Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mette Villingshøj is active.

Publication


Featured researches published by Mette Villingshøj.


Journal of Cellular Biochemistry | 2005

Analysis of the epidermal growth factor receptor specific transcriptome: Effect of receptor expression level and an activating mutation

Mikkel Wandahl Pedersen; Nina Pedersen; L. Damstrup; Mette Villingshøj; Søren Ulrik Salling Sønder; Klaus Rieneck; Lone Frier Bovin; Mogens Spang-Thomsen; Hans Skovgaard Poulsen

Overexpression or expression of activating mutations of the epidermal growth factor receptor (EGFR) is common in cancer and correlates with neoplastic progression. The present study employed Affymetrix® oligonucleotide arrays to profile genes induced by ligand‐activated EGFR with the receptor either moderately expressed or overexpressed at an in‐itself transforming level. These changes were compared to those induced by the naturally occurring constitutively active variant EGFRvIII. This study provides novel insight on the activities and mechanisms of EGFRvIII and EGFR mediated transformation, as genes encoding proteins with functions in promoting cell proliferation, invasion, antiapoptosis, and angiogenesis featured prominently in the EGFRvIII‐ and EGFR‐expressing cells. Surprisingly, it was found that ligand‐activated EGFR induced the expression of a large group of genes known to be inducible by interferons. Expression of this module was absent in the EGFRvIII‐expressing cell line and the parental cell line. Treatment with the specific EGFR inhibitor AG1478 indicated that the regulations were primary, receptor‐mediated events. Furthermore, activation of this module correlated with activation of STAT1 and STAT3. The results thus demonstrate that ligand‐activated EGFR at different expression levels results in different kinetics of signaling and induction of gene expression. In addition, the constitutively active variant EGFRvIII seems to activate only a subset of signal pathways and induce a subset of genes as compared to the ligand‐activated EGFR.


Experimental Cell Research | 2011

Maintenance of EGFR and EGFRvIII expressions in an in vivo and in vitro model of human glioblastoma multiforme

Marie-Thérése Stockhausen; Helle Broholm; Mette Villingshøj; Maria Kirchhoff; Tommy Gerdes; Karina Kristoffersen; Michael Kosteljanetz; Mogens Spang-Thomsen; Hans Skovgaard Poulsen

Glioblastoma multiforme (GBM) is the most common, and most aggressive primary brain tumor among adults. A vast majority of the tumors express high levels of the epidermal growth factor receptor (EGFR) as a consequence of gene amplification. Furthermore, gene amplification is often associated with mutation of EGFR, and the constitutive activated deletion variant EGFRvIII is the most common EGFR mutation found in GBM. Activated EGFR signaling, through overexpression and/or mutation, is involved in increased tumorigenic potential. As such, EGFR is an attractive target for GBM therapy. However, clinical studies with EGFR inhibitors have shown inconsistent results, and as such, further knowledge regarding the role of EGFR and EGFRvIII in GBM is needed. For this, an appropriate in vivo/in vitro tumor model is required. Here, we report the establishment of an experimental GBM model in which the expressions of EGFR and EGFRvIII are maintained both in xenograft tumors growing subcutaneously on mice and in cell cultures established in stem cell conditions. With this model it will be possible to further study the role of EGFR and EGFRvIII, and response to targeted therapy, in GBM.


International Journal of Cancer | 2008

EGFR induces expression of IRF-1 via STAT1 and STAT3 activation leading to growth arrest of human cancer cells

Peter Andreas Andersen; Mikkel Wandahl Pedersen; Anders Woetmann; Mette Villingshøj; Marie Thérèse Stockhausen; Niels Ødum; Hans Skovgaard Poulsen

Recently, we reported that epidermal growth factor receptor (EGFR) induce expression of a module of genes known to be inducible by interferons and particularly interferon‐γ. Here we show that the module is tightly regulated by EGFR in the 2 human cancer cell lines that overexpress EGFR, A431 and HN5. The module of genes included the tumor suppressor IRF‐1, which was used as a prototypical member to further investigate the regulation and function of the module. Ligand‐activated EGFR induce expression of IRF‐1 via phosphorylation of STAT1 and STAT3. In contrast, cells expressing the constitutively active cancer specific receptor EGFRvIII are unable to mediate phosphorylation of these STATs and thereby incapable of inducing IRF‐1. We also demonstrate that IRF‐1 is expressed in an EGF dose‐dependent manner, which correlates with inhibition of cell proliferation, and that the regulation of IRF‐1 is partially dependent on intracellular Src family kinase activity. Treatment with the dual specific Abl/c‐Src kinase inhibitor AZD0530 significantly reduces the growth inhibitory effect of high EGF concentrations, signifying that EGFR induced IRF‐1 is responsible for the observed growth inhibition. In addition, we show that media from these EGF treated cancer cells upregulate the activation marker CD69 on both B‐cells and T‐cells in peripheral blood. Taken together, these results suggest that cells acquiring sustained high activity of oncogenes such as EGFR are able to activate genes, whose products mediate growth arrest and activate immune effector cells, and which potentially could be involved in alerting the immune system in vivo leading to elimination of the transformed cells.


Cancer Biology & Therapy | 2013

Level of Notch activation determines the effect on growth and stem cell-like features in glioblastoma multiforme neurosphere cultures

Karina Kristoffersen; Mette Villingshøj; Hans Skovgaard Poulsen; Marie-Thérése Stockhausen

Background: Brain cancer stem-like cells (bCSC) are cancer cells with neural stem cell (NSC)-like properties found in glioblastoma multiforme (GBM) and they are assigned a central role in tumor initiation, progression and relapse. The Notch pathway is important for maintenance and cell fate decisions in the normal NSC population. Notch signaling is often deregulated in GBM and recent results suggest that this pathway plays a significant role in bCSC as well. We therefore wished to further elucidate the role of Notch activation in GBM-derived bCSC. Methods: Human-derived GBM xenograft cells were cultured as NSC-like neurosphere cultures. Notch modulation was accomplished either by blocking the pathway using the γ-secretase inhibitor DAPT or by activating it by transfecting the cells with the constitutive active Notch-1 receptor. Results: GBM neurosphere cultures with high endogenous Notch activation displayed sensitivity toward Notch inhibition with regard to tumorigenic features as demonstrated by increased G0/G1 population and reduced colony formation capacity. Of the NSC-like characteristics, only the primary sphere forming potential was affected, while no effect was observed on self-renewal or differentiation. In contrast, when Notch signaling was activated a decrease in the G0/G1 population and an enhanced capability of colony formation was observed, along with increased self-renewal and de-differentiation. Conclusion: Based on the presented results we propose that active Notch signaling plays a role for cell growth and stem cell-like features in GBM neurosphere cultures and that Notch-targeted anti-bCSC treatment could be feasible for GBM patients with high endogenous Notch pathway activation.


Cancer management and research | 2014

The impact of bevacizumab treatment on survival and quality of life in newly diagnosed glioblastoma patients.

Hans Skovgaard Poulsen; Thomas Urup; Signe Regner Michaelsen; Mikkel Staberg; Mette Villingshøj; Ulrik Lassen

Glioblastoma multiforme (GBM) remains one of the most devastating tumors, and patients have a median survival of 15 months despite aggressive local and systemic therapy, including maximal surgical resection, radiation therapy, and concomitant and adjuvant temozolomide. The purpose of antineoplastic treatment is therefore to prolong life, with a maintenance or improvement of quality of life. GBM is a highly vascular tumor and overexpresses the vascular endothelial growth factor A, which promotes angiogenesis. Preclinical data have suggested that anti-angiogenic treatment efficiently inhibits tumor growth. Bevacizumab is a humanized monoclonal antibody against vascular endothelial growth factor A, and treatment has shown impressive response rates in recurrent GBM. In addition, it has been shown that response is correlated to prolonged survival and improved quality of life. Several investigations in newly diagnosed GBM patients have been performed during recent years to test the hypothesis that newly diagnosed GBM patients should be treated with standard multimodality treatment, in combination with bevacizumab, in order to prolong life and maintain or improve quality of life. The results of these studies along with relevant preclinical data will be described, and pitfalls in clinical and paraclinical endpoints will be discussed.


Cellular Oncology | 2017

Inhibition of histone deacetylases sensitizes glioblastoma cells to lomustine

Mikkel Staberg; Signe Regner Michaelsen; Rikke D. Rasmussen; Mette Villingshøj; Hans Skovgaard Poulsen; Petra Hamerlik

PurposeGlioblastoma (GBM) ranks among the deadliest solid cancers worldwide and its prognosis has remained dismal, despite the use of aggressive chemo-irradiation treatment regimens. Limited drug delivery into the brain parenchyma and frequent resistance to currently available therapies are problems that call for a prompt development of novel therapeutic strategies. While only displaying modest efficacies as mono-therapy in pre-clinical settings, histone deacetylase inhibitors (HDACi) have shown promising sensitizing effects to a number of cytotoxic agents. Here, we sought to investigate the sensitizing effect of the HDACi trichostatin A (TSA) to the alkylating agent lomustine (CCNU), which is used in the clinic for the treatment of GBM.MethodsTwelve primary GBM cell cultures grown as neurospheres were used in this study, as well as one established GBM-derived cell line (U87 MG). Histone deacetylase (HDAC) expression levels were determined using quantitative real-time PCR and Western blotting. The efficacy of either CCNU alone or its combination with TSA was assessed using various assays, i.e., cell viability assays (MTT), cell cycle assays (flow cytometry, FACS), double-strand DNA break (DSB) quantification assays (microscopy/immunofluorescence) and expression profiling assays of proteins involved in apoptosis and cell stress (Western blotting and protein array).ResultsWe found that the HDAC1, 3 and 6 expression levels were significantly increased in GBM samples compared to non-neoplastic brain control samples. Additionally, we found that pre-treatment of GBM cells with TSA resulted in an enhancement of their sensitivity to CCNU, possibly via the accumulation of DSBs, decreased cell proliferation and viability rates, and an increased apoptotic rate.ConclusionFrom our data we conclude that the combined administration of TSA and CCNU eradicates GBM cells with a higher efficacy than either drug alone, thereby opening a novel avenue for the treatment of GBM.


Cancer Investigation | 2009

Improved Response by Co-targeting EGFR/EGFRvIII and Src Family Kinases in Human Cancer Cells

Peter Andreas Andersen; Mette Villingshøj; Hans Skovgaard Poulsen; Marie Thérèse Stockhausen

We hypothesized that co-targeting the epidermal growth factor receptor (EGFR) and Src with the EGFR inhibitor gefitinib and the Src inhibitor AZD0530 would increase growth inhibition and impede migration. Cells overexpressing EGFR were more sensitive to gefitinib than cells expressing mutated EGFR or normal levels of wild-type EGFR. Furthermore, cells with mutated EGFR responded to low doses of gefitinib with increased proliferation. AZD0530 was an effective inhibitor of proliferation and migration, irrespective of EGFR status. These results suggest that co-targeting EGFR and Src might be a valuable treatment approach for malignancies associated with altered expression of EGFR, EGFRvIII, and/or Src.


Cancer Cell International | 2016

Combined EGFR- and notch inhibition display additive inhibitory effect on glioblastoma cell viability and glioblastoma-induced endothelial cell sprouting in vitro

Mikkel Staberg; Signe Regner Michaelsen; Louise Stobbe Olsen; Mette K. Nedergaard; Mette Villingshøj; Marie Thérèse Stockhausen; Petra Hamerlik; Hans Skovgaard Poulsen

BackgroundFor Glioblastoma (GBM) patients, a number of anti-neoplastic strategies using specifically targeting drugs have been tested; however, the effects on survival have been limited. One explanation could be treatment resistance due to redundant signaling pathways, which substantiates the need for combination therapies. In GBM, both the epidermal growth factor receptor (EGFR) and the notch signaling pathways are often deregulated and linked to cellular growth, invasion and angiogenesis. Several studies have confirmed cross-talk and co-dependence of these pathways. Therefore, this study aimed at testing a combination treatment strategy using inhibitors targeting the notch and EGFR pathways.MethodsFor evaluation of cell viability a standard MTT assay was used. Western blotting (WB) and Q-RT-PCR were employed in order to assess the protein- and mRNA expression levels, respectively. In order to determine angiogenic processes, we used an endothelial spheroid sprouting assay. For assessment of secreted VEGF from GBM cells we performed a VEGF-quantikine ELISA.ResultsGBM cells were confirmed to express EGFR and Notch and to have the capacity to induce endothelial cell sprouting. Inhibition of EGFR and Notch signaling was achieved using either Iressa (gefitinib) or the gamma-secretase inhibitor DAPT. Our data showed that DAPT combined with Iressa treatment displayed increased inhibitory effect on cell viability and abrogated expression and activation of major pro-survival pathways. Similarly, the combinational treatment significantly increased abrogation of GBM-induced endothelial cell sprouting suggesting reduced GBM angiogenesis.ConclusionThis study finds that simultaneous targeting of notch and EGFR signaling leads to enhanced inhibitory effects on GBM-induced angiogenesis and cell viability, thereby stressing the importance of further evaluation of this targeting approach in a clinical setting.


Neuro-oncology | 2018

VEGF-C sustains VEGFR2 activation under bevacizumab therapy and promotes glioblastoma maintenance

Signe Regner Michaelsen; Mikkel Staberg; Henriette Pedersen; Kamilla E. Jensen; Wiktor Majewski; Helle Broholm; Mette K. Nedergaard; Christopher Meulengracht; Thomas Urup; Mette Villingshøj; Slávka Lukacova; Jane Skjøth-Rasmussen; Jannick Brennum; Andreas Kjær; Ulrik Lassen; Marie-Thérése Stockhausen; Hans Skovgaard Poulsen; Petra Hamerlik

Abstract Background Glioblastoma ranks among the most lethal cancers, with current therapies offering only palliation. Paracrine vascular endothelial growth factor (VEGF) signaling has been targeted using anti-angiogenic agents, whereas autocrine VEGF/VEGF receptor 2 (VEGFR2) signaling is poorly understood. Bevacizumab resistance of VEGFR2-expressing glioblastoma cells prompted interrogation of autocrine VEGF-C/VEGFR2 signaling in glioblastoma. Methods Autocrine VEGF-C/VEGFR2 signaling was functionally investigated using RNA interference and exogenous ligands in patient-derived xenograft lines and primary glioblastoma cell cultures in vitro and in vivo. VEGF-C expression and interaction with VEGFR2 in a matched pre- and post-bevacizumab treatment cohort were analyzed by immunohistochemistry and proximity ligation assay. Results VEGF-C was expressed by patient-derived xenograft glioblastoma lines, primary cells, and matched surgical specimens before and after bevacizumab treatment. VEGF-C activated autocrine VEGFR2 signaling to promote cell survival, whereas targeting VEGF-C expression reprogrammed cellular transcription to attenuate survival and cell cycle progression. Supporting potential translational significance, targeting VEGF-C impaired tumor growth in vivo, with superiority to bevacizumab treatment. Conclusions Our results demonstrate VEGF-C serves as both a paracrine and an autocrine pro-survival cytokine in glioblastoma, promoting tumor cell survival and tumorigenesis. VEGF-C permits sustained VEGFR2 activation and tumor growth, where its inhibition appears superior to bevacizumab therapy in improving tumor control.


Cancer Biology & Therapy | 2014

Inhibition of Notch signaling alters the phenotype of orthotopic tumors formed from glioblastoma multiforme neurosphere cells but does not hamper intracranial tumor growth regardless of endogene Notch pathway signature

Karina Kristoffersen; Mette K. Nedergaard; Mette Villingshøj; Rehannah Borup; Helle Broholm; Andreas Kjær; Hans Skovgaard Poulsen; Marie-Thérése Stockhausen

Background Brain cancer stem-like cells (bCSC) are cancer cells with neural stem cell (NSC)-like properties found in the devastating brain tumor glioblastoma multiforme (GBM). bCSC are proposed a central role in tumor initiation, progression, treatment resistance and relapse and as such present a promising target in GBM research. The Notch signaling pathway is often deregulated in GBM and we have previously characterized GBM-derived bCSC cultures based on their expression of the Notch-1 receptor and found that it could be used as predictive marker for the effect of Notch inhibition. The aim of the present project was therefore to further elucidate the significance of Notch pathway activity for the tumorigenic properties of GBM-derived bCSC. Methods Human-derived GBM xenograft cells previously established as NSC-like neurosphere cultures were used. Notch inhibition was accomplished by exposing the cells to the gamma-secretase inhibitor DAPT prior to gene expression analysis and intracranial injection into immunocompromised mice. Results By analyzing the expression of several Notch pathway components, we found that the cultures indeed displayed different Notch pathway signatures. However, when DAPT-treated neurosphere cells were injected into the brain of immunocompromised mice, no increase in survival was obtained regardless of Notch pathway signature and Notch inhibition. We did however observe a decrease in the expression of the stem cell marker Nestin, an increase in the proliferative marker Ki-67 and an increased number of abnormal vessels in tumors formed from DAPT-treated, high Notch-1 expressing cultures, when compared with the control. Conclusion Based on the presented results we propose that Notch inhibition partly induces differentiation of bCSC, and selects for a cell type that more strongly induces angiogenesis if the treatment is not sustained. However, this more differentiated cell type might prove to be more sensitive to conventional therapies.

Collaboration


Dive into the Mette Villingshøj's collaboration.

Top Co-Authors

Avatar

Hans Skovgaard Poulsen

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Signe Regner Michaelsen

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar

Mikkel Staberg

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar

Petra Hamerlik

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar

Helle Broholm

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar

Mette K. Nedergaard

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar

Karina Kristoffersen

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar

Mogens Spang-Thomsen

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar

Andreas Kjær

University of Copenhagen

View shared research outputs
Researchain Logo
Decentralizing Knowledge