Network


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

Hotspot


Dive into the research topics where Ellen Geerdens is active.

Publication


Featured researches published by Ellen Geerdens.


Nature Genetics | 2013

Exome sequencing identifies mutation in CNOT3 and ribosomal genes RPL5 and RPL10 in T-cell acute lymphoblastic leukemia

Kim De Keersmaecker; Zeynep Kalender Atak; Ning Li; Carmen Vicente; Stephanie Patchett; Tiziana Girardi; Valentina Gianfelici; Ellen Geerdens; Emmanuelle Clappier; Michaël Porcu; Idoya Lahortiga; Rossella Luca; Jiekun Yan; Gert Hulselmans; Hilde Vranckx; Roel Vandepoel; Bram Sweron; Kris Jacobs; Nicole Mentens; Iwona Wlodarska; Barbara Cauwelier; Jacqueline Cloos; Jean Soulier; Anne Uyttebroeck; Claudia Bagni; Bassem A. Hassan; Peter Vandenberghe; Arlen W. Johnson; Stein Aerts; Jan Cools

T-cell acute lymphoblastic leukemia (T-ALL) is caused by the cooperation of multiple oncogenic lesions. We used exome sequencing on 67 T-ALLs to gain insight into the mutational spectrum in these leukemias. We detected protein-altering mutations in 508 genes, with an average of 8.2 mutations in pediatric and 21.0 mutations in adult T-ALL. Using stringent filtering, we predict seven new oncogenic driver genes in T-ALL. We identify CNOT3 as a tumor suppressor mutated in 7 of 89 (7.9%) adult T-ALLs, and its knockdown causes tumors in a sensitized Drosophila melanogaster model. In addition, we identify mutations affecting the ribosomal proteins RPL5 and RPL10 in 12 of 122 (9.8%) pediatric T-ALLs, with recurrent alterations of Arg98 in RPL10. Yeast and lymphoid cells expressing the RPL10 Arg98Ser mutant showed a ribosome biogenesis defect. Our data provide insights into the mutational landscape of pediatric versus adult T-ALL and identify the ribosome as a potential oncogenic factor.


PLOS Genetics | 2013

Comprehensive analysis of transcriptome variation uncovers known and novel driver events in T-cell acute lymphoblastic leukemia

Zeynep Kalender Atak; Valentina Gianfelici; Gert Hulselmans; Kim De Keersmaecker; Arun George Devasia; Ellen Geerdens; Nicole Mentens; Sabina Chiaretti; Kaat Durinck; Anne Uyttebroeck; Peter Vandenberghe; Iwona Wlodarska; Jacqueline Cloos; Robin Foà; Franki Speleman; Jan Cools; Stein Aerts

RNA-seq is a promising technology to re-sequence protein coding genes for the identification of single nucleotide variants (SNV), while simultaneously obtaining information on structural variations and gene expression perturbations. We asked whether RNA-seq is suitable for the detection of driver mutations in T-cell acute lymphoblastic leukemia (T-ALL). These leukemias are caused by a combination of gene fusions, over-expression of transcription factors and cooperative point mutations in oncogenes and tumor suppressor genes. We analyzed 31 T-ALL patient samples and 18 T-ALL cell lines by high-coverage paired-end RNA-seq. First, we optimized the detection of SNVs in RNA-seq data by comparing the results with exome re-sequencing data. We identified known driver genes with recurrent protein altering variations, as well as several new candidates including H3F3A, PTK2B, and STAT5B. Next, we determined accurate gene expression levels from the RNA-seq data through normalizations and batch effect removal, and used these to classify patients into T-ALL subtypes. Finally, we detected gene fusions, of which several can explain the over-expression of key driver genes such as TLX1, PLAG1, LMO1, or NKX2-1; and others result in novel fusion transcripts encoding activated kinases (SSBP2-FER and TPM3-JAK2) or involving MLLT10. In conclusion, we present novel analysis pipelines for variant calling, variant filtering, and expression normalization on RNA-seq data, and successfully applied these for the detection of translocations, point mutations, INDELs, exon-skipping events, and expression perturbations in T-ALL.


Blood | 2014

JAK3 mutants transform hematopoietic cells through JAK1 activation, causing T-cell acute lymphoblastic leukemia in a mouse model

Sandrine Degryse; Charles E. de Bock; Luk Cox; Sofie Demeyer; Olga Gielen; Nicole Mentens; Kris Jacobs; Ellen Geerdens; Valentina Gianfelici; Gert Hulselmans; Mark Fiers; Stein Aerts; J Meijerink; Thomas Tousseyn; Jan Cools

JAK3 is a tyrosine kinase that associates with the common γ chain of cytokine receptors and is recurrently mutated in T-cell acute lymphoblastic leukemia (T-ALL). We tested the transforming properties of JAK3 pseudokinase and kinase domain mutants using in vitro and in vivo assays. Most, but not all, JAK3 mutants transformed cytokine-dependent Ba/F3 or MOHITO cell lines to cytokine-independent proliferation. JAK3 pseudokinase mutants were dependent on Jak1 kinase activity for cellular transformation, whereas the JAK3 kinase domain mutant could transform cells in a Jak1 kinase-independent manner. Reconstitution of the IL7 receptor signaling complex in 293T cells showed that JAK3 mutants required receptor binding to mediate downstream STAT5 phosphorylation. Mice transplanted with bone marrow progenitor cells expressing JAK3 mutants developed a long-latency transplantable T-ALL-like disease, characterized by an accumulation of immature CD8(+) T cells. In vivo treatment of leukemic mice with the JAK3 selective inhibitor tofacitinib reduced the white blood cell count and caused leukemic cell apoptosis. Our data show that JAK3 mutations are drivers of T-ALL and require the cytokine receptor complex for transformation. These results warrant further investigation of JAK1/JAK3 inhibitors for the treatment of T-ALL.


Blood | 2012

Mutation of the receptor tyrosine phosphatase PTPRC (CD45) in T-cell acute lymphoblastic leukemia

Michaël Porcu; Maria Kleppe; Valentina Gianfelici; Ellen Geerdens; Kim De Keersmaecker; Marco Tartaglia; Robin Foà; Jean Soulier; Barbara Cauwelier; Anne Uyttebroeck; Elizabeth Macintyre; Peter Vandenberghe; Vahid Asnafi; Jan Cools

The protein tyrosine phosphatase CD45, encoded by the PTPRC gene, is well known as a regulator of B- and T-cell receptor signaling. In addition, CD45 negatively regulates JAK family kinases downstream of cytokine receptors. Here, we report the presence of CD45 inactivating mutations in T-cell acute lymphoblastic leukemia. Loss-of-function mutations of CD45 were detected in combination with activating mutations in IL-7R, JAK1, or LCK, and down-regulation of CD45 expression caused increased signaling downstream of these oncoproteins. Furthermore, we demonstrate that down-regulation of CD45 expression sensitizes T cells to cytokine stimulation, as observed by increased JAK/STAT signaling, whereas overexpression of CD45 decreases cytokine-induced signaling. Taken together, our data identify a tumor suppressor role for CD45 in T-cell acute lymphoblastic leukemia.


PLOS ONE | 2012

High Accuracy Mutation Detection in Leukemia on a Selected Panel of Cancer Genes

Zeynep Kalender Atak; Kim De Keersmaecker; Valentina Gianfelici; Ellen Geerdens; Roel Vandepoel; Daphnie Pauwels; Michaël Porcu; Idoya Lahortiga; Vanessa Brys; Willy G. Dirks; Hilmar Quentmeier; Jacqueline Cloos; Harry Cuppens; Anne Uyttebroeck; Peter Vandenberghe; Jan Cools; Stein Aerts

With the advent of whole-genome and whole-exome sequencing, high-quality catalogs of recurrently mutated cancer genes are becoming available for many cancer types. Increasing access to sequencing technology, including bench-top sequencers, provide the opportunity to re-sequence a limited set of cancer genes across a patient cohort with limited processing time. Here, we re-sequenced a set of cancer genes in T-cell acute lymphoblastic leukemia (T-ALL) using Nimblegen sequence capture coupled with Roche/454 technology. First, we investigated how a maximal sensitivity and specificity of mutation detection can be achieved through a benchmark study. We tested nine combinations of different mapping and variant-calling methods, varied the variant calling parameters, and compared the predicted mutations with a large independent validation set obtained by capillary re-sequencing. We found that the combination of two mapping algorithms, namely BWA-SW and SSAHA2, coupled with the variant calling algorithm Atlas-SNP2 yields the highest sensitivity (95%) and the highest specificity (93%). Next, we applied this analysis pipeline to identify mutations in a set of 58 cancer genes, in a panel of 18 T-ALL cell lines and 15 T-ALL patient samples. We confirmed mutations in known T-ALL drivers, including PHF6, NF1, FBXW7, NOTCH1, KRAS, NRAS, PIK3CA, and PTEN. Interestingly, we also found mutations in several cancer genes that had not been linked to T-ALL before, including JAK3. Finally, we re-sequenced a small set of 39 candidate genes and identified recurrent mutations in TET1, SPRY3 and SPRY4. In conclusion, we established an optimized analysis pipeline for Roche/454 data that can be applied to accurately detect gene mutations in cancer, which led to the identification of several new candidate T-ALL driver mutations.


International Journal of Cancer | 2014

Identification of a novel, recurrent MBTD1-CXorf67 fusion in low-grade endometrial stromal sarcoma.

Barbara Dewaele; Joanna Przybyl; Anna Quattrone; Julio Finalet Ferreiro; Vanessa Vanspauwen; Ellen Geerdens; Valentina Gianfelici; Zeynep Kalender; Agnieszka Wozniak; Philippe Moerman; Raf Sciot; Sabrina Croce; Frédéric Amant; Peter Vandenberghe; Jan Cools; Maria Debiec-Rychter

Endometrial stromal sarcomas (ESSs) are a genetically heterogeneous group of rare uterine neoplasms that are commonly driven by recurrent gene rearrangements. In conventional low‐grade ESS, JAZF1‐SUZ12, PHF1‐JAZF1, EPC1‐PHF1 and MEAF6‐PHF1, and recently described ZC3H7‐BCOR chimeric fusions have been reported in > 50% of cases. Conversely, oncogenic t(10;17)(q22;p13) translocation yields YWHAE‐FAM22A/B chimeric proteins that are associated with histologically high‐grade and clinically more aggressive ESS. Integrating whole‐transcriptome paired‐end RNA sequencing with fluorescence in situ hybridization (FISH) and banding cytogenetics, we identified MBTD1 (malignant brain tumor domain‐containing 1) and CXorf67 (chromosome X open reading frame 67) as the genes involved in the novel reciprocal t(X;17)(p11.2;q21.33) translocation in two independent low‐grade ESS of classical histology. The presence of the MBTD1‐CXorf67 fusion transcript was validated in both cases using reverse‐transcription polymerase chain reaction followed by Sanger sequencing. A specific FISH assay was developed to detect the novel t(X;17) translocation in formalin‐fixed paraffin‐embedded material, and resulted in identification of an additional low‐grade ESS case positive for the MBTD1‐CXorf67 fusion among 25 uterine stromal tumors [14 ESS and 11 undifferentiated endometrial sarcomas (UESs)] that were negative for JAZF1 and YWHAE rearrangements. Gene expression profiles of seven ESS (including three with YWHAE and two with JAZF1 rearrangements) and four UES without specific chromosomal aberrations indicated clustering of tumors with MBTD1‐CXorf67 fusion together with low‐grade JAZF1‐associated ESS. The chimeric MBTD1‐CXorf67 fusion identifies yet another cytogenetically distinct subgroup of low‐grade ESS and offers the opportunity to shed light on the functions of two poorly characterized genes.


Haematologica | 2015

Targeted sequencing identifies associations between IL7R-JAK mutations and epigenetic modulators in T-cell acute lymphoblastic leukemia

Carmen Vicente; Claire Schwab; Michaël Broux; Ellen Geerdens; Sandrine Degryse; Sofie Demeyer; Idoya Lahortiga; Alannah Elliott; Lucy Chilton; Roberta La Starza; Cristina Mecucci; Peter Vandenberghe; Nicholas Goulden; Ajay Vora; Anthony V. Moorman; Jean Soulier; Christine J. Harrison; Emmanuelle Clappier; Jan Cools

T-cell acute lymphoblastic leukemia is caused by the accumulation of multiple oncogenic lesions, including chromosomal rearrangements and mutations. To determine the frequency and co-occurrence of mutations in T-cell acute lymphoblastic leukemia, we performed targeted re-sequencing of 115 genes across 155 diagnostic samples (44 adult and 111 childhood cases). NOTCH1 and CDKN2A/B were mutated/deleted in more than half of the cases, while an additional 37 genes were mutated/deleted in 4% to 20% of cases. We found that IL7R-JAK pathway genes were mutated in 27.7% of cases, with JAK3 mutations being the most frequent event in this group. Copy number variations were also detected, including deletions of CREBBP or CTCF and duplication of MYB. FLT3 mutations were rare, but a novel extracellular mutation in FLT3 was detected and confirmed to be transforming. Furthermore, we identified complex patterns of pairwise associations, including a significant association between mutations in IL7R-JAK genes and epigenetic regulators (WT1, PRC2, PHF6). Our analyses showed that IL7R-JAK genetic lesions did not confer adverse prognosis in T-cell acute lymphoblastic leukemia cases enrolled in the UK ALL2003 trial. Overall, these results identify interconnections between the T-cell acute lymphoblastic leukemia genome and disease biology, and suggest a potential clinical application for JAK inhibitors in a significant proportion of patients with T-cell acute lymphoblastic leukemia.


Haematologica | 2014

The Notch driven long non-coding RNA repertoire in T-cell acute lymphoblastic leukemia

Kaat Durinck; Annelynn Wallaert; Inge Vande Walle; Wouter Van Loocke; Pieter-Jan Volders; Suzanne Vanhauwaert; Ellen Geerdens; Yves Benoit; Nadine Van Roy; Bruce Poppe; Jean Soulier; Jan Cools; Pieter Mestdagh; Jo Vandesompele; Pieter Rondou; Pieter Van Vlierberghe; Tom Taghon; Franki Speleman

Genetic studies in T-cell acute lymphoblastic leukemia have uncovered a remarkable complexity of oncogenic and loss-of-function mutations. Amongst this plethora of genetic changes, NOTCH1 activating mutations stand out as the most frequently occurring genetic defect, identified in more than 50% of T-cell acute lymphoblastic leukemias, supporting a role as an essential driver for this gene in T-cell acute lymphoblastic leukemia oncogenesis. In this study, we aimed to establish a comprehensive compendium of the long non-coding RNA transcriptome under control of Notch signaling. For this purpose, we measured the transcriptional response of all protein coding genes and long non-coding RNAs upon pharmacological Notch inhibition in the human T-cell acute lymphoblastic leukemia cell line CUTLL1 using RNA-sequencing. Similar Notch dependent profiles were established for normal human CD34+ thymic T-cell progenitors exposed to Notch signaling activity in vivo. In addition, we generated long non-coding RNA expression profiles (array data) from ex vivo isolated Notch active CD34+ and Notch inactive CD4+CD8+ thymocytes and from a primary cohort of 15 T-cell acute lymphoblastic leukemia patients with known NOTCH1 mutation status. Integration of these expression datasets with publicly available Notch1 ChIP-sequencing data resulted in the identification of long non-coding RNAs directly regulated by Notch activity in normal and malignant T cells. Given the central role of Notch in T-cell acute lymphoblastic leukemia oncogenesis, these data pave the way for the development of novel therapeutic strategies that target hyperactive Notch signaling in human T-cell acute lymphoblastic leukemia.


Haematologica | 2015

Hedgehog pathway mutations in T-cell acute lymphoblastic leukemia.

Antonis Dagklis; Daphnie Pauwels; Idoya Lahortiga; Ellen Geerdens; Emilie Bittoun; Barbara Cauwelier; Thomas Tousseyn; Anne Uyttebroeck; Johan Maertens; Gregor Verhoef; Peter Vandenberghe; Jan Cools

Hedgehog signaling plays an important role in both embryonic pattern formation and in proliferation, survival and differentiation of adult tissues, including T cells.[1][1],[2][2] We report here the presence of rare acquired mutations in genes encoding components of the Hedgehog signaling pathway in


Leukemia | 2017

RPL5 on 1p22.1 is recurrently deleted in multiple myeloma and its expression is linked to bortezomib response

Isabel J.F. Hofman; M van Duin; E De Bruyne; Laura Fancello; George Mulligan; Ellen Geerdens; Emanuela Garelli; Cecilia Mancini; Heidi Lemmens; Michel Delforge; Peter Vandenberghe; I. Wlodarska; Anna Aspesi; Lucienne Michaux; Karin Vanderkerken; Pieter Sonneveld; K De Keersmaecker

Chromosomal region 1p22 is deleted in ⩾20% of multiple myeloma (MM) patients, suggesting the presence of an unidentified tumor suppressor. Using high-resolution genomic profiling, we delimit a 58 kb minimal deleted region (MDR) on 1p22.1 encompassing two genes: ectopic viral integration site 5 (EVI5) and ribosomal protein L5 (RPL5). Low mRNA expression of EVI5 and RPL5 was associated with worse survival in diagnostic cases. Patients with 1p22 deletion had lower mRNA expression of EVI5 and RPL5, however, 1p22 deletion status is a bad predictor of RPL5 expression in some cases, suggesting that other mechanisms downregulate RPL5 expression. Interestingly, RPL5 but not EVI5 mRNA levels were significantly lower in relapsed patients responding to bortezomib and; both in newly diagnosed and relapsed patients, bortezomib treatment could overcome their bad prognosis by raising their progression-free survival to equal that of patients with high RPL5 expression. In conclusion, our genetic data restrict the MDR on 1p22 to EVI5 and RPL5 and although the role of these genes in promoting MM progression remains to be determined, we identify RPL5 mRNA expression as a biomarker for initial response to bortezomib in relapsed patients and subsequent survival benefit after long-term treatment in newly diagnosed and relapsed patients.

Collaboration


Dive into the Ellen Geerdens's collaboration.

Top Co-Authors

Avatar

Jan Cools

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Peter Vandenberghe

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Anne Uyttebroeck

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Stein Aerts

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Sofie Demeyer

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Zeynep Kalender Atak

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carmen Vicente

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Gert Hulselmans

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Iwona Wlodarska

Université catholique de Louvain

View shared research outputs
Researchain Logo
Decentralizing Knowledge