Network


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

Hotspot


Dive into the research topics where Frédérique Coppée is active.

Publication


Featured researches published by Frédérique Coppée.


Proceedings of the National Academy of Sciences of the United States of America | 2007

DUX4, a candidate gene of facioscapulohumeral muscular dystrophy, encodes a transcriptional activator of PITX1

Manjusha Dixit; Eugénie Ansseau; Alexandra Tassin; Sara T. Winokur; Rongye Shi; Hong Qian; Sébastien Sauvage; Christel Matteotti; Anne Marie Van Acker; Oberdan Leo; Denise A. Figlewicz; Marietta Barro; Dalila Laoudj-Chenivesse; Alexandra Belayew; Frédérique Coppée; Yi-Wen Chen

Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant disorder linked to contractions of the D4Z4 repeat array in the subtelomeric region of chromosome 4q. By comparing genome-wide gene expression data from muscle biopsies of patients with FSHD to those of 11 other neuromuscular disorders, paired-like homeodomain transcription factor 1 (PITX1) was found specifically up-regulated in patients with FSHD. In addition, we showed that the double homeobox 4 gene (DUX4) that maps within the D4Z4 repeat unit was up-regulated in patient myoblasts at both mRNA and protein level. We further showed that the DUX4 protein could activate transient expression of a luciferase reporter gene fused to the Pitx1 promoter as well as the endogenous Pitx1 gene in transfected C2C12 cells. In EMSAs, DUX4 specifically interacted with a 30-bp sequence 5′-CGGATGCTGTCTTCTAATTAGTTTGGACCC-3′ in the Pitx1 promoter. Mutations of the TAAT core affected Pitx1-LUC activation in C2C12 cells and DUX4 binding in vitro. Our results suggest that up-regulation of both DUX4 and PITX1 in FSHD muscles may play critical roles in the molecular mechanisms of the disease.


Neuromuscular Disorders | 2007

The DUX4 gene at the FSHD1A locus encodes a pro-apoptotic protein.

Valeria Kowaljow; Aline Marcowycz; Eugénie Ansseau; Cecilia Conde; Sébastien Sauvage; Christel Matteotti; Cristina Arias; E. Daniel Corona; Nicolás G. Nuñez; Oberdan Leo; Ruddy Wattiez; Denise A. Figlewicz; Dalila Laoudj-Chenivesse; Alexandra Belayew; Frédérique Coppée; Alberto Luis Rosa

Facioscapulohumeral muscular dystrophy (FSHD) patients carry contractions of the D4Z4-tandem repeat array on chromosome 4q35. Decrease in D4Z4 copy number is thought to alter a chromatin structure and activate expression of neighboring genes. D4Z4 contains a putative double-homeobox gene called DUX4. We identified DUX4 mRNAs in cells transfected with genomic fragments containing the DUX4 gene. Using RT-PCR we also recognized expressed DUX4 mRNAs in primary FSHD myoblasts. Polyclonal antibodies raised against specific DUX4 peptides detected the DUX4 protein in cells transfected with D4Z4 elements. DUX4 localizes in the nucleus of cells transfected with CMV-DUX4 expression vectors. A DUX4-related protein is endogenously expressed in nuclei of adult and fetal human rhabdomyosarcoma cell lines. Overexpression of DUX4 induces cell death, induces caspase 3/7 activity and alters emerin distribution at the nuclear envelope. We propose that DUX4-mediated cell death contributes to the pathogenic pathway in FSHD.


The EMBO Journal | 2008

An isogenetic myoblast expression screen identifies DUX4‐mediated FSHD‐associated molecular pathologies

Darko Bosnakovski; Zhaohui Xu; Eun Ji Gang; Cristi L. Galindo; Mingju Liu; Tugba Simsek; Harold R. Garner; Siamak Agha-Mohammadi; Alexandra Tassin; Frédérique Coppée; Alexandra Belayew; Rita R.C. Perlingeiro; Michael Kyba

Facioscapulohumeral muscular dystrophy (FSHD) is caused by an unusual deletion with neomorphic activity. This deletion derepresses genes in cis; however which candidate gene causes the FSHD phenotype, and through what mechanism, is unknown. We describe a novel genetic tool, inducible cassette exchange, enabling rapid generation of isogenetically modified cells with conditional and variable transgene expression. We compare the effects of expressing variable levels of each FSHD candidate gene on myoblasts. This screen identified only one gene with overt toxicity: DUX4 (double homeobox, chromosome 4), a protein with two homeodomains, each similar in sequence to Pax3 and Pax7. DUX4 expression recapitulates key features of the FSHD molecular phenotype, including repression of MyoD and its target genes, diminished myogenic differentiation, repression of glutathione redox pathway components, and sensitivity to oxidative stress. We further demonstrate competition between DUX4 and Pax3/Pax7: when either Pax3 or Pax7 is expressed at high levels, DUX4 is no longer toxic. We propose a hypothesis for FSHD in which DUX4 expression interferes with Pax7 in satellite cells, and inappropriately regulates Pax targets, including myogenic regulatory factors, during regeneration.


Annals of Neurology | 2011

DUX4, a candidate gene for facioscapulohumeral muscular dystrophy, causes p53-dependent myopathy in vivo

Lindsay M. Wallace; Sara E. Garwick; Wenyan Mei; Alexandra Belayew; Frédérique Coppée; Katherine J. Ladner; Denis C. Guttridge; Jing Yang; Scott Q. Harper

Facioscapulohumeral muscular dystrophy (FSHD) is associated with D4Z4 repeat contraction on human chromosome 4q35. This genetic lesion does not result in complete loss or mutation of any gene. Consequently, the pathogenic mechanisms underlying FSHD have been difficult to discern. In leading FSHD pathogenesis models, D4Z4 contractions are proposed to cause epigenetic changes, which ultimately increase expression of genes with myopathic potential. Although no gene has been conclusively linked to FSHD development, recent evidence supports a role for the D4Z4‐encoded DUX4 gene in FSHD. In this study, our objective was to test the in vivo myopathic potential of DUX4.


PLOS ONE | 2011

The FSHD Atrophic Myotube Phenotype Is Caused by DUX4 Expression

Céline Vanderplanck; Eugénie Ansseau; Sébastien Charron; Nadia Stricwant; Alexandra Tassin; Dalila Laoudj-Chenivesse; S.D. Wilton; Frédérique Coppée; Alexandra Belayew

Background Facioscapulohumeral muscular dystrophy (FSHD) is linked to deletions in 4q35 within the D4Z4 repeat array in which we identified the double homeobox 4 (DUX4) gene. We found stable DUX4 mRNAs only derived from the most distal D4Z4 unit and unexpectedly extended to the flanking pLAM region that provided an intron and a polyadenylation signal. DUX4 encodes a transcription factor expressed in FSHD but not control primary myoblasts or muscle biopsies. The DUX4 protein initiates a large transcription deregulation cascade leading to muscle atrophy and oxidative stress, which are FSHD key features. Methodology/Principal Findings We now show that transfection of myoblasts with a DUX4 expression vector leads to atrophic myotube formation associated with the induction of E3 ubiquitin ligases (MuRF1 and Atrogin1/MAFbx) typical of muscle atrophy. DUX4 induces expression of downstream targets deregulated in FSHD such as mu-crystallin and TP53. We developed specific siRNAs and antisense oligonucleotides (AOs) targeting the DUX4 mRNA. Addition of these antisense agents to primary FSHD myoblast cultures suppressed DUX4 protein expression and affected expression of the above-mentioned markers. Conclusions/Significance These results constitute a proof of concept for the development of therapeutic approaches for FSHD targeting DUX4 expression.


Journal of Cellular and Molecular Medicine | 2013

DUX4 expression in FSHD muscle cells: How could such a rare protein cause a myopathy?

Alexandra Tassin; Dalila Laoudj-Chenivesse; Céline Vanderplanck; Marietta Barro; Sébastien Charron; Eugénie Ansseau; Yi-Wen Chen; J. Mercier; Frédérique Coppée; Alexandra Belayew

Facioscapulohumeral muscular dystrophy (FSHD) is one of the most frequent hereditary muscle disorders. It is linked to contractions of the D4Z4 repeat array in 4q35. We have characterized the double homeobox 4 (DUX4) gene in D4Z4 and its mRNA transcribed from the distal D4Z4 unit to a polyadenylation signal in the flanking pLAM region. It encodes a transcription factor expressed in FSHD but not healthy muscle cells which initiates a gene deregulation cascade causing differentiation defects, muscle atrophy and oxidative stress. PITX1 was the first identified DUX4 target and encodes a transcription factor involved in muscle atrophy. DUX4 was found expressed in only 1/1000 FSHD myoblasts. We have now shown it was induced upon differentiation and detected in about 1/200 myotube nuclei. The DUX4 and PITX1 proteins presented staining gradients in consecutive myonuclei which suggested a diffusion as known for other muscle nuclear proteins. Both protein half‐lifes were regulated by the ubiquitin‐proteasome pathway. In addition, we could immunodetect the DUX4 protein in FSHD muscle extracts. As a model, we propose the DUX4 gene is stochastically activated in a small number of FSHD myonuclei. The resulting mRNAs are translated in the cytoplasm around an activated nucleus and the DUX4 proteins diffuse to adjacent nuclei where they activate target genes such as PITX1. The PITX1 protein can further diffuse to additional myonuclei and expand the transcriptional deregulation cascade initiated by DUX4. Together the diffusion and the deregulation cascade would explain how a rare protein could cause the muscle defects observed in FSHD.


Human Genetics | 2012

Facioscapulohumeral muscular dystrophy (FSHD): an enigma unravelled?

Mark Richards; Frédérique Coppée; Nicholas Stuart Tudor Thomas; Alexandra Belayew; Meena Upadhyaya

Facioscapulohumeral muscular dystrophy (FSHD) is the third most common muscular dystrophy after the dystrophinopathies and myotonic dystrophy and is associated with a typical pattern of muscle weakness. Most patients with FSHD carry a large deletion in the polymorphic D4Z4 macrosatellite repeat array at 4q35 and present with 1–10 repeats whereas non-affected individuals possess 11–150 repeats. An almost identical repeat array is present at 10q26 and the high sequence identity between these two arrays can cause difficulties in molecular diagnosis. Each 3.3-kb D4Z4 unit contains a DUX4 (double homeobox 4) gene that, among others, is activated upon contraction of the 4q35 repeat array due to the induction of chromatin remodelling of the 4qter region. A number of 4q subtelomeric sequence variants are now recognised, although FSHD only occurs in association with three ‘permissive’ haplotypes, each of which is associated with a polyadenylation signal located immediately distal of the last D4Z4 unit. The resulting poly-A tail appears to stabilise DUX4 mRNAs transcribed from this most distal D4Z4 unit in FSHD muscle cells. Synthesis of both the DUX4 transcripts and protein in FSHD muscle cells induces significant cell toxicity. DUX4 is a transcription factor that may target several genes which results in a deregulation cascade which inhibits myogenesis, sensitises cells to oxidative stress and induces muscle atrophy, thus recapitulating many of the key molecular features of FSHD.


Journal of Biomolecular Screening | 2006

A new peptidic vector for molecular imaging of apoptosis, identified by phage display technology.

Catherine Laumonier; Jérôme Segers; Sophie Laurent; Alain Michel; Frédérique Coppée; Alexandra Belayew; Luce Vander Elst; Robert N. Muller

Phosphatidylserine (PS) exposure on the cell surface is an early marker of apoptosis. To select PS binding peptides as vectors of contrast agents to image apoptosis, a phage library has been exposed to perfused mouse livers. Phages not retained on control livers during the first perfusions were used for selections on apoptotic livers in a second series of perfusions. Four selected phages were further evaluated for binding to PS-coated enzyme-linked immunosorbent assay (ELISA) plates. They presented an apparent affinity constant (Ka app) for PS ranging from 6.08 × 1010 M to 1.62 × 1011M. These phages did not bind to phosphatidylcholine, and competition with annexin V confirmed their specific interaction with PS. The phage with the highest affinity-bound PS in ELISA with a Ka app = (1.6 ± 0.2) × 1011M. It carried the TLVSSL peptide that was synthesized. Specific competition with annexin V and with the synthetic peptide was performed and confirms the specificity of the interaction. (Journal of Biomolecular Screening 2006:537-545)


PLOS ONE | 2009

DUX4c Is Up-Regulated in FSHD. It Induces the MYF5 Protein and Human Myoblast Proliferation

Eugénie Ansseau; Dalila Laoudj-Chenivesse; Aline Marcowycz; Alexandra Tassin; Céline Vanderplanck; Sébastien Sauvage; Marietta Barro; Isabelle Mahieu; Axelle Leroy; India Leclercq; Véronique Mainfroid; Denise A. Figlewicz; Vincent Mouly; Gillian Butler-Browne; Alexandra Belayew; Frédérique Coppée

Facioscapulohumeral muscular dystrophy (FSHD) is a dominant disease linked to contractions of the D4Z4 repeat array in 4q35. We have previously identified a double homeobox gene (DUX4) within each D4Z4 unit that encodes a transcription factor expressed in FSHD but not control myoblasts. DUX4 and its target genes contribute to the global dysregulation of gene expression observed in FSHD. We have now characterized the homologous DUX4c gene mapped 42 kb centromeric of the D4Z4 repeat array. It encodes a 47-kDa protein with a double homeodomain identical to DUX4 but divergent in the carboxyl-terminal region. DUX4c was detected in primary myoblast extracts by Western blot with a specific antiserum, and was induced upon differentiation. The protein was increased about 2-fold in FSHD versus control myotubes but reached 2-10-fold induction in FSHD muscle biopsies. We have shown by Western blot and by a DNA-binding assay that DUX4c over-expression induced the MYF5 myogenic regulator and its DNA-binding activity. DUX4c might stabilize the MYF5 protein as we detected their interaction by co-immunoprecipitation. In keeping with the known role of Myf5 in myoblast accumulation during mouse muscle regeneration DUX4c over-expression activated proliferation of human primary myoblasts and inhibited their differentiation. Altogether, these results suggested that DUX4c could be involved in muscle regeneration and that changes in its expression could contribute to the FSHD pathology.


Clinical Endocrinology | 1995

Large goitre as a maladaptation to iodine deficiency

Jacques Emile Dumont; André M. Ermans; Carine Maenhaut; Frédérique Coppée; John B. Stanbury

Goitre is prevalent in areas of iodine deficiency. A number of years ago one of us proposed that goitre is an adaptation to this deficiency (Stanbury et al., 1954), but this concept has been generalized and overinterpreted. The true adaptation to iodine deficiency is an increase in iodide clearance which is a consequence of higher blood flow and greatly enhaliced trapping of iodide. It was implicit in our concept that thyroid hyperplasia might be a means to that end and it is this latter extrapolation that we wish to analyse and refine. First, endemic goitre is often large and filled with colloid. Such goitres, because of insufficient iodination of thyroglobulin and leakage of iodide, aggravate the iodine deficiency. Secondly, there are several examples, both human and experimental, of excellent adaptation by higher trapping of iodide without significant thyroid enlargement. In fact, the ideally adapted thyroid would grow by a factor of no more than 2 (i.e. a small goitre) and comprise small hyperplastic follicles. The biochemistry of thyroglobulin synthesis, iodide trapping and organification, and thyroid hormone secretion may explain disordered and useless overgrowth of the well established endemic goitre, which in many aspects is maladapted.

Collaboration


Dive into the Frédérique Coppée's collaboration.

Top Co-Authors

Avatar

Alexandra Belayew

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Eugénie Ansseau

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Alexandra Tassin

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Céline Vanderplanck

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Catherine Ledent

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Jacques Emile Dumont

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Armelle Wauters

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Céline Lancelot

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Sébastien Charron

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge