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: Molecular Oncology, Wiley, Vol. 10, No. 7 ( 2016-08), p. 1099-1117
    Abstract: Mifepristone triggers the unfolded protein response (UPR) in ovarian cancer cells. Mifepristone‐induced UPR is mediated by increased mRNA translation rate. Mifepristone increases autophagic flux in ovarian cancer cells. Mifepristone is lethal to ovarian cancer cells when combined with chloroquine. Mifepristone kills ovarian cancer cells in combination with proteasome inhibitors.
    Type of Medium: Online Resource
    ISSN: 1574-7891 , 1878-0261
    Language: English
    Publisher: Wiley
    Publication Date: 2016
    detail.hit.zdb_id: 2322586-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 19, No. 19_Supplement ( 2013-10-01), p. A23-A23
    Abstract: Ovarian cancer is rare; yet, it is the deadliest gynecologic malignancy with a 5-year survival rate that has only improved 9% in the past 30 years and a dismal overall survival rate that has remained stagnant for over 50 years. Although most patients diagnosed with advanced disease undergo remission after optimal surgical cytoreduction and platinum (Pt)-based chemotherapy, microscopic foci of cells defined as minimal residual disease (MRD) manage to survive within the peritoneal cavity and recreate the disease (recurrence) that develops a more aggressive phenotype for which current therapies almost always fail. Thus, it is critical to take advantage of the time the patient is in remission to attack the cells constituting the MRD by disengaging their repopulation capacity. Evidence suggests the feasibility of this consolidation therapeutic approach using cytostatic agents. Previously, we demonstrated that the repopulation of cells escaping Pt or Pt-taxane chemotherapy can be abrogated using steroids with antiprogestin/antiglucocorticoid activity, such as mifepristone (MF). We found that MF (i) blocks ovarian cancer cell growth by inducing G1 cell cycle arrest mediated by the up-regulation of cyclin dependent kinase inhibitors p21 and p27, leading to the blockage of the activity of cyclin dependent kinase 2 (Cdk2) and, consequently, lack of DNA synthesis and S phase progression; (ii) induces cell death in combination with blockage of the PI3K/Akt survival pathway; and (iii) it does not require progesterone receptors to exert its anti-growth effect. To further elucidate the cytotoxic mechanism triggered by MF in ovarian cancer cells, we performed genomic and proteomic screenings using ovarian cancer cell lines of different genetic backgrounds and sensitivities to Pt. We exposed OV2008 (wt p53, Pt sensitive), OV2008/C13 (wt p53/Pt resistant), or SK-OV-3 (null p53, Pt semi-resistant) cells to cytostatic concentrations of MF for 24, 48 or 72 h. Total RNAs and whole protein extracts were subjected to cDNA microarray analysis and two-dimensional nano-liquid chromatography (2D-nanoLC)-mass spectrometry (MS/MS), respectively. The combined use of cDNA microarray and 2D nanoLC-MS/MS to discover mRNAs and proteins differentially regulated by MF led to the identification of key proteins involved in the regulation of the unfolded protein response (UPR), which were further validated by Western blotting. We identified the master ER chaperone glucose regulated protein (GRP) 78 (also referred to as BiP/HSPA5) highly up-regulated by MF. Also the ER stress sensors protein kinase RNA-like ER kinase (PERK) and inositol-requiring kinase 1 (IRE1alpha) were up-regulated by MF. Activation of PERK and IRE1alpha signaling pathway usually leads to attenuated global protein synthesis thus contributing to cell cycle arrest. We found the translation repressor protein 4E-BP1 augmented by MF suggesting reduced protein translation capability. We also found highly up-regulated the transcription factor C/EBP homologous protein (CHOP), the induction of which is usually linked to cell death. In summary, we identified the UPR as a target for MF activity. We hypothesize that the UPR integrates the cytotoxicity of MF towards ovarian cancer cells when used as monotherapy or in combination therapies triggering, respectively, cell cycle arrest (cytostasis) or cell death (lethality) depending on the degree of cellular stress generated. Citation Format: Maria B. Hapon, Carlos D. Gamarra-Luques, Alicia A. Goyeneche, Eduardo A. Callegari, Kathleen M. Eyster, Carlos M. Telleria. Induction of the unfolded protein response in ovarian cancer cells exposed to cytostatic concentrations of antiprogestin/antiglucocorticoid mifepristone. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research: From Concept to Clinic; Sep 18-21, 2013; Miami, FL. Philadelphia (PA): AACR; Clin Cancer Res 2013;19(19 Suppl):Abstract nr A23.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2014
    In:  Journal of Ovarian Research Vol. 7, No. 1 ( 2014-12)
    In: Journal of Ovarian Research, Springer Science and Business Media LLC, Vol. 7, No. 1 ( 2014-12)
    Type of Medium: Online Resource
    ISSN: 1757-2215
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2014
    detail.hit.zdb_id: 2455679-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    Elsevier BV ; 1999
    In:  Steroids Vol. 64, No. 11 ( 1999-11), p. 760-766
    In: Steroids, Elsevier BV, Vol. 64, No. 11 ( 1999-11), p. 760-766
    Type of Medium: Online Resource
    ISSN: 0039-128X
    Language: English
    Publisher: Elsevier BV
    Publication Date: 1999
    detail.hit.zdb_id: 1498762-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    SAGE Publications ; 2013
    In:  Cancer Growth and Metastasis Vol. 6 ( 2013-01), p. CGM.S11124-
    In: Cancer Growth and Metastasis, SAGE Publications, Vol. 6 ( 2013-01), p. CGM.S11124-
    Abstract: We have previously shown that the antiprogestin and antiglucocorticoid mifepristone inhibits the growth of ovarian cancer cells. In this work, we hypothesized that cellular stress caused by mifepristone is limited to cytostasis and that cell killing is avoided as a consequence of the persistent activity of the PI3K/Akt survival pathway. To investigate the role of this pathway in mifepristone-induced growth inhibition, human ovarian cancer cells of various histological subtypes and genetic backgrounds were exposed to cytostatic doses of mifepristone in the presence or absence of the PI3K inhibitor, LY294002. The activation of Akt in ovarian cancer cells, as marked by its phosphorylation on Ser473, was not modified by cytostatic concentrations of mifepristone, but it was blocked upon treatment with LY294002. The combination mifepristone/LY294002, but not the individual drugs, killed ovarian cancer cells via apoptosis, as attested by genomic DNA fragmentation and cleavage of caspase-3, and the concomitant downregulation of antiapoptotic proteins Bcl-2 and XIAP. From a pharmacological standpoint, when assessing cell growth inhibition using a median-dose analysis algorithm, the interaction between mifepristone and LY294002 was synergistic. The lethality caused by the combination mifepristone/LY294004 in 2-dimensional cell cultures was recapitulated in organized, 3-dimensional spheroids. This study demonstrates that mifepristone and LY294002 when used individually cause cell growth arrest; yet, when combined, they cause lethality.
    Type of Medium: Online Resource
    ISSN: 1179-0644 , 1179-0644
    Language: English
    Publisher: SAGE Publications
    Publication Date: 2013
    detail.hit.zdb_id: 2594639-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2012
    In:  BMC Cancer Vol. 12, No. 1 ( 2012-12)
    In: BMC Cancer, Springer Science and Business Media LLC, Vol. 12, No. 1 ( 2012-12)
    Abstract: Advanced ovarian cancer is treated with cytoreductive surgery and combination platinum- and taxane-based chemotherapy. Although most patients have acute clinical response to this strategy, the disease ultimately recurs. In this work we questioned whether the synthetic steroid mifepristone, which as monotherapy inhibits the growth of ovarian cancer cells, is capable of preventing repopulation of ovarian cancer cells if given after a round of lethal cisplatin-paclitaxel combination treatment. Methods We established an in vitro approach wherein ovarian cancer cells with various sensitivities to cisplatin or paclitaxel were exposed to a round of lethal doses of cisplatin for 1 h plus paclitaxel for 3 h. Thereafter, cells were maintained in media with or without mifepristone, and short- and long-term cytotoxicity was assessed. Results Four days after treatment the lethality of cisplatin-paclitaxel was evidenced by reduced number of cells, increased hypodiploid DNA content, morphological features of apoptosis, DNA fragmentation, and cleavage of caspase-3, and of its downstream substrate PARP. Short-term presence of mifepristone either enhanced or did not modify such acute lethality. Seven days after receiving cisplatin-paclitaxel, cultures showed signs of relapse with escaping colonies that repopulated the plate in a time-dependent manner. Conversely, cultures exposed to cisplatin-paclitaxel followed by mifepristone not only did not display signs of repopulation following initial chemotherapy, but they also had their clonogenic capacity drastically reduced when compared to cells repopulating after cisplatin-paclitaxel. Conclusions Cytostatic concentrations of mifepristone after exposure to lethal doses of cisplatin and paclitaxel in combination blocks repopulation of remnant cells surviving and escaping the cytotoxic drugs.
    Type of Medium: Online Resource
    ISSN: 1471-2407
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2012
    detail.hit.zdb_id: 2041352-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Elsevier BV ; 1995
    In:  The Journal of Steroid Biochemistry and Molecular Biology Vol. 52, No. 6 ( 1995-6), p. 567-573
    In: The Journal of Steroid Biochemistry and Molecular Biology, Elsevier BV, Vol. 52, No. 6 ( 1995-6), p. 567-573
    Type of Medium: Online Resource
    ISSN: 0960-0760
    Language: English
    Publisher: Elsevier BV
    Publication Date: 1995
    detail.hit.zdb_id: 1482780-3
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Epidemiology, Biomarkers & Prevention Vol. 19, No. 10_Supplement ( 2010-10-01), p. A113-A113
    In: Cancer Epidemiology, Biomarkers & Prevention, American Association for Cancer Research (AACR), Vol. 19, No. 10_Supplement ( 2010-10-01), p. A113-A113
    Abstract: The overall goal of our research is to study the feasibility of “repositioning” antiprogestin compounds originally designed for contraceptive purposes for ovarian cancer therapeutics. Ovarian cancer is the most deadly disease of the reproductive tract in women. When the disease is diagnosed, in most cases abnormal growths have already progressed beyond the confines of the ovaries and into the nearby fallopian tubes, uterus, and various other sites within the peritoneal cavity. As a result, the majority of patients diagnosed with ovarian cancer require surgery followed by platinum-based chemotherapy. Yet the efficacy of this therapy is hindered by the elevated toxicity of platinum derivatives, the development of mechanisms to evade drug toxicity, and the repopulation of cells between treatment intervals. Thus, finding therapeutic interventions to overcome the limitations of platinum-based therapies is of critical clinical relevance. Our laboratory published that the prototypical antiprogestin mifepristone has potent anti-ovarian cancer activity in vitro and in vivo (1); is cytostatic at pharmacological doses in platinum-sensitive and platinum-resistant ovarian cancer cells, and regardless of p53 tumor suppressor expression (1-2); and when intertwined among courses of cisplatin treatment, prevents repopulation of ovarian cancer cells (3). We now provide data demonstrating that mifepristone potentiates the lethality of cisplatin. We show that the toxicity of cisplatin can be enhanced 4 fold when associated to cytostatic concentrations of antiprogestin mifepristone. We also demonstrate that mifepristone potentiates the long-term effect of short-term exposure of OV2008 cells to sub-lethal doses of cisplatin. Finally we provide evidence that mifepristone enhances cisplatin toxicity in platinum sensitive OV2008 (wt p53) and A2780 (wt p53) lines, their platinum-resistant siblings, OV2008/C13 (wt p53) and A2780/CP70 (mut p53), and semi-platinum-resistant SK-OV-3 (null p53). In summary, we demonstrate that the prototypical antiprogestin mifepristone potentiates cisplatin toxicity regardless of platinum sensitivity and p53 genetic background. Our studies provide proof-of-principle that antiprogestin-of which mifepristone is FDA approved for reproductive medicine-can be “repurposed” for another modality-of-use as part of the chemotherapeutic armamentarium for ovarian cancer patients. The strategy of adding an antiprogestin such as mifepristone to the platinum-based chemotherapeutic schedule in ovarian cancer should allow reducing the number of platinum cycles or the dose of the platinum agent without losing efficacy in terms of tumor growth inhibition yet reducing undesirable side effects. References: 1. Goyeneche AA, Caron RW, Telleria CM. Mifepristone inhibits ovarian cancer cell growth in vitro and in vivo. Clin Cancer Res. 2007 Jun 1;13(11):3370-9. 2. Freeburg EM, Goyeneche AA, Seidel EE, Telleria CM. Resistance to cisplatin does not affect sensitivity of human ovarian cancer cell lines to mifepristone cytotoxicity. Cancer Cell Int. 2009;9:4. 3. Freeburg EM, Goyeneche AA, Telleria CM. Mifepristone abrogates repopulation of ovarian cancer cells in between courses of cisplatin treatment. Int J Oncol. 2009 Mar;34(3):743-55. Citation Information: Cancer Epidemiol Biomarkers Prev 2010;19(10 Suppl):A113.
    Type of Medium: Online Resource
    ISSN: 1055-9965 , 1538-7755
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2036781-8
    detail.hit.zdb_id: 1153420-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 2098-2098
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 2098-2098
    Abstract: We have previously shown that the antiprogestin/antiglucocorticoid mifepristone inhibits the growth of ovarian cancer cells. In this work, we hypothesized that cellular stress caused by mifepristone is limited to cytostasis and that cell killing is avoided because of a compensatory activation of the PI3K/Akt survival pathway.To investigate the role of this pathway in mifepristone-induced growth inhibition, human ovarian cancer cells of various histological subtypes and genetic backgrounds were exposed to cytostatic doses of mifepristone in the presence or absence of the PI3K inhibitor, LY294002. Mifepristone-induced growth arrest of ovarian cancer cells was associated with phosphorylation of Akt (Ser473), which was prevented by treatment with LY294002. The combination mifepristone/LY294002, but not the individual drugs, killed ovarian cancer cells via apoptosis, as attested by genomic DNA fragmentation and cleavage of caspase-3, and the concomitant downregulation of anti-apoptotic proteins Bcl-2 and XIAP. From a pharmacological standpoint, when assessing cell growth inhibition using a median-dose analysis algorithm, the interaction between mifepristone and LY294002 was synergistic. The lethality caused by the combination mifepristone/LY294004 in two dimensional cell cultures was recapitulated in organized, tri-dimensional spheroids. This study demonstrates that mifepristone and LY294002, when used individually cause cell growth arrest, yet when combined, they cause lethality. This work further provides proof-of-concept that in combination, targeted cytostatic therapies can be an efficient tool to engage cancer cells in a death program. Citation Format: Stacy L. Wempe, Carlos D. Gamarra-Luques, Carlos M. Telleria. Synergistic lethality of mifepristone and LY294002 in ovarian cancer cells. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2098. doi:10.1158/1538-7445.AM2013-2098
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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 ...
  • 10
    In: International Journal of Molecular Sciences, MDPI AG, Vol. 23, No. 19 ( 2022-09-21), p. 11070-
    Abstract: Although the COVID-19 disease has developed into a worldwide pandemic, its pathophysiology remains to be fully understood. Insulin-degrading enzyme (IDE), a zinc-metalloprotease with a high affinity for insulin, has been found in the interactomes of multiple SARS-CoV-2 proteins. However, the relevance of IDE in the innate and adaptative immune responses elicited by circulating peripheral blood mononuclear cells is unknown. Here, we show that IDE is highly expressed on the surface of circulating monocytes, T-cells (both CD4+ and CD4−), and, to a lower extent, in B-cells from healthy controls. Notably, IDE’s surface expression was upregulated on monocytes from COVID-19 patients at diagnosis, and it was increased in more severe patients. However, IDE’s surface expression was downregulated (relative to healthy controls) 3 months after hospital discharge in all the studied immune subsets, with this effect being more pronounced in males than in females, and thus it was sex-dependent. Additionally, IDE levels in monocytes, CD4+ T-cells, and CD4− T-cells were inversely correlated with circulating insulin levels in COVID-19 patients (both at diagnosis and after hospital discharge). Of note, high glucose and insulin levels downregulated IDE surface expression by ~30% in the monocytes isolated from healthy donors, without affecting its expression in CD4+ T-cells and CD4− T-cells. In conclusion, our studies reveal the sex- and metabolism-dependent regulation of IDE in monocytes, suggesting that its regulation might be important for the recruitment of immune cells to the site of infection, as well as for glucometabolic control, in COVID-19 patients.
    Type of Medium: Online Resource
    ISSN: 1422-0067
    Language: English
    Publisher: MDPI AG
    Publication Date: 2022
    detail.hit.zdb_id: 2019364-6
    SSG: 12
    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...