Network


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

Hotspot


Dive into the research topics where Arjan Buist is active.

Publication


Featured researches published by Arjan Buist.


The Journal of Neuroscience | 2006

Vesicular Glutamate Transporter VGLUT2 Expression Levels Control Quantal Size and Neuropathic Pain

Diederik Moechars; Matthew C. Weston; Sandra Leo; Zsuzsanna Callaerts-Vegh; Ilse Goris; Guy Daneels; Arjan Buist; Miroslav Cik; P. van der Spek; Stefan U. Kass; Theo Meert; Rudi D'Hooge; Christian Rosenmund; R. Mark Hampson

Uptake of l-glutamate into synaptic vesicles is mediated by vesicular glutamate transporters (VGLUTs). Three transporters (VGLUT1–VGLUT3) are expressed in the mammalian CNS, with partial overlapping expression patterns, and VGLUT2 is the most abundantly expressed paralog in the thalamus, midbrain, and brainstem. Previous studies have shown that VGLUT1 is necessary for glutamatergic transmission in the hippocampus, but the role of VGLUT2 in excitatory transmission is unexplored in glutamatergic neurons and in vivo. We examined the electrophysiological and behavioral consequences of loss of either one or both alleles of VGLUT2. We show that targeted deletion of VGLUT2 in mice causes perinatal lethality and a 95% reduction in evoked glutamatergic responses in thalamic neurons, although hippocampal synapses function normally. Behavioral analysis of heterozygous VGLUT2 mice showed unchanged motor function, learning and memory, acute nociception, and inflammatory pain, but acquisition of neuropathic pain, maintenance of conditioned taste aversion, and defensive marble burying were all impaired. Reduction or loss of VGLUT2 in heterozygous and homozygous VGLUT2 knock-outs led to a graded reduction in the amplitude of the postsynaptic response to single-vesicle fusion in thalamic neurons, indicating that the vesicular VGLUT content is critically important for quantal size and demonstrating that VGLUT2-mediated reduction of excitatory drive affects specific forms of sensory processing.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Characterization of an orphan G protein-coupled receptor localized in the dorsal root ganglia reveals adenine as a signaling molecule

Eckhard Bender; Arjan Buist; Mirek Jurzak; Xavier Langlois; Geert Baggerman; Peter Verhasselt; Martine Ercken; Hong-Qing Guo; Cindy Wintmolders; Ilse Van den Wyngaert; Irma Van Oers; Liliane Schoofs; Walter Luyten

The cloning of novel G protein-coupled receptors and the search for their natural ligands, a process called reverse pharmacology, is an excellent opportunity to discover novel hormones and neurotransmitters. Based on a degenerate primer approach we have cloned a G protein-coupled receptor whose mRNA expression profile indicates highest expression in the dorsal root ganglia, specifically in the subset of small neurons, suggesting a role in nociception. In addition, moderate expression was found in lung, hypothalamus, peripheral blood leukocytes, and ovaries. Guided by a receptor-activation bioassay, we identified adenine as the endogenous ligand, which activated the receptor potently and with high structural stringency. Therefore, we propose to name this receptor as the adenine receptor. Hormonal functions have already been demonstrated for adenine derivatives like 6-benzylaminopurine in plants and 1-methyladenine in lower animals. Here, we demonstrate that adenine functions as a signaling molecule in mammals. This finding adds a third family besides P1 and P2 receptors to the class of purinergic receptors.


Journal of Biological Chemistry | 1997

Comparative Kinetic Analysis and Substrate Specificity of the Tandem Catalytic Domains of the Receptor-like Protein-tyrosine Phosphatase α

Li Wu; Arjan Buist; Jeroen den Hertog; Zhong Yin Zhang

The catalytic activity and substrate specificity of protein-tyrosine phosphatase α (PTPα) is primarily controlled by the membrane proximal catalytic domain (D1). The membrane distal (D2) domain of PTPα by itself is a genuine PTPase, possessing catalytic activity comparable to that of D1 using aryl phosphates as substrates. Surprisingly, kcat and kcat/Km for the D2-catalyzed hydrolysis of phosphotyrosine-containing peptides are several orders of magnitude reduced in comparison with those of D1. Substitution of the putative general acid/base Glu-690 in D2 by an Asp, which is invariably found in the WPD motifs in all cytoplasmic PTPases and all the D1 domains of receptor-like PTPases, only increases the kcat for D2 by 4-fold. Thus the much reduced D2 activity toward peptide substrates may be due to structural differences in the active sites other than the general acid/base. Alternatively, the D2 domain may have a functional active site with a highly stringent substrate specificity. PTPα display modest peptide substrate selectivity and are sensitive to charges adjacent to phosphotyrosine. In the sequence context of DADEpYLIPQQG (where pY stands for phosphotyrosine), the minimal sizes recognized by PTPα are either ADEpYLI or DADEpY-NH2.


Acta Neuropathologica | 2016

Heterotypic seeding of Tau fibrillization by pre-aggregated Abeta provides potent seeds for prion-like seeding and propagation of Tau-pathology in vivo.

Bruno Barbosa de Vasconcelos; Ilie-Cosmin Stancu; Arjan Buist; Peng Wang; Alexandre Vanoosthuyse; Kristof Van Kolen; An Verheyen; Pascal Kienlen-Campard; Jean-Noël Octave; Peter Baatsen; Diederik Moechars; Ilse Dewachter

Genetic, clinical, histopathological and biomarker data strongly support Beta-amyloid (Aβ) induced spreading of Tau-pathology beyond entorhinal cortex (EC), as a crucial process in conversion from preclinical cognitively normal to Alzheimer‘s Disease (AD), while the underlying mechanism remains unclear. In vivo preclinical models have reproducibly recapitulated Aβ-induced Tau-pathology. Tau pathology was thereby also induced by aggregated Aβ, in functionally connected brain areas, reminiscent of a prion-like seeding process. In this work we demonstrate, that pre-aggregated Aβ can directly induce Tau fibrillization by cross-seeding, in a cell-free assay, comparable to that demonstrated before for alpha-synuclein and Tau. We furthermore demonstrate, in a well-characterized cellular Tau-aggregation assay that Aβ-seeds cross-seeded Tau-pathology and strongly catalyzed pre-existing Tau-aggregation, reminiscent of the pathogenetic process in AD. Finally, we demonstrate that heterotypic seeded Tau by pre-aggregated Aβ provides efficient seeds for induction and propagation of Tau-pathology in vivo. Prion-like, heterotypic seeding of Tau fibrillization by Aβ, providing potent seeds for propagating Tau pathology in vivo, as demonstrated here, provides a compelling molecular mechanism for Aβ-induced propagation of Tau-pathology, beyond regions with pre-existing Tau-pathology (entorhinal cortex/locus coeruleus). Cross-seeding along functional connections could thereby resolve the initial spatial dissociation between amyloid- and Tau-pathology, and preferential propagation of Tau-pathology in regions with pre-existing ‘silent’ Tau-pathology, by conversion of a ‘silent’ Tau pathology to a ‘spreading’ Tau-pathology, observed in AD.


Journal of Biological Chemistry | 2000

Identification of p130cas as an in vivo substrate of receptor protein-tyrosine phosphatase alpha.

Arjan Buist; Christophe Blanchetot; Leon G.J. Tertoolen; Jeroen den Hertog

We have employed a substrate trapping strategy to identify physiological substrates of the receptor protein-tyrosine phosphatase α (RPTPα). Here we report that a substrate-trapping mutant of the RPTPα membrane proximal catalytic domain (D1), RPTPα-D1-C433S, specifically bound to tyrosine-phosphorylated proteins from pervanadate-treated cells. The membrane distal catalytic domain of RPTPα (D2) and mutants thereof did not bind to tyrosine-phosphorylated proteins. The pattern of tyrosine-phosphorylated proteins that bound to RPTPα-D1-C433S varied between cell lines, but a protein of approximately 130 kDa was pulled down from every cell line. This protein was identified as p130 cas . Tyrosine-phosphorylated p130 cas from fibronectin-stimulated NIH3T3 cells bound to RPTPα-D1-C433S as well, suggesting that p130 cas is a physiological substrate of RPTPα. RPTPα dephosphorylated p130 cas in vitro, and RPTPα co-localized with a subpopulation of p130 cas to the plasma membrane. Co-transfection experiments with activated SrcY529F, p130 cas , and RPTPα or inactive, mutant RPTPα indicated that RPTPα dephosphorylated p130 cas in vivo. Tyrosine-phosphorylated epidermal growth factor receptor was not dephosphorylated by RPTPα under these conditions, suggesting that p130 cas is a specific substrate of RPTPα in living cells. In conclusion, our results provide evidence that p130 cas is a physiological substrate of RPTPα in vivo.


Journal of Biomolecular Screening | 2016

Development of a Scalable, High-Throughput-Compatible Assay to Detect Tau Aggregates Using iPSC-Derived Cortical Neurons Maintained in a Three-Dimensional Culture Format.

X. Medda; Liesbeth Mertens; Sofie Versweyveld; Annick Diels; L. Barnham; Alexis Bretteville; Arjan Buist; An Verheyen; Ines Royaux; Andreas Ebneth; Alfredo Cabrera-Socorro

Tau aggregation is the pathological hallmark that best correlates with the progression of Alzheimer’s disease (AD). The presence of neurofibrillary tangles (NFTs), formed of hyperphosphorylated tau, leads to neuronal dysfunction and loss, and is directly associated with the cognitive decline observed in AD patients. The limited success in targeting β-amyloid pathologies has reinforced the hypothesis of blocking tau phosphorylation, aggregation, and/or spreading as alternative therapeutic entry points to treat AD. Identification of novel therapies requires disease-relevant and scalable assays capable of reproducing key features of the pathology in an in vitro setting. Here we use induced pluripotent stem cells (iPSCs) as a virtually unlimited source of human cortical neurons to develop a robust and scalable tau aggregation model compatible with high-throughput screening (HTS). We downscaled cell culture conditions to 384-well plate format and used Matrigel to introduce an extra physical protection against cell detachment that reduces shearing stress and better recapitulates pathological conditions. We complemented the assay with AlphaLISA technology for the detection of tau aggregates in a high-throughput-compatible format. The assay is reproducible across users and works with different commercially available iPSC lines, representing a highly translational tool for the identification of novel treatments against tauopathies, including AD.


Journal of Biological Chemistry | 2016

The Dynamics and Turnover of Tau Aggregates in Cultured Cells INSIGHTS INTO THERAPIES FOR TAUOPATHIES

Jing L. Guo; Arjan Buist; Alberto Soares; Kathleen Callaerts; Sara Calafate; Frederik Stevenaert; Joshua P. Daniels; Bryan E. Zoll; Alex Crowe; Kurt R. Brunden; Diederik Moechars; Virginia M.-Y. Lee

Filamentous tau aggregates, the hallmark lesions of Alzheimer disease (AD), play key roles in neurodegeneration. Activation of protein degradation systems has been proposed to be a potential strategy for removing pathological tau, but it remains unclear how effectively tau aggregates can be degraded by these systems. By applying our previously established cellular model system of AD-like tau aggregate induction using preformed tau fibrils, we demonstrate that tau aggregates induced in cells with regulated expression of full-length mutant tau can be gradually cleared when soluble tau expression is suppressed. This clearance is at least partially mediated by the autophagy-lysosome pathway, although both the ubiquitin-proteasome system and the autophagy-lysosome pathway are deficient in handling large tau aggregates. Importantly, residual tau aggregates left after the clearance phase leads to a rapid reinstatement of robust tau pathology once soluble tau expression is turned on again. Moreover, we succeeded in generating monoclonal cells persistently carrying tau aggregates without obvious cytotoxicity. Live imaging of GFP-tagged tau aggregates showed that tau inclusions are dynamic structures constantly undergoing “fission” and “fusion,” which facilitate stable propagation of tau pathology in dividing cells. These findings provide a greater understanding of cell-to-cell transmission of tau aggregates in dividing cells and possibly neurons.


PLOS ONE | 2015

Using Human iPSC-Derived Neurons to Model TAU Aggregation

An Verheyen; Annick Diels; Joyce Dijkmans; Tutu Oyelami; Giulia Meneghello; Liesbeth Mertens; Sofie Versweyveld; M. Borgers; Arjan Buist; Pieter J. Peeters; Miroslav Cik

Alzheimer’s disease and frontotemporal dementia are amongst the most common forms of dementia characterized by the formation and deposition of abnormal TAU in the brain. In order to develop a translational human TAU aggregation model suitable for screening, we transduced TAU harboring the pro-aggregating P301L mutation into control hiPSC-derived neural progenitor cells followed by differentiation into cortical neurons. TAU aggregation and phosphorylation was quantified using AlphaLISA technology. Although no spontaneous aggregation was observed upon expressing TAU-P301L in neurons, seeding with preformed aggregates consisting of the TAU-microtubule binding repeat domain triggered robust TAU aggregation and hyperphosphorylation already after 2 weeks, without affecting general cell health. To validate our model, activity of two autophagy inducers was tested. Both rapamycin and trehalose significantly reduced TAU aggregation levels suggesting that iPSC-derived neurons allow for the generation of a biologically relevant human Tauopathy model, highly suitable to screen for compounds that modulate TAU aggregation.


Stem cell reports | 2018

Genetically Engineered iPSC-Derived FTDP-17 MAPT Neurons Display Mutation-Specific Neurodegenerative and Neurodevelopmental Phenotypes

An Verheyen; Annick Diels; Joke Reumers; Kirsten Van Hoorde; Ilse Van den Wyngaert; Constantin van Outryve d’Ydewalle; An De Bondt; Jacobine Kuijlaars; Louis De Muynck; Ronald de Hoogt; Alexis Bretteville; Steffen Jaensch; Arjan Buist; Alfredo Cabrera-Socorro; Selina Wray; Andreas Ebneth; Peter Walter Maria Roevens; Ines Royaux; Pieter J. Peeters

Summary Tauopathies such as frontotemporal dementia (FTD) remain incurable to date, partially due to the lack of translational in vitro disease models. The MAPT gene, encoding the microtubule-associated protein tau, has been shown to play an important role in FTD pathogenesis. Therefore, we used zinc finger nucleases to introduce two MAPT mutations into healthy donor induced pluripotent stem cells (iPSCs). The IVS10+16 mutation increases the expression of 4R tau, while the P301S mutation is pro-aggregant. Whole-transcriptome analysis of MAPT IVS10+16 neurons reveals neuronal subtype differences, reduced neural progenitor proliferation potential, and aberrant WNT/SHH signaling. Notably, these neurodevelopmental phenotypes could be recapitulated in neurons from patients carrying the MAPT IVS10+16 mutation. Moreover, the additional pro-aggregant P301S mutation revealed additional phenotypes, such as an increased calcium burst frequency, reduced lysosomal acidity, tau oligomerization, and neurodegeneration. This series of iPSCs could serve as a platform to unravel a potential link between pathogenic 4R tau and FTD.


Cell Reports | 2015

Synaptic Contacts Enhance Cell-to-Cell Tau Pathology Propagation

Sara Calafate; Arjan Buist; Katarzyna Miskiewicz; Vinoy Vijayan; Guy Daneels; Bart De Strooper; Joris de Wit; Patrik Verstreken; Diederik Moechars

Collaboration


Dive into the Arjan Buist's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ilie-Cosmin Stancu

Catholic University of Leuven

View shared research outputs
Top Co-Authors

Avatar

Ilse Dewachter

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Jean-Noël Octave

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Pascal Kienlen-Campard

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Peng Wang

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge