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)  (10)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Molecular Cancer Therapeutics Vol. 17, No. 1_Supplement ( 2018-01-01), p. A212-A212
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 17, No. 1_Supplement ( 2018-01-01), p. A212-A212
    Abstract: Intrahepatic cholangiocarcinoma (iCCA) is a form of deadly malignancy with limited treatment options. Gain-of-function mutations in KRAS are one of the most frequent mutations, found in ~25% of human CCA patients. MEK inhibitors have been developed and show some activity against solid tumors with Ras mutants. However, whether MEK inhibitors are effective against KRas mutant CCAs remains unknown. In the current study, we established a new mouse iCCA model (NICD/KRas) by expressing activated forms of Notch (NICD) and KRas (KRasV12D). We investigated the therapeutic potential of MEK inhibitors in vitro using human CCA cell lines and in vivo using KRasV12/NICD iCCA preclinical model. We found that in general, CCA cell lines with KRas mutations are more sensitive to MEK inhibitor U0126. Treatment of U0126 in KRas mutant CCA cells leads to increased apoptosis and decreased expression of pro-apoptosis gene Survivin. Furthermore, we found that treating KRasV12/NICD tumor-bearing mice with MEK inhibitor PD0325901 (PD901) resulted in stable disease. At molecular levels, PD901 efficiently inhibited p-ERK expression KRasV12/NICD tumor cells, leading to increased apoptosis. In summary, our studies demonstrate that KRasV12/NICD mice represent a novel and useful preclinical model to study KRas-driven CCA development. Our data further support the usefulness of MEK inhibitors for treatment of KRas mutant CCA in patients. Citation Format: Mingjie Dong, Xianqiong Liu, Katja Evert, Kirsten Utpatel, Shanshan Zhang, Li Che, John Gordan, Diego Calvisi, Matthias evert, Yan Liu, Xin Chen. Preclinical efficacy of MEK inhibition in a K-Ras driven cholangiocarcinoma preclinical model [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr A212.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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. 72, No. 8_Supplement ( 2012-04-15), p. 984-984
    Abstract: The evolutionary conserved Hippo-pathway negatively regulates organ size control by phosphorylation and cytoplasmic retention of the transcriptional co-activator yes-associated protein (YAP). Recent studies demonstrated that deletion of essential Hippo-pathway constituents (e.g., Mst-1/2 and WW45) or overexpression of YAP lead to the development of liver cancer. However, the underlying molecular tumor-supporting mechanisms in carcinogenesis have not been defined so far. Overexpression and nuclear accumulation of YAP in nearly 70% of all human hepatocellular carcinomas (HCC) significantly correlated with tumor cell proliferation and dedifferentiation. In human HCC cell lines, siRNA-mediated inhibition of YAP significantly reduced tumor cell viability, and migration/invasion. Based on transcriptomic profiling approaches, the Notch ligand Jagged-1 (Jag-1) was identified as YAP-dependent target gene in HCC cells and in primary murine hepatocytes of transgenic animals expressing constitutively active YAPS127A. Inhibition of YAP reduced the protein levels of Jag-1, cleaved Notch receptor (NICD), and Hes-1, while YAP overexpression increased the amounts of all factors. As detected for YAP knock down, transfection of gene-specific siRNA targeting Jag-1 diminished HCC cell viability and migration. Overexpression and concomitant inhibition of Jag-1 abolished Hes-1 expression and YAP-induced HCC cell viability. By applying different mutant isoforms of YAP (e.g., YAPS127A and YAP5SA-delta-C - dominant negative isoform), TEAD4 but not TEAD1 was identified as the transcription factor required for YAP-dependent regulation of Jag-1 and Hes-1. Furthermore, the WNT/beta-catenin pathway, a putative inducer of Jag-1, did not influence the YAP-dependent modulation of Jag-1. Knock down experiments revealed Mst-2 and Lats-2 as negative regulators of YAP activity and Jag-1/Hes-1 expression. The amounts of YAP, Jag-1, and Hes-1 transcripts as well as proteins significantly correlated with each other in human HCC tissues. Most importantly, increased concentrations of all factors significantly associated with poor prognosis of HCC patients. These data demonstrate that high-level expression of YAP in HCC cells induces tumor growth and tumor cell dissemination in part through activation of the Jag-1/Notch pathway in a TEAD4-dependent and beta-catenin-independent manner. This regulatory cross-talk between Hippo- and Notch-signalling defines a group of HCC patients with poor overall survival. 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 984. doi:1538-7445.AM2012-984
    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 ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 1 ( 2019-01-01), p. 403-413
    Abstract: Intrahepatic cholangiocarcinoma (ICC) is an aggressive cancer type, lacking effective therapies and associated with a dismal prognosis. Palbociclib is a selective CDK4/6 inhibitor, which has been shown to suppress cell proliferation in many experimental cancer models. Recently, we demonstrated that pan-mTOR inhibitors, such as MLN0128, effectively induce apoptosis, although have limited efficacy in restraining proliferation of ICC cells. Here, we tested the hypothesis that palbociclib, due to its antproliferative properties in many cancer types, might synergize with MLN0128 to impair ICC growth. Experimental Design: Human ICC cell lines and the AKT/YapS127A ICC mouse model were used to test the therapeutic efficacy of palbociclib and MLN0128, either alone or in combination. Results: Administration of palbociclib suppressed in vitro ICC cell growth by inhibiting cell-cycle progression. Concomitant administration of palbociclib and MLN0128 led to a pronounced, synergistic growth constraint of ICC cell lines. Furthermore, while treatment with palbociclib or MLN0128 alone resulted in tumor growth reduction in AKT/YapS127A mice, a remarkable tumor regression was achieved when the two drugs were administered simultaneously. Mechanistically, palbociclib was found to potentiate MLN0128 mTOR inhibition activity, whereas MLN0128 prevented the upregulation of cyclin D1 induced by palbociclib treatment. Conclusions: Our study indicates the synergistic activity of palbociclib and MLN0128 in inhibiting ICC cell proliferation. Thus, combination of CDK4/6 and mTOR inhibitors might represent a novel, promising, and effective therapeutic approach against human ICC. See related commentary by Malumbres, p. 6
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Clinical Cancer Research Vol. 16, No. 14_Supplement ( 2010-07-15), p. B29-B29
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 16, No. 14_Supplement ( 2010-07-15), p. B29-B29
    Abstract: Introduction: Hepatocellular carcinoma (HCC) and cholangiocellular carcinoma (CCC) are deadly malignant liver tumors lacking of efficient treatment options. Akt/mTOR and Ras/MAPK pathways have been implicated in hepatic carcinogenesis. Our study is to define the genetic interactions between Akt/mTOR and Ras/MAPK pathways in liver carcinogenesis. Methods: Activation of Akt/mTOR and Ras/MAPK signaling was assayed in 62 human HCC and 25 CCC samples. An activated Akt (myr-Akt) was stably transfected into the mouse liver alone or together with activated N-Ras (RasV12) via hydrodynamic gene delivery. Molecular and biochemical features of liver lesions were analyzed. A primary cell line (Akt/Ras) was isolated from a tumor co-injected with Akt and Ras protooncogenes. Akt/Ras cells and human HCC cell lines were treated with different mTOR inhibitors, including NVP-BEZ235, PP242 and Rapamcyin, as well as the MEK inhibitor AZD6244. Results: Using human HCC samples, we demonstrated the coordinated activation of Akt/mTOR and Ras/MAPK pathways in subsets of HCC and CCC characterized by poor prognosis. We found that coexpression of myr-Akt with RasV12 synergizes to promote liver tumor development in mice, leading to HCC and CCC formation within 4 to 6 weeks post injection. At cellular level, tumors from Akt/N-Ras co-injection cells are characterized by high proliferation and low apoptotic index, and disrupted expression of genes involved in cell cycle regulation. At the molecular level, liver tumors show high levels of activated Akt/mTOR and Erk cascades. A primary cell line (Akt/Ras) was isolated from a tumor and used to test the response to different mTOR and MEK inhibitors. We found that dual PI3K/mTOR inhibitor NVP-BEZ235 has superior activity inhibiting Akt/Ras cell proliferation with IC50 around 2nM. It also efficiently inhibits phospho-S6, phospho-4EBP1 and cyclin D1 expression in Akt/Ras cells. Furthermore, NVP-BEZ235 synergizes with the MEK inhibitor AZD6244 to dramatically reduce the growth of the Akt/Ras cell line. Equivalent results were obtained in human HCC cell lines with the same inhibitors. Conclusion: Coordinated activation of Akt/mTOR and Ras/MAPK signaling is the hallmark of aggressive subtypes of human HCC and CCC. Activated Akt cooperates with activated Ras/MAPK to rapidly promote liver tumor formation in mice. The Akt/Ras mouse model is a useful pre-clinical system to test anti-neoplastic therapeutic approaches against HCC and CCC in vivo. In addition, our preliminary studies suggest that the combination of a dual PI3K/mTOR inhibitor with a MEK inhibitor may represent a promising novel targeted therapy for treating human HCC harboring activated Akt/mTOR and Ras/MAPK pathways. Citation Information: Clin Cancer Res 2010;16(14 Suppl):B29.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 2248-2248
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 2248-2248
    Abstract: AKT and Ras pathways are frequently concurrently hyperactivated in cancer. We have shown that co-expression of constitutively active forms of AKT (Myr-AKT) and activated Ras (N-RasV12) can rapidly induce liver tumors in mice. mTORC1 is the main downstream effector of AKT. mTORC1 phosphorylates rpS6 and 4EBP1, eukaryotic initiation factor eIF4E binding protein. Rapamycin is a well-characterized drug that partially inhibits mTORC1 activity. It can efficiently inhibit the phosphorylation of rpS6 but not the phosphorylation of 4EBP1. An unphophorylable mutant of 4EBP1, 4EBP1A4, is able to continuously bind to eIF4E and to inhibit eIF4E activity. Whether both of the two downstream effectors of mTORC1, rpS6 and 4EBP1, are required for hepatocarcinogenesis is unknown. The purpose of this study is to investigate the role of these two downstream effectors in hepatocarcinogenesis induced by co-activation of AKT and Ras. To determine the role of rpS6 in AKT/Ras-driven hepatocarcinogenesis, we treated AKT/Ras mice with rapamycin or vehicle daily for 7 weeks. After 7 weeks of vehicle treatment, all of the AKT/RAS/Veh mice developed large liver tumors. Lesions occupied ∼70-80% of the liver parenchyma. Histologically, the lesions consisted of large hepatocellular carcinonas (HCCs) and adenomas (HCAs). Tumor cells showed high proliferative activity, as indicated by Ki-67 staining. By contrast, after 7 weeks of Rapamycin treatment, none of the AKT/RAS/Rapa mice developed tumor. Small clusters of lipid-rich preneoplastic cells occupied 5-15% of the liver tissue. Few hepatocytes and non-parenchymal cells showed proliferative activity. Western blot analysis confirmed that Rapamycin inhibited the activation of rpS6 but had no effect on phosphorylated/inactivated levels of 4EBP1. These result indicated that Rapamycin treatment efficiently inhibited the progression of AKT/RAS induced liver tumor via inhibiting the rpS6 pathway. To determine the role of 4EBP1 in AKT/Ras-driven hepatocarcinogenesis, we overexpressed 4EBP1A4 or 4EBP1WT along with AKT and Ras into the mouse liver. Seven weeks after hydrodynamic injection, AKT/Ras/4EBP1WT mice developed large liver tumors which were equivalent to those developed in AKT/RAS/Veh mice. However, only few very small nodules developed in the livers of AKT/Ras/4EBP1A4 mice. The nodules consisted only of HCA and represented less than 5% of the lesional tissue. Notably, the proliferation in tumor cells of AKT/Ras/4EBP1A4 was more robust than in AKT/RAS/Rapa mice. The AKT/Ras/4EBP1A4 mice finally developed large liver tumors around 20 weeks after hydrodynamic injection. These results indicated that 4EBP1A4 efficiently delayed the AKT/RAS induced liver tumor. Altogether, our result indicates that the two main downstream effectors of mTORC1, rpS6 and 4EBP1, are both required for hepatocarcinogenesis induced by co-activation of AKT and Ras. 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 2248. doi:1538-7445.AM2012-2248
    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) ; 2006
    In:  Cancer Research Vol. 66, No. 3 ( 2006-02-01), p. 1833-1843
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 66, No. 3 ( 2006-02-01), p. 1833-1843
    Abstract: It has been shown that combined high local hyperinsulinism and hyperglycemia after low-number islet transplantation into the livers of streptozotocin-diabetic rats lead to the development of hepatocellular neoplasms but a substantial cocarcinogenic effect of genotoxic streptozotocin could not be ruled out completely. Thus, we herein investigated this model in BB/Pfd rats (n = 805; nine experimental groups), which develop spontaneous autoimmune diabetes similar to human type 1 diabetes. After low-number islet transplantation (n = 450), the liver acini downstream of the islets show insulin-induced alterations: massive glycogen and/or fat accumulation, translocation of the insulin receptor, decrease in glucose-6-phosphatase activity, increase in expression of insulin-like growth factor (IGF)-I, IGF-II/mannose-6-phosphate receptor, insulin receptor substrate-1, Raf-1, and Mek-1, corresponding to clear cell preneoplastic foci of altered hepatocytes known from chemical hepatocarcinogenesis and identical to that in streptozotocin-diabetic Lewis rats. After 6 months, many altered liver acini progressed to other types of preneoplasias often accompanied by an overexpression of the glutathione-S transferase (placental form), IGF-I receptor, and transforming growth factor (TGF)-α. After 12 to 15 and 15 to 18 months, 52% and 100% of the animals showed one or multiple hepatocellular adenomas or hepatocellular carcinomas (HCCs), respectively. Conclusively, this study identifies combined hyperinsulinism and hyperglycemia as a carcinogenic mechanism for the development of HCCs in diabetic rats. Hepatocarcinogenesis is independent from additional genotoxic compounds (i.e., streptozotocin), but is primarily triggered by increased intracellular insulin signaling via pathways associated with cell growth and proliferation, such as the Ras-Raf-mitogen-activated protein kinase pathway and the IGF system, and secondarily involves other growth factors, such as TGF-α. (Cancer Res 2006; 66(3): 1833-43)
    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 ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 64, No. 21 ( 2004-11-01), p. 8093-8100
    Abstract: Low-number transplantation of pancreatic islets into the livers of diabetic rats leads to transformation of the downstream liver acini into clear-cell foci of altered hepatocytes (FAHs). These FAHs correspond to the glycogen-storing (clear-cell) phenotype of hepatocellular preneoplasias and develop into hepatocellular adenomas (HCAs) and hepatocellular carcinomas (HCCs) within 6 to 24 months. In addition, they show metabolic alterations that resemble well-known insulin effects, most likely constituting the result of the local hyperinsulinemia. Thus, we investigated FAHs, HCAs, and HCCs for altered expression of insulin receptor, insulin receptor substrate-1 (IRS-1), Raf-1 and Mek-1. Light and electron microscopic immunohistochemistry revealed a translocation of insulin receptor from the plasma membrane (normal tissue) into the cytoplasm in clear-cell FAHs and an increase in insulin receptor expression in HCAs and HCCs. FAHs also showed an increase in IRS-1 gene expression, investigated by in situ hybridization and quantitative reverse transcription-PCR. IRS-1, Raf-1, and Mek-1 proteins were strongly overexpressed in FAHs and tumors, as compared with the unaltered liver tissue. These overexpressions were closely linked to the clear-cell phenotype of preneoplastic and neoplastic hepatocytes, because basophilic FAHs (later stages) and basophilic tumors showed no overexpressions. In this endocrine model of hepatocarcinogenesis, severe alterations of insulin signaling were induced by the pathological local action of islet hormones in the livers and may substantially contribute to the carcinogenic process.
    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 ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 1880-1880
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 1880-1880
    Abstract: Aberrant metabolism, including increased de novo lipogenesis, is one of the hallmarks of cancer. Fatty Acid Synthase (Fasn) catalyzes the de novo synthesis of long-chain fatty acids from acetyl-CoA and malonyl-CoA. Increased Fasn expression has been reported in multiple tumor types, and inhibition of Fasn expression has been shown to have tumor-suppressing activity. However, how increased de novo lipogenesis contributes to tumor initiation and progression, especially in vivo, remains unknown. In our previous studies, we showed that overexpression of the activated form of AKT (myr-AKT) induced de novo lipogenesis, hepatocyte proliferation and, eventually, liver cancer formation in mice. The tumorigenesis process could be significantly accelerated via co-expression of activated form of Ras. In the AKTRas tumor model, tumors were predominantly ( & gt;80%) hepatocellular carcinoma (HCC), and the remaining ( & lt;20%) were intrahepatic cholangiocarcinoma (ICC). The ICCs were induced via hepatocyte-biliary epithelial cell metaplasia in a Notch dependent manner. Intriguingly, fat droplet formation, increased expression of Fasn and other lipogenesis pathway genes were identified in HCC lesions, but not ICC lesions. To define whether Fasn-mediated de novo lipogenesis is a key metabolic event downstream of mTORC1 during AKTRas-induced hepatocarcinogenesis, we utilized Fasn flox/flox mice, and co-expressed AKTRas in the Fasn KO hepatocytes. Of note, we found that ablation of Fasn completely inhibited AKT induced lipogenesis and hepatocyte proliferation in mice. However, loss of Fasn modestly delayed AKTRas-induced liver tumor development. Histological analysis revealed that these tumor lesions were predominantly ICCs. The ICC cells were highly proliferative, and did not express Fasn or any other lipogenic pathway gene. The results suggest that Fasn-mediated de novo lipogenesis is required for AKTRas-induced HCC formation, but this process is dispensable for ICC formation. In summary, our experiments support a critical role for Fasn as a downstream effector of mTORC1 in HCC pathogenesis. However, Fasn-mediated do novo fatty acid synthesis is not required in all tumor types. It is likely that other sources of fatty acids, presumably including those derived from the diet, can be utilized by cancer cells for membrane synthesis during cell proliferation. Citation Format: Xin Chen, Lei Li, Chunmei Wang, Matthias Evert, Diego F. Calvisi, Clay F. Semenkovich. Role of fatty acid synthase and do novo lipogenesis in liver cancer development in mice. [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 1880. doi:10.1158/1538-7445.AM2013-1880 Note: This abstract was not presented at the AACR Annual Meeting 2013 because the presenter was unable to attend.
    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 ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 65, No. 15 ( 2005-08-01), p. 7013-7022
    Abstract: In contrast to high local insulin levels obtained after low-number transplantation (n = 350) of islets of Langerhans into the livers of diabetic rats, low insulin levels after high-number transplantation (n = 1,000) do not suffice to induce hepatocarcinogenesis. Herein, we investigated the possible cocarcinogenic potential of high and, in particular, low insulin levels, combining this in vivo model with a chemical model of hepatocarcinogenesis after administration of N-nitrosomorpholine (NNM). In three main experiments, different schemes of single or continuous NNM administration were combined with different transplantation procedures in diabetic or nondiabetic animals, i.e., low-number and high-number islet transplantation, transplantation of polystyrene particles, and sham transplantation. Animals were sacrificed between 3 and 53 weeks after transplantation procedures. Evidence for the cocarcinogenic effects of NNM and insulin was provided in each main experiment. NNM treatment after low-number islet transplantation resulted in an increase in the number of preneoplastic hepatocellular foci, and a significant increase in the number and an earlier appearance of hepatocellular adenomas and carcinomas compared with controls. Most intriguing was the increase in preneoplastic foci after combined NNM treatment and high-number islet transplantation, proving that insulin, even in lower doses, has at least cocarcinogenic effects on the downstream hepatocytes and thus promotes an otherwise initiated hepatocarcinogenic process. Conclusively, intrahepatic transplantation of pancreatic islets acts as a strong cocarcinogenic factor together with NNM in streptozotocin-diabetic rats.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2005
    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: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 4300-4300
    Abstract: Background & Aims: The Mouse Double Minute homolog 4 (MDM4) is one of the main negative p53 regulators in mammalian cells and mutational inactivation of p53 is a rare event in Western hepatocellular carcinomas (HCCs). 1q gains promote the upregulation of MDM4 in HCC, leading to a negative regulation of p53 activity. However, additional mechanisms might be involved that explain the increased MDM4 activity observed in HCCs with balanced MDM4 gene locus. Here we aimed at the identification of post-transcriptional mechanisms involved in the upregulation of MDM4 in HCC. Methods: To investigate a potential role of PI3K-AKT-mTOR signaling on MDM4 activity, HCC cell lines were treated with small molecule inhibitors and siRNAs and expression changes as well as functional analyses were recorded. Expression changes of central pathway components were investigated in human liver samples (normal liver, peritumorous liver tissue, and HCC). An AKT transgenic mouse model was used to show the involvement of AKT pathway in the regulation of MDM4 in vivo. Results: The inhibition of both PI3K-AKT and mTOR signaling pathways resulted in reduced MDM4 protein levels in HCC cell lines, which was associated with the transcriptional activation of p53-target genes. Biochemical assays revealed that both AKT-mediated phosphorylation and ubiquitin-specific protease 2a (USP2a)-mediated deubiquitination protected MDM4 from proteasomal in human HCC cell lines. In addition, AKT transgenic mice showed increased MDM4 protein levels indicating that AKT signaling is involved in the stabilization of MDM4 protein in vivo. Furthermore, the Eukaryotic translational Elongation Factor 1 alpha 2 (EEF1A2), which is frequently upregulated in human HCC, sustained the PI3K-AKT-mTOR cascade both in vitro and in vivo. In human HCCs, a strong positive correlation between the overexpression of EEF1A2, pAKT, USP2a, and MDM4 was observed, which was associated with shorter survival of HCC patients. Conclusions: Our data demonstrate that the EEF1A2/PI3K/AKT/mTOR cascade inactivates wild-type p53 in human HCC through the stabilization of the MDM4 protooncogene via a post-transcriptional mechanism involving an AKT-mediated phosphorylation of MDM4 and USP2a-mediated de-ubiquitination. Since the sustained activation of the EEF1A2/PI3K/AKT/mTOR/MDM4 axis has impact on the survival probability of HCC patients, it may thus represent a promising therapeutic target. Citation Format: Rossella Pellegrino, Diego F. Calvisi, Olaf Neumann, Xin Chen, Chunmei Wang, Bernhard Radlwimmer, Sara Ladu, Frank Dombrowski, Matthias Evert, Peter Schirmacher, Thomas Longerich. The EEF1A2-PI3K-AKT-mTOR axis supports the protumorigenic function of MDM4 in human hepatocellular carcinoma. [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 4300. doi:10.1158/1538-7445.AM2013-4300
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...