Network


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

Hotspot


Dive into the research topics where Inge Vande Walle is active.

Publication


Featured researches published by Inge Vande Walle.


Nature | 2011

A novel tumour-suppressor function for the Notch pathway in myeloid leukaemia

Apostolos Klinakis; Camille Lobry; Omar Abdel-Wahab; Philmo Oh; Hiroshi Haeno; Silvia Buonamici; Inge Vande Walle; Severine Cathelin; Thomas Trimarchi; Elisa Araldi; Cynthia Liu; Sherif Ibrahim; M. Beran; Jiri Zavadil; Argiris Efstratiadis; Tom Taghon; Franziska Michor; Ross L. Levine; Iannis Aifantis

Notch signalling is a central regulator of differentiation in a variety of organisms and tissue types. Its activity is controlled by the multi-subunit γ-secretase (γSE) complex. Although Notch signalling can play both oncogenic and tumour-suppressor roles in solid tumours, in the haematopoietic system it is exclusively oncogenic, notably in T-cell acute lymphoblastic leukaemia, a disease characterized by Notch1-activating mutations. Here we identify novel somatic-inactivating Notch pathway mutations in a fraction of patients with chronic myelomonocytic leukaemia (CMML). Inactivation of Notch signalling in mouse haematopoietic stem cells (HSCs) results in an aberrant accumulation of granulocyte/monocyte progenitors (GMPs), extramedullary haematopoieisis and the induction of CMML-like disease. Transcriptome analysis revealed that Notch signalling regulates an extensive myelomonocytic-specific gene signature, through the direct suppression of gene transcription by the Notch target Hes1. Our studies identify a novel role for Notch signalling during early haematopoietic stem cell differentiation and suggest that the Notch pathway can play both tumour-promoting and -suppressive roles within the same tissue.


Blood | 2014

ABT-199 mediated inhibition of BCL-2 as a novel therapeutic strategy in T-cell acute lymphoblastic leukemia

Sofie Peirs; Filip Matthijssens; Steven Goossens; Inge Vande Walle; Katia Ruggero; Charles E. de Bock; Sandrine Degryse; Kirsten Canté-Barrett; Delphine Briot; Emmanuelle Clappier; Tim Lammens; Barbara De Moerloose; Yves Benoit; Bruce Poppe; Jules P.P. Meijerink; Jan Cools; Jean Soulier; Terence H. Rabbitts; Tom Taghon; Franki Speleman; Pieter Van Vlierberghe

T-cell acute lymphoblastic leukemia (T-ALL) is a high-risk subtype of acute lymphoblastic leukemia (ALL) with gradually improved survival through introduction of intensified chemotherapy. However, therapy-resistant or refractory T-ALL remains a major clinical challenge. Here, we evaluated B-cell lymphoma (BCL)-2 inhibition by the BH3 mimetic ABT-199 as a new therapeutic strategy in human T-ALL. The T-ALL cell line LOUCY, which shows a transcriptional program related to immature T-ALL, exhibited high in vitro and in vivo sensitivity for ABT-199 in correspondence with high levels of BCL-2. In addition, ABT-199 showed synergistic therapeutic effects with different chemotherapeutic agents including doxorubicin, l-asparaginase, and dexamethasone. Furthermore, in vitro analysis of primary patient samples indicated that some immature, TLX3- or HOXA-positive primary T-ALLs are highly sensitive to BCL-2 inhibition, whereas TAL1 driven tumors mostly showed poor ABT-199 responses. Because BCL-2 shows high expression in early T-cell precursors and gradually decreases during normal T-cell differentiation, differences in ABT-199 sensitivity could partially be mediated by distinct stages of differentiation arrest between different molecular genetic subtypes of human T-ALL. In conclusion, our study highlights BCL-2 as an attractive molecular target in specific subtypes of human T-ALL that could be exploited by ABT-199.


Journal of Virology | 2005

Human Immunodeficiency Virus Nef Induces Rapid Internalization of the T-Cell Coreceptor CD8αβ

Veronique Stove; Inge Vande Walle; Evelien Naessens; Elisabeth Coene; Christophe P. Stove; Jean Plum; Bruno Verhasselt

ABSTRACT Human immunodeficiency virus (HIV) Nef is a membrane-associated protein decreasing surface expression of CD4, CD28, and major histocompatibility complex class I on infected cells. We report that Nef strongly down-modulates surface expression of the β-chain of the CD8αβ receptor by accelerated endocytosis, while CD8 α-chain expression is less affected. By mutational analysis of the cytoplasmic tail of the CD8 β-chain, an FMK amino acid motif was shown to be critical for Nef-induced endocytosis. Although independent of CD4, endocytosis of the CD8 β-chain was abrogated by the same mutations in Nef that affect CD4 down-regulation, suggesting common molecular interactions. The ability to down-regulate the human CD8 β-chain was conserved in HIV-1, HIV-2, and simian immunodeficiency virus SIVmac239 Nef and required an intact AP-2 complex. The Nef-mediated internalization of receptors, such as CD4, major histocompatibility complex class I, CD28, and CD8αβ, may contribute to the subversion of the host immune system and progression towards AIDS.


Blood | 2009

An early decrease in Notch activation is required for human TCR-αβ lineage differentiation at the expense of TCR-γδ T cells

Inge Vande Walle; Greet De Smet; Magda De Smedt; Bart Vandekerckhove; Georges Leclercq; Jean Plum; Tom Taghon

Although well characterized in the mouse, the role of Notch signaling in the human T-cell receptor alphabeta (TCR-alphabeta) versus TCR-gammadelta lineage decision is still unclear. Although it is clear in the mouse that TCR-gammadelta development is less Notch dependent compared with TCR-alphabeta differentiation, retroviral overexpression studies in human have suggested an opposing role for Notch during human T-cell development. Using the OP9-coculture system, we demonstrate that changes in Notch activation are differentially required during human T-cell development. High Notch activation promotes the generation of T-lineage precursors and gammadelta T cells but inhibits differentiation toward the alphabeta lineage. Reducing the amount of Notch activation rescues alphabeta-lineage differentiation, also at the single-cell level. Gene expression analysis suggests that this is mediated by differential sensitivities of Notch target genes in response to changes in Notch activation. High Notch activity increases DTX1, NRARP, and RUNX3 expression, genes that are down-regulated during alphabeta-lineage differentiation. Furthermore, increased interleukin-7 levels cannot compensate for the Notch dependent TCR-gammadelta development. Our results reveal stage-dependent molecular changes in Notch signaling that are critical for normal human T-cell development and reveal fundamental molecular differences between mouse and human.


Blood | 2009

Notch signaling is required for proliferation but not for differentiation at a well-defined β-selection checkpoint during human T-cell development

Tom Taghon; Inge Vande Walle; Greet De Smet; Magda De Smedt; Georges Leclercq; Bart Vandekerckhove; Jean Plum

Notch signaling is absolutely required for beta-selection during mouse T-cell development, both for differentiation and proliferation. In this report, we investigated whether Notch has an equally important role during human T-cell development. We show that human CD34(+) thymocytes can differentiate into CD4(+)CD8beta(+) double positive (DP) thymocytes in the absence of Notch signaling. While these DP cells phenotypically resemble human beta-selected cells, they lack a T-cell receptor (TCR)-beta chain. Therefore, we characterized the beta-selection checkpoint in human T-cell development, using CD28 as a differential marker at the immature single positive CD4(+)CD3(-)CD8alpha(-) stage. Through intracellular TCR-beta staining and gene expression analysis, we show that CD4(+)CD3(-)CD8alpha(-)CD28(+) thymocytes have passed the beta-selection checkpoint, in contrast to CD4(+)CD3(-)CD8alpha(-)CD28(-) cells. These CD4(+)CD3(-)CD8alpha(-)CD28(+) thymocytes can efficiently differentiate into CD3(+)TCRalphabeta(+) human T cells in the absence of Notch signaling. Importantly, preselection CD4(+)CD3(-)CD8alpha(-)CD28(-) thymocytes can also differentiate into CD3(+)TCRalphabeta(+) human T cells without Notch activation when provided with a rearranged TCR-beta chain. Proliferation of human thymocytes, however, is clearly Notch-dependent. Thus, we have characterized the beta-selection checkpoint during human T-cell development and show that human thymocytes require Notch signaling for proliferation but not for differentiation at this stage of development.


Blood | 2011

Jagged2 acts as a Delta-like Notch ligand during early hematopoietic cell fate decisions

Inge Vande Walle; Greet De Smet; Martina Gärtner; Magda De Smedt; Els Waegemans; Bart Vandekerckhove; Georges Leclercq; Jean Plum; Irwin D. Bernstein; Cynthia J. Guidos; Bruno Kyewski; Tom Taghon

Notch signaling critically mediates various hematopoietic lineage decisions and is induced in mammals by Notch ligands that are classified into 2 families, Delta-like (Delta-like-1, -3 and -4) and Jagged (Jagged1 and Jagged2), based on structural homology with both Drosophila ligands Delta and Serrate, respectively. Because the functional differences between mammalian Notch ligands were still unclear, we have investigated their influence on early human hematopoiesis and show that Jagged2 affects hematopoietic lineage decisions very similarly as Delta-like-1 and -4, but very different from Jagged1. OP9 coculture experiments revealed that Jagged2, like Delta-like ligands, induces T-lineage differentiation and inhibits B-cell and myeloid development. However, dose-dependent Notch activation studies, gene expression analysis, and promoter activation assays indicated that Jagged2 is a weaker Notch1-activator compared with the Delta-like ligands, revealing a Notch1 specific signal strength hierarchy for mammalian Notch ligands. Strikingly, Lunatic-Fringe- mediated glycosylation of Notch1 potentiated Notch signaling through Delta-like ligands and also Jagged2, in contrast to Jagged1. Thus, our results reveal a unique role for Jagged1 in preventing the induction of T-lineage differentiation in hematopoietic stem cells and show an unexpected functional similarity between Jagged2 and the Delta-like ligands.


Ecological Modelling | 2001

Modelling short-term CO2 fluxes and long-term tree growth in temperate forests with ASPECTS

Daniel P. Rasse; Louis François; Marc Aubinet; Andrew S Kowalski; Inge Vande Walle; Eric Laitat; Jean-Claude Gérard

The net ecosystem exchange (NEE) of CO2 between temperate forests and the atmosphere governs both carbon removal from the atmosphere and forest growth. In recent years, many experiments have been conducted to determine temperate forest NEE. These data have been used by forest modellers to better understand the processes that govern CO2 fluxes, and estimate the evolution of these fluxes under changing environmental conditions. Nevertheless, it is not clear whether models capable of handling short-term processes, which are mostly source-driven, can provide an accurate estimate of long-term forest growth, which is potentially more influenced by sink- and phenology-related processes. To analyse the interactions between short- and long-term processes, we developed the ASPECTS model, which predicts long-term forest growth by integrating, over time, hourly NEE estimates. Validation data consisting of measurements of NEE by eddy-covariance and forest carbon reservoir estimates were obtained from mixed deciduous and evergreen experimental forests located in Belgium. ASPECTS accurately estimated both: (1) the NEE fluxes for several years of data; and (2) the amount of carbon contained in stems, branches, leaves, fine and coarse roots. Our simulations demonstrated that: (1) NEE measurements in Belgian forests are compatible with forest growth over the course of the 20th century; and (2) that forest history and long-term processes need to be considered for accurate simulation of short-term CO2 fluxes.


Immunological Reviews | 2015

Epigenetics in T‐cell acute lymphoblastic leukemia

Sofie Peirs; Joni Van der Meulen; Inge Vande Walle; Tom Taghon; Franki Speleman; Bruce Poppe; Pieter Van Vlierberghe

Normal T‐cell development is a strictly regulated process in which hematopoietic progenitor cells migrate from the bone marrow to the thymus and differentiate from early T‐cell progenitors toward mature and functional T cells. During this maturation process, cooperation between a variety of oncogenes and tumor suppressors can drive immature thymocytes into uncontrolled clonal expansion and cause T‐cell acute lymphoblastic leukemia (T‐ALL). Despite improved insights in T‐ALL disease biology and comprehensive characterization of its genetic landscape, clinical care remained largely similar over the past decades and still consists of high‐dose multi‐agent chemotherapy potentially followed by hematopoietic stem cell transplantation. Even with such aggressive treatment regimens, which are often associated with considerable side effects, clinical outcome is still extremely poor in a significant subset of T‐ALL patients as a result of therapy resistance or hematological relapses. Recent genetic studies have identified recurrent somatic alterations in genes involved in DNA methylation and post‐translational histone modifications in T‐ALL, suggesting that epigenetic homeostasis is critically required in restraining tumor development in the T‐cell lineage. In this review, we provide an overview of the epigenetic regulators that could be implicated in T‐ALL disease biology and speculate how the epigenetic landscape of T‐ALL could trigger the development of epigenetic‐based therapies to further improve the treatment of human T‐ALL.


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.


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.

Collaboration


Dive into the Inge Vande Walle'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Magda De Smedt

Ghent University Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge