Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (17)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 2949-2949
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 2949-2949
    Abstract: REV3L, the catalytic subunit of DNA ploymerse zeta, is well known to participate in error-prone translesion synthesis (TLS) with less stringent and lower processivity. Recent evidence demonstrated that REV3L is involved in carcinogenesis and tumor progression. However, the function of REV3L remains unclear in esophageal squamous cell cancer (ESCC). In this study, we examined REV3L expression in ESCC tissues and its association with clinicopathological parameters. REV3L was found to be significantly up-regulated in ESCC tissues, which is correlated with lymph node metastasis and clinical stages of the patients. To further investigate the potential role of REV3L in esophageal cancer, stable ESCC cell lines with the suppression of REV3L expression were established. Down regulation of REV3L expression led to a decrease in cell proliferation and invasive tendency partly through suppressing cyclinD1 and survivin expression, and an increase in cellular sensitivity to 5-Fu by inducing G1 phase arrest and apoptosis. Therefore, REV3L plays an important role in ESCC progression and chemoresistance, and is a potential diagnostic marker and therapeutic target for ESCC. Citation Format: Jundong Zhou, Sitao Zou, Wei-Qun Ding, Jinchang Wu. REV3L, the catalytic subunit of pol ζ, is involved in the progression and chemoresistance of esophageal squamous cell cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2949.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. CT170-CT170
    Abstract: D-0316 in patients with advanced T790M-positive EGFR-mutant non-small cell lung cancer who progressed on prior EGFR-TKI therapy: results from a phase II study (NCT03861156) Background: Despite initial response to EGFR-TKI, most patients (pts) develop resistance with the EGFR T790M mutation detectable in ~50% of patients treated with first-/second-generation EGFR-TKIs. D-0316 is a third-generation EGFR-TKI that is selective for both EGFR-TKI sensitizing and T790M resistance mutations in pts with non-small cell lung cancer (NSCLC). We report the results of a registered, single-arm, phase II study of D-0316 in NSCLC pts with EGFR T790M who progressed on previous treatment with first-line EGFR-TKIs. Methods: In this phase II, open-label, single-arm study, eligible pts were those who had confirmed locally advanced or metastatic NSCLC, and had disease progression after first-line EGFR-TKI and with T790M mutation. Pts were initially orally given D-0316 50 mg. However, considering the benefits and risks of the pts, the dose was modified to 100 mg once daily with a 21-day lead-in at 75 mg once daily. The primary endpoint was objective response rate (ORR) based on independent review committee (IRC) according to RECIST 1.1.Results: As of October 31, 2019, 176 pts were enrolled in the 50 mg phase, in which 90 pts had partial response, achieving an ORR of 51.1% (95%CI: 43.5-58.7). Despite the immature PFS, disease progression or death occurred in 60 pts (34.1%) and the median PFS was 8.4 months (95% CI: 8.0-NE). Between September 12, 2019 and July 29, 2020, 689 pts were screened and 290 pts (median age 62.5) were enrolled in China and received 100mg D-0316 with a 21-day lead-in at 75 mg. At data cutoff (October 18, 2020), the median duration of follow-up was 5.5 months. 188 of the 290 pts achieved confirmed partial responses by IRC. The ORR was 64.8% (95% CI: 59.0-70.3) and the disease control rate (DCR) was 95.2% (95% CI: 92.0-97.3). The ORR was consistent across in most subgroups. Among 34 pts with brain metastases at baseline, 18 pts achieved confirmed partial responses and the intracranial ORR was 52.9% (95% CI: 35.1-70.2). The PFS, DoR, and OS were premature. The most common treatment-related adverse events were thrombocytopenia (57.2%), headache (27.6%), leukopenia (23.4%), anemia (22.1%) and rash (20.7%). The most common grade 3 or higher treatment-related adverse events were thrombocytopenia (11.7%). One death was due to treatment-related adverse events (interstitial lung disease). Six interstitial lung diseases (2.1%) were observed during study treatment. Conclusion: D-0316 has showed strong anti-tumor activities and tolerable toxicity in pts with EGFR T790M-positive NSCLC who have progressed after EGFR-TKI treatment. Citation Format: Shun Lu, Yiping Zhang, Guojun Zhang, Jianying Zhou, Shundong Cang, Ying Cheng, Gang Wu, Peiguo Cao, Dongqing Lv, Xiangming Jin, Hong Jian, Chengshui Chen, Guanming Jiang, Panwen Tian, Kai Wang, Hui Zhao, Gongyan Chen, Qun Chen, Cuimin Ding, Junquan Yang, Renhua Guo, Guoping Sun, Bin Wang, Liyan Jiang, Wu Zhuang, Zhe Liu, Jian Fang, Yunpeng Liu, Jian Zhang, Jun Chen, Yueyin Pan, Qitao Yu, Min Zhao, Jiuwei Cui, Dianming Li, Tienan Yi, Zhuang Yu, Yan Yang, Yan Zhang, Xiuyi Zhi, Yunchao Huang, Rong Wu, Liangan Chen, Aimin Zang, Lejie Cao, Qingshan Li, Xiaoling Li, Yong Song, Donglin Wang, Shucai Zhang. D-0316 in patients with advanced T790M-positive EGFR-mutant non-small cell lung cancer who progressed on prior EGFR-TKI therapy: results from a phase II study (NCT03861156) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr CT170.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 11 ( 2017-06-01), p. 3082-3091
    Abstract: Cancer stem–like cells (CSC) are thought to drive tumor initiation, metastasis, relapse, and therapeutic resistance, but their specific pathogenic characters in many cancers, including non–small cell lung cancer (NSCLC), have yet to be well defined. Here, we develop findings that the growth factor HGF promotes CSC sphere formation in NSCLC cell populations. In patient-derived sphere-forming assays (PD-SFA) with HGF, CD49f and CD104 were defined as novel markers of lung CSC (LCSC). In particular, we isolated a subpopulation of CD166+CD49fhiCD104−Lin− LCSC present in all human specimens of NSCLC examined, regardless of their histologic subtypes or genetic driver mutations. This specific cell population was tumorigenic and capable of self-renewal, giving rise to tumor spheres in vitro and orthotopic lung tumors in immune-compromised mice. Mechanistic investigations established that NOTCH1 was preferentially expressed in this cell subpopulation and required for self-renewal via the transcription factor HES1. Through a distinct HES1-independent pathway, NOTCH1 also protected LCSCs from cisplatin-induced cell death. Notably, treatment with a γ-secretase inhibitor that blunts NOTCH1 function ablated self-renewing LCSC activity and restored platinum sensitivity in vitro and in vivo. Overall, our results define the pathogenic characters of a cancer stem–like subpopulation in lung cancer, the targeting of which may relieve platinum resistance in this disease. Cancer Res; 77(11); 3082–91. ©2017 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2005
    In:  Cancer Research Vol. 65, No. 8 ( 2005-04-15), p. 3389-3395
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 65, No. 8 ( 2005-04-15), p. 3389-3395
    Abstract: Clioquinol, a metal chelator, has been used for many years as an antimicrobial agent and more recently as a potential treatment for Alzheimer's disease. Because it binds copper and zinc, metals essential for the activity of the enzyme superoxide dismutase-1 (SOD1), a potential target for anticancer drug development, we investigated its effects on human cancer cells. Treatment with clioquinol reduced the viability of eight different human cancer cell lines in a concentration-dependent manner, with IC50 values in the low micromolar range. Biochemical analysis revealed that clioquinol induced cancer cell death through apoptotic pathways that require caspase activity. Although clioquinol induced modest inhibition of SOD1 activity in treated cells, comparable inhibition by a known SOD1 inhibitor, diethyldithiocarbamate, did not result in cytotoxicity. The addition of copper, iron, or zinc did not rescue cells from cliquinol-induced cytotoxicity but enhanced its killing, arguing against metal chelation as its major mechanism of action. To test if clioquinol might act as an ionophore, a fluorescent probe was used to monitor intracellular zinc concentrations. The addition of clioquinol resulted in elevated levels of intracellular zinc, indicating that clioquinol acts as a zinc ionophore. In an in vivo xenografts mouse model, clioquinol inhibited tumor growth of xenografts over a 6-week period, without inducing visible toxicity. Our results show that clioquinol has anticancer effects both in vitro and in vivo. Transition metal ionophores may be a subclass of metal chelators with anticancer activity deserving of further development.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2005
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 4194-4194
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 4194-4194
    Abstract: Background: Superoxide dismutase 1 (SOD1), the ubiquitously expressed and predominant superoxide dismutase, catalyzes the conversion of superoxide ions into hydrogen peroxide which will be further detoxified by catalase or glutathione peroxidase. The SOD1 gene has been reported to be over-expressed in human cancer cells, and targeting SOD1 has been proposed as a new strategy for cancer therapy. While the transcriptional regulation of the SOD1 gene has been well-characterized, the contribution of the SOD1 3′-UTR to SOD1 gene expression has not been previously examined in any model systems. Methods: The full length (325 bases) and different fragments of the SOD1 3′UTR were amplified from human cancer cells. Each of the amplified products was inserted into a pGL3-promoter reporter vector downstream of the luciferase cDNA. The reporter constructs were transfected into Panc-1 (pancreatic cancer), A2780 (ovarian cancer), and HepG2 (liver cancer) cells and luciferase activity was analyzed. The wild type pGL3-promoter vector and the antisense orientation of the SOD1 3′-UTR reporter construct served as controls. Luciferase mRNA stability was examined by reverse transcriptase real-time PCR. Western blot was performed to examine endogenous SOD1 protein expression levels. Results: The luciferase activity was 10 (HepG2) and 70 (A2780 and Panc-1) fold higher in cells transfected with the full-length SOD1 3′-UTR compared with those in control cells. The dramatically increased luciferase activity was likely attributed to enhanced luciferase mRNA stability as analyzed by real-time RT-PCR. Significantly reduced luciferase activities were found in cells transfected with the reporter constructs bearing different fragments of the SOD1 3′-UTR. Most noticeably, transfection with the reporter construct bearing a deletion of the first 200 bases of the SOD1 3′-UTR brought the reporter activity down to a level similar to that in controls. Further deletion analysis showed that the secondary structure of the SOD1 3′-UTR may also play a key role in SOD1 3′-UTR-mediated gene expression and the putative AU-rich elements are unlikely to confer the sustained SOD1 gene expression. Moreover, we found that the full-length SOD1 3′-UTR can serve as a decoy or competitor to attenuate endogenous SOD1 protein expression, indicating the involvement of trans-acting RNA binding factors in maintaining high expression of the SOD1 gene. Summary: The SOD1 3′-UTR maintains the high expression of the SOD1 gene in human cancer cells likely through increasing SOD1 mRNA stability. The interaction of the SOD1 3′-UTR with the trans-acting RNA binding proteins is required to maintain SOD1 gene expression. We are actively working on identifying these RNA binding proteins and their interacting cis-elements in the SOD1 3′-UTR in our model systems. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4194. doi:1538-7445.AM2012-4194
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 707-707
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 707-707
    Abstract: . DNA polymerase iota (Pol iota) is an error-prone DNA polymerase involved in translesion DNA synthesis (TLS) that may play a significant role in the accumulation of DNA mutations. Our previous studies identified the elevated expression of Pol iota in human esophageal squamous cell cancer (ESCC) tissues and revealed that Pol iota contributes to ESCCs’ progression. The present study aimed to investigate the molecular mechanism by which Pol iota enhances the invasiveness and metastasis of ESCC cells. We found that the expression of Pol iota was higher in ESCCs with lymph node metastasis compared to those without lymph node metastasis. Kaplan-Meier analysis revealed a negative correlation between the expression of Pol iota and patients’ prognosis. Furthermore, the expression of matrix metalloproteinase-2(MMP-2) and matrix metalloproteinase-9(MMP-9), both being essential regulators for cells invasion, were associated with that of Pol iota in ESCCs tissue samples. The wound healing and transwells assay revealed that over-expression of Pol iota enhances motility and invasiveness of ESCC cells. In vivo, up-regulation of Pol iota promoted the colonization of ESCC cells in the liver, lung and kidney. Signaling pathway analysis showed that Pol iota induces the expression of MMP-2/9 and enhances the ESCCs progression via JNK-AP-1 cascade. Altogether, our finding demonstrated the underlying mechanism by which Pol iota promotes ESCCs’ development. Citation Format: Shitao Zou, Jinchang Wu, Wei-Qun Ding, Jundong Zhou. High expression of Pol é promotes invasion and metastases in esophageal squamous cell carcinoma. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 707.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 5205-5205
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 5205-5205
    Abstract: DHA is a long-chain omega-3 polyunsaturated fatty acid that has anticancer properties, including the ability to suppress tumor angiogenesis. However, the precise mechanism of DHAs anti-angiogenic activity is not well defined. It is recognized that the intercommunication between cancer cells and their microenvironment is essential to tumor angiogenesis. Exosomes are nanometer-sized extracellular vesicles that are important mediators of intercellular communication and in recent years, the role of tumor-derived exosomes in cancer progression has been realized. However, ways to limit or alter their influence on cancer progression has not been demonstrated. At this time, very little is known about the role of breast cancer derived exosomes in contributing to breast cancer progression and whether these exosomes mediate DHAs anticancer activity. To investigate, breast cancer exosomes were collected from the conditioned media of MCF7 and MDA-MB-231 breast cancer cells engineered to express GFP-tagged CD63 after treatment with DHA. We observed an increase in exosome secretion from the DHA-treated cells. Total RNA was extracted from the DHA-treated and control MCF7-derived exosomes and analyzed by small RNA sequencing. The expression of 83 exosome microRNAs was altered by DHA ( & gt;2-fold) and several of the most abundant exosomal microRNAs (miR-23b, miR-27a/b, miR-21, and miR-320b) are known to have anti-angiogenic activity. When DHA-treated MCF7 cells were co-cultured with or the collected exosomes were directly applied to endothelial cell cultures, we observed an increase in the expression of these microRNAs in endothelial cells. Furthermore, overexpression of miR-23b and miR-320b in endothelial cells decreased the expression of their pro-angiogenic target genes (PLAU, AMOTL1, NRP1 and ETS2) and significantly inhibited tube formation by endothelial cells, an effect that could be reversed by inhibition of exosome secretion via Rab27A knockdown. These results suggest that the microRNAs transferred by exosomes mediate DHAs anti-angiogenic action. Our data demonstrates that DHA alters breast cancer exosome secretion and microRNA contents that leads to the inhibition of endothelial tube formation. These results provide insight into the future possibility of developing new cancer therapeutic strategies targeting exosome secretion and content transmission. Citation Format: Bethany N. Hannafon, Karla Carpenter, William Berry, Ralf Janknecht, William Dooley, Wei-Qun Ding. Docosahexaenoic acid (DHA) alters breast cancer exosome-mediated microRNA signaling. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5205. doi:10.1158/1538-7445.AM2015-5205
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Molecular Cancer Therapeutics Vol. 10, No. 11_Supplement ( 2011-11-12), p. B7-B7
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 10, No. 11_Supplement ( 2011-11-12), p. B7-B7
    Abstract: Background: Hypoxia inducible factor-1α (HIF-1α) is a therapeutic target in solid tumors because it activates transcription of the genes associated with tumor growth, angiogenesis and chemo/radio-resistance. Activation of PPARα has been demonstrated to inhibit tumor growth and angiogenesis, yet the mechanisms behind these actions remain obscure. The present study examined the effects of PPARα activation on HIF-1α mediated signaling in human cancer cells. Methods: The human cancer lines MCF-7 (breast) and A2780 (ovarian), and human endothelial line, EA.hy926 were used as model systems for this study. Hypoxia was achieved via a hypoxia chamber. The HRE-luciferase reporter gene and Western blot were applied to analyze HIF-1α expression and activity. RT-PCR and co-immunoprecipitation were performed to understand the molecular mechanisms of PPARα-mediated suppression of HIF-1α signaling. The level of vascular endothelial growth factor (VEGF) in cultured medium was determined using an ELISA, and in vitro angiogenesis was evaluated using the tube formation assay. Results: Incubation of cancer cells with 1 % oxygen for 16 hours significantly induced HIF-1α expression and activity. Treatment of the cells with PPARα agonists, but not a PPARγ agonist, prior to hypoxia diminished hypoxia-induced HIF-1α expression and activity. Consistently, a PPARα antagonist attenuated PPARα agonist-induced suppression of HIF-1α signaling. Over-expression of an HA-tagged HIF-1α and HIF-1α-P402/P564A mutant protein demonstrated that activation of PPARα promotes HIF-1 α degradation in our model system. This was further confirmed by the use of MG-132, a proteasome inhibitor, which reversed PPARα-mediated suppression of the HIF-1α signaling, and by RT-PCR analysis of HIF-1α mRNA, which was unchanged by PPARα activation. Using the co-immunoprecipitation technique, we found that activation of PPARα enhances the binding of hydroxylated HIF-1α to the Von Hippel-Lindau tumor suppressor, a protein known to mediate HIF-1α degradation through the ubiquitin-proteasome pathway. Consequential to the PPARα-mediated suppression of HIF-1α signaling, vascular endothelial growth factor secretion from cancer cells was found to be significantly reduced, and the tube formation by EA.hy926 cells was dramatically impaired. Summary: Activation of PPARα suppresses HIF-1α expression and activity by promoting HIF-1α degradation in cancer cells. PPARα agonists are potential anticancer agents that inhibit tumor angiogenesis via the suppression of HIF-1α signaling. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr B7.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 2048-2048
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 2048-2048
    Abstract: Tissue factor is recognized to be an important regulator of tumor angiogenesis and metastasis, independent of its participation in the blood coagulation pathway. The tissue factor gene is highly expressed only in breast carcinoma cells having high invasive potential, but the mechanisms regulating tissue factor gene expression in breast cancer cells remain unclear. This study examined the hypothesis that microRNA regulation of tissue factor expression is responsible for over-expression of the tissue factor gene in invasive breast cancer cells. Tissue factor was highly expressed in the highly invasive MDA-MB-231 line but was barely detectable in the less invasive MCF-7 line, as assayed by RT-PCR and western blot. A reporter gene assay showed that the tissue factor promoter can be activated in extracts prepared from MCF-7 cells. Forced expression of tissue factor cDNA was achieved in MCF-7 cells, implying that that tissue factor expression is regulated by the 3′-UTR of its transcript. Bioinformatics analysis suggested that microRNA species such as miR-19 (miR-19a and 19b) and miR-106b interact with the 3’-UTR of the tissue factor transcript and may regulate tissue factor protein translation and/or mRNA degradation. microRNA array confirmed that miR-19b is highly expressed in MCF-7 cells, as compared to MDA-MB-231 cells. A reporter gene assay using a TF-3′-UTR-luciferase construct indicated that the tissue factor 3′-UTR negatively regulates tissue factor expression, an effect that was completely reversed by deletion of the miR-19 binding site in the reporter construct. Application of an antisense microRNA for miR-19 enhanced tissue factor expression in MCF-7 cells. This study demonstrates that tissue factor expression in breast cancer cells is regulated by miR-19. The expression pattern of miR-19 in breast cancer tissues and its role in mediating angiogenesis and metastasis merit future investigation. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2048.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2004
    In:  Molecular Cancer Therapeutics Vol. 3, No. 9 ( 2004-09-01), p. 1109-1117
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 3, No. 9 ( 2004-09-01), p. 1109-1117
    Abstract: Docosahexaenoic acid (DHA, 22:6 n-3), a polyunsaturated fatty acid found in fish oil, exerts cytotoxic effects on cancer cells. Although DHA was toxic toward five human cancer cell lines (MCF-7, MDA-MB-231, SiHa, Raji, and DHL-4), the lines were not uniformly sensitive. DHL-4, a bcl-2 overexpressing lymphoid line, was the most sensitive (IC50, 5.2 μmol/L) and the cervical cancer cell line, SiHa, was the most resistant (IC50, & gt;300 μmol/L). Lipid peroxidation has been cited by others as an important component of DHA toxicity, and we confirmed that vitamin E prevents the cytotoxic effects of DHA. Lipid peroxidation was greater following DHA treatment of the sensitive DHL-4 cells than in the resistant SiHa cells, as assessed by thiobarbituric acid reactive substance generation. DHL-4 cells treated with DHA for 20 hours showed a 3.5-fold increase in thiobarbituric acid reactive substances, whereas SiHa cells showed no increase. Reverse transcription-PCR analysis detected a down-regulation of the expression of the major antioxidant enzyme, superoxide dismutase (SOD) 1, in DHL-4 cells but not in SiHa cells after DHA treatment. Knockdown of SOD1 expression in SiHa cells with small interfering RNA significantly enhanced lipid peroxidation and cytotoxicity on exposure to DHA. These results show that DHL-4 cells are highly sensitive to the cytotoxic effect of DHA and that regulation of SOD1 expression may play an important role in determining the sensitivity of different tumor cells to the cytotoxic effects of DHA.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2004
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages