GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: The Lancet, Elsevier BV, Vol. 401, No. 10388 ( 2023-05), p. 1584-1594
    Type of Medium: Online Resource
    ISSN: 0140-6736
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 2067452-1
    detail.hit.zdb_id: 3306-6
    detail.hit.zdb_id: 1476593-7
    SSG: 5,21
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 9 ( 2017-05-01), p. 2453-2463
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 9 ( 2017-05-01), p. 2453-2463
    Abstract: DNA repair capacity is critical for survival of cancer cells upon therapeutic DNA damage and thus is an important determinant of susceptibility to chemotherapy in cancer patients. In this study, we identified a novel function of nuclear receptor NR1D1 in DNA repair, which enhanced chemosensitivity in breast cancer cells. NR1D1 inhibited both nonhomologous end joining and homologous recombination double-strand breaks repair, and delayed the clearance of γH2AX DNA repair foci that formed after treatment of doxorubicin. PARylation of NR1D1 by PARP1 drove its recruitment to damaged DNA lesions. Deletion of the ligand binding domain of NR1D1 that interacted with PARP1, or treatment of 6-(5H)-phenanthridinone, an inhibitor of PARP1, suppressed the recruitment of NR1D1 to DNA damaged sites, indicating PARylation as a critical step for the NR1D1 recruitment. NR1D1 inhibited recruitment of the components of DNA damage response complex such as SIRT6, pNBS1, and BRCA1 to DNA lesions. Downregulation of NR1D1 in MCF7 cells resulted in resistance to doxorubicin, both in vitro and in vivo. Analysis of four public patient data sets indicated that NR1D1 expression correlates positively with clinical outcome in breast cancer patients who received chemotherapy. Our findings suggest that NR1D1 and its ligands provide therapeutic options that could enhance the outcomes of chemotherapy in breast cancer patients. Cancer Res; 77(9); 2453–63. ©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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 3610-3610
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 3610-3610
    Abstract: The most accepted therapeutic regimen for treating ER-alpha breast cancer is Tamoxifen. Yet over 30% of patients gain resistance. Recently, lipid metabolism has been rising as a new mode of cancer drug resistance. In order for a rapid growth, cancer cells show a strong avidity for lipid which they obtain by de novo lipogenesis or increasing the uptake of exogenous lipids. Lipid metabolism may confer drug resistance, but little is known about its association with endocrine resistance. Thus, we aimed to investigate the association of lipid metabolism in the context of tamoxifen resistance. First, fatty acid translocase and membrane-associated fatty acid binding protein, both of which are responsible for fatty acid uptake, were overexpressed in MCF7/TAMR-8 and T47D/TR-1, TR-2 cell lines. Also the mRNA level of fatty acid activating enzyme, long-chain fatty-acid-coenzyme A, were elevated as well. Next, through Oil-red-O staining and Nile red staining followed by FACS analysis, we examined whether tamoxifen-resistant cells actually increase fatty acid uptake. When supplemented with free fatty acids, tamoxifen-resistant cell lines showed higher efficiency in fatty acid uptake compared to the control cell lines. Subsequently when the amount of triglyceride accumulation was measured, more TG were present in resistant cells at the basal level and after free fatty acid treatment. Taken together, these results suggest lipid uptake by cancer cells may play a role in driving tamoxifen-resistance in breast cancer. Citation Format: Sewon Hwang, Mi-Ock Lee. Cellular fatty acid uptake is enhanced in tamoxifen-resistant breast cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3610.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 18 ( 2023-09-15), p. 3045-3058
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 18 ( 2023-09-15), p. 3045-3058
    Abstract: Potentiating antitumor immunity is a promising therapeutic approach for treating a variety of cancers, including breast cancer. One potential strategy to promote antitumor immunity is targeting DNA damage response. Given that the nuclear receptor NR1D1 (also known as REV-ERBα) inhibits DNA repair in breast cancer cells, we explored the role of NR1D1 in antitumor CD8+ T-cell responses. First, deletion of Nr1d1 in MMTV-PyMT transgenic mice resulted in increased tumor growth and lung metastasis. Orthotopic allograft experiments suggested that loss of Nr1d1 in tumor cells rather than in stromal cells played a prominent role in increasing tumor progression. Comprehensive transcriptome analyses revealed that biological processes including type I IFN signaling and T cell–mediated immune responses were associated with NR1D1. Indeed, the expression of type I IFNs and infiltration of CD8+ T cells and natural killer cells in tumors were suppressed in Nr1d1−/−;MMTV-PyMT mice. Mechanistically, NR1D1 promoted DNA damage–induced accumulation of cytosolic DNA fragments and activated cGAS-STING signaling, which increased the production of type I IFNs and downstream chemokines CCL5 and CXCL10. Pharmacologic activation of NR1D1 by its ligand, SR9009, enhanced type I IFN–mediated antitumor immunity accompanied by the suppression of tumor progression and lung metastasis. Taken together, these findings reveal the critical role of NR1D1 in enhancing antitumor CD8+ T-cell responses, suggesting that NR1D1 may be a good therapeutic target for breast cancer. Significance: NR1D1 suppresses breast cancer progression and lung metastasis by enhancing antitumor immunity via cGAS-STING pathway activation, which provides potential immunotherapeutic strategies for breast cancer.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Wiley ; 2016
    In:  The Journal of Dermatology Vol. 43, No. 11 ( 2016-11), p. 1381-1382
    In: The Journal of Dermatology, Wiley, Vol. 43, No. 11 ( 2016-11), p. 1381-1382
    Type of Medium: Online Resource
    ISSN: 0385-2407 , 1346-8138
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2016
    detail.hit.zdb_id: 2222121-9
    Location Call Number Limitation 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. 1637-1637
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1637-1637
    Abstract: The Liver X Receptor (LXR) is a well-established ligand-regulated transcription factor involved in cholesterol homeostasis, lipid metabolism, and inflammation. Recent studies have reported a strong antiproliferative effect of LXRs in multiple types of cancer including prostate, colon, lung, and breast cancer. However, little is known about its association with cancer metastasis. Therefore we aimed to investigate the potential of LXR ligands in the context of breast cancer cell migration, and its underlying molecular mechanism. We first performed 2D-migration assay in 4T1 breast cancer cells. Treatment of an LXR antagonist significantly reduced the rate of 4T1 cell migration. However, LXR agonists such as T0901317, GW3965 did not have much effect. Next, we investigated the molecular mechanism of cell migration. Since the degree of invasion and metastasis of breast carcinoma is well correlated with elevated expression of metastasis-associated protein 1 (MTA1), we examined whether MTA1 expression could be regulated by LXR ligands. Interestingly, when LXR agonists were treated, increased protein expression of MTA1 and its downstream target protein HIF-1α was observed in MCF7, MDA-MB-231, and 4T1-Luc breast cancer cell lines. Transient expression of LXRα demonstrated the same results as well. Furthermore, the treat of 22(S)-HC down-regulated MTA1 and HIF-1α protein expressions. Together, our findings indicate that LXR influences migratory pattern of breast cancer cells, which may affect metastatic property of breast cancer cells via LXR-dependent regulation of MTA1 and HIF-1α expression. Citation Format: Sewon Hwang, Tae Young Na, Hyelin Na, Minho Lee, Mi-Ock Lee. 22(S)-Hydroxycholesterol, an antagonist of LXR, inhibits breast cancer cell migration. [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 1637.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Wiley ; 2017
    In:  The Journal of Dermatology Vol. 44, No. 4 ( 2017-04), p. 470-471
    In: The Journal of Dermatology, Wiley, Vol. 44, No. 4 ( 2017-04), p. 470-471
    Type of Medium: Online Resource
    ISSN: 0385-2407 , 1346-8138
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2017
    detail.hit.zdb_id: 2222121-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...