRNA) as recent therapeutic targets for gene-silencing. Curr Drug Targets 2007; 8: 469-82.
    11. Xiu-Lian D, Kui W, Ya K et al. Apotransferrin is internalized and distributed in the same way as holotransferrin in K562 cells. J Cell Physiol 2004; 201: 45-54.
    12. Giannetti AM, Halbrooks PJ, Mason AB et al. The molecular mechanism for receptor-stimulated iron release from the plasma iron transport protein transferrin. Structure (Camb) 2005; 13: 1613-23.
    13. Basar I, Ayhan A, Bircan K et al. Transferrin receptor activity as a marker in transitional cell carcinoma of the bladder. Br J Urol 1991; 67: 165-8.
    14. Kircheis R, Blessing T, Brunner S et al. Tumor targeting with surface-shielded ligand--polycation DNA complexes. J Control Release 2001; 72: 165-70.
    15. Ogris M, Wagner E. Tumor-targeted gene transfer with DNA polyplexes. Somat Cell Mol Genet 2002; 27: 85-95.
    16. Acker T, Plate KH. Hypoxia and hypoxia inducible factors (HIF) as important regulators of tumor physiology. Cancer Treat Res 2004; 117: 219-48.
    17. Brahimi-Horn C, Berra E, Pouyssegur J. Hypoxia: the tumor's gateway to progression along the angiogenic pathway. Trends Cell Biol 2001; 11: S32-6.
    18. Goda N, Dozier SJ, Johnson RS. HIF-1 in cell cycle regulation, apoptosis, and tumor progression. Antioxid Redox Signal 2003; 5: 467-73.
    19. Hammond EM, Giaccia AJ. The role of p53 in hypoxia-induced apoptosis. Biochem Biophys Res Commun 2005; 331: 718-25.
    20. Kunz M, Ibrahim SM. Molecular responses to hypoxia in tumor cells. Mol Cancer 2003; 2: 23.
    21. Jones DT, Trowbridge IS, Harris AL. Effects of transferrin receptor blockade on cancer cell proliferation and hypoxia-inducible factor function and their differential regulation by ascorbate. Cancer Res 2006; 66: 2749-56.
    1. Liu G, Wong-Staal F, Li QX. Development of new RNAi therapeutics. Histol Histopathol 2007; 22: 211-7.
    2. Wagner E. Programmed drug delivery: nanosystems for tumor targeting. Expert Opin Biol Ther 2007; 7: 587-93.
    3. Chakraborty C. Potentiality of small interfering RNAs (siRNA) as recent therapeutic targets for gene-silencing. Curr Drug Targets 2007; 8: 469-82.
    4. Vorhies JS, Nemunaitis J. Nonviral delivery vehicles for use in short hairpin RNA-based cancer therapies. Expert Rev Anticancer Ther 2007; 7: 373-82.
    5. Scherr M, Eder M. Gene silencing by small regulatory RNAs in mammalian cells. Cell Cycle 2007; 6: 444-9.
    6. Ogris M, Wagner E. Tumor-targeted gene transfer with DNA polyplexes. Somat Cell Mol Genet 2002; 27: 85-95.
    7. Park YM, Shin BA, Oh IJ. Poly(L-lactic acid)/polyethylenimine nanoparticles as plasmid DNA carriers. Arch Pharm Res 2008; 31: 96-102.
    8. Sakae M, Ito T, Yoshihara C et al. Highly efficient in vivo gene transfection by plasmid/PEI complexes coated by anionic PEG derivatives bearing carboxyl groups and RGD peptide. Biomed Pharmacother 2008.
    9. Duan Y, Zheng J, Han S et al. A tumor targeted gene vector modified with G250 monoclonal antibody for gene therapy. J Control Release 2008.
    10. Kircheis R, Kichler A, Wallner G et al. Coupling of cell-binding ligands to polyethylenimine for targeted gene delivery. Gene Ther 1997; 4: 409-18.
    11. Asai T, Trinh R, Ng PP et al. A human biotin acceptor domain allows site-specific conjugation of an enzyme to an antibody-avidin fusion protein for targeted drug delivery. Biomol Eng 2005; 21: 145-55.
    12. E MT, Hewitt J, K CU et al. Identification of the epitope of a monoclonal antibody that disrupts binding of human transferrin to the human transferrin receptor. Febs J 2005; 272: 6344-53.
    13. Daniels TR, Delgado T, Rodriguez JA et al. The transferrin receptor part I: Biology andtargeting with cytotoxic antibodies for the treatment of cancer. Clin Immunol 2006; 121: 144-58.
    14. Testa U, Pelosi E, Peschle C. The transferrin receptor. Crit Rev Oncog 1993; 4: 241-76.
    15. Kircheis R, Blessing T, Brunner S et al. Tumor targeting with surface-shielded ligand--polycation DNA complexes. J Control Release 2001; 72: 165-70.
    1. Cummins DL, Cummins JM, Pantle H et al. Cutaneous malignant melanoma. Mayo Clin Proc 2006; 81: 500-7.
    2. Demierre MF. Epidemiology and prevention of cutaneous melanoma. Curr Treat Options Oncol 2006; 7: 181-6.
    3. Ranieri JM, Wagner JD, Wenck S et al. The prognostic importance of sentinel lymph node biopsy in thin melanoma. Ann Surg Oncol 2006; 13: 927-32.
    4. Naylor MF, Chen WR, Teague TK et al. In situ photoimmunotherapy: a tumour-directed treatment for melanoma. Br J Dermatol 2006; 155: 1287-92.
    5. Atkins MB. Cytokine-based therapy and biochemotherapy for advanced melanoma. Clin Cancer Res 2006; 12: 2353s-8s.
    6. Chin L, Garraway LA, Fisher DE. Malignant melanoma: genetics and therapeutics in the genomic era. Genes Dev 2006; 20: 2149-82.
    7. Gheysen G, Vanholme B. RNAi from plants to nematodes. Trends Biotechnol 2007; 25: 89-92.
    8. Gruber J, Boese G, Tuschl T et al. RNA interference by osmotic lysis of pinosomes: liposome-independent transfection of siRNAs into mammalian cells. Biotechniques 2004; 37: 96-102.
    9. Wall NR, Shi Y. Small RNA: can RNA interference be exploited for therapy? Lancet 2003; 362: 1401-3.
    10. Ameyar-Zazoua M, Guasconi V, Ait-Si-Ali S. siRNA as a route to new cancer therapies. Expert Opin Biol Ther 2005; 5: 221-4.
    11. Campbell TN, Choy FY. RNA interference: past, present and future. Curr Issues Mol Biol 2005; 7: 1-6.
    12. Bi F, Liu N, Fan D. Small interfering RNA: a new tool for gene therapy. Curr Gene Ther 2003; 3: 411-7.
    13. Cheng JC, Moore TB, Sakamoto KM. RNA interference and human disease. Mol Genet Metab 2003; 80: 121-8.
    14. Fuchs U, Damm-Welk C, Borkhardt A. Silencing of disease-related genes by small interfering RNAs. Curr Mol Med 2004; 4: 507-17.
    15. Ait-Si-Ali S, Guasconi V, Harel-Bellan A. [RNA interference and its possible use incancer therapy]. Bull Cancer 2004; 91: 15-8.
    16. Acker T, Plate KH. Hypoxia and hypoxia inducible factors (HIF) as important regulators of tumor physiology. Cancer Treat Res 2004; 117: 219-48.
    17. Acker T, Plate KH. Role of hypoxia in tumor angiogenesis-molecular and cellular angiogenic crosstalk. Cell Tissue Res 2003; 314: 145-55.
    18. Airley R, Loncaster J, Davidson S et al. Glucose transporter glut-1 expression correlates with tumor hypoxia and predicts metastasis-free survival in advanced carcinoma of the cervix. Clin Cancer Res 2001; 7: 928-34.
    19. Belozerov VE, Van Meir EG. Hypoxia inducible factor-1: a novel target for cancer therapy. Anticancer Drugs 2005; 16: 901-9.
    20. Brown JM. Tumor microenvironment and the response to anticancer therapy. Cancer Biol Ther 2002; 1: 453-8.
    21. Clottes E. [Hypoxia-inducible factor 1: regulation, involvement in carcinogenesis and target for anticancer therapy]. Bull Cancer 2005; 92: 119-27.
    22. Brahimi-Horn MC, Pouyssegur J. Harnessing the hypoxia-inducible factor in cancer and ischemic disease. Biochem Pharmacol 2006.
    23. Powis G, Kirkpatrick L. Hypoxia inducible factor-1alpha as a cancer drug target. Mol Cancer Ther 2004; 3: 647-54.
    24. Jiang H, Feng Y. Hypoxia-inducible factor 1alpha (HIF-1alpha) correlated with tumor growth and apoptosis in ovarian cancer. Int J Gynecol Cancer 2006; 16 Suppl 1: 405-12.
    25. Maxwell PH, Dachs GU, Gleadle JM et al. Hypoxia-inducible factor-1 modulates gene expression in solid tumors and influences both angiogenesis and tumor growth. Proc Natl Acad Sci U S A 1997; 94: 8104-9.
    26. Urano N, Fujiwara Y, Doki Y et al. Overexpression of hypoxia-inducible factor-1 alpha in gastric adenocarcinoma. Gastric Cancer 2006; 9: 44-9.
    27. Erler JT, Cawthorne CJ, Williams KJ et al. Hypoxia-mediated down-regulation of Bid and Bax in tumors occurs via hypoxia-inducible factor 1-dependent and -independent mechanisms and contributes to drug resistance. Mol Cell Biol 2004; 24: 2875-89.
    28. Greijer AE, van der Wall E. The role of hypoxia inducible factor 1 (HIF-1) in hypoxia induced apoptosis. J Clin Pathol 2004; 57: 1009-14.
    1. Wykoff CC, Pugh CW, Harris AL et al. The HIF pathway: implications for patterns of gene expression in cancer. Novartis Found Symp 2001; 240: 212-25; discussion 25-31.
    2. Lee JW, Bae SH, Jeong JW et al. Hypoxia-inducible factor (HIF-1)alpha: its protein stability and biological functions. Exp Mol Med 2004; 36: 1-12.
    3. Greco O, Marples B, Joiner MC et al. How to overcome (and exploit) tumor hypoxia for targeted gene therapy. J Cell Physiol 2003; 197: 312-25.
    4. Bacon AL, Harris AL. Hypoxia-inducible factors and hypoxic cell death in tumour physiology. Ann Med 2004; 36: 530-9.
    5. Brahimi-Horn C, Berra E, Pouyssegur J. Hypoxia: the tumor's gateway to progression along the angiogenic pathway. Trends Cell Biol 2001; 11: S32-6.
    6. Blagosklonny MV. Hypoxia-inducible factor: Achilles' heel of antiangiogenic cancer therapy (review). Int J Oncol 2001; 19: 257-62.
    7. Gu J, Milligan J, Huang LE. Molecular mechanism of hypoxia-inducible factor 1alpha -p300 interaction. A leucine-rich interface regulated by a single cysteine. J Biol Chem 2001; 276: 3550-4.
    8. Kasper LH, Boussouar F, Boyd K et al. Two transactivation mechanisms cooperate for the bulk of HIF-1-responsive gene expression. Embo J 2005; 24: 3846-58.
    9. Lando D, Peet DJ, Whelan DA et al. Asparagine hydroxylation of the HIF transactivation domain a hypoxic switch. Science 2002; 295: 858-61.
    10. Tanimoto K, Makino Y, Pereira T et al. Mechanism of regulation of the hypoxia-inducible factor-1 alpha by the von Hippel-Lindau tumor suppressor protein. Embo J 2000; 19: 4298-309.
    11. Pereira T, Zheng X, Ruas JL et al. Identification of residues critical for regulation of protein stability and the transactivation function of the hypoxia-inducible factor-1alpha by the von Hippel-Lindau tumor suppressor gene product. J Biol Chem 2003; 278: 6816-23.
    12. Brahimi-Horn MC, Pouyssegur J. Harnessing the hypoxia-inducible factor in cancer and ischemic disease. Biochem Pharmacol 2006.
    13. Belozerov VE, Van Meir EG. Hypoxia inducible factor-1: a novel target for cancer therapy. Anticancer Drugs 2005; 16: 901-9.
    14. Brahimi-Horn C, Pouyssegur J. The role of the hypoxia-inducible factor in tumor metabolism growth and invasion. Bull Cancer 2006; 93: E73-80.
    15. Goda N, Dozier SJ, Johnson RS. HIF-1 in cell cycle regulation, apoptosis, and tumor progression. Antioxid Redox Signal 2003; 5: 467-73.
    16. Minet E, Michel G, Remacle J et al. Role of HIF-1 as a transcription factor involved in embryonic development, cancer progression and apoptosis (review). Int J Mol Med 2000; 5: 253-9.
    17. Richardson DR. Molecular mechanisms of iron uptake by cells and the use of iron chelators for the treatment of cancer. Curr Med Chem 2005; 12: 2711-29.
    18. Pugh CW, Ratcliffe PJ. The von Hippel-Lindau tumor suppressor, hypoxia-inducible factor-1 (HIF-1) degradation, and cancer pathogenesis. Semin Cancer Biol 2003; 13: 83-9.
    19. Giatromanolaki A, Harris AL. Tumour hypoxia, hypoxia signaling pathways and hypoxia inducible factor expression in human cancer. Anticancer Res 2001; 21: 4317-24.
    20. Koukourakis MI, Giatromanolaki A, Simopoulos C et al. Lactate dehydrogenase 5 (LDH5) relates to up-regulated hypoxia inducible factor pathway and metastasis in colorectal cancer. Clin Exp Metastasis 2005; 22: 25-30.
    21. Berra E, Pages G, Pouyssegur J. MAP kinases and hypoxia in the control of VEGF expression. Cancer Metastasis Rev 2000; 19: 139-45.
    22. Bicknell R, Harris AL. Novel angiogenic signaling pathways and vascular targets. Annu Rev Pharmacol Toxicol 2004; 44: 219-38.
    23. Choi KS, Bae MK, Jeong JW et al. Hypoxia-induced angiogenesis during carcinogenesis. J Biochem Mol Biol 2003; 36: 120-7.
    24. Mazure NM, Brahimi-Horn MC, Berta MA et al. HIF-1: master and commander of the hypoxic world. A pharmacological approach to its regulation by siRNAs. Biochem Pharmacol 2004; 68: 971-80.
    25. Maxwell PH. HIF-1's relationship to oxygen: simple yet sophisticated. Cell Cycle 2004;3: 156-9.
    26. Greijer AE, van der Wall E. The role of hypoxia inducible factor 1 (HIF-1) in hypoxia induced apoptosis. J Clin Pathol 2004; 57: 1009-14.
    27. Zhou J, Schmid T, Schnitzer S et al. Tumor hypoxia and cancer progression. Cancer Lett 2006; 237: 10-21.
    28. Acker T, Plate KH. Role of hypoxia in tumor angiogenesis-molecular and cellular angiogenic crosstalk. Cell Tissue Res 2003; 314: 145-55.
    29. Clottes E. [Hypoxia-inducible factor 1: regulation, involvement in carcinogenesis and target for anticancer therapy]. Bull Cancer 2005; 92: 119-27.
    30. Acker T, Plate KH. Hypoxia and hypoxia inducible factors (HIF) as important regulators of tumor physiology. Cancer Treat Res 2004; 117: 219-48.
    31. Schindl M, Schoppmann SF, Samonigg H et al. Overexpression of hypoxia-inducible factor 1alpha is associated with an unfavorable prognosis in lymph node-positive breast cancer. Clin Cancer Res 2002; 8: 1831-7.
    32. Birner P, Schindl M, Obermair A et al. Overexpression of hypoxia-inducible factor 1alpha is a marker for an unfavorable prognosis in early-stage invasive cervical cancer. Cancer Res 2000; 60: 4693-6.
    33. Hui EP, Chan AT, Pezzella F et al. Coexpression of hypoxia-inducible factors 1alpha and 2alpha, carbonic anhydrase IX, and vascular endothelial growth factor in nasopharyngeal carcinoma and relationship to survival. Clin Cancer Res 2002; 8: 2595-604.
    34. Zhong H, De Marzo AM, Laughner E et al. Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res 1999; 59: 5830-5.
    35. Zhong H, Hanrahan C, van der Poel H et al. Hypoxia-inducible factor 1alpha and 1beta proteins share common signaling pathways in human prostate cancer cells. Biochem Biophys Res Commun 2001; 284: 352-6.
    36. Talks KL, Turley H, Gatter KC et al. The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated macrophages. Am J Pathol 2000; 157: 411-21.
    37. Bos R, van Diest PJ, van der Groep P et al. Protein expression of B-cell lymphomagene 6 (BCL-6) in invasive breast cancer is associated with cyclin D1 and hypoxia-inducible factor-1alpha (HIF-1alpha). Oncogene 2003; 22: 8948-51.
    38. Wiesener MS, Seyfarth M, Warnecke C et al. Paraneoplastic erythrocytosis associated with an inactivating point mutation of the von Hippel-Lindau gene in a renal cell carcinoma. Blood 2002; 99: 3562-5.
    39. Wiesener MS, Munchenhagen PM, Berger I et al. Constitutive activation of hypoxia-inducible genes related to overexpression of hypoxia-inducible factor-1alpha in clear cell renal carcinomas. Cancer Res 2001; 61: 5215-22.
    40. Volm M, Koomagi R. Hypoxia-inducible factor (HIF-1) and its relationship to apoptosis and proliferation in lung cancer. Anticancer Res 2000; 20: 1527-33.
    41. Takahashi R, Tanaka S, Hiyama T et al. Hypoxia-inducible factor-1alpha expression and angiogenesis in gastrointestinal stromal tumor of the stomach. Oncol Rep 2003; 10: 797-802.
    42. Buchler P, Reber HA, Buchler M et al. Hypoxia-inducible factor 1 regulates vascular endothelial growth factor expression in human pancreatic cancer. Pancreas 2003; 26: 56-64.
    43. Berta M, Brahimi-Horn C, Pouyssegur J. [Regulation of the Hypoxia-Inducible Factor-1alpha (HIF-1alpha): a breath of fresh air in hypoxia research]. J Soc Biol 2004; 198: 113-20.
    44. Chen L, Shibasaki F. [Angiogenesis and metastasis of carcinoma]. Clin Calcium 2006; 16: 81-7.
    45. Dachs GU, Stratford IJ. The molecular response of mammalian cells to hypoxia and the potential for exploitation in cancer therapy. Br J Cancer Suppl 1996; 27: S126-32.
    46. Marti HH. Angiogenesis--a self-adapting principle in hypoxia. Exs 2005: 163-80.
    47. L'Allemain G. [The hypoxia-inducible factor HIF as a new target in cancer research]. Bull Cancer 2002; 89: 257-60.
    48. Mukhopadhyay D, Datta K. Multiple regulatory pathways of vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) expression in tumors. Semin Cancer Biol 2004; 14: 123-30.
    49. Palayoor ST, Tofilon PJ, Coleman CN. Ibuprofen-mediated reduction ofhypoxia-inducible factors HIF-1alpha and HIF-2alpha in prostate cancer cells. Clin Cancer Res 2003; 9: 3150-7.
    50. Palayoor ST, Burgos MA, Shoaibi A et al. Effect of radiation and ibuprofen on normoxic renal carcinoma cells overexpressing hypoxia-inducible factors by loss of von Hippel-Lindau tumor suppressor gene function. Clin Cancer Res 2004; 10: 4158-64.
    51. Mabjeesh NJ, Escuin D, LaVallee TM et al. 2ME2 inhibits tumor growth and angiogenesis by disrupting microtubules and dysregulating HIF. Cancer Cell 2003; 3: 363-75.
    52. Mabjeesh NJ, Willard MT, Frederickson CE et al. Androgens stimulate hypoxia-inducible factor 1 activation via autocrine loop of tyrosine kinase receptor/phosphatidylinositol 3'-kinase/protein kinase B in prostate cancer cells. Clin Cancer Res 2003; 9: 2416-25.
    53. Lu H, Forbes RA, Verma A. Hypoxia-inducible factor 1 activation by aerobic glycolysis implicates the Warburg effect in carcinogenesis. J Biol Chem 2002; 277: 23111-5.
    54. Garber K. Energy boost: the Warburg effect returns in a new theory of cancer. J Natl Cancer Inst 2004; 96: 1805-6.
    55. Dang CV, Lewis BC, Dolde C et al. Oncogenes in tumor metabolism, tumorigenesis, and apoptosis. J Bioenerg Biomembr 1997; 29: 345-54.
    56. Robey IF, Lien AD, Welsh SJ et al. Hypoxia-inducible factor-1alpha and the glycolytic phenotype in tumors. Neoplasia 2005; 7: 324-30.
    57. Semenza GL, Artemov D, Bedi A et al. 'The metabolism of tumours': 70 years later. Novartis Found Symp 2001; 240: 251-60; discussion 60-4.
    58. Schwarzer R, Tondera D, Arnold W et al. REDD1 integrates hypoxia-mediated survival signaling downstream of phosphatidylinositol 3-kinase. Oncogene 2005; 24: 1138-49.
    59. Blancher C, Harris AL. The molecular basis of the hypoxia response pathway: tumour hypoxia as a therapy target. Cancer Metastasis Rev 1998; 17: 187-94.
    60. Carmeliet P, Dor Y, Herbert JM et al. Role of HIF-1alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 1998; 394: 485-90.
    61. Horiuchi A, Imai T, Shimizu M et al. Hypoxia-induced changes in the expression of VEGF, HIF-1 alpha and cell cycle-related molecules in ovarian cancer cells. Anticancer Res 2002; 22: 2697-702.
    62. Kuijper A, van der Groep P, van der Wall E et al. Expression of hypoxia-inducible factor 1 alpha and its downstream targets in fibroepithelial tumors of the breast. Breast Cancer Res 2005; 7: R808-18.
    63. Acs G, Zhang PJ, McGrath CM et al. Hypoxia-inducible erythropoietin signaling in squamous dysplasia and squamous cell carcinoma of the uterine cervix and its potential role in cervical carcinogenesis and tumor progression. Am J Pathol 2003; 162: 1789-806.
    64. Brune B, Zhou J, von Knethen A. Nitric oxide, oxidative stress, and apoptosis. Kidney Int Suppl 2003: S22-4.
    65. Bussink J, Kaanders JH, van der Kogel AJ. Tumor hypoxia at the micro-regional level: clinical relevance and predictive value of exogenous and endogenous hypoxic cell markers. Radiother Oncol 2003; 67: 3-15.
    66. Brune B, von Knethen A, Sandau KB. Transcription factors p53 and HIF-1alpha as targets of nitric oxide. Cell Signal 2001; 13: 525-33.
    67. Hammond EM, Giaccia AJ. The role of p53 in hypoxia-induced apoptosis. Biochem Biophys Res Commun 2005; 331: 718-25.
    68. Hopfl G, Wenger RH, Ziegler U et al. Rescue of hypoxia-inducible factor-1alpha-deficient tumor growth by wild-type cells is independent of vascular endothelial growth factor. Cancer Res 2002; 62: 2962-70.
    69. Fan LF, Diao LM, Chen DJ et al. [Expression of HIF-1 alpha and its relationship to apoptosis and proliferation in lung cancer]. Ai Zheng 2002; 21: 254-8.
    70. Piret JP, Lecocq C, Toffoli S et al. Hypoxia and CoCl2 protect HepG2 cells against serum deprivation- and t-BHP-induced apoptosis: a possible anti-apoptotic role for HIF-1. Exp Cell Res 2004; 295: 340-9.
    71. Ghafar MA, Anastasiadis AG, Chen MW et al. Acute hypoxia increases the aggressive characteristics and survival properties of prostate cancer cells. Prostate 2003; 54: 58-67.
    72. Comerford KM, Wallace TJ, Karhausen J et al. Hypoxia-inducible factor-1-dependent regulation of the multidrug resistance (MDR1) gene. Cancer Res 2002; 62: 3387-94.
    73. Wartenberg M, Gronczynska S, Bekhite MM et al. Regulation of the multidrug resistance transporter P-glycoprotein in multicellular prostate tumor spheroids by hyperthermia and reactive oxygen species. Int J Cancer 2005; 113: 229-40.
    74. Xia S, Yu SY, Yuan XL et al. [Effects of hypoxia on expression of P-glycoprotein and multidrug resistance protein in human lung adenocarcinoma A549 cell line]. Zhonghua Yi Xue Za Zhi 2004; 84: 663-6.
    75. Comerford KM, Cummins EP, Taylor CT. c-Jun NH2-terminal kinase activation contributes to hypoxia-inducible factor 1alpha-dependent P-glycoprotein expression in hypoxia. Cancer Res 2004; 64: 9057-61.
    76. Hyun JY, Chun YS, Kim TY et al. Hypoxia-inducible factor 1alpha- mediated resistance to phenolic anticancer. Chemotherapy 2004; 50: 119-26.
    77. Kung AL, Zabludoff SD, France DS et al. Small molecule blockade of transcriptional coactivation of the hypoxia-inducible factor pathway. Cancer Cell 2004; 6: 33-43.
    78. Isaacs JS, Jung YJ, Mimnaugh EG et al. Hsp90 regulates a von Hippel Lindau-independent hypoxia-inducible factor-1 alpha-degradative pathway. J Biol Chem 2002; 277: 29936-44.
    79. Sun X, Kanwar JR, Leung E et al. Gene transfer of antisense hypoxia inducible factor-1 alpha enhances the therapeutic efficacy of cancer immunotherapy. Gene Ther 2001; 8: 638-45.
    80. Ruan H, Wang J, Hu L et al. Killing of brain tumor cells by hypoxia-responsive element mediated expression of BAX. Neoplasia 1999; 1: 431-7.
    81. Ruan H, Deen DF. Use of hypoxia-regulated gene expression in tumor-specific gene therapy. Curr Opin Investig Drugs 2001; 2: 839-43.
    82. Laughner E, Taghavi P, Chiles K et al. HER2 (neu) signaling increases the rate of hypoxia-inducible factor 1alpha (HIF-1alpha) synthesis: novel mechanism for HIF-1-mediated vascular endothelial growth factor expression. Mol Cell Biol 2001; 21: 3995-4004.
    83. Mie Lee Y, Kim SH, Kim HS et al. Inhibition of hypoxia-induced angiogenesis byFK228, a specific histone deacetylase inhibitor, via suppression of HIF-1alpha activity. Biochem Biophys Res Commun 2003; 300: 241-6.
    84. Mabjeesh NJ, Post DE, Willard MT et al. Geldanamycin induces degradation of hypoxia-inducible factor 1alpha protein via the proteosome pathway in prostate cancer cells. Cancer Res 2002; 62: 2478-82.
    85. Sodhi A, Montaner S, Patel V et al. The Kaposi's sarcoma-associated herpes virus G protein-coupled receptor up-regulates vascular endothelial growth factor expression and secretion through mitogen-activated protein kinase and p38 pathways acting on hypoxia-inducible factor 1alpha. Cancer Res 2000; 60: 4873-80.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700