Network


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

Hotspot


Dive into the research topics where Ruben Boon is active.

Publication


Featured researches published by Ruben Boon.


The EMBO Journal | 2016

Relief of hypoxia by angiogenesis promotes neural stem cell differentiation by targeting glycolysis

Christian Lange; Miguel Turrero García; Ilaria Decimo; Francesco Bifari; Guy Eelen; Annelies Quaegebeur; Ruben Boon; Hui Zhao; Bram Boeckx; Junlei Chang; Christine Wu; Ferdinand le Noble; Diether Lambrechts; Mieke Dewerchin; Calvin J. Kuo; Wieland B. Huttner; Peter Carmeliet

Blood vessels are part of the stem cell niche in the developing cerebral cortex, but their in vivo role in controlling the expansion and differentiation of neural stem cells (NSCs) in development has not been studied. Here, we report that relief of hypoxia in the developing cerebral cortex by ingrowth of blood vessels temporo‐spatially coincided with NSC differentiation. Selective perturbation of brain angiogenesis in vessel‐specific Gpr124 null embryos, which prevented the relief from hypoxia, increased NSC expansion at the expense of differentiation. Conversely, exposure to increased oxygen levels rescued NSC differentiation in Gpr124 null embryos and increased it further in WT embryos, suggesting that niche blood vessels regulate NSC differentiation at least in part by providing oxygen. Consistent herewith, hypoxia‐inducible factor (HIF)‐1α levels controlled the switch of NSC expansion to differentiation. Finally, we provide evidence that high glycolytic activity of NSCs is required to prevent their precocious differentiation in vivo. Thus, blood vessel function is required for efficient NSC differentiation in the developing cerebral cortex by providing oxygen and possibly regulating NSC metabolism.


Nature Communications | 2017

Proline metabolism supports metastasis formation and could be inhibited to selectively target metastasizing cancer cells.

Ilaria Elia; Dorien Broekaert; Stefan Christen; Ruben Boon; Enrico Radaelli; Martin F. Orth; Catherine M. Verfaillie; Thomas G. P. Grunewald; Sarah-Maria Fendt

Metastases are the leading cause of mortality in patients with cancer. Metastasis formation requires cancer cells to adapt their cellular phenotype. However, how metabolism supports this adaptation of cancer cells is poorly defined. We use 2D versus 3D cultivation to induce a shift in the cellular phenotype of breast cancer cells. We discover that proline catabolism via proline dehydrogenase (Prodh) supports growth of breast cancer cells in 3D culture. Subsequently, we link proline catabolism to in vivo metastasis formation. In particular, we find that PRODH expression and proline catabolism is increased in metastases compared to primary breast cancers of patients and mice. Moreover, inhibiting Prodh is sufficient to impair formation of lung metastases in the orthotopic 4T1 and EMT6.5 mouse models, without adverse effects on healthy tissue and organ function. In conclusion, we discover that Prodh is a potential drug target for inhibiting metastasis formation.


Stem cell reports | 2015

Efficient Recombinase-Mediated Cassette Exchange in hPSCs to Study the Hepatocyte Lineage Reveals AAVS1 Locus-Mediated Transgene Inhibition

Laura Ordovas; Ruben Boon; Mariaelena Pistoni; Yemiao Chen; Esther Wolfs; Wenting Guo; Rangarajan Sambathkumar; Sylwia Bobis-Wozowicz; Nicky Helsen; Jolien Vanhove; Pieter Berckmans; Qing Cai; Kim Vanuytsel; Kristel Eggermont; Veerle Vanslembrouck; Béla Z. Schmidt; Susanna Raitano; Ludo Van Den Bosch; Yaakov Nahmias; Toni Cathomen; Tom Struys; Catherine M. Verfaillie

Summary Tools for rapid and efficient transgenesis in “safe harbor” loci in an isogenic context remain important to exploit the possibilities of human pluripotent stem cells (hPSCs). We created hPSC master cell lines suitable for FLPe recombinase-mediated cassette exchange (RMCE) in the AAVS1 locus that allow generation of transgenic lines within 15 days with 100% efficiency and without random integrations. Using RMCE, we successfully incorporated several transgenes useful for lineage identification, cell toxicity studies, and gene overexpression to study the hepatocyte lineage. However, we observed unexpected and variable transgene expression inhibition in vitro, due to DNA methylation and other unknown mechanisms, both in undifferentiated hESC and differentiating hepatocytes. Therefore, the AAVS1 locus cannot be considered a universally safe harbor locus for reliable transgene expression in vitro, and using it for transgenesis in hPSC will require careful assessment of the function of individual transgenes.


Journal of Hepatology | 2016

Stem cell-derived hepatocytes: A novel model for hepatitis E virus replication

Nicky Helsen; Yannick Debing; Jan Paeshuyse; Kai Dallmeier; Ruben Boon; Mar Coll; Pau Sancho-Bru; Christel Claes; Johan Neyts; Catherine M. Verfaillie

BACKGROUND & AIMS Yearly, approximately 20million people become infected with the hepatitis E virus (HEV) resulting in over 3million cases of acute hepatitis. Although HEV-mediated hepatitis is usually self-limiting, severe cases of fulminant hepatitis as well as chronic infections have been reported, resulting annually in an estimated 60,000 deaths. We studied whether pluripotent stem cell (PSC)-derived hepatocytes, mesodermal and/or neuroprogenitor cells support HEV replication. METHODS Human PSC were differentiated towards hepatocyte-like cells, mesodermal cells and neuroprogenitors and subsequently infected with HEV. Infection and replication of HEV was analyzed by qRT-PCR, RNA in situ hybridization, negative strand RT-PCR, production of infectious virions and transfection with a transient HEV reporter replicon. RESULTS PSC-derived hepatocytes supported the complete replication cycle of HEV, as demonstrated by the intracellular presence of positive and negative strand HEV RNA and the production of infectious virions. The replication of the virus in these cells was inhibited by the antiviral drugs ribavirin and interferon-α2b. In contrast to PSC-derived hepatocytes, PSC-derived mesodermal cells and neuroprogenitors only supported HEV replication upon transfection with a HEV subgenomic replicon. CONCLUSION We demonstrate that PSC can be used to study the hepatotropism of HEV infection. The complete replication cycle of HEV can be recapitulated in infected PSC-derived hepatocytes. By contrast other germ layer cells support intracellular replication but are not infectable with HEV. Thus the early steps in the viral cycle are the main determinant governing HEV tissue tropism. PSC-hepatocytes offer a physiological relevant tool to study the biology of HEV infection and replication and may aid in the design of therapeutic strategies.


Stem Cell Research | 2015

Highly proliferative primitive fetal liver hematopoietic stem cells are fueled by oxidative metabolic pathways

Javed Karim Manesia; Zhuofei Xu; Dorien Broekaert; Ruben Boon; Alex van Vliet; Guy Eelen; Thomas Vanwelden; Steve Stegen; Nick van Gastel; Alberto Pascual-Montano; Sarah-Maria Fendt; Geert Carmeliet; Peter Carmeliet; Satish Khurana; Catherine M. Verfaillie

Hematopoietic stem cells (HSCs) in the fetal liver (FL) unlike adult bone marrow (BM) proliferate extensively, posing different metabolic demands. However, metabolic pathways responsible for the production of energy and cellular building blocks in FL HSCs have not been described. Here, we report that FL HSCs use oxygen dependent energy generating pathways significantly more than their BM counterparts. RNA-Seq analysis of E14.5 FL versus BM derived HSCs identified increased expression levels of genes involved in oxidative phosphorylation (OxPhos) and the citric acid cycle (TCA). We demonstrated that FL HSCs contain more mitochondria than BM HSCs, which resulted in increased levels of oxygen consumption and reactive oxygen species (ROS) production. Higher levels of DNA repair and antioxidant pathway gene expression may prevent ROS-mediated (geno)toxicity in FL HSCs. Thus, we here for the first time highlight the underestimated importance of oxygen dependent pathways for generating energy and building blocks in FL HSCs.


Nature Communications | 2017

HDAC6 inhibition reverses axonal transport defects in motor neurons derived from FUS-ALS patients

Wenting Guo; Ruben Boon; Philip Van Damme; Ludo Van Den Bosch; Werend Boesmans; Natasja Geens; Jolien Steyaert; Laura Fumagalli; Pieter Vanden Berghe; Matthew Jarpe; Laura Ordovas; Thomas Vanwelden; Catherine M. Verfaillie; Wim Robberecht; Cynthia Lefebvre-Omar; Susanne Petri; Marc Welters; Maximilian Naujock; Abdulsamie Patel; Tine Tricot; Delphine Bohl; Pieter Baatsen; Florian Wegner; Veronick Benoy; Jared Sterneckert; Tijs Vandoorne

Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disorder due to selective loss of motor neurons (MNs). Mutations in the fused in sarcoma (FUS) gene can cause both juvenile and late onset ALS. We generated and characterized induced pluripotent stem cells (iPSCs) from ALS patients with different FUS mutations, as well as from healthy controls. Patient-derived MNs show typical cytoplasmic FUS pathology, hypoexcitability, as well as progressive axonal transport defects. Axonal transport defects are rescued by CRISPR/Cas9-mediated genetic correction of the FUS mutation in patient-derived iPSCs. Moreover, these defects are reproduced by expressing mutant FUS in human embryonic stem cells (hESCs), whereas knockdown of endogenous FUS has no effect, confirming that these pathological changes are mutant FUS dependent. Pharmacological inhibition as well as genetic silencing of histone deacetylase 6 (HDAC6) increase α-tubulin acetylation, endoplasmic reticulum (ER)–mitochondrial overlay, and restore the axonal transport defects in patient-derived MNs.Amyotrophic lateral sclerosis (ALS) leads to selective loss of motor neurons. Using motor neurons derived from induced pluripotent stem cells from patients with ALS and FUS mutations, the authors demonstrate that axonal transport deficits that are observed in these cells can be rescued by HDAC6 inhibition.


Metabolic Engineering | 2017

Dual loss of succinate dehydrogenase (SDH) and complex I activity is necessary to recapitulate the metabolic phenotype of SDH mutant tumors

Doriane Lorendeau; Gianmarco Rinaldi; Ruben Boon; Pieter Spincemaille; Kristine Metzger; Christian Jäger; Stefan Christen; Xiangyi Dong; Sabine Kuenen; Karin Voordeckers; Patrik Verstreken; David Cassiman; Pieter Vermeersch; Catherine M. Verfaillie; Karsten Hiller; Sarah-Maria Fendt

Mutations in succinate dehydrogenase (SDH) are associated with tumor development and neurodegenerative diseases. Only in tumors, loss of SDH activity is accompanied with the loss of complex I activity. Yet, it remains unknown whether the metabolic phenotype of SDH mutant tumors is driven by loss of complex I function, and whether this contributes to the peculiarity of tumor development versus neurodegeneration. We addressed this question by decoupling loss of SDH and complex I activity in cancer cells and neurons. We found that sole loss of SDH activity was not sufficient to recapitulate the metabolic phenotype of SDH mutant tumors, because it failed to decrease mitochondrial respiration and to activate reductive glutamine metabolism. These metabolic phenotypes were only induced upon the additional loss of complex I activity. Thus, we show that complex I function defines the metabolic differences between SDH mutation associated tumors and neurodegenerative diseases, which could open novel therapeutic options against both diseases.


Stem cell reports | 2016

H3K27me3 Does Not Orchestrate the Expression of Lineage-Specific Markers in hESC-Derived Hepatocytes In Vitro

Jolien Vanhove; Mariaelena Pistoni; Marc Welters; Kristel Eggermont; Veerle Vanslembrouck; Nicky Helsen; Ruben Boon; Mustapha Najimi; Etienne Sokal; Philippe Collas; J. Willem Voncken; Catherine M. Verfaillie

Summary Although pluripotent stem cells can be differentiated into the hepatocyte lineages, such cells retain an immature phenotype. As the chromatin state of regulatory regions controls spatiotemporal gene expression during development, we evaluated changes in epigenetic histone marks in lineage-specific genes throughout in vitro hepatocyte differentiation from human embryonic stem cells (hESCs). Active acetylation and methylation marks at promoters and enhancers correlated with progressive changes in gene expression. However, repression-associated H3K27me3 marks at these control regions showed an inverse correlation with gene repression during transition from hepatic endoderm to a hepatocyte-like state. Inhibitor of Enhancer of Zeste Homolog 2 (EZH2) reduced H3K27me3 decoration but did not improve hepatocyte maturation. Thus, H3K27me3 at regulatory regions does not regulate transcription and appears dispensable for hepatocyte lineage differentiation of hESCs in vitro.


Journal of Visualized Experiments | 2016

Rapid and Efficient Generation of Recombinant Human Pluripotent Stem Cells by Recombinase-mediated Cassette Exchange in the AAVS1 Locus

Laura Ordovas; Ruben Boon; Mariaelena Pistoni; Yemiao Chen; Rangarajan Sambathkumar; Nicky Helsen; Jolien Vanhove; Pieter Berckmans; Qing Cai; Kim Vanuytsel; Susanna Raitano; Catherine M. Verfaillie

Even with the revolution of gene-targeting technologies led by CRISPR-Cas9, genetic modification of human pluripotent stem cells (hPSCs) is still time consuming. Comparative studies that use recombinant lines with transgenes integrated into safe harbor loci could benefit from approaches that use site-specific targeted recombinases, like Cre or FLPe, which are more rapid and less prone to off-target effects. Such methods have been described, although they do not significantly outperform gene targeting in most aspects. Using Zinc-finger nucleases, we previously created a master cell line in the AAVS1 locus of hPSCs that contains a GFP-Hygromycin-tk expressing cassette, flanked by heterotypic FRT sequences. Here, we describe the procedures to perform FLPe recombinase-mediated cassette exchange (RMCE) using this line. The master cell line is transfected with a RMCE donor vector, which contains a promoterless Puromycin resistance, and with FLPe recombinase. Application of both a positive (Puromycin) and negative (FIAU) selection program leads to the selection of RMCE without random integrations. RMCE generates fully characterized pluripotent polyclonal transgenic lines in 15 d with 100% efficiency. Despite the recently described limitations of the AAVS1 locus, the ease of the system paves the way for hPSC transgenesis in isogenic settings, is necessary for comparative studies, and enables semi-high-throughput genetic screens for gain/loss of function analysis that would otherwise be highly time consuming.


Stem cell reports | 2018

SOX10 Single Transcription Factor-Based Fast and Efficient Generation of Oligodendrocytes from Human Pluripotent Stem Cells

Juan Antonio García-León; Manoj Kumar; Ruben Boon; David Chau; Jennifer One; Esther Wolfs; Kristel Eggermont; Pieter Berckmans; Nilhan Gunhanlar; Femke M.S. de Vrij; Bas Lendemeijer; Benjamin Pavie; Nikky Corthout; Steven A. Kushner; José Carlos Dávila; Ivo Lambrichts; Wei Shou Hu; Catherine M. Verfaillie

Summary Scarce access to primary samples and lack of efficient protocols to generate oligodendrocytes (OLs) from human pluripotent stem cells (hPSCs) are hampering our understanding of OL biology and the development of novel therapies. Here, we demonstrate that overexpression of the transcription factor SOX10 is sufficient to generate surface antigen O4-positive (O4+) and myelin basic protein-positive OLs from hPSCs in only 22 days, including from patients with multiple sclerosis or amyotrophic lateral sclerosis. The SOX10-induced O4+ population resembles primary human OLs at the transcriptome level and can myelinate neurons in vivo. Using in vitro OL-neuron co-cultures, myelination of neurons by OLs can also be demonstrated, which can be adapted to a high-throughput screening format to test the response of pro-myelinating drugs. In conclusion, we provide an approach to generate OLs in a very rapid and efficient manner, which can be used for disease modeling, drug discovery efforts, and potentially for therapeutic OL transplantation.

Collaboration


Dive into the Ruben Boon's collaboration.

Top Co-Authors

Avatar

Catherine M. Verfaillie

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Nicky Helsen

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Jolien Vanhove

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Laura Ordovas

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Mariaelena Pistoni

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Pieter Berckmans

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Rangarajan Sambathkumar

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Mar Coll

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Aernout Luttun

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Kim Vanuytsel

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge