Network


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

Hotspot


Dive into the research topics where José Van der Heyden is active.

Publication


Featured researches published by José Van der Heyden.


European Journal of Cancer and Clinical Oncology | 1986

Recombinant tumor necrosis factor: its effect and its synergism with interferon-γ on a variety of normal and transformed human cell lines

Lucie Fransen; José Van der Heyden; Roos Ruysschaert; Walter Fiers

Tumor Necrosis Factor (TNF), released by induced macrophages, causes tumor necrosis in animals, and preferentially kills transformed cells in vitro. Using pure, recombinant human TNF, we report here its cytotoxic action on several human transformed and non-transformed cell lines. Furthermore, remarkable synergism between TNF and interferon-gamma (IFN-gamma) was observed in a large number of human cell lines, especially breast, cervix and colon carcinomas. Some other human cell lines, not sensitive to TNF alone, became highly sensitive when IFN-gamma was present as well. We could not demonstrate a synergism between TNF and IFN-gamma on any of the lymphoma/leukemia cell lines tested. All normal human, non-transformed diploid cell lines were insensitive to TNF even in the presence of IFN-gamma. This study also confirms the observation that inhibition of protein synthesis by metabolic drugs (e.g. actinomycin D) remarkably enhances the sensitivity of several target cell lines to cytolysis by TNF.


Nature Cell Biology | 2001

Design and application of a cytokine-receptor-based interaction trap.

Sven Eyckerman; Annick Verhee; José Van der Heyden; Irma Lemmens; Xaveer Van Ostade; Joël Vandekerckhove; Jan Tavernier

Ligand-induced clustering of type I cytokine receptor subunits leads to trans-phosphorylation and activation of associated cytosolic janus kinases (JAKs). In turn, JAKs phosphorylate tyrosine residues in the receptor tails, leading to recruitment and activation of signalling molecules. Among these, signal transducers and activators of transcription (STATs) are important in the direct transmission of signals to the nucleus. Here, we show that incorporation of an interaction trap in a signalling-deficient receptor allows the identification of protein–protein interactions, using a STAT-dependent complementation assay. Mammalian protein–protein interaction trap (MAPPIT) adds to existing yeast two-hybrid procedures, as originally explored by Fields and Song, and permits the detection of both modification-independent and of phosphorylation-dependent interactions in intact human cells. We also demonstrate that MAPPIT can be used to screen complex complementary DNA libraries, and using this approach, we identify cytokine-inducible SH2-containing protein (CIS) and suppressor of cytokine signalling-2 (SOCS-2) as interaction partners of the phosphotyrosine 402 (Tyr 402)-binding motif in the erythropoietin receptor (EpoR). Importantly, this approach places protein–protein interactions in their normal physiological context, and is especially applicable to the in situ analysis of signal transduction pathways.


Molecular and Cellular Biology | 2006

Inquiring into the Differential Action of Interferons (IFNs): an IFN-α2 Mutant with Enhanced Affinity to IFNAR1 Is Functionally Similar to IFN-β

Diego Jaitin; Laila C. Roisman; Eva Jaks; Martynas Gavutis; Jacob Piehler; José Van der Heyden; Gilles Uzé; Gideon Schreiber

ABSTRACT Alpha and beta interferons (IFN-α and IFN-β) are multifunctional cytokines that exhibit differential activities through a common receptor composed of the subunits IFNAR1 and IFNAR2. Here we combined biophysical and functional studies to explore the mechanism that allows the alpha and beta IFNs to act differentially. For this purpose, we have engineered an IFN-α2 triple mutant termed the HEQ mutant that mimics the biological properties of IFN-β. Compared to wild-type (wt) IFN-α2, the HEQ mutant confers a 30-fold higher binding affinity towards IFNAR1, comparable to that measured for IFN-β, resulting in a much higher stability of the ternary complex as measured on model membranes. The HEQ mutant, like IFN-β, promotes a differentially higher antiproliferative effect than antiviral activity. Both bring on a down-regulation of the IFNAR2 receptor upon induction, confirming an increased ternary complex stability of the plasma membrane. Oligonucleotide microarray experiments showed similar gene transcription profiles induced by the HEQ mutant and IFN-β and higher levels of gene induction or repression than those for wt IFN-α2. Thus, we show that the differential activities of IFN-β are directly related to the binding affinity for IFNAR1. Conservation of the residues mutated in the HEQ mutant within IFN-α subtypes suggests that IFN-α has evolved to bind IFNAR1 weakly, apparently to sustain differential levels of biological activities compared to those induced by IFN-β.


Cellular Immunology | 1986

Recombinant tumor necrosis factor: species specificity for a variety of human and murine transformed cell lines

Lucie Fransen; Marie-Rose Ruysschaert; José Van der Heyden; Walter Fiers

Tumor necrosis factor (TNF) exhibits cytotoxic or cytostatic activity on a wide range of animal and human transformed cell lines. Using pure, recombinant human and mouse TNF, we examined the degree of species specificity of the in vitro TNF activity on a variety of human and murine transformed cell lines. This species specificity was studied for the TNF activity alone or in synergism with IFN-gamma. Recombinant human and mouse TNF behave remarkably similarly regarding the in vitro cytolytic/cytostatic activity. However, a certain degree of species-specific preference could be revealed as human cell lines needed a higher concentration of recombinant mouse TNF than of recombinant human TNF to attain a similar effect, while on mouse cells the reverse was true. Also, synergism with IFN-gamma seemed more effective when the target cell was treated with homologous TNF.


Retrovirology | 2010

Definition of the interacting interfaces of Apobec3G and HIV-1 Vif using MAPPIT mutagenesis analysis.

Delphine Lavens; Frank Peelman; José Van der Heyden; Isabel Uyttendaele; Dominiek Catteeuw; Bertrand Van Schouwbroeck; Julia Kurth; Sabine Hallenberger; Reginald Clayton; Jan Tavernier

The host restriction factor Apobec3G is a cytidine deaminase that incorporates into HIV-1 virions and interferes with viral replication. The HIV-1 accessory protein Vif subverts Apobec3G by targeting it for proteasomal degradation. We propose a model in which Apobec3G N-terminal domains symmetrically interact via a head-to-head interface containing residues 122 RLYYFW 127. To validate this model and to characterize the Apobec3G–Apobec3G and the Apobec3G–Vif interactions, the mammalian protein–protein interaction trap two-hybrid technique was used. Mutations in the head-to-head interface abrogate the Apobec3G–Apobec3G interaction. All mutations that inhibit Apobec3G–Apobec3G binding also inhibit the Apobec3G–Vif interaction, indicating that the head-to head interface plays an important role in the interaction with Vif. Only the D128K, P129A and T32Q mutations specifically affect the Apobec3G–Vif association. In our model, D128, P129 and T32 cluster at the edge of the head-to-head interface, possibly forming a Vif binding site composed of two Apobec3G molecules. We propose that Vif either binds at the Apobec3G head-to-head interface or associates with an RNA-stabilized Apobec3G oligomer.


European Journal of Immunology | 2001

Neutralizing monoclonal antibodies can potentiate IL-5 signaling.

Lennart Zabeau; José Van der Heyden; Daniël Broekaert; Annick Verhee; Joël Vandekerckhove; Sheng-Jiun Wu; Irwin M. Chaiken; Peter C. Heinrich; Jan Tavernier

IL‐5 is a major determinant in the survival, differentiation and effector‐functions of eosinophils. It mediates its effect upon binding and activation of a membrane bound receptor (R), composed of a ligand‐specific α‐chain and a β‐chain, shared with the receptors for IL‐3 and granulocyte‐macrophage colony‐stimulating factor. We have generated and mapped the epitopes of three monoclonal antibodies (mAb) directed against this cytokine: the strong neutralizing mAb 5A5 and 1E1, and the very weak neutralizing mAb H30. We found that H30 as well as 5A5 can increase proliferation above the level induced by human (h)IL‐5 alone, in a JAK‐2‐dependent manner, and at every sub‐optimal hIL‐5 concentration analyzed. This effect is dependent on mAb‐mediated cross‐linking of IL‐5R complexes, and is only observed on cell lines expressing a hybrid human/mouse IL‐5Rα‐chain. We discuss these findings in view of the stoichiometric and topological requirements for an activated IL‐5R. Since humanized anti‐IL‐5 mAb are currently in clinical testing, our findings imply that such mAb should be carefully evaluated for their potentiating effects.


Science Signaling | 2002

Design and Use of a Mammalian Protein-Protein Interaction Trap (MAPPIT)

Sven Eyckerman; Irma Lemmens; Sam Lievens; José Van der Heyden; Annick Verhee; Joël Vandekerckhove; Jan Tavernier

Identifying the interaction partners of a protein is a straightforward way to gain insight into the proteins function and to position it in an interaction network such as a signal transduction pathway. Various techniques have been developed to serve this purpose, and some are specifically designed to study posttranslational modifications in mammalian proteins and to clarify their normal physiological context. However, several intrinsic constraints limit the use of these technologies, and most are not suitable for screening for new interacting partners. In the Mammalian Protein-Protein Interaction Trap (MAPPIT) Protocol described here, knowledge of cytokine receptor signaling has been used to design a versatile genetic tool that can be used analytically and for detection of new protein-protein interactions in mammalian cells.


Journal of Virological Methods | 2008

MAPPIT (MAmmalian Protein-Protein Interaction Trap) as a tool to study HIV reverse transcriptase dimerization in intact human cells

Els Pattyn; Delphine Lavens; José Van der Heyden; Annick Verhee; Sam Lievens; Irma Lemmens; Sabine Hallenberger; Dirk Jochmans; Jan Tavernier

The high mutation rate of Human Immunodeficiency Virus (HIV) leads to the rapid derivation of compound-resistant virus strains and thus necessitates the identification and development of compounds with alternative mode of actions. MAPPIT (MAmmalian Protein-Protein Interaction Trap) is a highly efficient tool to study protein-protein interactions in intact human cells and is applied to study the dimerization process of the HIV reverse transcriptase complex. Highly specific signals for the p66/p51 and p66/p66 interactions could readily be detected. Specificity was established further by introducing mutations in either subunit. Treatment with efavirenz resulted in an increased MAPPIT signal, with an EC50 value of 64nM for the p66/p51 interaction, and allowed detection of the p51/p51 homodimerization, confirming the context-dependent asymmetric contribution of both subunits. These results show that MAPPIT can be used as a novel screening tool for anti-HIV compounds in intact human cells.


Methods of Molecular Biology | 2012

ArrayMAPPIT: a screening platform for human protein interactome analysis.

Sam Lievens; Nele Vanderroost; Dieter Defever; José Van der Heyden; Jan Tavernier

Mammalian protein-protein interaction trap (MAPPIT) is a two-hybrid technology to identify and characterize interactions of proteins with other proteins or organic molecules in living mammalian (human) cells. The method relies on complementation of a modified cytokine receptor complex. Protein interaction restores the signalling competence of the complex, which is monitored through the activation of a reporter gene. Here, we describe a protocol that has been recently developed to increase the utility of MAPPIT as a tool to identify novel interactions. In the ArrayMAPPIT assay, a collection of prey proteins which is arrayed in high-density microtiter plates is efficiently screened for interaction partners using reverse transfection into a bait-expressing cell pool.


Molecular & Cellular Proteomics | 2016

Proteome-scale binary interactomics in human cells

Sam Lievens; José Van der Heyden; Delphine Masschaele; Leentje De Ceuninck; Ioanna Petta; Surya Gupta; Veronic De Puysseleyr; Virginie Vauthier; Irma Lemmens; Dries De Clercq; Dieter Defever; Nele Vanderroost; Anne-Sophie De Smet; Sven Eyckerman; Serge Van Calenbergh; Lennart Martens; Karolien De Bosscher; Claude Libert; David E. Hill; Marc Vidal; Jan Tavernier

Because proteins are the main mediators of most cellular processes they are also prime therapeutic targets. Identifying physical links among proteins and between drugs and their protein targets is essential in order to understand the mechanisms through which both proteins themselves and the molecules they are targeted with act. Thus, there is a strong need for sensitive methods that enable mapping out these biomolecular interactions. Here we present a robust and sensitive approach to screen proteome-scale collections of proteins for binding to proteins or small molecules using the well validated MAPPIT (Mammalian Protein-Protein Interaction Trap) and MASPIT (Mammalian Small Molecule-Protein Interaction Trap) assays. Using high-density reverse transfected cell microarrays, a close to proteome-wide collection of human ORF clones can be screened for interactors at high throughput. The versatility of the platform is demonstrated through several examples. With MAPPIT, we screened a 15k ORF library for binding partners of RNF41, an E3 ubiquitin protein ligase implicated in receptor sorting, identifying known and novel interacting proteins. The potential related to the fact that MAPPIT operates in living human cells is illustrated in a screen where the protein collection is scanned for interactions with the glucocorticoid receptor (GR) in its unliganded versus dexamethasone-induced activated state. Several proteins were identified the interaction of which is modulated upon ligand binding to the GR, including a number of previously reported GR interactors. Finally, the screening technology also enables detecting small molecule target proteins, which in many drug discovery programs represents an important hurdle. We show the efficiency of MASPIT-based target profiling through screening with tamoxifen, a first-line breast cancer drug, and reversine, an investigational drug with interesting dedifferentiation and antitumor activity. In both cases, cell microarray screens yielded known and new potential drug targets highlighting the utility of the technology beyond fundamental biology.

Collaboration


Dive into the José Van der Heyden's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gilles Uzé

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge