Network


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

Hotspot


Dive into the research topics where Lucas Treps is active.

Publication


Featured researches published by Lucas Treps.


The EMBO Journal | 2014

Aberrant methylation of tRNAs links cellular stress to neuro-developmental disorders

Sandra Blanco; Sabine Dietmann; Joana V. Flores; Shobbir Hussain; Claudia Kutter; Peter Humphreys; Margus Lukk; Patrick Lombard; Lucas Treps; Martyna Popis; Stefanie Kellner; Sabine M. Hölter; Lillian Garrett; Wolfgang Wurst; Lore Becker; Thomas Klopstock; Helmut Fuchs; Valérie Gailus-Durner; Martin Hrabĕ de Angelis; Ragnhildur Káradóttir; Mark Helm; Jernej Ule; Joseph G. Gleeson; Duncan T. Odom; Michaela Frye

Mutations in the cytosine‐5 RNA methyltransferase NSun2 cause microcephaly and other neurological abnormalities in mice and human. How post‐transcriptional methylation contributes to the human disease is currently unknown. By comparing gene expression data with global cytosine‐5 RNA methylomes in patient fibroblasts and NSun2‐deficient mice, we find that loss of cytosine‐5 RNA methylation increases the angiogenin‐mediated endonucleolytic cleavage of transfer RNAs (tRNA) leading to an accumulation of 5′ tRNA‐derived small RNA fragments. Accumulation of 5′ tRNA fragments in the absence of NSun2 reduces protein translation rates and activates stress pathways leading to reduced cell size and increased apoptosis of cortical, hippocampal and striatal neurons. Mechanistically, we demonstrate that angiogenin binds with higher affinity to tRNAs lacking site‐specific NSun2‐mediated methylation and that the presence of 5′ tRNA fragments is sufficient and required to trigger cellular stress responses. Furthermore, the enhanced sensitivity of NSun2‐deficient brains to oxidative stress can be rescued through inhibition of angiogenin during embryogenesis. In conclusion, failure in NSun2‐mediated tRNA methylation contributes to human diseases via stress‐induced RNA cleavage.


Cancer Cell | 2016

Inhibition of the Glycolytic Activator PFKFB3 in Endothelium Induces Tumor Vessel Normalization, Impairs Metastasis, and Improves Chemotherapy

Anna Rita Cantelmo; Lena Christin Conradi; Aleksandra Brajic; Jermaine Goveia; Joanna Kalucka; Andreas Pircher; Pallavi Chaturvedi; Johanna Hol; Bernard Thienpont; Laure Anne Teuwen; Sandra Schoors; Bram Boeckx; Joris Vriens; Anna Kuchnio; Koen Veys; Bert Cruys; Lise Finotto; Lucas Treps; Tor Espen Stav-Noraas; Francesco Bifari; Peter Stapor; Kim R. Kampen; Katrien De Bock; Guttorm Haraldsen; Luc Schoonjans; Ton J. Rabelink; Guy Eelen; Bart Ghesquière; Jalees Rehman; Diether Lambrechts

Abnormal tumor vessels promote metastasis and impair chemotherapy. Hence, tumor vessel normalization (TVN) is emerging as an anti-cancer treatment. Here, we show that tumor endothelial cells (ECs) have a hyper-glycolytic metabolism, shunting intermediates to nucleotide synthesis. EC haplo-deficiency or blockade of the glycolytic activator PFKFB3 did not affect tumor growth, but reduced cancer cell invasion, intravasation, and metastasis by normalizing tumor vessels, which improved vessel maturation and perfusion. Mechanistically, PFKFB3 inhibition tightened the vascular barrier by reducing VE-cadherin endocytosis in ECs, and rendering pericytes more quiescent and adhesive (via upregulation of N-cadherin) through glycolysis reduction; it also lowered the expression of cancer cell adhesion molecules in ECs by decreasing NF-κB signaling. PFKFB3-blockade treatment also improved chemotherapy of primary and metastatic tumors.


Atherosclerosis | 2016

Endothelial cell metabolism: A novel player in atherosclerosis? Basic principles and therapeutic opportunities

Andreas Pircher; Lucas Treps; Natalia Bodrug; Peter Carmeliet

Atherosclerosis is a leading cause of morbidity and mortality in Western society. Despite improved insight into disease pathogenesis and therapeutic options, additional treatment strategies are required. Emerging evidence highlights the relevance of endothelial cell (EC) metabolism for angiogenesis, and indicates that EC metabolism is perturbed when ECs become dysfunctional to promote atherogenesis. In this review, we overview the latest insights on EC metabolism and discuss current knowledge on how atherosclerosis deregulates EC metabolism, and how maladaptation of deregulated EC metabolism can contribute to atherosclerosis progression. We will also highlight possible therapeutic avenues, based on targeting EC metabolism.


Pharmacological Reviews | 2016

Manipulating Angiogenesis by Targeting Endothelial Metabolism: Hitting the Engine Rather than the Drivers—A New Perspective?

Lucas Treps; Lena-Christin Conradi; Ulrike Harjes; Peter Carmeliet

Excessive angiogenesis (i.e., the formation of new blood vessels) contributes to different pathologies, among them cancer and ocular disorders. Conversely, dysfunction of endothelial cells (ECs) contributes to cardiovascular complications, as is the case in diabetes. Inhibition of pathologic angiogenesis in blinding eye disease and cancer by targeting growth factors such as vascular endothelial growth factor has become an accepted therapeutic strategy. However, recent studies also unveiled the emerging importance of EC metabolism in controlling angiogenesis. In this overview, we will discuss recent insights in the metabolic regulation of angiogenesis, focusing on the best-characterized metabolic pathways, and highlight deregulation of EC metabolism in cancer and diabetes. We will give an outlook on how targeting EC metabolism can be used for blocking pathologic angiogenesis and for normalizing EC dysfunction.


The EMBO Journal | 2017

Endothelial cell metabolism in health and disease: impact of hypoxia

Brian W. Wong; Elke Marsch; Lucas Treps; Myriam Baes; Peter Carmeliet

In contrast to the general belief, endothelial cell (EC) metabolism has recently been identified as a driver rather than a bystander effect of angiogenesis in health and disease. Indeed, different EC subtypes present with distinct metabolic properties, which determine their function in angiogenesis upon growth factor stimulation. One of the main stimulators of angiogenesis is hypoxia, frequently observed in disease settings such as cancer and atherosclerosis. It has long been established that hypoxic signalling and metabolism changes are highly interlinked. In this review, we will provide an overview of the literature and recent findings on hypoxia‐driven EC function and metabolism in health and disease. We summarize evidence on metabolic crosstalk between different hypoxic cell types with ECs and suggest new metabolic targets.


Physiological Reviews | 2018

Endothelial Cell Metabolism

Guy Eelen; Pauline de Zeeuw; Lucas Treps; Ulrike Harjes; Brian W. Wong; Peter Carmeliet

Endothelial cells (ECs) are more than inert blood vessel lining material. Instead, they are active players in the formation of new blood vessels (angiogenesis) both in health and (life-threatening) diseases. Recently, a new concept arose by which EC metabolism drives angiogenesis in parallel to well-established angiogenic growth factors (e.g., vascular endothelial growth factor). 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3-driven glycolysis generates energy to sustain competitive behavior of the ECs at the tip of a growing vessel sprout, whereas carnitine palmitoyltransferase 1a-controlled fatty acid oxidation regulates nucleotide synthesis and proliferation of ECs in the stalk of the sprout. To maintain vascular homeostasis, ECs rely on an intricate metabolic wiring characterized by intracellular compartmentalization, use metabolites for epigenetic regulation of EC subtype differentiation, crosstalk through metabolite release with other cell types, and exhibit EC subtype-specific metabolic traits. Importantly, maladaptation of EC metabolism contributes to vascular disorders, through EC dysfunction or excess angiogenesis, and presents new opportunities for anti-angiogenic strategies. Here we provide a comprehensive overview of established as well as newly uncovered aspects of EC metabolism.


Physiology | 2017

Central Role of Metabolism in Endothelial Cell Function and Vascular Disease

Laura Bierhansl; Lena-Christin Conradi; Lucas Treps; Mieke Dewerchin; Peter Carmeliet

The importance of endothelial cell (EC) metabolism and its regulatory role in the angiogenic behavior of ECs during vessel formation and in the function of different EC subtypes determined by different vascular beds has been recognized only in the last few years. Even more importantly, apart from a role of nitric oxide and reactive oxygen species in EC dysfunction, deregulations of EC metabolism in disease only recently received increasing attention. Although comprehensive metabolic characterization of ECs still needs further investigation, the concept of targeting EC metabolism to treat vascular disease is emerging. In this overview, we summarize EC-specific metabolic pathways, describe the current knowledge on their deregulation in vascular diseases, and give an outlook on how vascular endothelial metabolism can serve as a target to normalize deregulated endothelium.


The Journal of Pathology | 2018

EnLIGHTenment of tumor vessel normalization and immunotherapy in glioblastoma: LIGHT normalizes blood vessels and improves immune cell infiltration

Lucas Treps

Glioblastoma multiforme (GBM) is a highly vascularized and aggressive brain tumor. Despite aggressive standard care, GBM remains predominantly fatal; hence, new innovative therapies are required. Recent research published in the Journal of Pathology has identified the CGKRK peptide as a promising tool with which to specifically target the tumor vasculature from high‐grade glioma. This tumor vessel‐homing peptide was fused to the tumor necrosis factor superfamily member LIGHT/TNFSF14, and injected intravenously into murine orthotopic GBM models. After treatment, the tumor vasculature appeared to be less abnormal, with normalized features such as increased endothelial barrier integrity, pericyte contractility, and tumor perfusion. Moreover, CGKRK–LIGHT induced the appearance of high endothelial venules (HEVs), which are specialized structures that play a role in lymphocyte trafficking and have been shown to increase T‐cell infiltration in solid tumors. Combining CGKRK–LIGHT with anti‐angiogenic and immune checkpoint blockade treatments boosted HEV induction and cytotoxic T‐cell infiltration, leading to a reduction in tumor burden. In this Commentary, I highlight the therapeutic opportunities provided by and the current limitations of LIGHT‐vascular targeting peptide as a new approach to target GBM and enhance tumor vessel delivery and immunotherapy efficacy. Copyright


Nucleic Acids Research | 2018

EndoDB: a database of endothelial cell transcriptomics data

Shawez Khan; Federico Taverna; Katerina Rohlenova; Lucas Treps; Vincent Geldhof; Laura de Rooij; Liliana Sokol; Andreas Pircher; Lena-Christin Conradi; Joanna Kalucka; Luc Schoonjans; Guy Eelen; Mieke Dewerchin; Tobias Karakach; Xuri Li; Jermaine Goveia; Peter Carmeliet

Abstract Endothelial cells (ECs) line blood vessels, regulate homeostatic processes (blood flow, immune cell trafficking), but are also involved in many prevalent diseases. The increasing use of high-throughput technologies such as gene expression microarrays and (single cell) RNA sequencing generated a wealth of data on the molecular basis of EC (dys-)function. Extracting biological insight from these datasets is challenging for scientists who are not proficient in bioinformatics. To facilitate the re-use of publicly available EC transcriptomics data, we developed the endothelial database EndoDB, a web-accessible collection of expert curated, quality assured and pre-analyzed data collected from 360 datasets comprising a total of 4741 bulk and 5847 single cell endothelial transcriptomes from six different organisms. Unlike other added-value databases, EndoDB allows to easily retrieve and explore data of specific studies, determine under which conditions genes and pathways of interest are deregulated and assess reprogramming of metabolism via principal component analysis, differential gene expression analysis, gene set enrichment analysis, heatmaps and metabolic and transcription factor analysis, while single cell data are visualized as gene expression color-coded t-SNE plots. Plots and tables in EndoDB are customizable, downloadable and interactive. EndoDB is freely available at https://vibcancer.be/software-tools/endodb, and will be updated to include new studies.


Cell Metabolism | 2018

Quiescent Endothelial Cells Upregulate Fatty Acid β-Oxidation for Vasculoprotection via Redox Homeostasis

Joanna Kalucka; Laura Bierhansl; Nadine Vasconcelos Conchinha; Rindert Missiaen; Ilaria Elia; Ulrike Bruning; Samantha Scheinok; Lucas Treps; Anna Rita Cantelmo; Charlotte Dubois; Pauline de Zeeuw; Jermaine Goveia; Annalisa Zecchin; Federico Taverna; Francisco Morales-Rodriguez; Aleksandra Brajic; Lena-Christin Conradi; Sandra Schoors; Ulrike Harjes; Kim Vriens; Gregor-Alexander Pilz; Rongyuan Chen; Richard M. Cubbon; Bernard Thienpont; Bert Cruys; Brian W. Wong; Bart Ghesquière; Mieke Dewerchin; Katrien De Bock; Xavier Sagaert

Little is known about the metabolism of quiescent endothelial cells (QECs). Nonetheless, when dysfunctional, QECs contribute to multiple diseases. Previously, we demonstrated that proliferating endothelial cells (PECs) use fatty acid β-oxidation (FAO) for de novo dNTP synthesis. We report now that QECs are not hypometabolic, but upregulate FAO >3-fold higher than PECs, not to support biomass or energy production but to sustain the tricarboxylic acid cycle for redox homeostasis through NADPH regeneration. Hence, endothelial loss of FAO-controlling CPT1A in CPT1AΔEC mice promotes EC dysfunction (leukocyte infiltration, barrier disruption) by increasing endothelial oxidative stress, rendering CPT1AΔEC mice more susceptible to LPS and inflammatory bowel disease. Mechanistically, Notch1 orchestrates the use of FAO for redox balance in QECs. Supplementation of acetate (metabolized to acetyl-coenzyme A) restores endothelial quiescence and counters oxidative stress-mediated EC dysfunction in CPT1AΔEC mice, offering therapeutic opportunities. Thus, QECs use FAO for vasculoprotection against oxidative stress-prone exposure.

Collaboration


Dive into the Lucas Treps's collaboration.

Top Co-Authors

Avatar

Peter Carmeliet

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Lena-Christin Conradi

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Jermaine Goveia

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Joanna Kalucka

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Mieke Dewerchin

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Andreas Pircher

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Anna Rita Cantelmo

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Bart Ghesquière

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Federico Taverna

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Guy Eelen

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge