Targeting HDACs in Pancreatic Neuroendocrine Tumor Models

Standard

Targeting HDACs in Pancreatic Neuroendocrine Tumor Models. / Schmitz, Rosa Lynn; Weissbach, Julia; Kleilein, Jan; Bell, Jessica; Hüttelmaier, Stefan; Viol, Fabrice; Clauditz, Till; Grabowski, Patricia; Laumen, Helmut; Rosendahl, Jonas; Michl, Patrick; Schrader, Jörg; Krug, Sebastian.

In: CELLS-BASEL, Vol. 10, No. 6, 06.06.2021, p. 1408.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Schmitz, RL, Weissbach, J, Kleilein, J, Bell, J, Hüttelmaier, S, Viol, F, Clauditz, T, Grabowski, P, Laumen, H, Rosendahl, J, Michl, P, Schrader, J & Krug, S 2021, 'Targeting HDACs in Pancreatic Neuroendocrine Tumor Models', CELLS-BASEL, vol. 10, no. 6, pp. 1408. https://doi.org/10.3390/cells10061408

APA

Schmitz, R. L., Weissbach, J., Kleilein, J., Bell, J., Hüttelmaier, S., Viol, F., Clauditz, T., Grabowski, P., Laumen, H., Rosendahl, J., Michl, P., Schrader, J., & Krug, S. (2021). Targeting HDACs in Pancreatic Neuroendocrine Tumor Models. CELLS-BASEL, 10(6), 1408. https://doi.org/10.3390/cells10061408

Vancouver

Schmitz RL, Weissbach J, Kleilein J, Bell J, Hüttelmaier S, Viol F et al. Targeting HDACs in Pancreatic Neuroendocrine Tumor Models. CELLS-BASEL. 2021 Jun 6;10(6):1408. https://doi.org/10.3390/cells10061408

Bibtex

@article{229814d6ff8b44d5980dc946348ee42b,
title = "Targeting HDACs in Pancreatic Neuroendocrine Tumor Models",
abstract = "Compared to pancreatic adenocarcinoma (PDAC), pancreatic neuroendocrine tumors (PanNET) represent a rare and heterogeneous tumor entity. In addition to surgical resection, several therapeutic approaches, including biotherapy, targeted therapy or chemotherapy are applicable. However, primary or secondary resistance to current therapies is still challenging. Recent genome-wide sequencing efforts in PanNET identified a large number of mutations in pathways involved in epigenetic modulation, including acetylation. Therefore, targeting epigenetic modulators in neuroendocrine cells could represent a new therapeutic avenue. Detailed information on functional effects and affected signaling pathways upon epigenetic targeting in PanNETs, however, is missing. The primary human PanNET cells NT-3 and NT-18 as well as the murine insulinoma cell lines beta-TC-6 (mouse) and RIN-T3 (rat) were treated with the non-selective histone-deacetylase (HDAC) inhibitor panobinostat (PB) and analyzed for functional effects and affected signaling pathways by performing Western blot, FACS and qPCR analyses. Additionally, NanoString analysis of more than 500 potentially affected targets was performed. In vivo immunohistochemistry (IHC) analyses on tumor samples from xenografts and the transgenic neuroendocrine Rip1Tag2-mouse model were investigated. PB dose dependently induced cell cycle arrest and apoptosis in neuroendocrine cells in human and murine species. HDAC inhibition stimulated redifferentiation of human primary PanNET cells by increasing mRNA-expression of somatostatin receptors (SSTRs) and insulin production. In addition to hyperacetylation of known targets, PB mediated pleitropic effects via targeting genes involved in the cell cycle and modulation of the JAK2/STAT3 axis. The HDAC subtypes are expressed ubiquitously in the existing cell models and in human samples of metastatic PanNET. Our results uncover epigenetic HDAC modulation using PB as a promising new therapeutic avenue in PanNET, linking cell-cycle modulation and pathways such as JAK2/STAT3 to epigenetic targeting. Based on our data demonstrating a significant impact of HDAC inhibition in clinical relevant in vitro models, further validation in vivo is warranted.",
author = "Schmitz, {Rosa Lynn} and Julia Weissbach and Jan Kleilein and Jessica Bell and Stefan H{\"u}ttelmaier and Fabrice Viol and Till Clauditz and Patricia Grabowski and Helmut Laumen and Jonas Rosendahl and Patrick Michl and J{\"o}rg Schrader and Sebastian Krug",
year = "2021",
month = jun,
day = "6",
doi = "10.3390/cells10061408",
language = "English",
volume = "10",
pages = "1408",
journal = "CELLS-BASEL",
issn = "2073-4409",
publisher = "MDPI Multidisciplinary Digital Publishing Institute",
number = "6",

}

RIS

TY - JOUR

T1 - Targeting HDACs in Pancreatic Neuroendocrine Tumor Models

AU - Schmitz, Rosa Lynn

AU - Weissbach, Julia

AU - Kleilein, Jan

AU - Bell, Jessica

AU - Hüttelmaier, Stefan

AU - Viol, Fabrice

AU - Clauditz, Till

AU - Grabowski, Patricia

AU - Laumen, Helmut

AU - Rosendahl, Jonas

AU - Michl, Patrick

AU - Schrader, Jörg

AU - Krug, Sebastian

PY - 2021/6/6

Y1 - 2021/6/6

N2 - Compared to pancreatic adenocarcinoma (PDAC), pancreatic neuroendocrine tumors (PanNET) represent a rare and heterogeneous tumor entity. In addition to surgical resection, several therapeutic approaches, including biotherapy, targeted therapy or chemotherapy are applicable. However, primary or secondary resistance to current therapies is still challenging. Recent genome-wide sequencing efforts in PanNET identified a large number of mutations in pathways involved in epigenetic modulation, including acetylation. Therefore, targeting epigenetic modulators in neuroendocrine cells could represent a new therapeutic avenue. Detailed information on functional effects and affected signaling pathways upon epigenetic targeting in PanNETs, however, is missing. The primary human PanNET cells NT-3 and NT-18 as well as the murine insulinoma cell lines beta-TC-6 (mouse) and RIN-T3 (rat) were treated with the non-selective histone-deacetylase (HDAC) inhibitor panobinostat (PB) and analyzed for functional effects and affected signaling pathways by performing Western blot, FACS and qPCR analyses. Additionally, NanoString analysis of more than 500 potentially affected targets was performed. In vivo immunohistochemistry (IHC) analyses on tumor samples from xenografts and the transgenic neuroendocrine Rip1Tag2-mouse model were investigated. PB dose dependently induced cell cycle arrest and apoptosis in neuroendocrine cells in human and murine species. HDAC inhibition stimulated redifferentiation of human primary PanNET cells by increasing mRNA-expression of somatostatin receptors (SSTRs) and insulin production. In addition to hyperacetylation of known targets, PB mediated pleitropic effects via targeting genes involved in the cell cycle and modulation of the JAK2/STAT3 axis. The HDAC subtypes are expressed ubiquitously in the existing cell models and in human samples of metastatic PanNET. Our results uncover epigenetic HDAC modulation using PB as a promising new therapeutic avenue in PanNET, linking cell-cycle modulation and pathways such as JAK2/STAT3 to epigenetic targeting. Based on our data demonstrating a significant impact of HDAC inhibition in clinical relevant in vitro models, further validation in vivo is warranted.

AB - Compared to pancreatic adenocarcinoma (PDAC), pancreatic neuroendocrine tumors (PanNET) represent a rare and heterogeneous tumor entity. In addition to surgical resection, several therapeutic approaches, including biotherapy, targeted therapy or chemotherapy are applicable. However, primary or secondary resistance to current therapies is still challenging. Recent genome-wide sequencing efforts in PanNET identified a large number of mutations in pathways involved in epigenetic modulation, including acetylation. Therefore, targeting epigenetic modulators in neuroendocrine cells could represent a new therapeutic avenue. Detailed information on functional effects and affected signaling pathways upon epigenetic targeting in PanNETs, however, is missing. The primary human PanNET cells NT-3 and NT-18 as well as the murine insulinoma cell lines beta-TC-6 (mouse) and RIN-T3 (rat) were treated with the non-selective histone-deacetylase (HDAC) inhibitor panobinostat (PB) and analyzed for functional effects and affected signaling pathways by performing Western blot, FACS and qPCR analyses. Additionally, NanoString analysis of more than 500 potentially affected targets was performed. In vivo immunohistochemistry (IHC) analyses on tumor samples from xenografts and the transgenic neuroendocrine Rip1Tag2-mouse model were investigated. PB dose dependently induced cell cycle arrest and apoptosis in neuroendocrine cells in human and murine species. HDAC inhibition stimulated redifferentiation of human primary PanNET cells by increasing mRNA-expression of somatostatin receptors (SSTRs) and insulin production. In addition to hyperacetylation of known targets, PB mediated pleitropic effects via targeting genes involved in the cell cycle and modulation of the JAK2/STAT3 axis. The HDAC subtypes are expressed ubiquitously in the existing cell models and in human samples of metastatic PanNET. Our results uncover epigenetic HDAC modulation using PB as a promising new therapeutic avenue in PanNET, linking cell-cycle modulation and pathways such as JAK2/STAT3 to epigenetic targeting. Based on our data demonstrating a significant impact of HDAC inhibition in clinical relevant in vitro models, further validation in vivo is warranted.

U2 - 10.3390/cells10061408

DO - 10.3390/cells10061408

M3 - SCORING: Journal article

C2 - 34204116

VL - 10

SP - 1408

JO - CELLS-BASEL

JF - CELLS-BASEL

SN - 2073-4409

IS - 6

ER -