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
    Online Resource
    Online Resource
    Rockefeller University Press ; 2013
    In:  The Journal of Experimental Medicine Vol. 210, No. 13 ( 2013-12-16), p. 21013OIA63-
    In: The Journal of Experimental Medicine, Rockefeller University Press, Vol. 210, No. 13 ( 2013-12-16), p. 21013OIA63-
    Type of Medium: Online Resource
    ISSN: 0022-1007 , 1540-9538
    RVK:
    Language: English
    Publisher: Rockefeller University Press
    Publication Date: 2013
    detail.hit.zdb_id: 1477240-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Gut, BMJ
    Abstract: Large-scale genome sequencing efforts of human tumours identified epigenetic modifiers as one of the most frequently mutated gene class in human cancer. However, how these mutations drive tumour development and tumour progression are largely unknown. Here, we investigated the function of the histone demethylase KDM6A in gastrointestinal cancers, such as liver cancer and pancreatic cancer. Design Genetic alterations as well as expression analyses of KDM6A were performed in patients with liver cancer. Genetic mouse models of liver and pancreatic cancer coupled with Kdm6a-deficiency were investigated, transcriptomic and epigenetic profiling was performed, and in vivo and in vitro drug treatments were conducted. Results KDM6A expression was lost in 30% of patients with liver cancer. Kdm6a deletion significantly accelerated tumour development in murine liver and pancreatic cancer models. Kdm6a-deficient tumours showed hyperactivation of mTORC1 signalling, whereas endogenous Kdm6a re-expression by inducible RNA-interference in established Kdm6a-deficient tumours diminished mTORC1 activity resulting in attenuated tumour progression. Genome-wide transcriptional and epigenetic profiling revealed direct binding of Kdm6a to crucial negative regulators of mTORC1, such as Deptor, and subsequent transcriptional activation by epigenetic remodelling. Moreover, in vitro and in vivo genetic epistasis experiments illustrated a crucial function of Deptor and mTORC1 in Kdm6a-dependent tumour suppression. Importantly, KDM6A expression in human tumours correlates with mTORC1 activity and KDM6A-deficient tumours exhibit increased sensitivity to mTORC1 inhibition. Conclusion KDM6A is an important tumour suppressor in gastrointestinal cancers and acts as an epigenetic toggle for mTORC1 signalling. Patients with KDM6A-deficient tumours could benefit of targeted therapy focusing on mTORC1 inhibition.
    Type of Medium: Online Resource
    ISSN: 0017-5749 , 1468-3288
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 1492637-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 64, No. 17 ( 2004-09-01), p. 6058-6064
    Abstract: Molecular subtyping of human hepatocellular carcinoma (HCC) with potential mechanistic and therapeutic impact has not been achieved thus far. We have analyzed the mRNA expression patterns of 43 different human HCC samples and 3 HCC cell lines in comparison with normal adult liver using high-density cDNA microarrays. Two main groups of HCC, designated group A (65%) and group B (35%), were distinguished based on clustering of the most highly varying genes. Group A HCCs were characterized by induction of a number of interferon (IFN)-regulated genes, whereas group B was characterized mainly by down-regulation of several apoptosis-relevant and IFN-regulated genes. The number of apoptotic tumor cells and tumor-infiltrating lymphocytes was significantly higher in tumors of group A as compared with those of group B. Based on the expression pattern, group B was further subdivided into two subgroups, designated subgroup B1 (6 of 43 tumors, 14%) and subgroup B2 (9 of 43 tumors, 21%). A prominent characteristic of subgroup B1 was high overexpression of insulin-like growth factor (IGF)-II. All tested HCC cell lines expressed equally high concentrations of IGF-II transcripts and co-segregated with group B1 in clustering. IGF-II overexpression and induction of IFN-related genes were mutually exclusive, even when analysis was extended to other cancer expression profile studies. Moreover, IFN-γ treatment substantially reduced IGF-II expression in HCC cells. In conclusion, cDNA microarray analyses provided subtyping of HCCs that is related to intratumor inflammation and tumor cell apoptosis. This profiling may be of mechanistic and therapeutic impact because IGF-II overexpression has been linked to reduced apoptosis and increased proliferation and may be accessible to therapeutic intervention.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2004
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 18 ( 2016-09-15), p. 5550-5561
    Abstract: Hepatocellular carcinoma (HCC) represents the second leading cause of cancer-related deaths and is reported to be resistant to chemotherapy caused by tumor-initiating cells. These tumor-initiating cells express stem cell markers. An accumulation of tumor-initiating cells can be found in 2% to 50% of all HCC and is correlated with a poor prognosis. Mechanisms that mediate chemoresistance include drug export, increased metabolism, and quiescence. Importantly, the mechanisms that regulate quiescence in tumor-initiating cells have not been analyzed in detail so far. In this research we have developed a single cell tracking method to follow up the fate of tumor-initiating cells during chemotherapy. Thereby, we were able to demonstrate that mCXCL1 exerts cellular state-specific effects regulating the resistance to chemotherapeutics. mCXCL1 is the mouse homolog of the human IL8, a chemokine that correlates with poor prognosis in HCC patients. We found that mCXCL1 blocks differentiation of premalignant cells and activates quiescence in tumor-initiating cells. This process depends on the activation of the mTORC1 kinase. Blocking of the mTORC1 kinase induces differentiation of tumor-initiating cells and allows their subsequent depletion using the chemotherapeutic drug doxorubicin. Our work deciphers the mCXCL1–mTORC1 pathway as crucial in liver cancer stem cell maintenance and highlights it as a novel target in combination with conventional chemotherapy. Cancer Res; 76(18); 5550–61. ©2016 AACR.
    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 ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 1300-1300
    Abstract: The far upstream element (FUSE)-binding proteins (FBP)- 1, FBP-2, and FBP-3 represent a family of single-strand nucleic acid binding factors, regulating transcriptional as well as post-transcriptional processes. Elevated expression and pro-tumorigenic functions of all FBPs have been described for human liver cancer [1, 2]. First data indicated that FBP-1 affects microtubule dynamics through regulating MT-destabilizing factors in non-small cell lung cancer (NSCLC) [3] , however, comprehensive studies analyzing the expression and functional relevance of all FBPs in NSCLCs are missing so far. In order to define the expression of FBPs in lung cancer, FBP-transcript and protein levels were analysed in primary human NSCLC tissue samples. Semiquantitative real-time PCR as well as Tissue-Micro-Array (TMA) analyses revealed an elevated expression of FBP-1 and FBP-2 in most NSCLC tissue samples ( & gt;60%) in comparison to non-tumorous specimens. Interestingly, nuclear accumulation of FBP-1 significantly correlated with FBP-2 expression (r=0.33; p & lt;0.01), suggesting common regulatory mechanisms. In vitro, transient inhibition of FBP-1 by gene-specific siRNAs in NSCLC cell lines (Calu-6) was associated with decreased tumor cell viability (-76%; MTT assay), proliferation (-83%; BrdU assay), and increased apoptosis (2.8-fold; Nicoletti FACS assay). In contrast, inhibition of FBP-2 reduced cell viability of Calu-1 cells (-32%; MTT assay) but predominantly reduced tumor cell migration (-62%; two-dimensional scratch assay) as well as tumor cell invasion (-81%; sprouting assay) in all analyzed NSCLC cell lines (Calu-1, Calu-6, and A549), suggesting that both FBP isoforms facilitate partly distinct tumor-supporting effects. Surprisingly, efficient single-gene inhibition of FBP-1 or FBP-2 in A549 cells did not affect tumor cell viability. In contrast, the concomitant knock down of both FBP isoforms resulted in significantly decreased cell viability (-69%), suggesting that some FBP family members may compensate the loss of other members. Actually, FBP-2 negatively regulates FBP-1 expression in A549 cells, resulting in increased FBP-1 transcript and protein levels after FBP-2 inhibition. Therefore, functional compensation prevents A549 cells from anti-tumorigenic effects after FBP-2 knock down. In summary, this study provides evidence that coordinated overexpression of FBP-1 and FBP-2 is a frequent event in NSCLCs and that both factors are support tumor growth and NSCLC cell dissemination. Interestingly, partial functional redundance and mutual negative regulation of FBPs indicate that these factors may fine-tune the oncogenic behavior of NSCLC cells. References: [1] M Malz et al., (2009) Hepatology; [2] A Brauckhoff et al., (2011) J. Hepatol. [3] S Singer et al., (2009) Cancer Res 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 1300. doi:1538-7445.AM2012-1300
    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 ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1130-1130
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1130-1130
    Abstract: Background: Disturbance of hepatocyte polarity is frequently observed in liver cancer tissues; however, it is still controversially discussed if this phenotype represents the cause or the result of tumorigenesis. Due to their multi-functional properties, hepatocytes are characterized by distinct luminal, lateral and basal domains that are defined by specific protein complex. These cell protein complexes such as the apical Crumbs- (CRB3, Pals1, Patj), the lateral Par- (Par3, Par6, aPKC) and the basolateral Scribble-complex (Scrib, Dlg, Lgl) are essential for the formation of hepatocellular polarity. So far, it is unknown if and which protein complex constituents are involved in liver tumor development and how these factors induce possible downstream effector mechanisms. Methods: To identify complex proteins that affect hepatocellular polarity, a siRNA screen targeting all relevant polarity factors was performed in HCC cells that form bile canaliculi in vitro (HepG2). Localization of Scribble in HCC cell lines and human HCC tissues was analyzed by western immunoblotting and immunofluorescence. Expression vectors containing wildtype Scribble and an isoform lacking polarity complex binding capacity were cloned (Scrib-wt, Scrib-P305L) and stably transfected in HCC cells. The impact of these isoforms on PI3K/AKT signaling pathway activity and cell viability were analyzed. In addition, the interaction of Scrib-wt, Scrib-P305L with phosphatases regulating the PI3K/AKT pathway was tested by Co-IP analysis. Results: The siRNA screen revealed that Scribble strongly affects hepatocellular polarity. Scribble located near the membrane in polarized cells (e.g., HepG2, HuH-1) but in the cytoplasm in non-polarized cells (HLE, HLF). Scribble overexpression and cytoplasmic accumulation correlates with poor overall survival of HCC patients. In vitro, the inhibition of Scribble reduced and overexpression of cytoplasmic Scribble (Scrib-P305L) induced AKT phosphorylation. In addition, Scrib-P305L supported HCC cell viability in an AKT-dependent manner. Immunofluorescence revealed co-localization of Scribble with the AKT phosphatases PTEN and PHLPP1. Co-IP studies revealed a physical interaction between PHLPP1 and both Scribbles isoforms (wildtype and P305L mutant). Conclusion/Outlook: Our data demonstrate that the cytoplasmic accumulation of Scribble in HCC cells promotes tumor cell growth in an AKT-dependent manner probably through the interaction with AKT phosphatases. These results indicate that the disturbance of cell polarity (e.g. by Scribble overexpression) can foster oncogenic effects and therefore must be regarded as initiating event in hepatocarcinogenesis. Citation Format: Shan Wan, Anne-Sophie Meyer, Sofia Weiler, Teresa Lutz, Stephanie Roessler, Peter Schirmacher, Federico Pinna, Kai Breuhahn. Subcellular localization of the cell polarity protein Scribble defines its oncogenic activity in hepatocellular 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 1130.
    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
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 3089-3089
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 3089-3089
    Abstract: Overexpression and nuclear enrichment of the Hippo pathway-regulated transcriptional co-activator Yes-Associated Protein (YAP) is frequently detected in human hepatocellular carcinoma (HCC) and correlates with poor clinical outcome of cancer patients1. In mice, inducible overexpression of the constitutively active isoform YAPS127A induces hepatocellular proliferation leading to hepatomegaly followed by tumor formation within 12-15 weeks. How oncogenic YAP and its paralogue TAZ (syn. WWTR1, WW Domain Containing Transcription Regulator 1) induce uncontrolled proliferation in liver cells is not known, yet. Analysis of expression profiling data, derived from human liver cancer cells after siRNA-mediated inhibition of endogenous YAP, revealed that the Minichromosome Maintenance (MCMs) family members MCM2-7 were positively regulated. MCMs are key components of the pre-replication complex and involved in the formation of the replication fork, which is essential for efficient DNA duplication followed by mitosis2. We confirmed that silencing of YAP and TAZ by independent siRNAs reduced the mRNA and protein expression of MCM2-7 in liver cancer cell lines. In vitro analyses revealed that transcription factors of the TEAD family were the most relevant YAP binding partners needed for the transcriptional regulation of MCMs. Using chromatin immunoprecipitation and Dual Luciferase Reporter Assay, we showed that YAP, TAZ and TEAD4 directly bound the MCM promoter. First in vitro data suggested that viability of liver cancer cells, subjected to drug induced replication stress, was exclusively reduced in cells with reduced YAP, TAZ or MCM protein expression. Administration of the YAP/TAZ/TEAD inhibitor Verteporfin reduced the expression of all MCMs in liver cancer cells. Vice versa, the overexpression of YAPS127A in transgenic mice showed significantly increased levels of the proliferation marker Ki67 and MCM2-7. Expression data from 242 HCC patients illustrated an association between YAP and MCM mRNA levels, with elevated MCMs significantly correlating with worse overall survival and early cancer recurrence3. Immunohistochemical stains of HCC tissue micro-arrays revealed a highly significant correlation between Ki67 expression, nuclear YAP overexpression and nuclear MCM2-7 enrichment. In summary, our results strongly suggest that YAP and TAZ facilitate their tumor-supporting properties through the regulation of MCM2-6, a complete protein complex. We conclude that combined inhibition of YAP and TAZ or perturbation of MCM activity might represent an efficient therapeutic approach for the treatment of a subgroup of HCC patients. 1Tschaharganeh D, et al., Gastroenterol. 2013; 2Deegan TD, et al., Curr Opin Struct Biol. 2016; 3Roessler S, et al., Cancer Res. 2010. Citation Format: Maria Knaub, Sofia Weiler, Stefan Thomann, Peter Schirmacher, Kai Breuhahn. YAP and TAZ induce MCM protein expression to facilitate tumor-supporting properties in liver cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3089.
    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 ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 24 ( 2020-12-15), p. 5502-5514
    Abstract: The oncogene yes-associated protein (YAP) controls liver tumor initiation and progression via cell extrinsic functions by creating a tumor-supporting environment in conjunction with cell autonomous mechanisms. However, how YAP controls organization of the microenvironment and in particular the vascular niche, which contributes to liver disease and hepatocarcinogenesis, is poorly understood. To investigate heterotypic cell communication, we dissected murine and human liver endothelial cell (EC) populations into liver sinusoidal endothelial cells (LSEC) and continuous endothelial cells (CEC) through histomorphological and molecular characterization. In YAPS127A-induced tumorigenesis, a gradual replacement of LSECs by CECs was associated with dynamic changes in the expression of genes involved in paracrine communication. The formation of new communication hubs connecting CECs and LSECs included the hepatocyte growth factor (Hgf)/c-Met signaling pathway. In hepatocytes and tumor cells, YAP/TEA domain transcription factor 4 (TEAD4)–dependent transcriptional induction of osteopontin (Opn) stimulated c-Met expression in EC with CEC phenotype, which sensitized these cells to the promigratory effects of LSEC-derived Hgf. In human hepatocellular carcinoma, the presence of a migration-associated tip-cell signature correlated with poor clinical outcome and the loss of LSEC marker gene expression. The occurrence of c-MET–expressing CECs in human liver cancer samples was confirmed at the single-cell level. In summary, YAP-dependent changes of the liver vascular niche comprise the formation of heterologous communication hubs in which tumor cell–derived factors modify the cross-talk between LSECs and CECs via the HGF/c-MET axis. Significance: YAP-dependent changes of the liver vascular niche comprise the formation of heterologous communication hubs in which tumor cell-derived factors modify the cross-talk between EC subpopulations.
    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 ...
  • 9
    Online Resource
    Online Resource
    Wiley ; 2012
    In:  International Journal of Cancer Vol. 130, No. 3 ( 2012-02-01), p. 575-583
    In: International Journal of Cancer, Wiley, Vol. 130, No. 3 ( 2012-02-01), p. 575-583
    Type of Medium: Online Resource
    ISSN: 0020-7136
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2012
    detail.hit.zdb_id: 218257-9
    detail.hit.zdb_id: 1474822-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: International Journal of Oncology, Spandidos Publications, Vol. 48, No. 4 ( 2016-4), p. 1679-1687
    Type of Medium: Online Resource
    ISSN: 1019-6439 , 1791-2423
    RVK:
    Language: English
    Publisher: Spandidos Publications
    Publication Date: 2016
    detail.hit.zdb_id: 2079608-0
    detail.hit.zdb_id: 1154403-X
    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...