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
Filter
  • American Association for Cancer Research (AACR)  (6)
  • 1
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 18, No. 9 ( 2019-09-01), p. 1615-1627
    Abstract: In B-cell acute lymphoblastic leukemia (B-ALL), activation of Notch signaling leads to cell-cycle arrest and apoptosis. We aimed to harness knowledge acquired by understanding a mechanism of Notch-induced cell death to elucidate a therapeutically viable target in B-ALL. To this end, we identified that Notch activation suppresses Polo-like kinase 1 (PLK1) in a B-ALL–specific manner. We identified that PLK1 is expressed in all subsets of B-ALL and is highest in Philadelphia-like (Ph-like) ALL, a high-risk subtype of disease. We biochemically delineated a mechanism of Notch-induced PLK1 downregulation that elucidated stark regulation of p53 in this setting. Our findings identified a novel posttranslational cascade initiated by Notch in which CHFR was activated via PARP1-mediated PARylation, resulting in ubiquitination and degradation of PLK1. This led to hypophosphorylation of MDM2Ser260, culminating in p53 stabilization and upregulation of BAX. shRNA knockdown or pharmacologic inhibition of PLK1 using BI2536 or BI6727 (volasertib) in B-ALL cell lines and patient samples led to p53 stabilization and cell death. These effects were seen in primary human B-ALL samples in vitro and in patient-derived xenograft models in vivo. These results highlight PLK1 as a viable therapeutic target in B-ALL. Efficacy of clinically relevant PLK1 inhibitors in B-ALL patient-derived xenograft mouse models suggests that use of these agents may be tailored as an additional therapeutic strategy in future clinical studies.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 101-101
    Abstract: Colorectal cancer (CRC) is the second highest cause of cancer deaths in the United States according to Center for Disease Control. Increasing evidences suggest that a diet rich in fruits and vegetables will reduce incidences of colorectal adenomas. We hypothesize that the natural compounds present in fruits and vegetables, along with their metabolites generated by the gut microbiota, could be responsible for cancer prevention by targeting the cell cycle proteins. Previous studies conducted in the laboratory have identified 2,4,6- Trihydroxybenzoic acid (2,4,6-THBA), one of the degradation products of flavonoids, as a potential chemo-preventive agent mediating its effect through inhibition of cyclin dependent kinases (CDKs). Transport of monocarboxylic acids like 2,4,6-THBA is mediated by the Solute Ligand Carrier (SLC) family of transporters; in particular SLC5A8 and SLC5A12 have been implicated as tumor suppressor proteins that are down-regulated/non-functional in various cancers. In this study, we investigated the ability of the monocarboxylic acid transporter SLC5A8 to transport 2,4,6-THBA into the mammalian cells. Our pilot studies show that the cells expressing SLC5A8 transporters have the capacity to transport 2,4,6-THBA leading to upregulation of p21 and p27 as measured by western blots and qPCR. This was associated with decreased cell number as well as decreased colony formation as measured through clonogenic assays. We suggest that chemo-preventive actions of dietary compounds containing flavonoids may occur through 2,4,6-THBA, contributing largely to the prevention of CRC. Citation Format: Ranjini Sankaranarayanan, Chaitanya K. Valiveti, Severine van Slambrouck, Siddharth S. Kesharwani, D. Ramesh Kumar, Teresa Seefeldt, Hemachand Tummala, Jayarama B. Gunaje. Studies on the flavonoid metabolite 2,4,6-trihydroxybenzoic acid as a CDK inhibitor and anti-proliferative agent: Potential role in cancer prevention [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 101.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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) ; 2018
    In:  Cancer Research Vol. 78, No. 21 ( 2018-11-01), p. 6346-6346
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 21 ( 2018-11-01), p. 6346-6346
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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) ; 2006
    In:  Cancer Research Vol. 66, No. 17 ( 2006-09-01), p. 8421-8429
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 66, No. 17 ( 2006-09-01), p. 8421-8429
    Abstract: Transcription factor NF-E2-related factor 2 (Nrf2) regulates antioxidant response element (ARE)–mediated expression and coordinated induction of chemoprotective proteins in response to chemical stress. In this report, we investigated Nrf2 response to low and high dose UVB irradiation. Low dose (7.5 J/m2) UVB exposure of mouse hepatoma, mouse keratinocyte, and human skin fibroblast cells led to the nuclear accumulation of Nrf2 and up-regulation of ARE-mediated gene expression. On the contrary, and intriguingly, high dose (20 J/m2) UVB exposure of cells led to the nuclear exclusion of Nrf2 and down-regulation of chemoprotective gene expression with possible implications in UVB carcinogenesis. We investigated the mechanism by which high dose UVB induced the nuclear exclusion of Nrf2. Prior treatment with nuclear export inhibitor, leptomycin B, abrogated the UVB-induced nuclear exclusion of Nrf2, indicating that the decrease of Nrf2 in the nucleus was due to the nuclear export of Nrf2. High dose UVB increased the phosphorylation of Nrf2Y568 which stimulated the nuclear export of Nrf2. Mutation of Nrf2Y568 to phenylalanine and src kinase inhibitor PP2 abrogated/reduced the UVB-induced phosphorylation of Nrf2Y568 and nuclear exclusion of Nrf2. Transfection with src family member Fyn small interfering RNA resulted in the nuclear accumulation of Nrf2 and an increase in the expression and UVB induction of ARE-mediated gene expression. UVB exposure also induced the nuclear localization of Fyn. These results suggest that high dose UVB induced the activation/nuclear localization of Fyn which led to increased phosphorylation of Nrf2Y568 and enhanced nuclear export of Nrf2. This resulted in nuclear exclusion of Nrf2 and down-regulation of ARE-mediated chemoprotective gene expression. (Cancer Res 2006; 66(17): 8421-9)
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2006
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 6353-6353
    Abstract: Increasing evidence supports the concept that tumor-initiating cancer stem cells (CSCs) are responsible for the relapse and reconstitution of formerly treated tumors. Previously, our lab identified a potent and selective inhibitor of colorectal CSCs, named G2.2, from a library of non-saccharide glycosaminoglycan mimetics (NSGMs). To further improve upon the potency, pharmacokinetics, and drug-like properties of G2.2, cholesterol-modified analogs were rationally designed using computational molecular modeling and molecular dynamics studies. A small group of designed NSGMs, labeled as G2C, G5C and G8C, were synthesized for biological evaluation. These agents and the parent NSGM G2.2 were studied for their ability to inhibit the growth of primary spheroids from a panel of 15 human colorectal cancer (CRC) cells representative of the consensus molecular subtypes of human CRC. Spheroid formation frequencies (SFFs) of cell lines were measured using limiting dilution assays (LDAs), and IC50 of inhibition was evaluated from dose-response curves for monolayer growth and primary spheroid growth in the presence of NSGMs. Selectivity of CSC targeting was assessed from the ratio of IC50,monolayer to IC50,spheroid. Pharmacokinetics (PK) and bioavailability in rodents were deduced following IV and PO administrations. In-vivo therapeutic potential of the NSGMs was studied in CSC-induced xenograft models aided by ex-vivo CSC phenotype characterization. Each NSGM preferentially targeted 3D spheroidal growth in comparison to monolayer cellular growth by 12- to 450-fold, suggesting excellent CSC selectivity. Cholesterol modification of G2.2 enhanced in-vitro spheroid inhibition potency across the entire panel of cell lines on average by 3- to 21-fold. G8C showed the highest inhibition potency with an IC50 of 1 to 10 μM across cell lines. Cells with higher SFF were several-fold more sensitive to NSGMs, suggesting that basal expression of CSCs could predict NSGM response. Robust reductions in tumor volumes were observed in mice treated with G5C (100 mg/kg i.p. 3x a week) following an intial-treatment with oxaliplatin and 5-fluorouracil (weekly x3) compared to vehicle controls. This reduction was concomitant with a 3.9-fold reduction in colonic crypt stemness marker LGR5+ cells and 2.6-fold decrease in xenograft-derived tertiary spheroids. No gross or organ specific toxicity was found in animals treated with G5C. Finally, cholesterol modification greatly improved PK and oral bioavailability of parent G2.2. Overall, cholesterol modification of CSC-selective G2.2 was found to enhance in vitro potency against spheroidal growth without compromising selectivity toward tumor initiating CSCs. The G2.2 analogs offered robust tumor reductions in vivo with essentially no off target toxicity, and serve as promising and novel potential cancer therapeutics. Citation Format: Connor P. O'Hara, Rio S. Boothello, Shravan Morla, Alberto Vera, Daniel K. Afosah, Nehru V. Sankaranarayanan, Balaji Nagarajan, Chetna Sharon, Bhaumik B. Patel, Umesh R. Desai. Cholesterol modification enhances potency and pharmacokinetic properties of a selective cancer stem cell targeting agent [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 6353.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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 ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 1618-1618
    Abstract: Introduction: Recent clinical trials suggest that use of erythropoietin to treat chemotherapy-induced anemia results in enhanced tumor progression and impaired survival in certain cancer patients. Rationalizing these observations with Epo's pleiotropic functionality and the lack of compelling evidence for Epo receptor (EpoR) involvement, we hypothesized the existence of a novel Epo receptor. Capturing this premise in terms of a holistic in silico strategy, we analyzed the secreted human proteome for receptors possessing structural, regulatory and functional features consistent with Epo binding and subsequent tumorigenic signaling. Our results provided EphB4 as the most likely candidate molecule. Methods: To determine the possible association between EphB4 and Epo-mediated tumor growth, we examined several ovarian (A2780, HeyA8-MDR, SKOV3ip1) and breast (MDA-231) cancer cell lines. In addition, we selected A2780 ovarian cancer cells to develop stable clones in which either EpoR or EphB4 was silenced using shRNA (A2780 shEpoR, A2780 shEphB4). Competitive and kinetic binding studies were performed using 125I- Epo. Proliferation, migration, and invasion assays were performed as described previously. To assess the role of EpoR and EphB4 in Epo-induced tumor growth, we silenced EpoR and/or EphB4 with specific siRNAs loaded into DOPC nanoliposomes. Statistical significance was established at p & lt; 0.05. Results: Binding studies with 125I- Epo revealed Epo binding to EphB4 in a low-affinity fashion. Exposure of shEpoR cells to soluble EpoR showed non-competitive inhibition; however, exposure to soluble EphB4 competitively inhibited 125I- Epo binding. These results demonstrate the specificity of Epo binding to the EphB4 receptor. At pharmacologically relevant doses, Epo treatment of shEpoR cells led to activation of the Stat-3 pathway. In addition, in vitro functional assays revealed significant effects of Epo on proliferation (p=0.003), migration (p=0.006), and invasion (p=0.02) of shEpoR cells, but not shEphB4 cells. In orthotopic ovarian (Hey-A8 MDR, SKOV3ip1, A2780) and breast (MDA-231) cancer models, treatment with Epo resulted in increased tumor growth compared to untreated animals (1.1 vs. 1.6 g, p=0.05; 0.7 vs. 1.75 g, p=0.03; 0.6 vs. 1.5 g, p=0.02; .012 vs 0.4 g, p=0.006), respectively. In vivo, (A2780 and MDA-231 models), EpoR siRNA did not affect Epo-stimulated tumor growth. In contrast, EphB4 siRNA-DOPC completely blocked Epo-stimulated tumor growth. Conclusions: Collectively, these results point to EphB4 as an alternative Epo receptor that mediates Epo-induced tumor growth by activating Stat-3. These findings offer exciting theranostic avenues for management of anemic cancer patients. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 1618. doi:10.1158/1538-7445.AM2011-1618
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...