Network


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

Hotspot


Dive into the research topics where Grischa Toedt is active.

Publication


Featured researches published by Grischa Toedt.


Journal of Clinical Investigation | 2008

BRAF gene duplication constitutes a mechanism of MAPK pathway activation in low-grade astrocytomas

Stefan M. Pfister; Wibke G. Janzarik; Marc Remke; Aurélie Ernst; Wiebke Werft; Natalia Becker; Grischa Toedt; Andrea Wittmann; Christian P. Kratz; Heike Olbrich; Rezvan Ahmadi; Barbara Thieme; Stefan Joos; Bernhard Radlwimmer; Andreas E. Kulozik; Torsten Pietsch; Christel Herold-Mende; Astrid Gnekow; Guido Reifenberger; Andrey Korshunov; Wolfram Scheurlen; Heymut Omran; Peter Lichter

The molecular pathogenesis of pediatric astrocytomas is still poorly understood. To further understand the genetic abnormalities associated with these tumors, we performed a genome-wide analysis of DNA copy number aberrations in pediatric low-grade astrocytomas by using array-based comparative genomic hybridization. Duplication of the BRAF protooncogene was the most frequent genomic aberration, and tumors with BRAF duplication showed significantly increased mRNA levels of BRAF and a downstream target, CCND1, as compared with tumors without duplication. Furthermore, denaturing HPLC showed that activating BRAF mutations were detected in some of the tumors without BRAF duplication. Similarly, a marked proportion of low-grade astrocytomas from adult patients also had BRAF duplication. Both the stable silencing of BRAF through shRNA lentiviral transduction and pharmacological inhibition of MEK1/2, the immediate downstream phosphorylation target of BRAF, blocked the proliferation and arrested the growth of cultured tumor cells derived from low-grade gliomas. Our findings implicate aberrant activation of the MAPK pathway due to gene duplication or mutation of BRAF as a molecular mechanism of pathogenesis in low-grade astrocytomas and suggest inhibition of the MAPK pathway as a potential treatment.


Nucleic Acids Research | 2012

ELM—the database of eukaryotic linear motifs

Holger Dinkel; Sushama Michael; Robert J. Weatheritt; Norman E. Davey; Kim Van Roey; Brigitte Altenberg; Grischa Toedt; Bora Uyar; Markus Seiler; Aidan Budd; Lisa Jödicke; Marcel Andre Dammert; Christian Schroeter; Maria Hammer; Tobias Schmidt; Peter Jehl; Caroline McGuigan; Magdalena Dymecka; Claudia Chica; Katja Luck; Allegra Via; Andrew Chatr-aryamontri; Niall J. Haslam; Gleb Grebnev; Richard J. Edwards; Michel O. Steinmetz; Heike Meiselbach; Francesca Diella; Toby J. Gibson

Linear motifs are short, evolutionarily plastic components of regulatory proteins and provide low-affinity interaction interfaces. These compact modules play central roles in mediating every aspect of the regulatory functionality of the cell. They are particularly prominent in mediating cell signaling, controlling protein turnover and directing protein localization. Given their importance, our understanding of motifs is surprisingly limited, largely as a result of the difficulty of discovery, both experimentally and computationally. The Eukaryotic Linear Motif (ELM) resource at http://elm.eu.org provides the biological community with a comprehensive database of known experimentally validated motifs, and an exploratory tool to discover putative linear motifs in user-submitted protein sequences. The current update of the ELM database comprises 1800 annotated motif instances representing 170 distinct functional classes, including approximately 500 novel instances and 24 novel classes. Several older motif class entries have been also revisited, improving annotation and adding novel instances. Furthermore, addition of full-text search capabilities, an enhanced interface and simplified batch download has improved the overall accessibility of the ELM data. The motif discovery portion of the ELM resource has added conservation, and structural attributes have been incorporated to aid users to discriminate biologically relevant motifs from stochastically occurring non-functional instances.


Clinical Cancer Research | 2006

Identification of Gains on 1q and Epidermal Growth Factor Receptor Overexpression as Independent Prognostic Markers in Intracranial Ependymoma

Frank Mendrzyk; Andrey Korshunov; Axel Benner; Grischa Toedt; Stefan M. Pfister; Bernhard Radlwimmer; Peter Lichter

Purpose: Pathogenesis of ependymomas is still poorly understood and molecular markers for risk-adapted patient stratification are not available. Our aim was to screen for novel genomic imbalances and prognostic markers in ependymal tumors. Experimental Design: We analyzed 68 sporadic tumors by matrix-based comparative genomic hybridization using DNA microarrays containing >6,400 genomic DNA fragments. Novel recurrent genomic gains were validated by fluorescence in situ hybridization using a tissue microarray consisting of 170 intracranial ependymomas. Candidate genes were also tested for mRNA expression by quantitative real-time PCR, and protein expression was determined by immunohistochemistry on the tissue microarray. Results: Chromosomal gain of 1q correlated with pediatric patients (P = 0.004), intracranial ependymomas (P = 0.05), and tumors of grade III (P = 0.002). Gain of 1q21.1-32.1 was associated with tumor recurrence in intracranial ependymomas (P < 0.001). Furthermore, gain of 1q25 as determined by fluorescence in situ hybridization represented an independent prognostic marker for either recurrence-free survival (P < 0.001) or overall survival (P = 0.003). Recurrent gains at 5p15.33 covering hTERT were validated by immunohistochemistry, and elevated protein levels correlated with adverse prognosis (P = 0.01). In addition to frequent gains and high-level amplification of epidermal growth factor receptor (EGFR) at 7p11.2, immunohistochemistry revealed protein overexpression to be correlated with poor prognosis (P = 0.002). EGFR protein status subdivides intracranial grade II ependymomas into two different risk groups (P = 0.03) as shown by multivariate analysis. Conclusions: Thus, the states of 1q25 and EGFR represent independent prognostic markers for intracranial ependymomas to identify patient subgroups with different risk profiles in further clinical investigations. Moreover, EGFR might serve as therapeutic target for more specific chemotherapy applications.


Journal of Clinical Oncology | 2006

Gene Expression Signature Predicting Pathologic Complete Response With Gemcitabine, Epirubicin, and Docetaxel in Primary Breast Cancer

Olaf Thuerigen; Andreas Schneeweiss; Grischa Toedt; Patrick Warnat; Meinhard Hahn; Heidi Kramer; Benedikt Brors; Christian Rudlowski; Axel Benner; Florian Schuetz; Bjoern Tews; Roland Eils; Hans Peter Sinn; Christof Sohn; Peter Lichter

PURPOSE Primary systemic therapy (PST) with gemcitabine (G), epirubicin (E), and docetaxel (Doc) has resulted in a pathologic complete response (pCR) in 26% of primary breast cancer patients. This study was aimed at the identification of a gene expression signature in diagnostic core biopsy tissue samples that predicts pCR. PATIENTS AND METHODS Core biopsy samples from patients with operable primary breast cancer, T2-4N0-2M0, enrolled onto two phase I and II trials evaluating GEDoc (n = 48) and GE sequentially followed by Doc (GEsDoc; n = 52) as PST were snap frozen and subjected to RNA expression profiling. A signature predicting pCR was discovered in the training set (GEsDoc) applying a support vector machine algorithm, and performance of this classifier was validated on the independent test set (GEDoc) by receiver operator characteristics analysis. RESULTS We identified a signature consisting of 512 genes, which was enriched in genes involved in transforming growth factor beta and RAS-mediated signaling pathways, that predicts pCR with a sensitivity of 78%, a specificity of 90%, and an overall accuracy of 88% (95% CI, 75% to 95%). Apart from our signature, only HER2 overexpression was an independent predictor of pCR in multivariate analysis. CONCLUSION In conclusion, our gene expression signature allows prediction of pCR to PST containing G, E, and Doc with unprecedented high overall accuracy and robustness.


Hepatology | 2007

Etiology‐dependent molecular mechanisms in human hepatocarcinogenesis

Christof Schlaeger; T Longerich; Claudia Schiller; Peter Bewerunge; Arianeb Mehrabi; Grischa Toedt; Jörg Kleeff; Volker Ehemann; Roland Eils; Peter Lichter; Peter Schirmacher; Bernhard Radlwimmer

Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide and is characterized by aggressive tumor behavior coupled with poor prognosis. Various etiologies have been linked to HCC development, most prominently chronic hepatitis B and C virus infections as well as chronic alcohol consumption. In approximately 10% of HCCs, the etiology remains cryptic; however, recent epidemiological data suggest that most of these cryptogenic HCCs develop due to nonalcoholic steatohepatitis. To identify etiology‐dependent DNA copy number aberrations and genes relevant to hepatocarcinogenesis, we performed array‐based comparative genomic hybridization of 63 HCCs of well‐defined etiology and 4 HCC cell lines followed by gene expression profiling and functional analyses of candidate genes. For a 10‐megabase chromosome region on 8q24, we observed etiology‐dependent copy number gains and MYC overexpression in viral and alcohol‐related HCCs, resulting in up‐regulation of MYC target genes. Cryptogenic HCCs showed neither 8q24 gains, nor MYC overexpression, nor target gene activation, suggesting that tumors of this etiology develop by way of a distinct MYC‐independent pathomechanism. Furthermore, we detected several etiology‐independent small chromosome aberrations, including amplification of MDM4 on 1q32.1 and frequent gains of EEF1A2 on 20q13.33. Both genes were overexpressed in approximately half the HCCs examined, and gene silencing reduced cell viability as well as proliferation and increased apoptosis rates in HCC cell lines. Conclusion: Our findings suggest that MDM4 and EEF1A2 act as etiology‐independent oncogenes in a significant percentage of HCCs. (HEPATOLOGY 2008.)


Current Biology | 2012

A Proteome-wide Screen for Mammalian SxIP Motif-Containing Microtubule Plus-End Tracking Proteins

Kai Jiang; Grischa Toedt; Susana Montenegro Gouveia; Norman E. Davey; Shasha Hua; Babet van der Vaart; Ilya Grigoriev; Jesper Larsen; Lotte B. Pedersen; Karel Bezstarosti; Mariana Lince-Faria; Jeroen Demmers; Michel O. Steinmetz; Toby J. Gibson; Anna Akhmanova

Microtubule plus-end tracking proteins (+TIPs) are structurally and functionally diverse factors that accumulate at the growing microtubule plus-ends, connect them to various cellular structures, and control microtubule dynamics [1, 2]. EB1 and its homologs are +TIPs that can autonomously recognize growing microtubule ends and recruit to them a variety of other proteins. Numerous +TIPs bind to end binding (EB) proteins through natively unstructured basic and serine-rich polypeptide regions containing a core SxIP motif (serine-any amino acid-isoleucine-proline) [3]. The SxIP consensus sequence is short, and the surrounding sequences show high variability, raising the possibility that undiscovered SxIP containing +TIPs are encoded in mammalian genomes. Here, we performed a proteome-wide search for mammalian SxIP-containing +TIPs by combining biochemical and bioinformatics approaches. We have identified a set of previously uncharacterized EB partners that have the capacity to accumulate at the growing microtubule ends, including protein kinases, a small GTPase, centriole-, membrane-, and actin-associated proteins. We show that one of the newly identified +TIPs, CEP104, interacts with CP110 and CEP97 at the centriole and is required for ciliogenesis. Our study reveals the complexity of the mammalian +TIP interactome and provides a basis for investigating the molecular crosstalk between microtubule ends and other cellular structures.


Journal of Clinical Oncology | 2011

FSTL5 Is a Marker of Poor Prognosis in Non-WNT/Non-SHH Medulloblastoma

Marc Remke; Thomas Hielscher; Andrey Korshunov; Paul A. Northcott; Sebastian Bender; Marcel Kool; Frank Westermann; Axel Benner; Huriye Cin; Marina Ryzhova; Dominik Sturm; Hendrik Witt; Daniel Haag; Grischa Toedt; Andrea Wittmann; Anna Schöttler; André O. von Bueren; Andreas von Deimling; Stefan Rutkowski; Wolfram Scheurlen; Andreas E. Kulozik; Michael D. Taylor; Peter Lichter; Stefan M. Pfister

PURPOSE Integrated genomics approaches have revealed at least four distinct biologic variants of medulloblastoma: WNT (wingless), SHH (sonic hedgehog), group C, and group D. Because of the remarkable clinical heterogeneity of group D tumors and the dismal prognosis of group C patients, it is vital to identify molecular biomarkers that will allow early and effective treatment stratification in these non-WNT/non-SHH tumors. PATIENTS AND METHODS We combined transcriptome and DNA copy-number analyses for 64 primary medulloblastomas. Bioinformatic tools were used to discover marker genes of molecular variants. Differentially expressed transcripts were evaluated for prognostic value in the screening cohort. The prognostic power of follistatin-like 5 (FSTL5) immunopositivity was tested for 235 nonoverlapping medulloblastoma samples on two independent tissue microarrays. RESULTS Comprehensive analyses of transcriptomic and genetic alterations delineate four distinct variants of medulloblastoma. Stable subgroup separation was achieved by using the 300 transcripts that varied the most. Distinct expression patterns of FSTL5 in each molecular subgroup were confirmed by quantitative real-time polymerase chain reaction. Immunopositivity of FSTL5 identified a large cohort of patients (84 of 235 patients; 36%) at high risk for relapse and death. Importantly, more than 50% of non-WNT/non-SHH tumors displayed FSTL5 negativity, delineating a large patient cohort with a good prognosis who would otherwise be considered intermediate or high-risk on the basis of current molecular subgrouping. CONCLUSION FSTL5 expression denoted a dismal prognosis both within and across medulloblastoma subgroups. The addition of FSTL5 immunohistochemistry to existing molecular stratification schemes constitutes a reliable and cost-effective tool for prognostication in future clinical trials of medulloblastoma.


Genes, Chromosomes and Cancer | 2005

Detection of chromosomal imbalances in retinoblastoma by matrix-based comparative genomic hybridization

Boris Zielinski; Sandrine Gratias; Grischa Toedt; Frank Mendrzyk; Daniel E. Stange; Bernhard Radlwimmer; Dietmar R. Lohmann; Peter Lichter

The genetic hallmark of retinoblastoma is mutation or deletion of the RB1 gene, whereas other genetic alterations that are also required are largely unknown. To screen for genomic imbalances on a genomewide level, we studied a series of 17 primary retinoblastomas by matrix‐based comparative genomic hybridization (matrix‐CGH). The matrix‐CGH chip contained 6,000 immobilized genomic DNA fragments covering the human genome, with an average resolution of about 500 kb. The most frequent imbalances detected were gains on chromosome arms 1q (12 of 17), 6p (10 of 17), 2p (5 of 17), and 19q (4 of 17) and loss on 16q (7 of 17). Candidate regions could be narrowed to small intervals by the identified minimally overlapping regions on 1q22, 1q32.1q32.2, 2p24.1, and 6p21.33–p21.31. Furthermore, two as‐yet‐unknown high‐level amplifications were detected, each in a single patient, on chromosome bands 1p34.2 and 1p33. Thus, this study identified new chromosomal regions and therefore potential candidate genes that may play a role in retinoblastoma.


Neurology | 2007

DNA microarray analysis identifies candidate regions and genes in unexplained mental retardation

Hartmut Engels; Antje Brockschmidt; Alexander Hoischen; Christina Landwehr; Kristin Bosse; Constanze Walldorf; Grischa Toedt; Bernhard Radlwimmer; Peter Propping; P. Lichter; Ruthild G. Weber

Objective: Because in most patients with mental retardation (MR), who constitute 2 to 3% of the population, the etiology remains unknown, we wanted to identify novel chromosomal candidate regions and genes associated with the MR phenotype. Methods: We screened for microimbalances in 60 clinically well-characterized patients with unexplained MR mostly combined with congenital anomalies. Genome-wide array-based comparative genomic hybridization was performed on DNA microarrays with an average resolution of <0.5 Mb. We verified every nonpolymorphic array clone outside the diagnostic thresholds by fluorescence in situ hybridization and performed breakpoint analyses on confirmed imbalances. Results: Six presumably causal microimbalances were detected, five of which have not been reported. Microdeletions were found in five patients with MR and distinctive facial features, who also had neurologic findings (three cases), brain anomalies (two cases), and growth retardation (two cases), in chromosomal bands 6q11.1-q13 (10.8 Mb), Xq21.31-q21.33 (4.0 Mb), 1q24.1-q24.2 (3.8 Mb), 19p13.12 (2.1 Mb), and 4p12-p13 (1.1 Mb). One microduplication was detected in 22q11.2 (2.8 Mb) including the DiGeorge syndrome critical region in a patient with mild MR, microcephaly at birth, and dysmorphisms. Three imbalances were shown to be de novo and two inherited. The Xq21 microdeletion in a boy with borderline intellectual functioning was inherited from a normal mother; the 22q11.2 microduplication was inherited from a normal father and was present in two affected siblings. Conclusion: We could identify novel microimbalances as the probable cause of mental retardation in 10% of patients with unclear etiology. The gene content of the microimbalances was found to correlate with phenotype severity. Precise breakpoint analyses allowed the identification of deleted genes presumably causing mental retardation.


Blood | 2009

High-resolution genomic profiling of childhood T-ALL reveals frequent copy-number alterations affecting the TGF-β and PI3K-AKT pathways and deletions at 6q15-16.1 as a genomic marker for unfavorable early treatment response

Marc Remke; Stefan M. Pfister; Corinne Kox; Grischa Toedt; Natalia Becker; Axel Benner; Wiebke Werft; Stephen Breit; Shuangyou Liu; Felix Engel; Andrea Wittmann; Martin Zimmermann; Martin Stanulla; Martin Schrappe; Wolf Dieter Ludwig; Claus R. Bartram; Bernhard Radlwimmer; Martina U. Muckenthaler; Peter Lichter; Andreas E. Kulozik

Precursor T-cell acute lymphoblastic leukemia (T-ALL) in children represents a clinical challenge, because relapses are usually fatal. It is thus necessary to identify high-risk patients as early as possible to effectively individualize treatment. We aimed to define novel molecular risk markers in T-ALL and performed array-based comparative genomic hybridization (array-CGH) and expression analyses in 73 patients. We show that DNA copy-number changes are common in T-ALL and affect 70 of 73 (96%) patients. Notably, genomic imbalances predicted to down-regulate the TGF-beta or up-regulate the PI3K-AKT pathways are identified in 25 of 73 (34%) and 21 of 73 (29%) patients, suggesting that these pathways play key roles in T-ALL leukemogenesis. Furthermore, we identified a deletion at 6q15-16.1 in 9 of 73 (12%) of the patients, which predicts poor early treatment response. This deletion includes the CASP8AP2 gene, whose expression is shown to be down-regulated. The interaction of CASP8AP2 with CASP8 plays a crucial role in apoptotic regulation, suggesting a functional link between the clinical effect of the deletion and the molecular mode of action. The data presented here implicate the TGF-beta and PI3K-AKT pathways in T-ALL leukemogenesis and identify a subgroup of patients with CASP8AP2 deletions and poor early treatment response.

Collaboration


Dive into the Grischa Toedt's collaboration.

Top Co-Authors

Avatar

Peter Lichter

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Bernhard Radlwimmer

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Axel Benner

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Meinhard Hahn

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Frank Mendrzyk

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Stefan M. Pfister

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Andrey Korshunov

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Natalia Becker

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Toby J. Gibson

European Bioinformatics Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge