Network


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

Hotspot


Dive into the research topics where Kaat Durinck is active.

Publication


Featured researches published by Kaat Durinck.


Nature Methods | 2013

The need for transparency and good practices in the qPCR literature

Stephen A. Bustin; Vladimir Benes; Jeremy A. Garson; Jan Hellemans; Jim F. Huggett; Mikael Kubista; Reinhold Mueller; Tania Nolan; Michael W. Pfaffl; Gregory L. Shipley; Carl T. Wittwer; Peter Schjerling; Philip J. R. Day; Mónica Abreu; Begoña Aguado; Jean-François Beaulieu; Anneleen Beckers; Sara Bogaert; John A. Browne; Fernando Carrasco-Ramiro; Liesbeth Ceelen; Kate L. Ciborowski; Pieter Cornillie; Stephanie Coulon; Ann Cuypers; Sara De Brouwer; Leentje De Ceuninck; Jurgen De Craene; Hélène De Naeyer; Ward De Spiegelaere

Two surveys of over 1,700 publications whose authors use quantitative real-time PCR (qPCR) reveal a lack of transparent and comprehensive reporting of essential technical information. Reporting standards are significantly improved in publications that cite the Minimum Information for Publication of Quantitative Real-Time PCR Experiments (MIQE) guidelines, although such publications are still vastly outnumbered by those that do not.


Blood | 2015

The H3K27me3 demethylase UTX is a gender-specific tumor suppressor in T-cell acute lymphoblastic leukemia

Joni Van der Meulen; Viraj Sanghvi; Konstantinos Mavrakis; Kaat Durinck; Fang Fang; Filip Matthijssens; Pieter Rondou; Monica Rosen; Tim Pieters; Peter Vandenberghe; Eric Delabesse; Tim Lammens; Barbara De Moerloose; Björn Menten; Nadine Van Roy; Bruno Verhasselt; Bruce Poppe; Yves Benoit; Tom Taghon; Ari Melnick; Franki Speleman; Hans-Guido Wendel; Pieter Van Vlierberghe

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive form of leukemia that is mainly diagnosed in children and shows a skewed gender distribution toward males. In this study, we report somatic loss-of-function mutations in the X-linked histone H3K27me3 demethylase ubiquitously transcribed X (UTX) chromosome, in human T-ALL. Interestingly, UTX mutations were exclusively present in male T-ALL patients and allelic expression analysis revealed that UTX escapes X-inactivation in female T-ALL lymphoblasts and normal T cells. Notably, we demonstrate in vitro and in vivo that the H3K27me3 demethylase UTX functions as a bona fide tumor suppressor in T-ALL. Moreover, T-ALL driven by UTX inactivation exhibits collateral sensitivity to pharmacologic H3K27me3 inhibition. All together, our results show how a gender-specific and therapeutically relevant defect in balancing H3K27 methylation contributes to T-cell leukemogenesis.


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.


Experimental Hematology | 2015

Novel biological insights in T-cell acute lymphoblastic leukemia

Kaat Durinck; Steven Goossens; Sofie Peirs; Annelynn Wallaert; Wouter Van Loocke; Filip Matthijssens; Tim Pieters; Tim Lammens; Pieter Rondou; Nadine Van Roy; Barbara De Moerloose; Yves Benoit; Jody J. Haigh; Frank Speleman; Bruce Poppe; Pieter Van Vlierberghe

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive type of blood cancer that accounts for about 15% of pediatric and 25% of adult acute lymphoblastic leukemia (ALL) cases. It is considered as a paradigm for the multistep nature of cancer initiation and progression. Genetic and epigenetic reprogramming events, which transform T-cell precursors into malignant T-ALL lymphoblasts, have been extensively characterized over the past decade. Despite our comprehensive understanding of the genomic landscape of human T-ALL, leukemia patients are still treated by high-dose multiagent chemotherapy, potentially followed by hematopoietic stem cell transplantation. Even with such aggressive treatment regimens, which are often associated with considerable acute and long-term side effects, about 15% of pediatric and 40% of adult T-ALL patients still relapse, owing to acquired therapy resistance, and present with very dismal survival perspectives. Unfortunately, the molecular mechanisms by which residual T-ALL tumor cells survive chemotherapy and act as a reservoir for leukemic progression and hematologic relapse remain poorly understood. Nevertheless, it is expected that enhanced molecular understanding of T-ALL disease biology will ultimately facilitate a targeted therapy driven approach that can reduce chemotherapy-associated toxicities and improve survival of refractory T-ALL patients through personalized salvage therapy. In this review, we summarize recent biological insights into the molecular pathogenesis of T-ALL and speculate how the genetic landscape of T-ALL could trigger the development of novel therapeutic strategies for the treatment of human T-ALL.


Nature Communications | 2015

ZEB2 drives immature T-cell lymphoblastic leukaemia development via enhanced tumour-initiating potential and IL-7 receptor signalling

Steven Goossens; Enrico Radaelli; Odile Blanchet; Kaat Durinck; Joni Van der Meulen; Sofie Peirs; Tom Taghon; Cedric Tremblay; Magdaline Costa; Morvarid Farhang Ghahremani; Jelle De Medts; Sonia Bartunkova; Katharina Haigh; Claire Schwab; Natalie Farla; Tim Pieters; Filip Matthijssens; Nadine Van Roy; J. Adam Best; Kim Deswarte; Pieter Bogaert; Catherine L. Carmichael; Adam Samuel Rickard; Santi Suryani; Lauryn S. Bracken; Raed Alserihi; Kirsten Canté-Barrett; Lieven Haenebalcke; Emmanuelle Clappier; Pieter Rondou

Early T-cell precursor leukaemia (ETP-ALL) is a high-risk subtype of human leukaemia that is poorly understood at the molecular level. Here we report translocations targeting the zinc finger E-box-binding transcription factor ZEB2 as a recurrent genetic lesion in immature/ETP-ALL. Using a conditional gain-of-function mouse model, we demonstrate that sustained Zeb2 expression initiates T-cell leukaemia. Moreover, Zeb2-driven mouse leukaemia exhibit some features of the human immature/ETP-ALL gene expression signature, as well as an enhanced leukaemia-initiation potential and activated Janus kinase (JAK)/signal transducers and activators of transcription (STAT) signalling through transcriptional activation of IL7R. This study reveals ZEB2 as an oncogene in the biology of immature/ETP-ALL and paves the way towards pre-clinical studies of novel compounds for the treatment of this aggressive subtype of human T-ALL using our Zeb2-driven mouse model.


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.


Leukemia | 2013

Novel TAL1 targets beyond protein-coding genes: identification of TAL1-regulated microRNAs in T-cell acute lymphoblastic leukemia

Nádia C. Correia; Kaat Durinck; Ana Paula Leite; Maté Ongenaert; Pieter Rondou; Franki Speleman; Francisco J. Enguita; João T. Barata

Novel TAL1 targets beyond protein-coding genes: identification of TAL1-regulated microRNAs in T-cell acute lymphoblastic leukemia


Nature Communications | 2016

GATA3 induces human T-cell commitment by restraining Notch activity and repressing NK-cell fate

Inge Vande Walle; Anne-Catherine Dolens; Kaat Durinck; Katrien De Mulder; Wouter Van Loocke; Sagar S. Damle; Els Waegemans; Jelle De Medts; Imke Velghe; Magda De Smedt; Bart Vandekerckhove; Tessa Kerre; Jean Plum; Georges Leclercq; Ellen V. Rothenberg; Pieter Van Vlierberghe; Frank Speleman; Tom Taghon

The gradual reprogramming of haematopoietic precursors into the T-cell fate is characterized by at least two sequential developmental stages. Following Notch1-dependent T-cell lineage specification during which the first T-cell lineage genes are expressed and myeloid and dendritic cell potential is lost, T-cell specific transcription factors subsequently induce T-cell commitment by repressing residual natural killer (NK)-cell potential. How these processes are regulated in human is poorly understood, especially since efficient T-cell lineage commitment requires a reduction in Notch signalling activity following T-cell specification. Here, we show that GATA3, in contrast to TCF1, controls human T-cell lineage commitment through direct regulation of three distinct processes: repression of NK-cell fate, upregulation of T-cell lineage genes to promote further differentiation and restraint of Notch activity. Repression of the Notch1 target gene DTX1 hereby is essential to prevent NK-cell differentiation. Thus, GATA3-mediated positive and negative feedback mechanisms control human T-cell lineage commitment.


Leukemia | 2016

Long noncoding RNA signatures define oncogenic subtypes in T-cell acute lymphoblastic leukemia

Annelynn Wallaert; Kaat Durinck; W Van Loocke; I Van de Walle; Filip Matthijssens; Pieter-Jan Volders; F Avila Cobos; Dries Rombaut; Pieter Rondou; Pieter Mestdagh; Jo Vandesompele; Bruce Poppe; Tom Taghon; Jean Soulier; P Van Vlierberghe; F. Speleman

Long noncoding RNA signatures define oncogenic subtypes in T-cell acute lymphoblastic leukemia


Leukemia | 2015

Characterization of the genome-wide TLX1 binding profile in T-cell acute lymphoblastic leukemia

Kaat Durinck; W Van Loocke; J Van der Meulen; I Van de Walle; Maté Ongenaert; Pieter Rondou; Annelynn Wallaert; C E de Bock; N. Van Roy; Bruce Poppe; Jan Cools; Jean Soulier; Tom Taghon; F. Speleman; P Van Vlierberghe

The TLX1 transcription factor is critically involved in the multi-step pathogenesis of T-cell acute lymphoblastic leukemia (T-ALL) and often cooperates with NOTCH1 activation during malignant T-cell transformation. However, the exact molecular mechanism by which these T-cell specific oncogenes cooperate during transformation remains to be established. Here, we used chromatin immunoprecipitation followed by sequencing to establish the genome-wide binding pattern of TLX1 in human T-ALL. This integrative genomics approach showed that ectopic TLX1 expression drives repression of T cell-specific enhancers and mediates an unexpected transcriptional antagonism with NOTCH1 at critical target genes, including IL7R and NOTCH3. These phenomena coordinately trigger a TLX1-driven pre-leukemic phenotype in human thymic precursor cells, reminiscent of the thymus regression observed in murine TLX1 tumor models, and create a strong genetic pressure for acquiring activating NOTCH1 mutations as a prerequisite for full leukemic transformation. In conclusion, our results uncover a functional antagonism between cooperative oncogenes during the earliest phases of tumor development and provide novel insights in the multi-step pathogenesis of TLX1-driven human leukemia.

Collaboration


Dive into the Kaat Durinck's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bruce Poppe

Ghent University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yves Benoit

Ghent University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge