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: Journal of Clinical Anesthesia, Elsevier BV, Vol. 72 ( 2021-09), p. 110314-
    Type of Medium: Online Resource
    ISSN: 0952-8180
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 1500489-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: BMC Cancer, Springer Science and Business Media LLC, Vol. 12, No. 1 ( 2012-12)
    Abstract: Cancer stem cells (CSC) are believed to play a crucial role in cancer recurrence due to their resistance to conventional chemotherapy and capacity for self-renewal. Recent studies have reported that salinomycin, a livestock antibiotic, selectively targets breast cancer stem cells 100-fold more effectively than paclitaxel. In our study we sought to determine the effects of salinomycin on head and neck squamous cell carcinoma (HNSCC) stem cells. Methods MTS and TUNEL assays were used to study cell proliferation and apoptosis as a function of salinomycin exposure in JLO-1, a putative HNSCC stem cell culture. MTS and trypan blue dye exclusion assays were performed to investigate potential drug interactions between salinomycin and cisplatin or paclitaxel. Stem cell-like phenotype was measured by mRNA expression of stem cell markers, sphere-forming capacity, and matrigel invasion assays. Immunoblotting was also used to determine expression of epithelial-mesenchymal transition (EMT) markers and Akt phosphorylation. Arrays by Illumina, Inc. were used to profile microRNA expression as a function of salinomycin dose. Results In putative HNSCC stem cells, salinomycin was found to significantly inhibit cell viability, induce a 71.5% increase in levels of apoptosis, elevate the Bax/Bcl-2 ratio, and work synergistically with cisplatin and paclitaxel in inducing cell death. It was observed that salinomycin significantly inhibited sphere forming-capability and repressed the expression of CD44 and BMI-1 by 3.2-fold and 6.2-fold, respectively. Furthermore, salinomycin reduced invasion of HNSCC stem cells by 2.1 fold. Contrary to expectations, salinomycin induced the expression of EMT markers Snail, vimentin, and Zeb-1, decreased expression of E-cadherin, and also induced phosphorylation of Akt and its downstream targets GSK3-β and mTOR. Conclusions These results demonstrate that in HNSCC cancer stem cells, salinomycin can cause cell death and decrease stem cell properties despite activation of both EMT and Akt.
    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 ...
  • 3
    In: Otology & Neurotology, Ovid Technologies (Wolters Kluwer Health), Vol. 33, No. 9 ( 2012-12), p. 1640-1647
    Type of Medium: Online Resource
    ISSN: 1531-7129
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2012
    detail.hit.zdb_id: 2058738-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: International Journal of Molecular Sciences, MDPI AG, Vol. 20, No. 1 ( 2019-01-07), p. 193-
    Abstract: Cancer stem cells (CSCs) have been shown as a distinct population of cancer cells strongly implicated with resistance to conventional chemotherapy. Metformin, the most widely prescribed drug for diabetes, was reported to target cancer stem cells in various cancers. In this study, we sought to determine the effects of metformin on head and neck squamous cell carcinoma (HNSCC). CSCs and non-stem HNSCC cells were treated with metformin and cisplatin alone, and in combination, and cell proliferation levels were measured through MTS assays. Next, potential targets of metformin were explored through computational small molecule binding analysis. In contrast to the reported effects of metformin on CSCs in other cancers, our data suggests that metformin protects HNSCC CSCs against cisplatin in vitro. Treatment with metformin resulted in a dose-dependent induction of the stem cell genes CD44, BMI-1, OCT-4, and NANOG. On the other hand, we observed that metformin successfully decreased the proliferation of non-stem HNSCC cells. Computational drug–protein interaction analysis revealed mitochondrial complex III to be a likely target of metformin. Based on our results, we present the novel hypothesis that metformin targets complex III to reduce reactive oxygen species (ROS) levels, leading to the differential effects observed on non-stem cancer cells and CSCs.
    Type of Medium: Online Resource
    ISSN: 1422-0067
    Language: English
    Publisher: MDPI AG
    Publication Date: 2019
    detail.hit.zdb_id: 2019364-6
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: PLoS ONE, Public Library of Science (PLoS), Vol. 7, No. 2 ( 2012-2-27), p. e32459-
    Type of Medium: Online Resource
    ISSN: 1932-6203
    Language: English
    Publisher: Public Library of Science (PLoS)
    Publication Date: 2012
    detail.hit.zdb_id: 2267670-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 3378-3378
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 3378-3378
    Abstract: The cancer stem cell hypothesis posits that within a tumor exists a distinct subpopulation of cells responsible for tumor initiation, progression, and maintenance. These cancer stem cells (CSCs) tend to be resistant to conventional chemotherapy and, like normal stem cells, possess the ability to self-renew and differentiate, thereby effecting tumor recurrence. Metformin, the most widely prescribed drug for the treatment of diabetes, has received attention in recent years as a potential anticancer agent capable of targeting cancer stem cells through such means as inhibiting cell proliferation and abrogating chemo-resistance. In the current study, we sought to determine the effects of Metformin on a putative head and neck squamous cell carcinoma (HNSCC) cancer stem cell culture. In contrast to the findings of previous studies, our data suggests that Metformin promotes properties of a cancer stem cell phenotype in HNSCC in vitro. Treatment with Metformin resulted in a dose-dependent induction of the stem cell genes CD44, BMI-1, Oct-4, and Nanog, as measured by qPCR. These results were supported by immunofluorescence data. Metformin treatment promoted self-renewal capacity of HNSCC stem cells, as demonstrated by the increase in size and number of tumorspheres formed in non-adherent and non-differentiating conditions. At various doses, treatment with Metformin alone had no effect on cell proliferation, as measured by MTS assay. Furthermore, when administered in combination with cisplatin, Metformin significantly protected against cisplatin-induced cell death, as demonstrated by MTS and TUNEL assays. Immunoblot experiments demonstrating a decrease in Akt phosphorylation upon treatment with Metformin suggests Metformin-mediated cytoprotection is independent of the Akt pathway. Other possible mechanisms are currently under investigation, including Metformin's ability to regulate autophagy. Although further in vivo studies are necessary, taken together, our findings suggest that Metformin may not be an effective therapeutic option for patients with HNSCC. 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 3378. doi:1538-7445.AM2012-3378
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 4368-4368
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 4368-4368
    Abstract: Cancer stem cells are believed to play a crucial role in cancer recurrence due to their resistance to conventional chemotherapy and capacity for self-renewal. Gupta and colleagues recently reported that salinomycin, a livestock antibiotic, selectively targets breast cancer stem cells. In our study we sought to determine the effect of salinomycin on head and neck squamous cell carcinoma (HNSCC) stem cells. We observed that salinomycin inhibits self-renewal capacity in HNSCC stem cells, as shown by a dose-dependent decrease in sphere formation. In addition, treatment with salinomycin resulted in a decrease in mRNA expression of the stem cell markers CD44 and BMI-1, both of which are critical for maintaining tumorigenicity in HNSCC. In combination with the chemotherapeutic agents cisplatin and doxorubicin, salinomycin synergistically killed HNSCC cancer stem cells more effectively compared to either drug alone, as demonstrated by cell proliferation and TUNEL assays. High-throughput analysis revealed a set of differentially expressed microRNAs in salinomycin-treated HNSCC stem cells, warranting further investigation into the possibility of microRNA-mediated pathways that are regulated by salinomycin. Taken together, our findings indicate promise for using salinomycin or related derivatives as a novel treatment for HNSCC, although further studies are needed to confirm efficacy in vivo. 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 4368. doi:10.1158/1538-7445.AM2011-4368
    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 ...
  • 8
    In: BMC Research Notes, Springer Science and Business Media LLC, Vol. 4, No. 1 ( 2011-12)
    Abstract: Recent studies indicate an increase in tumor progression and recurrence in head and neck squamous cell carcinomas (HNSCC) of cancer patients taking recombinant human erythropoietin (rhEpo) for anemia. This study was undertaken to investigate the potential role of rhEpo in invasion, proliferation, and cisplatin-induced cell death in HNSCC cell lines. Methods The following experiments were performed with two HNSCC cell lines, UMSCC-10B and UMSCC-22B. Presence of EpoR in both cell lines was determined by western blot and quantitative PCR. Colorimetric MTS assays and clonogenic assays were used to study the effect of rhEpo at pharmacologically relevant doses on cell proliferation. Matrigel invasion assays were performed in order to determine effects of exogenous rhEpo on invasive abilities. Clonogenic assays were also used to study potential cytoprotective effects of rhEpo against cisplatin. Immunoblotting was done to analyze the effect of rhEpo on Akt phosphorylation. Finally, MTS and TUNEL assays were performed to test our hypothesis that Akt activation by PI3K was involved in rhEpo-mediated cisplatin resistance. Results HNSCC cell lines were shown to express Epo receptor (EpoR). RhEpo increased invasion 1.8-fold in UMSCC-10B and 2.6-fold in UMSCC-22B compared to control. RhEpo at 10 U/ml increased cell proliferation by 41% and 53% in UMSCC-10B and UMSCC-22B, respectively, and colony formation by 1.5-fold and 1.8-fold. UMSCC-10B treated with cisplatin and exposed to rhEpo at 1 and 10 U/ml resulted in a 1.7-fold and 3.0-fold increase in colony number compared to control, respectively. UMSCC-22B treated with cisplatin and rhEpo at 1 or 10 U/ml resulted in ~2.5-fold increase in colony number. A TUNEL assay demonstrated a 30.5% and 76.5% increase in survival in UMSCC-10B and UMSCC-22B cells, respectively, in cisplatin and rhEpo-treated cells compared to cisplatin alone. MTS assay showed similar cytoprotective effects. Western blot revealed increased phosphorylation of Akt upon exposure of HNSCC cell lines to rhEpo. MTS assay and TUNEL analyses implicate Akt as a likely contributor to regulation of rhEpo-mediated cytoprotection. Conclusions The results demonstrate that, in HNSCC cells expressing functional EpoR, rhEpo promotes invasion, cell proliferation, and induces resistance to cisplatin, which may contribute to tumor progression.
    Type of Medium: Online Resource
    ISSN: 1756-0500
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2011
    detail.hit.zdb_id: 2413336-X
    Location Call Number Limitation 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. 4262-4262
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 4262-4262
    Abstract: The cancer stem cell (CSC) hypothesis proposes that only the CSCs within a tumor possess the ability to self-renew and to generate new tumors. Under this model, CSCs are presumed responsible for cellular invasion and metastasis, as well as the recurrence of cancers following chemotherapy. In head and neck squamous cell carcinoma (HNSCC), cells which are CD44 positive and lineage negative (CD44+/Lin-) have been shown to possess properties of CSCs. These cells form tumors in NOD/SCID mice at a much greater efficiency compared to CD44-/Lin- cells, and express the proto-oncogene BMI-1. More recently, it has been shown in pancreatic cancer that a distinct subpopulation of CSCs expressing the chemokine receptor CXCR4 is essential for tumor metastasis. Here we demonstrate the existence of a highly invasive CXCR4+ subpopulation in HNSCC cancer stem cells and elucidate the pathways through which this subpopulation could be regulated. For this study putative CSC cultures derived from primary HNSCC tissue were used from which CXCR4-expressing subpopulations were isolated. A matrigel invasion assay revealed that the CXCR4+ fraction of CSCs was significantly more invasive compared to the CXCR4- fraction. We also surveyed the mRNA expression of these two fractions by qRT-PCR, and found that expression of the proteinases MMP2 and MMP9 was markedly higher in CXCR4+ cells. Interestingly, treatment with nicotine increased the expression of CXCR4 and its ligand SDF-1 in the parental CSC culture, suggesting a possible mechanism by which CXCR4 could be regulated within the CSC population. Nicotine, a major component of tobacco, is known to have proliferative, anti-apoptotic, and angiogenic effects, and has been shown to promote epithelial-to-mesenchymal transition in a variety of human cancers. Mortality from HNSCC remains high due primarily to cancer invasion of vital structures and development of distant metastases. Our findings shed light on the mechanisms by which CXCR4 confers an invasive and metastatic CSC phenotype. In addition, the observed effects of nicotine could provide novel insights towards the regulation of the CXCR4/SDF-1 axis in HNSCC, for which tobacco use has been identified as the leading risk factor. Continued efforts in mapping the molecular mechanisms surrounding CXCR4-mediated invasion and metastasis of CSCs could lead to new and effective treatment strategies for HNSCC. 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 4262.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 4275-4275
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 4275-4275
    Abstract: Recently, the growth and metastasis of tumors, including head and neck squamous cell carcinomas (HNSCC), have been attributed to a subpopulation within tumors known as cancer stem cells (CSC). Studies have shown that HNSCC cells that are CD44+ are putative CSC and have the capability to self-renew, differentiate and preferentially express BMI-1. The polycomb group protein BMI-1 is a transcriptional repressor that silences the genes responsible for the expression of the cell cycle inhibitor genes p16 and p19. The gene is essential for maintaining the self-renewal capability of both normal and CSC. Very recent studies have shown that BMI-1 may also be implicated in other critical roles in cancer cell behavior including its role in the invasiveness of cancer cells. In this study, we demonstrate that in HNSCC CSC, BMI-1 is critical for cellular invasion. Knockdown of BMI-1 in CD44+ cells results in significant inhibition of invasion while overexpression of BMI-1 in CD44- cells promotes invasion. Furthermore, we show that this enhaced invasion conferred by BMI-1 is through induction of epithelial-to-mesenchymal transition (EMT) genes and the down-regulation of E-cadherin. In addition, we present evidence that the proteinases MMP2 and MMP9 are induced upon overexpression of BMI-1. CSC are also characterized by being chemoresistant, in part through their enhanced ability to extrude drugs because of their high expression of drug transporters. In this study, we demonstrate another mechanism for the resistance of CSC to DNA-damaging agents. We present evidence that another function of BMI-1 in CSC is its ability to regulate apoptosis. Knockdown of BMI1-1 in CSC using siRNA resulted in increased levels of cisplatin-induced apoptosis compared to cells transfected with scrambled siRNA. Our findings lend support to previous studies that have implicated BMI-1 in the regulation of pro-survival genes as well as the DNA damage response pathway. Our findings have revealed that BMI-1 is not only essential for self-renewal and tumorigenesis, but also that it has critical functions in cancer stem cell drug resistance and invasion, and most likely, metastasis. Since BMI-1 controls many of the properties of CSC and is preferentially expressed in HNSCC CSC, this oncogene may serve not only as a tumor marker, but also as an ideal therapeutic target. 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 4275.
    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
    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...