Network


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

Hotspot


Dive into the research topics where Michael Dewaele is active.

Publication


Featured researches published by Michael Dewaele.


Nature Medicine | 2012

MDM4 is a key therapeutic target in cutaneous melanoma

Agnieszka Gembarska; Flavie Luciani; Clare G Fedele; Elisabeth A. Russell; Michael Dewaele; Stephanie Villar; Aleksandra Zwolinska; Sue Haupt; Job de Lange; Dana Yip; James S. Goydos; Jody J. Haigh; Ygal Haupt; Lionel Larue; Aart G. Jochemsen; Hubing Shi; Gatien Moriceau; Roger S. Lo; Ghanem Elias Ghanem; Mark Shackleton; Federico Bernal; Jean-Christophe Marine

The inactivation of the p53 tumor suppressor pathway, which often occurs through mutations in TP53 (encoding tumor protein 53) is a common step in human cancer. However, in melanoma—a highly chemotherapy-resistant disease—TP53 mutations are rare, raising the possibility that this cancer uses alternative ways to overcome p53-mediated tumor suppression. Here we show that Mdm4 p53 binding protein homolog (MDM4), a negative regulator of p53, is upregulated in a substantial proportion (∼65%) of stage I–IV human melanomas and that melanocyte-specific Mdm4 overexpression enhanced tumorigenesis in a mouse model of melanoma induced by the oncogene Nras. MDM4 promotes the survival of human metastatic melanoma by antagonizing p53 proapoptotic function. Notably, inhibition of the MDM4-p53 interaction restored p53 function in melanoma cells, resulting in increased sensitivity to cytotoxic chemotherapy and to inhibitors of the BRAF (V600E) oncogene. Our results identify MDM4 as a key determinant of impaired p53 function in human melanoma and designate MDM4 as a promising target for antimelanoma combination therapy.


Autophagy | 2011

Ins(1,4,5)P3 receptor-mediated Ca2+ signaling and autophagy induction are interrelated

Jean-Paul Decuypere; Kirsten Welkenhuyzen; Tomas Luyten; Raf Ponsaerts; Michael Dewaele; Jordi Molgó; Patrizia Agostinis; Ludwig Missiaen; Humbert De Smedt; Jan B. Parys; Geert Bultynck

The role of intracellular Ca2+ signaling in starvation-induced autophagy remains unclear. Here, we examined Ca2+ dynamics during starvation-induced autophagy and the underlying molecular mechanisms. Tightly correlating with autophagy stimulation, we observed a remodeling of the Ca2+ signalosome. First, short periods of starvation (1 to 3 h) caused a prominent increase of the ER Ca2+-store content and enhanced agonist-induced Ca2+ release. The mechanism involved the upregulation of intralumenal ER Ca2+-binding proteins, calreticulin and Grp78/BiP, which increased the ER Ca2+-buffering capacity and reduced the ER Ca2+ leak. Second, starvation led to Ins(1,4,5)P3R sensitization. Immunoprecipitation experiments showed that during starvation Beclin 1, released from Bcl-2, first bound with increasing efficiency to Ins(1,4,5)P3Rs; after reaching a maximal binding after 3 h, binding, however, decreased again. The interaction site of Beclin 1 was determined to be present in the N-terminal Ins(1,4,5)P3-binding domain of the Ins(1,4,5)P3R. The starvation-induced Ins(1,4,5)P3R sensitization was abolished in cells treated with BECN1 siRNA, but not with ATG5 siRNA, pointing toward an essential role of Beclin 1 in this process. Moreover, recombinant Beclin 1 sensitized Ins(1,4,5)P3Rs in 45Ca2+-flux assays, indicating a direct regulation of Ins(1,4,5)P3R activity by Beclin 1. Finally, we found that Ins(1,4,5)P3R-mediated Ca2+ signaling was critical for starvation-induced autophagy stimulation, since the Ca2+ chelator BAPTA-AM as well as the Ins(1,4,5)P3R inhibitor xestospongin B abolished the increase in LC3 lipidation and GFP-LC3-puncta formation. Hence, our results indicate a tight and essential interrelation between intracellular Ca2+ signaling and autophagy stimulation as a proximal event in response to starvation.


Nature Communications | 2015

Decoding the regulatory landscape of melanoma reveals TEADS as regulators of the invasive cell state

Annelien Verfaillie; Hana Imrichova; Zeynep Kalender Atak; Michael Dewaele; Florian Rambow; Gert Hulselmans; Christiaens; Dmitry Svetlichnyy; Flavie Luciani; Van den Mooter L; Claerhout S; Mark Fiers; Fabrice Journé; Ghanem Elias Ghanem; Carl Herrmann; Georg Halder; Jean-Christophe Marine; Stein Aerts

Transcriptional reprogramming of proliferative melanoma cells into a phenotypically distinct invasive cell subpopulation is a critical event at the origin of metastatic spreading. Here we generate transcriptome, open chromatin and histone modification maps of melanoma cultures; and integrate this data with existing transcriptome and DNA methylation profiles from tumour biopsies to gain insight into the mechanisms underlying this key reprogramming event. This shows thousands of genomic regulatory regions underlying the proliferative and invasive states, identifying SOX10/MITF and AP-1/TEAD as regulators, respectively. Knockdown of TEADs shows a previously unrecognized role in the invasive gene network and establishes a causative link between these transcription factors, cell invasion and sensitivity to MAPK inhibitors. Using regulatory landscapes and in silico analysis, we show that transcriptional reprogramming underlies the distinct cellular states present in melanoma. Furthermore, it reveals an essential role for the TEADs, linking it to clinically relevant mechanisms such as invasion and resistance.


Journal of Cellular and Molecular Medicine | 2011

Autophagy pathways activated in response to PDT contribute to cell resistance against ROS damage

Michael Dewaele; Wim Martinet; Noemi Rubio; Tom Verfaillie; Peter de Witte; Jacques Piette; Patrizia Agostinis

Reactive oxygen species (ROS) concurrently instigate apoptosis and autophagy pathways, but the link between these processes remains unclear. Because cytotoxic ROS formation is exploited in anticancer therapy, such as in photodynamic therapy (PDT), a better understanding of the complex interplay between autophagy and apoptosis is urgently required. Previously, we reported that ROS generated by PDT with an endoplasmic reticulum (ER)‐associated sensitizer leads to loss of ER‐Ca2+ homeostasis, ER stress and apoptosis. Here we show that PDT prompted Akt‐mTOR (mammalian target of rapamycin) pathway down‐regulation and stimulated macroautophagy (MA) in cancer and normal cells. Overexpression of the antioxidant enzyme glutathione peroxidase‐4 reversed mTOR down‐regulation and blocked MA progression and apoptosis. Attenuating MA using Atg5 knockdown or 3‐methyladenine, reduced clearance of oxidatively damaged proteins and increased apoptosis, thus revealing a cytoprotective role of MA in PDT. Paradoxically, genetic loss of MA improved clearance of oxidized proteins and reduced photokilling. We found that up‐regulation of chaperone‐mediated autophagy (CMA) in unstressed Atg5−/− cells compensated for MA loss and increased cellular resistance to PDT. CMA‐deficient cells were significantly sensitized to photokilling but were protected against the ER stressor thapsigargin. These results disclose a stress‐specific recruitment of autophagy pathways with cytoprotective function and unravel CMA as the dominant defence mechanism against PDT.


Journal of Clinical Investigation | 2016

Antisense oligonucleotide–mediated MDM4 exon 6 skipping impairs tumor growth

Michael Dewaele; Tommaso Tabaglio; Karen Willekens; Marco Bezzi; Shun Xie Teo; Diana H.P. Low; Cheryl M. Koh; Florian Rambow; Mark Fiers; Aljosja Rogiers; Enrico Radaelli; Muthafar Al-Haddawi; Soo Yong Tan; Els Hermans; Frédéric Amant; Hualong Yan; Manikandan Lakshmanan; Ratnacaram Chandrahas Koumar; Soon Thye Lim; Frederick A. Derheimer; Robert M. Campbell; Zahid Q. Bonday; Vinay Tergaonkar; Mark Shackleton; Christine Blattner; Jean-Christophe Marine; Ernesto Guccione

MDM4 is a promising target for cancer therapy, as it is undetectable in most normal adult tissues but often upregulated in cancer cells to dampen p53 tumor-suppressor function. The mechanisms that underlie MDM4 upregulation in cancer cells are largely unknown. Here, we have shown that this key oncogenic event mainly depends on a specific alternative splicing switch. We determined that while a nonsense-mediated, decay-targeted isoform of MDM4 (MDM4-S) is produced in normal adult tissues as a result of exon 6 skipping, enhanced exon 6 inclusion leads to expression of full-length MDM4 in a large number of human cancers. Although this alternative splicing event is likely regulated by multiple splicing factors, we identified the SRSF3 oncoprotein as a key enhancer of exon 6 inclusion. In multiple human melanoma cell lines and in melanoma patient-derived xenograft (PDX) mouse models, antisense oligonucleotide-mediated (ASO-mediated) skipping of exon 6 decreased MDM4 abundance, inhibited melanoma growth, and enhanced sensitivity to MAPK-targeting therapeutics. Additionally, ASO-based MDM4 targeting reduced diffuse large B cell lymphoma PDX growth. As full-length MDM4 is enhanced in multiple human tumors, our data indicate that this strategy is applicable to a wide range of tumor types. We conclude that enhanced MDM4 exon 6 inclusion is a common oncogenic event and has potential as a clinically compatible therapeutic target.


Free Radical Biology and Medicine | 2014

p38MAPK-regulated induction of p62 and NBR1 after photodynamic therapy promotes autophagic clearance of ubiquitin aggregates and reduces reactive oxygen species levels by supporting Nrf2–antioxidant signaling

Noemi Rubio; Julien Verrax; Michael Dewaele; Tom Verfaillie; Terje Johansen; Jacques Piette; Patrizia Agostinis

Emerging evidence indicates that oxidative stress instigates the formation of ubiquitin (Ub) aggregates, substrates of autophagy, through a process requiring the ubiquitin binding adaptors p62/SQSTM1 and NBR1. Here, we have investigated the role of p62 and NBR1 in cell survival after hypericin-mediated photodynamic therapy (Hyp-PDT), a procedure known to incite robust reactive oxygen species (ROS)-based endoplasmic reticulum stress and autophagy pathways. We found that Hyp-PDT stimulated the formation of p62- and NBR1-associated Ub aggregates in normal and cancer cells, which were ultimately removed by autophagy, through a mechanism partially regulated by p38(MAPK). In line with this, genetic or pharmacological p38(MAPK) inhibition reduced p62 and NBR1 levels and aggregate formation and impaired Nrf2 activation, thus increasing photo-oxidative stress and cell death. p62-deficient cells, or cells lacking p62 and with reduced levels of NBR1 (through siRNA knockdown), also displayed reduced aggregate formation but exhibited attenuated ROS levels, reduced caspase activation, and improved survival after Hyp-PDT. The increased resistance to photo-oxidative stress exhibited by cells lacking p62 and/or NBR1 was overruled by the inhibition of p38(MAPK), which restored cytotoxic ROS levels, thus indicating the relevance of this signal in the control of cell viability. Taken together these findings provide evidence that in photodynamically treated cells a p38(MAPK)-regulated pathway coordinates the p62/NBR1-mediated clearance of cytosolic aggregates and mitigates PDT-induced proteotoxicity. They also reveal that a functional p38(MAPK)-Nrf2 signal is required to keep ROS levels in check and protect against PDT-induced proteotoxicity, independent of aggregate formation.


Clinical Cancer Research | 2017

Amplification of 1q32.1 refines the molecular classification of endometrial carcinoma

Jeroen Depreeuw; Ellen Stelloo; Elisabeth M. Osse; Carien L. Creutzberg; Remi A. Nout; Matthieu Moisse; Diego A. Garcia-Dios; Michael Dewaele; Karen Willekens; Jean-Christophe Marine; Xavier Matias-Guiu; Frédéric Amant; Diether Lambrechts; Tjalling Bosse

Purpose: Molecular classification of endometrial cancer identified distinct molecular subgroups. However, the largest subset of endometrial cancers remains poorly characterized and is referred to as the “nonspecific molecular profile” (NSMP) subgroup. Here, we aimed at refining the classification of this subgroup by profiling somatic copy-number aberrations (SCNAs). Experimental Design: SCNAs were analyzed in 141 endometrial cancers using whole-genome SNP arrays and pooled with 361 endometrial cancers from The Cancer Genome Atlas. Genomic Identification of Significant Targets in Cancer (GISTIC) identified statistically enriched SCNAs and penalized Cox regression assessed survival effects. The prognostic significance of relevant SCNAs was validated using multiplex ligation-dependent probe amplification in 840 endometrial cancers from the PORTEC-1/2 trials. Copy-number status of genes was correlated with gene expression to identify potential cancer drivers. One plausible oncogene was validated in vitro using antisense oligonucleotide-based strategy. Results: SCNAs affecting chromosome 1q32.1 significantly correlated with worse relapse-free survival (RFS) in the NSMP subgroup (HR, 2.12; 95% CI, 1.26–3.59; P = 0.005). This effect was replicated in NSMP endometrial cancers from PORTEC-1/2 (HR, 2.34; 95% CI, 1.17–4.70; P = 0.017). A new molecular classification including the 1q32.1 amplification improved risk prediction of recurrence. MDM4 gene expression strongly correlated with 1q32.1 amplification. Silencing MDM4 inhibited cell growth in cell lines carrying 1q32.1 amplification, but not in those without MDM4 amplification. Vice versa, increasing MDM4 expression in nonamplified cell lines stimulated cell proliferation. Conclusions: 1q32.1 amplification was identified as a prognostic marker for poorly characterized NSMP endometrial cancers, refining the molecular classification of this subgroup. We functionally validated MDM4 as a potential oncogenic driver in the 1q32.1 region. Clin Cancer Res; 23(23); 7232–41. ©2017 AACR.


Nature | 2018

Codon-specific translation reprogramming promotes resistance to targeted therapy

Francesca Rapino; Sylvain Delaunay; Florian Rambow; Zhaoli Zhou; Lars Tharun; Pascal De Tullio; Olga Sin; Kateryna Shostak; Sebastian Schmitz; Jolanda Piepers; Bart Ghesquière; Latifa Karim; Benoit Charloteaux; Diane Jamart; Alexandra Florin; Charles Lambert; Andrée Rorive; Guy Jerusalem; Eleonora Leucci; Michael Dewaele; Marc Vooijs; Sebastian A. Leidel; Michel Georges; Marianne Voz; Bernard Peers; Reinhard Büttner; Jean-Christophe Marine; Alain Chariot; Pierre Close

Reprogramming of mRNA translation has a key role in cancer development and drug resistance1. However, the molecular mechanisms that are involved in this process remain poorly understood. Wobble tRNA modifications are required for specific codon decoding during translation2,3. Here we show, in humans, that the enzymes that catalyse modifications of wobble uridine 34 (U34) tRNA (U34 enzymes) are key players of the protein synthesis rewiring that is induced by the transformation driven by the BRAFV600E oncogene and by resistance to targeted therapy in melanoma. We show that BRAFV600E-expressing melanoma cells are dependent on U34 enzymes for survival, and that concurrent inhibition of MAPK signalling and ELP3 or CTU1 and/or CTU2 synergizes to kill melanoma cells. Activation of the PI3K signalling pathway, one of the most common mechanisms of acquired resistance to MAPK therapeutic agents, markedly increases the expression of U34 enzymes. Mechanistically, U34 enzymes promote glycolysis in melanoma cells through the direct, codon-dependent, regulation of the translation of HIF1A mRNA and the maintenance of high levels of HIF1α protein. Therefore, the acquired resistance to anti-BRAF therapy is associated with high levels of U34 enzymes and HIF1α. Together, these results demonstrate that U34 enzymes promote the survival and resistance to therapy of melanoma cells by regulating specific mRNA translation.Enzymes that catalyse modifications of wobble uridine 34 tRNA are essential for the survival of melanoma cells that rely on HIF1α-dependent metabolism through codon-dependent regulation of the translation of HIF1A mRNA.


Cell | 2018

Toward Minimal Residual Disease-Directed Therapy in Melanoma.

Florian Rambow; Aljosja Rogiers; Oskar Marin-Bejar; Sara Aibar; Julia Femel; Michael Dewaele; Panagiotis Karras; Daniel Brown; Young Hwan Chang; Maria Debiec-Rychter; Carmen Adriaens; Enrico Radaelli; Pascal Wolter; Oliver Bechter; Reinhard Dummer; Mitchell P. Levesque; Adriano Piris; Dennie T. Frederick; Genevieve M. Boland; Keith T. Flaherty; Joost van den Oord; Thierry Voet; Stein Aerts; Amanda W. Lund; Jean-Christophe Marine

Many patients with advanced cancers achieve dramatic responses to a panoply of therapeutics yet retain minimal residual disease (MRD), which ultimately results in relapse. To gain insights into the biology of MRD, we applied single-cell RNA sequencing to malignant cells isolated from BRAF mutant patient-derived xenograft melanoma cohorts exposed to concurrent RAF/MEK-inhibition. We identified distinct drug-tolerant transcriptional states, varying combinations of which co-occurred within MRDs from PDXs and biopsies of patients on treatment. One of these exhibited a neural crest stem cell (NCSC) transcriptional program largely driven by the nuclear receptor RXRG. An RXR antagonist mitigated accumulation of NCSCs in MRD and delayed the development of resistance. These data identify NCSCs as key drivers of resistance and illustrate the therapeutic potential of MRD-directed therapy. They also highlight how gene regulatory network architecture reprogramming may be therapeutically exploited to limit cellular heterogeneity, a key driver of disease progression and therapy resistance.


British Journal of Dermatology | 2018

Dabrafenib plus trametinib in BRAF K601E-mutant melanoma

Aljosja Rogiers; D. Thomas; S. Vander Borght; J. J. van den Oord; Oliver Bechter; Michael Dewaele; Florian Rambow; J-C Marine; Pascal Wolter

About 40 - 50% of cutaneous melanomas have activating BRAF mutations that are reachable with targeted therapy and combined BRAF-MEK inhibition improves clinical outcomes in advanced BRAF V600E/K-mutant melanoma. Three combinations are FDA-approved for this indication: dabrafenib-trametinib, vemurafenib-cobimetinib and encorafenib-binimetinib. The K601E mutation comprises approximately 3% of BRAF mutations in patients with melanoma. This article is protected by copyright. All rights reserved.

Collaboration


Dive into the Michael Dewaele's collaboration.

Top Co-Authors

Avatar

Florian Rambow

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Jean-Christophe Marine

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Patrizia Agostinis

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Stein Aerts

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Annelien Verfaillie

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Flavie Luciani

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Gert Hulselmans

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Hana Imrichova

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Mark Fiers

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Zeynep Kalender Atak

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge