30 research outputs found

    Hmga2 is dispensable for pancreatic cancer development, metastasis, and therapy resistance.

    Get PDF
    Expression of the chromatin-associated protein HMGA2 correlates with progression, metastasis and therapy resistance in pancreatic ductal adenocarcinoma (PDAC). Hmga2 has also been identified as a marker of a transient subpopulation of PDAC cells that has increased metastatic ability. Here, we characterize the requirement for Hmga2 during growth, dissemination, and metastasis of PDAC in vivo using conditional inactivation of Hmga2 in well-established autochthonous mouse models of PDAC. Overall survival, primary tumour burden, presence of disseminated tumour cells in the peritoneal cavity or circulating tumour cells in the blood, and presence and number of metastases were not significantly different between mice with Hmga2-wildtype or Hmga2-deficient tumours. Treatment of mice with Hmga2-wildtype and Hmga2-deficient tumours with gemcitabine did not uncover a significant impact of Hmga2-deficiency on gemcitabine sensitivity. Hmga1 and Hmga2 overlap in their expression in both human and murine PDAC, however knockdown of Hmga1 in Hmga2-deficient cancer cells also did not decrease metastatic ability. Thus, Hmga2 remains a prognostic marker which identifies a metastatic cancer cell state in primary PDAC, however Hmga2 has limited if any direct functional impact on PDAC progression and therapy resistance

    Identification of epidermal Pdx1 expression discloses different roles of Notch1 and Notch2 in murine KrasG12D-induced skin carcinogenesis in vivo

    Get PDF
    Background The Ras and Notch signaling pathways are frequently activated during development to control many diverse cellular processes and are often dysregulated during tumorigenesis. To study the role of Notch and oncogenic Kras signaling in a progenitor cell population, Pdx1-Cre mice were utilized to generate conditional oncogenic KrasG12D mice with ablation of Notch1 and/or Notch2. Methodology/Principal Findings Surprisingly, mice with activated KrasG12D and Notch1 but not Notch2 ablation developed skin papillomas progressing to squamous cell carcinoma providing evidence for Pdx1 expression in the skin. Immunostaining and lineage tracing experiments indicate that PDX1 is present predominantly in the suprabasal layers of the epidermis and rarely in the basal layer. Further analysis of keratinocytes in vitro revealed differentiation-dependent expression of PDX1 in terminally differentiated keratinocytes. PDX1 expression was also increased during wound healing. Further analysis revealed that loss of Notch1 but not Notch2 is critical for skin tumor development. Reasons for this include distinct Notch expression with Notch1 in all layers and Notch2 in the suprabasal layer as well as distinctive p21 and β-catenin signaling inhibition capabilities. Conclusions/Significance Our results provide strong evidence for epidermal expression of Pdx1 as of yet not identified function. In addition, this finding may be relevant for research using Pdx1-Cre transgenic strains. Additionally, our study confirms distinctive expression and functions of Notch1 and Notch2 in the skin supporting the importance of careful dissection of the contribution of individual Notch receptors

    MALDI imaging mass spectrometry for in situ proteomic analysis of preneoplastic lesions in pancreatic cancer.

    No full text
    The identification of new biomarkers for preneoplastic pancreatic lesions (PanINs, IPMNs) and early pancreatic ductal adenocarcinoma (PDAC) is crucial due to the diseases high mortality rate upon late detection. To address this task we used the novel technique of matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) on genetically engineered mouse models (GEM) of pancreatic cancer. Various GEM were analyzed with MALDI IMS to investigate the peptide/protein-expression pattern of precursor lesions in comparison to normal pancreas and PDAC with cellular resolution. Statistical analysis revealed several discriminative m/z-species between normal and diseased tissue. Intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasm (IPMN) could be distinguished from normal pancreatic tissue and PDAC by 26 significant m/z-species. Among these m/z-species, we identified Albumin and Thymosin-beta 4 by liquid chromatography and tandem mass spectrometry (LC-MS/MS), which were further validated by immunohistochemistry, western blot, quantitative RT-PCR and ELISA in both murine and human tissue. Thymosin-beta 4 was found significantly increased in sera of mice with PanIN lesions. Upregulated PanIN expression of Albumin was accompanied by increased expression of liver-restricted genes suggesting a hepatic transdifferentiation program of preneoplastic cells. In conclusion we show that GEM of endogenous PDAC are a suitable model system for MALDI-IMS and subsequent LC-MS/MS analysis, allowing in situ analysis of small precursor lesions and identification of differentially expressed peptides and proteins

    <i>Pdx1-Cre;Kras;N1ko</i> mice develop skin tumors.

    No full text
    <p><b>A:</b> Kaplan-Meier tumor-free survival analysis of <i>Pdx1-Cre</i> mice. <b>B:</b> Table summarizing survival and skin tumor incidence observed in <i>Pdx1-Cre</i> mice. <b>C:</b> PCR results confirm <i>Notch1</i> deletion and <i>Kras<sup>G12D</sup></i> activation in pancreas and skin papilloma while non-recombined status in unaffected skin, liver and in WT control DNA. <b>D:</b> Examples of skin neoplasia observed: papillomas of neck-head and ear (i), sebaceous gland tumor (ii), cutaneous horns (iii, black arrowhead) and SCC (iii, white arrow). <b>E:</b> Hematoxilin and eosin staining (HE) of WT skin (i) and characteristic cutaneous histopathologies found in <i>Pdx1-Cre;Kras;N1ko mice</i>: hyperplasia (ii), skin papilloma (iii) and SCC (iv). <b>F:</b> X-Gal staining indicates Cre-mediated recombination in skin hyperplasia (left) and papillomas (right) of <i>Pdx1-Cre;Kras;N1ko</i>;<i>ROSA26R-LacZ</i> reporter mice. The scale bars represent 50 µm.</p

    Biochemical analysis of <i>K5;N1ko</i> and <i>K5;N2ko</i> keratinocytes.

    No full text
    <p><b>A:</b> Western blot analysis of primary keratinocytes isolated from different genotypes indicates correct <i>Notch1</i> and <i>Notch2</i> ablation and shows distinct modulation of β-catenin signaling and p21 expression. <b>B:</b> Quantitative RT-PCR show <i>Hes1</i> and <i>p21</i> transcripts levels in primary keratinocytes of the indicated genotypes. <b>C:</b> Luciferase reporter assay reveals that N1IC is a more potent inhibitor of β-catenin-LEF/TCF-sensitive TOP plasmid than N2IC. FOP plasmid is β-catenin-LEF/TCF-insensitive and serves as a specificity control. Both N1IC and N2IC induce Hes1 in a comparable manner as quantified using a Hes1-luc reporter.</p
    corecore