Network


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

Hotspot


Dive into the research topics where Jan J. Molenaar is active.

Publication


Featured researches published by Jan J. Molenaar.


Nature | 2012

Sequencing of neuroblastoma identifies chromothripsis and defects in neuritogenesis genes

Jan J. Molenaar; Jan Koster; Danny A. Zwijnenburg; Peter van Sluis; Linda J. Valentijn; Ida van der Ploeg; Mohamed Hamdi; Johan van Nes; Bart A. Westerman; Jennemiek van Arkel; Marli E. Ebus; Franciska Haneveld; Arjan Lakeman; Linda Schild; Piet Molenaar; Peter Stroeken; Max M. van Noesel; Ingrid Øra; Evan E. Santo; Huib N. Caron; Ellen M. Westerhout; Rogier Versteeg

Neuroblastoma is a childhood tumour of the peripheral sympathetic nervous system. The pathogenesis has for a long time been quite enigmatic, as only very few gene defects were identified in this often lethal tumour. Frequently detected gene alterations are limited to MYCN amplification (20%) and ALK activations (7%). Here we present a whole-genome sequence analysis of 87 neuroblastoma of all stages. Few recurrent amino-acid-changing mutations were found. In contrast, analysis of structural defects identified a local shredding of chromosomes, known as chromothripsis, in 18% of high-stage neuroblastoma. These tumours are associated with a poor outcome. Structural alterations recurrently affected ODZ3, PTPRD and CSMD1, which are involved in neuronal growth cone stabilization. In addition, ATRX, TIAM1 and a series of regulators of the Rac/Rho pathway were mutated, further implicating defects in neuritogenesis in neuroblastoma. Most tumours with defects in these genes were aggressive high-stage neuroblastomas, but did not carry MYCN amplifications. The genomic landscape of neuroblastoma therefore reveals two novel molecular defects, chromothripsis and neuritogenesis gene alterations, which frequently occur in high-risk tumours.


Nature Genetics | 2012

LIN28B induces neuroblastoma and enhances MYCN levels via let-7 suppression

Jan J. Molenaar; Raquel Domingo-Fernández; Marli E. Ebus; Sven Lindner; Jan Koster; Ksenjia Drabek; Pieter Mestdagh; Peter van Sluis; Linda J. Valentijn; Johan van Nes; Marloes Broekmans; Franciska Haneveld; Richard Volckmann; Isabella Bray; Lukas C. Heukamp; Annika Sprüssel; Theresa Thor; Kristina Kieckbusch; Ludger Klein-Hitpass; Matthias Fischer; Jo Vandesompele; Alexander Schramm; Max M. van Noesel; Luigi Varesio; Franki Speleman; Angelika Eggert; Raymond L. Stallings; Huib N. Caron; Rogier Versteeg; Johannes H. Schulte

LIN28B regulates developmental processes by modulating microRNAs (miRNAs) of the let-7 family. A role for LIN28B in cancer has been proposed but has not been established in vivo. Here, we report that LIN28B showed genomic aberrations and extensive overexpression in high-risk neuroblastoma compared to several other tumor entities and normal tissues. High LIN28B expression was an independent risk factor for adverse outcome in neuroblastoma. LIN28B signaled through repression of the let-7 miRNAs and consequently resulted in elevated MYCN protein expression in neuroblastoma cells. LIN28B–let-7–MYCN signaling blocked differentiation of normal neuroblasts and neuroblastoma cells. These findings were fully recapitulated in a mouse model in which LIN28B expression in the sympathetic adrenergic lineage induced development of neuroblastomas marked by low let-7 miRNA levels and high MYCN protein expression. Interference with this pathway might offer therapeutic perspectives.


Nature Genetics | 2015

Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations

Thomas F. Eleveld; Derek A. Oldridge; Virginie Bernard; Jan Koster; Leo Colmet Daage; Sharon J. Diskin; Linda Schild; Nadia Bessoltane Bentahar; Angela Bellini; Mathieu Chicard; Eve Lapouble; Valérie Combaret; Patricia Legoix-Né; Jean Michon; Trevor J. Pugh; Lori S. Hart; JulieAnn Rader; Edward F. Attiyeh; Jun S. Wei; Shile Zhang; Arlene Naranjo; Julie M. Gastier-Foster; Michael D. Hogarty; Shahab Asgharzadeh; Malcolm A. Smith; Jaime M. Guidry Auvil; Thomas B. K. Watkins; Danny A. Zwijnenburg; Marli E. Ebus; Peter van Sluis

The majority of patients with neuroblastoma have tumors that initially respond to chemotherapy, but a large proportion will experience therapy-resistant relapses. The molecular basis of this aggressive phenotype is unknown. Whole-genome sequencing of 23 paired diagnostic and relapse neuroblastomas showed clonal evolution from the diagnostic tumor, with a median of 29 somatic mutations unique to the relapse sample. Eighteen of the 23 relapse tumors (78%) showed mutations predicted to activate the RAS-MAPK pathway. Seven of these events were detected only in the relapse tumor, whereas the others showed clonal enrichment. In neuroblastoma cell lines, we also detected a high frequency of activating mutations in the RAS-MAPK pathway (11/18; 61%), and these lesions predicted sensitivity to MEK inhibition in vitro and in vivo. Our findings provide a rationale for genetic characterization of relapse neuroblastomas and show that RAS-MAPK pathway mutations may function as a biomarker for new therapeutic approaches to refractory disease.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Inactivation of CDK2 is synthetically lethal to MYCN over-expressing cancer cells

Jan J. Molenaar; Marli E. Ebus; Dirk Geerts; Jan Koster; Fieke Lamers; Linda J. Valentijn; Ellen M. Westerhout; Rogier Versteeg; Huib N. Caron

Two genes have a synthetically lethal relationship when the silencing or inhibiting of 1 gene is only lethal in the context of a mutation or activation of the second gene. This situation offers an attractive therapeutic strategy, as inhibition of such a gene will only trigger cell death in tumor cells with an activated second oncogene but spare normal cells without activation of the second oncogene. Here we present evidence that CDK2 is synthetically lethal to neuroblastoma cells with MYCN amplification and over-expression. Neuroblastomas are childhood tumors with an often lethal outcome. Twenty percent of the tumors have MYCN amplification, and these tumors are ultimately refractory to any therapy. Targeted silencing of CDK2 by 3 RNA interference techniques induced apoptosis in MYCN-amplified neuroblastoma cell lines, but not in MYCN single copy cells. Silencing of MYCN abrogated this apoptotic response in MYCN-amplified cells. Inversely, silencing of CDK2 in MYCN single copy cells did not trigger apoptosis, unless a MYCN transgene was activated. The MYCN induced apoptosis after CDK2 silencing was accompanied by nuclear stabilization of P53, and mRNA profiling showed up-regulation of P53 target genes. Silencing of P53 rescued the cells from MYCN-driven apoptosis. The synthetic lethality of CDK2 silencing in MYCN activated neuroblastoma cells can also be triggered by inhibition of CDK2 with a small molecule drug. Treatment of neuroblastoma cells with roscovitine, a CDK inhibitor, at clinically achievable concentrations induced MYCN-dependent apoptosis. The synthetically lethal relationship between CDK2 and MYCN indicates CDK2 inhibitors as potential MYCN-selective cancer therapeutics.


Cancer Research | 2008

Cyclin D1 and CDK4 Activity Contribute to the Undifferentiated Phenotype in Neuroblastoma

Jan J. Molenaar; Marli E. Ebus; Jan Koster; Peter van Sluis; Carel J. M. van Noesel; Rogier Versteeg; Huib N. Caron

Genomic aberrations of Cyclin D1 (CCND1), CDK4, and CDK6 in neuroblastoma indicate that dysregulation of the G(1) entry checkpoint is an important cell cycle aberration in this pediatric tumor. Here, we report that analysis of Affymetrix expression data of primary neuroblastic tumors shows an extensive overexpression of Cyclin D1, which correlates with histologic subgroups. Immunohistochemical analysis showed overexpression of Cyclin D1 in neuroblasts and low Cyclin D1 expression in all cell types in ganglioneuroma. This suggests an involvement of G(1)-regulating genes in neuronal differentiation processes which we further evaluated using RNA interference against Cyclin D1 and its kinase partners CDK4 and CDK6 in several neuroblastoma cell lines. The Cyclin D1 and CDK4 knockdown resulted in pRb pathway inhibition as shown by an almost complete disappearance of CDK4/CDK6-specific pRb phosphorylation, reduction of E2F transcriptional activity, and a decrease of Cyclin A protein levels. Phenotype analysis showed a significant reduction in cell proliferation, a G(1)-specific cell cycle arrest, and, moreover, an extensive neuronal differentiation. Affymetrix microarray profiling of small interfering RNA-treated cells revealed a shift in expression profile toward a neuronal phenotype. Several new potential downstream players are identified. We conclude that neuroblastoma functionally depend on overexpression of G(1)-regulating genes to maintain their undifferentiated phenotype.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Functional MYCN signature predicts outcome of neuroblastoma irrespective of MYCN amplification

Linda J. Valentijn; Jan Koster; Franciska Haneveld; Rachida Ait Aissa; Peter van Sluis; Marloes Broekmans; Jan J. Molenaar; Johan van Nes; Rogier Versteeg

Neuroblastoma is a pediatric tumor of the sympathetic nervous system. MYCN (V-myc myelocytomatosis viral-related oncogene, neuroblastoma derived [avian]) is amplified in 20% of neuroblastomas, and these tumors carry a poor prognosis. However, tumors without MYCN amplification also may have a poor outcome. Here, we identified downstream targets of MYCN by shRNA-mediated silencing MYCN in neuroblastoma cells. From these targets, 157 genes showed an expression profile correlating with MYCN mRNA levels in NB88, a series of 88 neuroblastoma tumors, and therefore represent in vivo relevant MYCN pathway genes. This 157-gene signature identified very poor prognosis tumors in NB88 and independent neuroblastoma cohorts and was more powerful than MYCN amplification or MYCN expression alone. Remarkably, this signature also identified poor outcome of a group of tumors without MYCN amplification. Most of these tumors have low MYCN mRNA levels but high nuclear MYCN protein levels, suggesting stabilization of MYCN at the protein level. One tumor has an MYC amplification and high MYC expression. Chip-on-chip analyses showed that most genes in this signature are directly regulated by MYCN. MYCN induces genes functioning in cell cycle and DNA repair while repressing neuronal differentiation genes. The functional MYCN-157 signature recognizes classical neuroblastoma with MYCN amplification, as well as a newly identified group marked by MYCN protein stabilization.


Nature Genetics | 2015

TERT rearrangements are frequent in neuroblastoma and identify aggressive tumors

Linda J. Valentijn; Jan Koster; Danny A. Zwijnenburg; Nancy E. Hasselt; Peter van Sluis; Richard Volckmann; Max M. van Noesel; Rani E. George; Godelieve A.M. Tytgat; Jan J. Molenaar; Rogier Versteeg

Whole-genome sequencing detected structural rearrangements of TERT in 17 of 75 high-stage neuroblastomas, with five cases resulting from chromothripsis. Rearrangements were associated with increased TERT expression and targeted regions immediately up- and downstream of TERT, positioning a super-enhancer close to the breakpoints in seven cases. TERT rearrangements (23%), ATRX deletions (11%) and MYCN amplifications (37%) identify three almost non-overlapping groups of high-stage neuroblastoma, each associated with very poor prognosis.


Genes, Chromosomes and Cancer | 2007

ArrayCGH-based classification of neuroblastoma into genomic subgroups†

Evi Michels; Jo Vandesompele; Katleen De Preter; Jasmien Hoebeeck; Joëlle Vermeulen; Alexander Schramm; Jan J. Molenaar; Björn Menten; Bárbara Marques; Raymond L. Stallings; Valérie Combaret; Christine Devalck; Anne De Paepe; Rogier Versteeg; Angelika Eggert; Genevieve Laureys; Nadine Van Roy; Frank Speleman

High‐resolution array comparative genomic hybridization (arrayCGH) profiling was performed on 75 primary tumors and 29 cell lines to gain further insight into the genetic heterogeneity of neuroblastoma and to refine genomic subclassification. Using a novel data‐mining strategy, three major and two minor genomic subclasses were delineated. Eighty‐three percent of tumors could be assigned to the three major genomic subclasses, corresponding to the three known clinically and biologically relevant subsets in neuroblastoma. The remaining subclasses represented (1) tumors with no/few copy number alterations or an atypical pattern of aberrations and (2) tumors with 11q13 amplification. Inspection of individual arrayCGH profiles showed that recurrent genomic imbalances were not exclusively associated with a specific subclass. Of particular notice were tumors with numerical imbalances typically observed in subtype 1 neuroblastoma, in association with genomic features of subtype 2A or 2B. A search for prognostically relevant genomic alterations disclosed 1q gain as a predictive marker for therapy failure within the group of subtype 2A and 2B tumors. In cell lines, a high incidence of 6q loss was observed, with a 3.87–5.32 Mb region of common loss within 6q25.1–6q25.2. Our study clearly illustrates the importance of genomic profiling in relation to tumor behavior in neuroblastoma. We propose that genome‐wide assessment of copy number alterations should ideally be included in the genetic workup of neuroblastoma. Further multicentric studies on large tumor series are warranted in order to improve therapeutic stratification in conjunction with other features such as age at diagnosis, tumor stage, and gene expression signatures.


Clinical Cancer Research | 2011

miRNA Expression Profiling Enables Risk Stratification in Archived and Fresh Neuroblastoma Tumor Samples

Katleen De Preter; Pieter Mestdagh; Joëlle Vermeulen; Fjoralba Zeka; Arlene Naranjo; Isabella Bray; Victoria Castel; Caifu Chen; Elżbieta Drożyńska; Angelika Eggert; Michael D. Hogarty; Ewa Izycka-Swieszewska; Wendy B. London; Rosa Noguera; Marta Piqueras; Kenneth Bryan; Benjamin Schowe; Peter van Sluis; Jan J. Molenaar; Alexander Schramm; Johannes H. Schulte; Raymond L. Stallings; Rogier Versteeg; Genevieve Laureys; Nadine Van Roy; Frank Speleman; Jo Vandesompele

Purpose: More accurate assessment of prognosis is important to further improve the choice of risk-related therapy in neuroblastoma (NB) patients. In this study, we aimed to establish and validate a prognostic miRNA signature for children with NB and tested it in both fresh frozen and archived formalin-fixed paraffin-embedded (FFPE) samples. Experimental Design: Four hundred-thirty human mature miRNAs were profiled in two patient subgroups with maximally divergent clinical courses. Univariate logistic regression analysis was used to select miRNAs correlating with NB patient survival. A 25-miRNA gene signature was built using 51 training samples, tested on 179 test samples, and validated on an independent set of 304 fresh frozen tumor samples and 75 archived FFPE samples. Results: The 25-miRNA signature significantly discriminates the test patients with respect to progression-free and overall survival (P < 0.0001), both in the overall population and in the cohort of high-risk patients. Multivariate analysis indicates that the miRNA signature is an independent predictor of patient survival after controlling for current risk factors. The results were confirmed in an external validation set. In contrast to a previously published mRNA classifier, the 25-miRNA signature was found to be predictive for patient survival in a set of 75 FFPE neuroblastoma samples. Conclusions: In this study, we present the largest NB miRNA expression study so far, including more than 500 NB patients. We established and validated a robust miRNA classifier, able to identify a cohort of high-risk NB patients at greater risk for adverse outcome using both fresh frozen and archived material. Clin Cancer Res; 17(24); 7684–92. ©2011 AACR.


Genes, Chromosomes and Cancer | 2003

Rearrangements and increased expression of cyclin D1 (CCND1) in neuroblastoma.

Jan J. Molenaar; Peter van Sluis; Kathy Boon; Rogier Versteeg; Huib N. Caron

Cyclin D1 regulates G1 cell cycle progression by controlling the phosphorylation of the retinoblastoma protein. This pathway is frequently deregulated in many malignancies. In neuroblastoma, however, no consistent G1 cell cycle checkpoint aberrations have been found. We examined the possible deregulation of cyclin D1 (CCND1) in this tumor. mRNA expression profiles of neuroblastoma generated by SAGE (Serial Analysis of Gene Expression) revealed a high expression of CCND1 in a subset of neuroblastoma cell lines and tumors. The CCND1 expression level can be 0.3% of the total cellular mRNA. Northern blot analysis of CCND1 expression showed a relative overexpression in 16 of 23 neuroblastoma cell lines and 10 of 15 tumor samples. In the majority of cases, the high CCND1 mRNA levels also led to high CCND1 protein levels. In the search for mechanisms causing this relative overexpression, we screened for amplifications and rearrangements of CCND1. Five amplifications were found in 202 neuroblastoma tumors and cell lines. Analysis of the 3′‐UTR of CCND1 showed a rearrangement in 1 of 96 tumors. These clonal aberrations of CCND1 together with the high expression suggest a role for deregulated CCND1 activity in neuroblastoma tumorigenesis.

Collaboration


Dive into the Jan J. Molenaar's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan Koster

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Huib N. Caron

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Linda Schild

University of Amsterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge