Network


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

Hotspot


Dive into the research topics where Alexandra Belayew is active.

Publication


Featured researches published by Alexandra Belayew.


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.


Gene | 1999

NUCLEOTIDE SEQUENCE OF THE PARTIALLY DELETED D4Z4 LOCUS IN A PATIENT WITH FSHD IDENTIFIES A PUTATIVE GENE WITHIN EACH 3.3 KB ELEMENT

J. Gabriels; Marie-Claire Beckers; Hao Ding; A. De Vriese; Stephane Plaisance; S.M. van der Maarel; George W. Padberg; Rune R. Frants; Jane E. Hewitt; D. Collen; Alexandra Belayew

Facioscapulohumeral muscular dystrophy (FSHD) is linked to the polymorphic D4Z4 locus on chromosome 4q35. In non-affected individuals, this locus comprises 10-100 tandem copies of members of the 3.3kb dispersed repeat family. Deletions leaving 1-8 such repeats have been associated with FSHD, for which no candidate gene has been identified. We have determined the complete nucleotide sequence of a 13.5kb EcoRI genomic fragment comprising the only two 3.3kb elements left in the affected D4Z4 locus of a patient with FSHD. Sequence analyses demonstrated that the two 3.3kb repeats were identical. They contain a putative promoter that was not previously detected, with a TACAA instead of a TATAA box, and a GC box. Transient expression of a luciferase reporter gene fused to 191bp of this promoter, demonstrated strong activity in transfected human rhabdomyosarcoma TE671 cells that was affected by mutations in the TACAA or GC box. In addition, these 3.3kb repeats include an open reading frame (ORF) starting 149bp downstream from the TACAA box and encoding a 391 residue protein with two homeodomains (DUX4). In-vitro transcription/translation of the ORF in a rabbit reticulocyte lysate yielded two (35)S Cys/ (35)S Met labeled products with apparent molecular weights of 38 and 75kDa on SDS-PAGE, corresponding to the DUX4 monomer and dimer, respectively. In conclusion, we propose that each of the 3.3kb elements in the partially deleted D4Z4 locus could include a DUX4 gene encoding a double homeodomain protein.


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.


The EMBO Journal | 1984

Isolation and characterization of the human prolactin gene

Anh T Truong; Colette Duez; Alexandra Belayew; André Renard; Raymond Pictet; Greame I Bell; Joseph Martial

Prolactin (PRL) and growth hormone (GH) genes derive from a common ancestor and still share some sequence homologies. Their expression in the pituitary gland is regulated in opposite directions by most of the many hormones acting on them. This provides an interesting system to study sequences involved in gene expression. Using a human PRL cDNA clone as a probe, we screened a human genomic DNA library in lambda phage and isolated a single recombinant comprising the whole hPRL gene. It was characterized by restriction endonuclease mapping and cDNA hybridization, by DNA heteroduplex analysis and by nucleotide sequencing. The hPRL gene is present as a single copy per haploid genome, is approximately 10 kb long and contains four introns, three of which interrupt the coding sequence at the same locations as in the known GH and PRL genes. The origin of transcription was determined by S1 mapping on prolactinoma mRNAs. The search for direct and inverted repeats, as well as dyad symmetries was carried out in the 900‐bp sequenced in the 5′‐flanking region. Sequence homologies between hPRL, hGH and rPRL were derived from computer drawn matrices for these upstream regions.


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.


Molecular and Cellular Biology | 1982

Genetic analysis of alpha-fetoprotein synthesis in mice.

Alexandra Belayew; S M Tilghman

The differential induction of alpha-fetoprotein (AFP) mRNA during liver regeneration in three inbred strains of mice was examined to determine the genetic and molecular bases for the differences in protein production. BALB/cJ, C3H/He, and C57BL/6 mice, previously identified as high, intermediate, and low AFP producers, respectively, were used. Liver AFP mRNA concentrations during normal development and after carbon tetrachloride administration were measured and shown to correlate exactly with the serum protein concentrations. By performing a series of genetic crosses, we identified two unlinked genetic loci that acted independently to affect the inducibility of AFP mRNA. The raf gene, previously identified by Olsson et al. (J. Exp. Med. 145:819-827, 1977), determines the adult basal level of AFP mRNA, and the Rif gene affects its inducibility during regeneration. By using a polymorphic restriction endonuclease site within the albumin-AFP structural gene region, we show that neither regulatory gene is closely linked to the structural genes. In addition, neither gene affects the concentration of albumin mRNA during development or liver regeneration.


Molecular and Cellular Endocrinology | 1991

Multihormonal regulation of the human prolactin gene expression from 5000 bp of its upstream sequence

Monique Berwaer; Philippe Monget; Bernard Peers; M. Mathy-Hartert; Eric Bellefroid; Julian R. E. Davis; Alexandra Belayew; Joseph Martial

We have cloned DNA sequences extending up to 6000 bp upstream from the first exon of the human prolactin (hPRL) gene. 5000 bp of these upstream sequences were fused to a CAT reporter gene and shown to provide tissue-specific transient expression in rat pituitary GH3 cells. Multihormonal response was found in this transient expression assay, leading to significant 2- to 5-fold induction by addition of 8-chlorophenylthio-cyclic AMP, thyrotropin-releasing hormone, epidermal growth factor, basic fibroblast growth factor, phorbol myristate acetate, a calcium channel agonist (Bay K-8644) and triiodothyronine. A 3-fold inhibition was observed in the presence of the glucocorticoid agonist dexamethasone. The sequence of the hPRL promoter was determined up to coordinate -3470. Computer similarity search between the rat and human sequences showed two highly conserved regions corresponding to the proximal and distal tissue specific enhancers described in both PRL promoters.


Experimental Neurology | 2008

DUX4c, an FSHD candidate gene, interferes with myogenic regulators and abolishes myoblast differentiation.

Darko Bosnakovski; Sarah Lamb; Tugba Simsek; Zhaohui Xu; Alexandra Belayew; Rita C.R. Perlingeiro; Michael Kyba

Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant neuromuscular disease. It maps to the D4Z4 repeat array at 4q35, and correlates with a repeat contraction which derepresses transcription of local genes. Which, if any, of these genes is pathogenic to muscle, and through what molecular mechanism is unknown. The present study investigates the function of one candidate gene, DUX4c, encoded by a truncated inverted D4Z4 element located 42 kb proximal to the D4Z4 repeats. Using a gain of function approach we tested DUX4c for toxicity and effects on differentiation in C2C12 myoblasts. DUX4c-expressing myoblasts appear morphologically normal but have reduced expression of myogenic regulators, including MyoD and Myf5. Consistent with this, DUX4c-expressing myoblasts are unable to differentiate into myotubes. Furthermore, overexpression of Myf5 or MyoD rescued myoblast differentiation, suggesting that reductions in expression of these regulators are the relevant DUX4c-induced physiological changes that inhibit differentiation. Our results suggest that upregulation of DUX4c can have a deleterious effect on muscle homeostasis and regeneration, and point to a possible role for DUX4c in the pathology of FSHD.

Collaboration


Dive into the Alexandra Belayew's collaboration.

Top Co-Authors

Avatar

Frédérique Coppée

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexandra Tassin

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Desire Collen

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Céline Vanderplanck

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Catherine Charles

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Frank Bulens

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge