Network


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

Hotspot


Dive into the research topics where Sophie Remacle is active.

Publication


Featured researches published by Sophie Remacle.


Nucleic Acids Research | 2008

An ultraconserved Hox–Pbx responsive element resides in the coding sequence of Hoxa2 and is active in rhombomere 4

Xavier Lampe; Omar Abdel O.A. Samad; Allan Guiguen; Christelle Matis; Sophie Remacle; Jacques J. Picard; Filippo M. Rijli; René Rezsohazy

The Hoxa2 gene has a fundamental role in vertebrate craniofacial and hindbrain patterning. Segmental control of Hoxa2 expression is crucial to its function and several studies have highlighted transcriptional regulatory elements governing its activity in distinct rhombomeres. Here, we identify a putative Hox–Pbx responsive cis-regulatory sequence, which resides in the coding sequence of Hoxa2 and is an important component of Hoxa2 regulation in rhombomere (r) 4. By using cell transfection and chromatin immunoprecipitation (ChIP) assays, we show that this regulatory sequence is responsive to paralogue group 1 and 2 Hox proteins and to their Pbx co-factors. Importantly, we also show that the Hox–Pbx element cooperates with a previously reported Hoxa2 r4 intronic enhancer and that its integrity is required to drive specific reporter gene expression in r4 upon electroporation in the chick embryo hindbrain. Thus, both intronic as well as exonic regulatory sequences are involved in Hoxa2 segmental regulation in the developing r4. Finally, we found that the Hox–Pbx exonic element is embedded in a larger 205-bp long ultraconserved genomic element (UCE) shared by all vertebrate genomes. In this respect, our data further support the idea that extreme conservation of UCE sequences may be the result of multiple superposed functional and evolutionary constraints.


American Journal of Human Genetics | 2007

The Nuclear Factor κB–Activator Gene PLEKHG5 Is Mutated in a Form of Autosomal Recessive Lower Motor Neuron Disease with Childhood Onset

Isabelle Maystadt; René Rezsohazy; Martine Barkats; Sandra Duque; Pascal Vannuffel; Sophie Remacle; Barbara Lambert; Mustapha Najimi; Etienne Sokal; Arnold Munnich; Louis Viollet; Christine Verellen-Dumoulin

Lower motor neuron diseases (LMNDs) include a large spectrum of clinically and genetically heterogeneous disorders. Studying a large inbred African family, we recently described a novel autosomal recessive LMND variant characterized by childhood onset, generalized muscle involvement, and severe outcome, and we mapped the disease gene to a 3.9-cM interval on chromosome 1p36. We identified a homozygous missense mutation (c.1940 T-->C [p.647 Phe-->Ser]) of the Pleckstrin homology domain-containing, family G member 5 gene, PLEKHG5. In transiently transfected HEK293 and MCF10A cell lines, we found that wild-type PLEKHG5 activated the nuclear factor kappa B (NF kappa B) signaling pathway and that both the stability and the intracellular location of mutant PLEKHG5 protein were altered, severely impairing the NF kappa B transduction pathway. Moreover, aggregates were observed in transiently transfected NSC34 murine motor neurons overexpressing the mutant PLEKHG5 protein. Both loss of PLEKHG5 function and aggregate formation may contribute to neurotoxicity in this novel form of LMND.


Molecular and Cellular Biology | 2004

Loss of Function but No Gain of Function Caused by Amino Acid Substitutions in the Hexapeptide of Hoxa1 In Vivo

Sophie Remacle; Leïla Abbas; Olivier De Backer; Nathalie Pacico; Anthony Gavalas; Françoise Gofflot; Jacques Picard; René Rezsohazy

ABSTRACT Homeodomain containing transcription factors of the Hox family play critical roles in patterning the anteroposterior embryonic body axis, as well as in controlling several steps of organogenesis. Several Hox proteins have been shown to cooperate with members of the Pbx family for the recognition and activation of identified target enhancers. Hox proteins contact Pbx via a conserved hexapeptide motif. Previous biochemical studies provided evidence that critical amino acid substitutions in the hexapeptide sequence of Hoxa1 abolish its interaction with Pbx. As a result, these substitutions also abolish Hoxa1 activity on known target enhancers in cellular models, suggesting that Hoxa1 activity relies on its capacity to interact with Pbx. Here, we show that mice with mutations in the Hoxa1 hexapeptide display hindbrain, cranial nerve, and skeletal defects highly reminiscent of those reported for the Hoxa1 loss of function. Since similar hexapeptide mutations in the mouse Hoxb8 and the Drosophila AbdA proteins result in activity modulation and gain of function, our data demonstrate that the functional importance of the hexapeptide in vivo differs according to the Hox proteins.


PLOS Biology | 2012

Hox Proteins Display a Common and Ancestral Ability to Diversify Their Interaction Mode with the PBC Class Cofactors

Bruno Hudry; Sophie Remacle; Marie-Claire Delfini; René Rezsohazy; Yacine Graba; Samir Merabet

Hox protein function during development and evolution relies on conserved multiple interaction modes with cofactors of the PBC and Meis families.


BMC Developmental Biology | 2012

Protein interactions of the transcription factor Hoxa1

Barbara Lambert; Julie Vandeputte; Sophie Remacle; Isabelle Bergiers; Nicolas Simonis; Jean-Claude Twizere; Marc Vidal; René Rezsohazy

BackgroundHox proteins are transcription factors involved in crucial processes during animal development. Their mode of action remains scantily documented. While other families of transcription factors, like Smad or Stat, are known cell signaling transducers, such a function has never been squarely addressed for Hox proteins.ResultsTo investigate the mode of action of mammalian Hoxa1, we characterized its interactome by a systematic yeast two-hybrid screening against ~12,200 ORF-derived polypeptides. Fifty nine interactors were identified of which 45 could be confirmed by affinity co-purification in animal cell lines. Many Hoxa1 interactors are proteins involved in cell-signaling transduction, cell adhesion and vesicular trafficking. Forty-one interactions were detectable in live cells by Bimolecular Fluorescence Complementation which revealed distinctive intracellular patterns for these interactions consistent with the selective recruitment of Hoxa1 by subgroups of partner proteins at vesicular, cytoplasmic or nuclear compartments.ConclusionsThe characterization of the Hoxa1 interactome presented here suggests unexplored roles for Hox proteins in cell-to-cell communication and cell physiology.


PLOS ONE | 2011

A Retinoic Acid Responsive Hoxa3 Transgene Expressed in Embryonic Pharyngeal Endoderm, Cardiac Neural Crest and a Subdomain of the Second Heart Field

Nata Y.S.-G Diman; Sophie Remacle; Nicolas Bertrand; Jacques J. Picard; Stéphane Zaffran; René Rezsohazy

A transgenic mouse line harbouring a β-galacdosidase reporter gene controlled by the proximal 2 kb promoter of Hoxa3 was previously generated to investigate the regulatory cues governing Hoxa3 expression in the mouse. Examination of transgenic embryos from embryonic day (E) 8.0 to E15.5 revealed regionally restricted reporter activity in the developing heart. Indeed, transgene expression specifically delineated cells from three distinct lineages: a subpopulation of the second heart field contributing to outflow tract myocardium, the cardiac neural crest cells and the pharyngeal endoderm. Manipulation of the Retinoic Acid (RA) signaling pathway showed that RA is required for correct expression of the transgene. Therefore, this transgenic line may serve as a cardiosensor line of particular interest for further analysis of outflow tract development.


Developmental Dynamics | 2007

Identification of Lmo1 as Part of a Hox- Dependent Regulatory Network for Hindbrain Patterning

Christelle Matis; Franck Oury; Sophie Remacle; Xavier Lampe; Françoise Gofflot; Jacques J. Picard; Filippo M. Rijli; René Rezsohazy

The embryonic functions of Hox proteins have been extensively investigated in several animal phyla. These transcription factors act as selectors of developmental programmes, to govern the morphogenesis of multiple structures and organs. However, despite the variety of morphogenetic processes Hox proteins are involved in, only a limited set of their target genes has been identified so far. To find additional targets, we used a strategy based upon the simultaneous overexpression of Hoxa2 and its cofactors Pbx1 and Prep in a cellular model. Among genes whose expression was upregulated, we identified LMO1, which codes for an intertwining LIM‐only factor involved in protein–DNA oligomeric complexes. By analysing its expression in Hox knockout mice, we show that Lmo1 is differentially regulated by Hoxa2 and Hoxb2, in specific columns of hindbrain neuronal progenitors. These results suggest that Lmo1 takes part in a Hox paralogue 2–dependent network regulating anteroposterior and dorsoventral hindbrain patterning. Developmental Dynamics 236:2675–2684, 2007.


Journal of Cellular Biochemistry | 2010

Pentapeptide insertion mutagenesis of the Hoxa1 protein: Mapping of transcription activation and DNA‐binding regulatory domains

Barbara Lambert; Julie Vandeputte; Pierre-Marie Desmet; Bernard Hallet; Sophie Remacle; René Rezsohazy

The mode of action of Hoxa1, like that of most Hox proteins, remains poorly characterized. In an effort to identify functional determinants contributing to the activity of Hoxa1 as a transcription factor, we generated 18 pentapeptide insertion mutants of the Hoxa1 protein and we assayed them in transfected cells for their activity on target enhancers from the EphA2 and Hoxb1 genes known to respond to Hoxa1 in the developing hindbrain. Only four mutants displayed a complete loss‐of‐function. Three of them contained an insertion in the homeodomain of Hoxa1, whereas the fourth loss‐of‐function mutant harbored an insertion in the very N‐terminal end of the protein. Transcription activation assays in yeast further revealed that the integrity of both the N‐terminal end and homeodomain is required for Hoxa1‐mediated transcriptional activation. Furthermore, an insertion in the serine–threonine–proline rich C‐terminal extremity of Hoxa1 induced an increase in activity in mammalian cells as well as in the yeast assay. The C‐terminal extremity thus modulates the transcriptional activation capacity of the protein. Finally, electrophoretic mobility shift assays revealed that the N‐terminal extremity of the protein also exerts a modulatory influence on DNA binding by Hoxa1–Pbx1a heterodimers. J. Cell. Biochem. 110: 484–496, 2010.


Animal Reproduction Science | 2008

Kinetics of fertilization and development, and sex ratio of bovine embryos produced using the semen of different bulls.

Mazen Alomar; Hélène Tasiaux; Sophie Remacle; Fabienne George; Delphine Paul; Isabelle Donnay


Nucleic Acids Research | 2002

Changing homeodomain residues 2 and 3 of Hoxa1 alters its activity in a cell-type and enhancer dependent manner

Sophie Remacle; Chloë Shaw-Jackson; Christelle Matis; Xavier Lampe; Jacques J. Picard; René Rezsohazy

Collaboration


Dive into the Sophie Remacle's collaboration.

Top Co-Authors

Avatar

René Rezsohazy

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Jacques J. Picard

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Barbara Lambert

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Christelle Matis

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Françoise Gofflot

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Xavier Lampe

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Christine Verellen-Dumoulin

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Delphine Paul

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Fabienne George

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Filippo M. Rijli

Université catholique de Louvain

View shared research outputs
Researchain Logo
Decentralizing Knowledge