Characterization of pancreatic glucagon-producing tumors and pituitary gland tumors in transgenic mice overexpressing MYCN in hGFAP-positive cells

Standard

Characterization of pancreatic glucagon-producing tumors and pituitary gland tumors in transgenic mice overexpressing MYCN in hGFAP-positive cells. / Fielitz, Kathrin; Althoff, Kristina; De Preter, Katleen; Nonnekens, Julie; Ohli, Jasmin; Elges, Sandra; Hartmann, Wolfgang; Klöppel, Günter; Knösel, Thomas; Schulte, Marc; Klein-Hitpass, Ludger; Beisser, Daniela; Reis, Henning; Eyking, Annette; Cario, Elke; Schulte, Johannes H; Schramm, Alexander; Schüller, Ulrich.

in: ONCOTARGET, Jahrgang 7, Nr. 46, 19.10.2016, S. 74415-74426.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Fielitz, K, Althoff, K, De Preter, K, Nonnekens, J, Ohli, J, Elges, S, Hartmann, W, Klöppel, G, Knösel, T, Schulte, M, Klein-Hitpass, L, Beisser, D, Reis, H, Eyking, A, Cario, E, Schulte, JH, Schramm, A & Schüller, U 2016, 'Characterization of pancreatic glucagon-producing tumors and pituitary gland tumors in transgenic mice overexpressing MYCN in hGFAP-positive cells', ONCOTARGET, Jg. 7, Nr. 46, S. 74415-74426. https://doi.org/10.18632/oncotarget.12766

APA

Fielitz, K., Althoff, K., De Preter, K., Nonnekens, J., Ohli, J., Elges, S., Hartmann, W., Klöppel, G., Knösel, T., Schulte, M., Klein-Hitpass, L., Beisser, D., Reis, H., Eyking, A., Cario, E., Schulte, J. H., Schramm, A., & Schüller, U. (2016). Characterization of pancreatic glucagon-producing tumors and pituitary gland tumors in transgenic mice overexpressing MYCN in hGFAP-positive cells. ONCOTARGET, 7(46), 74415-74426. https://doi.org/10.18632/oncotarget.12766

Vancouver

Bibtex

@article{b831aff50ec64b2a97a8382a549aea28,
title = "Characterization of pancreatic glucagon-producing tumors and pituitary gland tumors in transgenic mice overexpressing MYCN in hGFAP-positive cells",
abstract = "Amplification or overexpression of MYCN is involved in development and maintenance of multiple malignancies. A subset of these tumors originates from neural precursors, including the most aggressive forms of the childhood tumors, neuroblastoma and medulloblastoma. In order to model the spectrum of MYCN-driven neoplasms in mice, we transgenically overexpressed MYCN under the control of the human GFAP-promoter that, among other targets, drives expression in neural progenitor cells. However, LSL-MYCN;hGFAP-Cre double transgenic mice did neither develop neural crest tumors nor tumors of the central nervous system, but presented with neuroendocrine tumors of the pancreas and, less frequently, the pituitary gland. Pituitary tumors expressed chromogranin A and closely resembled human pituitary adenomas. Pancreatic tumors strongly produced and secreted glucagon, suggesting that they derived from glucagon- and GFAP-positive islet cells. Interestingly, 3 out of 9 human pancreatic neuroendocrine tumors expressed MYCN, supporting the similarity of the mouse tumors to the human system. Serial transplantations of mouse tumor cells into immunocompromised mice confirmed their fully transformed phenotype. MYCN-directed treatment by AuroraA- or Brd4-inhibitors resulted in significantly decreased cell proliferation in vitro and reduced tumor growth in vivo. In summary, we provide a novel mouse model for neuroendocrine tumors of the pancreas and pituitary gland that is dependent on MYCN expression and that may help to evaluate MYCN-directed therapies.",
author = "Kathrin Fielitz and Kristina Althoff and {De Preter}, Katleen and Julie Nonnekens and Jasmin Ohli and Sandra Elges and Wolfgang Hartmann and G{\"u}nter Kl{\"o}ppel and Thomas Kn{\"o}sel and Marc Schulte and Ludger Klein-Hitpass and Daniela Beisser and Henning Reis and Annette Eyking and Elke Cario and Schulte, {Johannes H} and Alexander Schramm and Ulrich Sch{\"u}ller",
year = "2016",
month = oct,
day = "19",
doi = "10.18632/oncotarget.12766",
language = "English",
volume = "7",
pages = "74415--74426",
journal = "ONCOTARGET",
issn = "1949-2553",
publisher = "IMPACT JOURNALS LLC",
number = "46",

}

RIS

TY - JOUR

T1 - Characterization of pancreatic glucagon-producing tumors and pituitary gland tumors in transgenic mice overexpressing MYCN in hGFAP-positive cells

AU - Fielitz, Kathrin

AU - Althoff, Kristina

AU - De Preter, Katleen

AU - Nonnekens, Julie

AU - Ohli, Jasmin

AU - Elges, Sandra

AU - Hartmann, Wolfgang

AU - Klöppel, Günter

AU - Knösel, Thomas

AU - Schulte, Marc

AU - Klein-Hitpass, Ludger

AU - Beisser, Daniela

AU - Reis, Henning

AU - Eyking, Annette

AU - Cario, Elke

AU - Schulte, Johannes H

AU - Schramm, Alexander

AU - Schüller, Ulrich

PY - 2016/10/19

Y1 - 2016/10/19

N2 - Amplification or overexpression of MYCN is involved in development and maintenance of multiple malignancies. A subset of these tumors originates from neural precursors, including the most aggressive forms of the childhood tumors, neuroblastoma and medulloblastoma. In order to model the spectrum of MYCN-driven neoplasms in mice, we transgenically overexpressed MYCN under the control of the human GFAP-promoter that, among other targets, drives expression in neural progenitor cells. However, LSL-MYCN;hGFAP-Cre double transgenic mice did neither develop neural crest tumors nor tumors of the central nervous system, but presented with neuroendocrine tumors of the pancreas and, less frequently, the pituitary gland. Pituitary tumors expressed chromogranin A and closely resembled human pituitary adenomas. Pancreatic tumors strongly produced and secreted glucagon, suggesting that they derived from glucagon- and GFAP-positive islet cells. Interestingly, 3 out of 9 human pancreatic neuroendocrine tumors expressed MYCN, supporting the similarity of the mouse tumors to the human system. Serial transplantations of mouse tumor cells into immunocompromised mice confirmed their fully transformed phenotype. MYCN-directed treatment by AuroraA- or Brd4-inhibitors resulted in significantly decreased cell proliferation in vitro and reduced tumor growth in vivo. In summary, we provide a novel mouse model for neuroendocrine tumors of the pancreas and pituitary gland that is dependent on MYCN expression and that may help to evaluate MYCN-directed therapies.

AB - Amplification or overexpression of MYCN is involved in development and maintenance of multiple malignancies. A subset of these tumors originates from neural precursors, including the most aggressive forms of the childhood tumors, neuroblastoma and medulloblastoma. In order to model the spectrum of MYCN-driven neoplasms in mice, we transgenically overexpressed MYCN under the control of the human GFAP-promoter that, among other targets, drives expression in neural progenitor cells. However, LSL-MYCN;hGFAP-Cre double transgenic mice did neither develop neural crest tumors nor tumors of the central nervous system, but presented with neuroendocrine tumors of the pancreas and, less frequently, the pituitary gland. Pituitary tumors expressed chromogranin A and closely resembled human pituitary adenomas. Pancreatic tumors strongly produced and secreted glucagon, suggesting that they derived from glucagon- and GFAP-positive islet cells. Interestingly, 3 out of 9 human pancreatic neuroendocrine tumors expressed MYCN, supporting the similarity of the mouse tumors to the human system. Serial transplantations of mouse tumor cells into immunocompromised mice confirmed their fully transformed phenotype. MYCN-directed treatment by AuroraA- or Brd4-inhibitors resulted in significantly decreased cell proliferation in vitro and reduced tumor growth in vivo. In summary, we provide a novel mouse model for neuroendocrine tumors of the pancreas and pituitary gland that is dependent on MYCN expression and that may help to evaluate MYCN-directed therapies.

U2 - 10.18632/oncotarget.12766

DO - 10.18632/oncotarget.12766

M3 - SCORING: Journal article

C2 - 27769070

VL - 7

SP - 74415

EP - 74426

JO - ONCOTARGET

JF - ONCOTARGET

SN - 1949-2553

IS - 46

ER -