Network


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

Hotspot


Dive into the research topics where Christoph Wiesner is active.

Publication


Featured researches published by Christoph Wiesner.


Cancer Letters | 2012

Acetylation of histone H3 prevents resistance development caused by chronic mTOR inhibition in renal cell carcinoma cells

Eva Juengel; Anna Dauselt; Jasmina Makarević; Christoph Wiesner; Igor Tsaur; Georg Bartsch; Axel Haferkamp; Roman A. Blaheta

Chronic mTOR inhibition may induce resistance development in renal cell carcinoma (RCC). We analyzed whether long-term exposure of RCC cells to the mTOR-inhibitor RAD001 evokes resistance and whether additional targeting histone deacetylases (HDAC) by valproic acid (VPA) overcomes RAD001 resistance. It is demonstrated that responsiveness to either drug alone is lost over time, evidenced by increased cell growth, proliferation and de-differentiation. However, drug sensitivity was conserved when RAD001 and VPA were applied in concert. Molecular analysis particularly revealed strong re-activation of Akt under chronic RAD001 or diminished histone H3 acetylation under chronic VPA single drug exposure. Combined drug application did not inactivate Akt but rather resulted in H3 acetylation remaining high while RCC cell growth was still reduced. siRNA experiments revealed that histone H3 acetylation is responsible for preserving drug sensitivity in RCCs.


Life Sciences | 2011

Inhibitory effects of the HDAC inhibitor valproic acid on prostate cancer growth are enhanced by simultaneous application of the mTOR inhibitor RAD001

Steffen Wedel; Lukasz Hudak; Jens-Michael Seibel; Eva Juengel; Igor Tsaur; Christoph Wiesner; Axel Haferkamp; Roman A. Blaheta

AIMS To analyze the combined impact of the histone deacetylase (HDAC) inhibitor valproic acid (VPA) and the mammalian target of rapamycin (mTOR) inhibitor RAD001 on prostate cancer cell growth. MAIN METHODS PC-3, DU-145 and LNCaP cells were treated with RAD001, VPA or with an RAD001-VPA combination for 3 or 5 days. Tumor cell growth, cell cycle progression and cell cycle regulating proteins were then investigated by MTT assay, flow cytometry and Western blotting, respectively. Effects of drug treatment on cell signaling pathways were determined. KEY FINDINGS Separate application of RAD001 or VPA distinctly reduced tumor cell growth and impaired cell cycle progression. Significant additive effects were evoked when both drugs were used in concert. Particularly, the cell cycle regulating proteins cdk1, cdk2, cdk4 and cyclin B were reduced, whereas p21 and p27 were enhanced by the RAD001-VPA combination. Signaling analysis revealed deactivation of EGFr, ERK1/2 and p70S6k. Phosphorylation of Akt was diminished in DU-145 but elevated in PC-3 and LNCaP cells. SIGNIFICANCE The RAD001-VPA combination exerted profound antitumor properties on a panel of prostate cancer cell lines. Therefore, simultaneous blockage of HDAC and mTOR related pathways should be considered when designing novel therapeutic strategies for treating prostate carcinoma.


Cancer Letters | 2011

The cdk1-cyclin B complex is involved in everolimus triggered resistance in the PC3 prostate cancer cell line

Igor Tsaur; Jasmina Makarević; Lukasz Hudak; Eva Juengel; M. Kurosch; Christoph Wiesner; Georg Bartsch; Sebastian Harder; Axel Haferkamp; Roman A. Blaheta

The growth potential of PC3 prostate cancer cells, sensible (PC3(par)) or resistant (PC3(res)) to the mTOR inhibitor everolimus (RAD001) was investigated. Cell growth and proliferation of PC3(res) was similar to that of PC3(par), and late apoptosis increased in PC3(par) but decreased in PC3(res) following treatment with low dosed everolimus. PC3(res) accumulated in the G2/M-phase, accompanied by cdk1, cdk2 and cyclin B elevation. Knocking down cdk1 or cyclin B distinctly blocked the growth activity of PC3(res). One reason for everolimus resistance may be up-regulation of the cdk1-cyclin B complex in prostate cancer cells, leading to enhanced progression towards G2/M.


Anti-Cancer Drugs | 2011

HDAC inhibition delays cell cycle progression of human bladder cancer cells in vitro.

Stefan Vallo; Wang Xi; Lukasz Hudak; Eva Juengel; Igor Tsaur; Christoph Wiesner; Axel Haferkamp; Roman A. Blaheta

Our aim was to analyze the impact of the histone deacetylase (HDAC)-inhibitor valproic acid (VPA) on bladder cancer cell growth in vitro. RT-4, TCCSUP, UMUC-3, and RT-112 bladder cancer cells were treated with VPA (0.125–1 mmol/l) without and with preincubation periods of 3 and 5 days. Controls remained untreated. Tumor cell growth, cell cycle progression, and cell cycle-regulating proteins were investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, flow cytometry, and western blotting, respectively. Effects of VPA on histone H3 and H4 acetylation and HDAC3 and HDAC4 were also determined. Without preincubation, no tumor cell growth reduction was observed with 0.125 and 0.25 mmol/l VPA in TCCSUP, UMUC-3, and RT-112 cells, whereas 0.5 and 1 mmol/l VPA diminished the cell number significantly. VPA (0.25 mmol/l) did exert tumor growth-blocking effects after a 3-day preincubation. To achieve antitumor effects with VPA (0.125 mmol/l), a 5-day preincubation was necessary. A 3-day or 5-day preincubation was also necessary to distinctly delay cell cycle progression, with maximum effects at VPA (1 mmol/l). After the 5-day preincubation, the cell cycle-regulating proteins cdk1, cdk2, cdk4, and cyclins B, D1, and E were reduced, whereas p27 was enhanced. Diminished HDAC3 and 4 expression induced by VPA was accompanied by elevated acetylation of H3 and H4. VPA exerted growth-blocking properties on a panel of bladder cancer cell lines, commensurate with dose and exposure time. Long-term application induced much stronger effects than did shorter application and should be considered when designing therapeutic strategies for treating bladder carcinoma.


The Prostate | 2012

Low dosed interferon alpha augments the anti-tumor potential of histone deacetylase inhibition on prostate cancer cell growth and invasion.

Lukasz Hudak; Patrick Tezeeh; Steffen Wedel; Jasmina Makarević; Eva Juengel; Igor Tsaur; Georg Bartsch; Christoph Wiesner; Axel Haferkamp; Roman A. Blaheta

We evaluated whether low‐dosed interferon alpha (IFNa) may augment the anti‐tumor potential of the histone deacetylase (HDAC)‐inhibitor valproic acid (VPA) on prostate cancer cells in vitro and in vivo. PC‐3, DU‐145, or LNCaP prostate cancer cells were treated with VPA (1 mM), IFNa (200 U/ml), or with the VPA‐IFNa combination. Tumor cell growth, cell cycle progression, and cell cycle regulating proteins were then investigated by the MTT assay, flow cytometry, and western blotting. Tumor cell adhesion to endothelium or to immobilized extracellular matrix proteins, as well as migratory properties of the cells, were evaluated. Integrin α and β adhesion molecules and alterations of cell signaling pathways were analyzed. Finally, effects of the drug treatment on prostate cancer growth in vivo were determined in the NOD/SCID mouse model. VPA reduced tumor cell adhesion, migration, and growth in vitro. A much stronger anti‐cancer potential was evoked by the VPA‐IFNa combination, although IFNa in itself did not block growth or adhesion. The same effect was seen when tumor growth was evaluated in vivo. Molecular analysis revealed distinct elevation of histone H3 acetylation caused by VPA which was further up‐regulated by VPA‐IFNa, whereas IFNa alone did not alter H3 acetylation. The combinatorial benefit became obvious in Akt phosphorylation, p21 and p27 and integrin α1, α3, and β1 expression. Application of low‐dosed IFNa to a VPA based regimen profoundly boosts the anti‐tumor properties of VPA. The combined use of VPA and low‐dosed IFNa may therefore be an innovative option in treating advanced prostate cancer. Prostate 72:1719–1735, 2012.


Experimental Biology and Medicine | 2013

HDAC inhibition suppresses bladder cancer cell adhesion to collagen under flow conditions

Eva Juengel; Sascha Meyer dos Santos; Tanja Schneider; Jasmina Makarević; Lukasz Hudak; Georg Bartsch; Axel Haferkamp; Christoph Wiesner; Roman A. Blaheta

The influence of the histone deacetylase (HDAC)-inhibitor, valproic acid (VPA), on bladder cancer cell adhesion in vitro was investigated in this paper. TCCSUP and RT-112 bladder cancer cells were treated with VPA (0.5 or 1 mM) twice or thrice weekly for 14 days. Controls remained untreated. Tumour cell interaction with immobilized collagen was evaluated by a flow-based adhesion assay using a shear force of 2 or 4 dyne/cm2. The effects of VPA on the integrin adhesion receptors α3, α5, β1, β3 and β4 were assessed by flow cytometry to determine integrin surface expression and by western blotting to determine the cytoplasmic integrin level. VPA of 0.5 mM and 1 mM significantly prevented binding of both RT-112 and TCCSUP cells to collagen, compared with the untreated controls. Adhesion was reduced to a higher extent when RT-112 (subjected to 2 dyne/cm2) or TCCSUP (subjected to 2 or 4 dyne/cm2) tumour cells were treated with VPA three times a week, compared to the two times a week protocol. VPA caused a significant up-regulation of the integrin α3, α5, β1, β3 and β4 subtypes on the TCCSUP cell surface membrane. In RT-112 cells, only integrin α5 was elevated on the cell surface following VPA exposure. Western blotting revealed an up-regulation of α3, α5, β3 and β4 integrins and down-regulation of the integrin β1 protein by VPA in TCCSUP. VPA also up-regulated α5 and down-regulated β1 integrin in RT-112 cells, but also reduced α3 and β3 in TCCSUP. VPA exerted adhesion-blocking properties on bladder cancer cells under physiologic flow conditions. The effects were accompanied by distinct modifications of the integrin expression profile, which differ depending on the cell lines used. Application of VPA might be an innovative option to prevent bladder cancer dissemination.


Review of Scientific Instruments | 2016

Beam transport and space charge compensation strategies (invited)

O. Meusel; M. Droba; Daniel Noll; K. Schulte; Philipp Schneider; Christoph Wiesner

The transport of intense ion beams is affected by the collective behavior of this kind of multi-particle and multi-species system. The space charge expressed by the generalized perveance dominates the dynamical process of thermalisation, which leads to emittance growth. To prevent changes of intrinsic beam properties and to reduce the intensity dependent focusing forces, space charge compensation seems to be an adequate solution. In the case of positively charged ion beams, electrons produced by residual gas ionization and secondary electrons provide the space charge compensation. The influence of the compensation particles on the beam transport and the local degree of space charge compensation is given by different beam properties as well as the ion beam optics. Especially for highly charged ion beams, space charge compensation in combination with poor vacuum conditions leads to recombination processes and therefore increased beam losses. Strategies for providing a compensation-electron reservoir at very low residual gas pressures will be discussed.


Clinical & Experimental Metastasis | 2011

Impact of combined HDAC and mTOR inhibition on adhesion, migration and invasion of prostate cancer cells

Steffen Wedel; Lukasz Hudak; Jens-Michael Seibel; Jasmina Makarević; Eva Juengel; Igor Tsaur; Christoph Wiesner; Axel Haferkamp; Roman A. Blaheta


European Physical Journal Plus | 2016

The Frankfurt neutron source FRANZ

Suha Alzubaidi; Ulrich Bartz; Markus Basten; Alexander Bechtold; L.P.Chau; Christine Claessens; Hannes Dinter; M. Droba; Christopher Fix; Hendrik Hähnel; Manuel Heilmann; O. Hinrichs; Simon Huneck; Batu Klump; Marcel Lotz; Dominik Mäder; O. Meusel; Daniel Noll; Tobias Nowottnick; Marcus Obermayer; Onur Payir; Nils Petry; Holger Podlech; U. Ratzinger; A. Schempp; Stefan Schmidt; Philipp Schneider; Anja Seibel; Malte Schwarz; W. Schweizer


Archive | 2012

FRANZ - ACCELERATOR TEST BENCH AND NEUTRON SOURCE

O. Meusel; L.P.Chau; Manuel Heilmann; Holger Podlech; U. Ratzinger; K.Volk; Christoph Wiesner

Collaboration


Dive into the Christoph Wiesner's collaboration.

Top Co-Authors

Avatar

O. Meusel

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

U. Ratzinger

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

M. Droba

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Daniel Noll

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eva Juengel

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Roman A. Blaheta

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

L.P.Chau

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Lukasz Hudak

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Hannes Dinter

Goethe University Frankfurt

View shared research outputs
Researchain Logo
Decentralizing Knowledge