Network


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

Hotspot


Dive into the research topics where Nathalie Allaman-Pillet is active.

Publication


Featured researches published by Nathalie Allaman-Pillet.


The EMBO Journal | 2006

A unique set of SH3-SH3 interactions controls IB1 homodimerization.

Ole Kristensen; Sylvie Guenat; Imran Dar; Nathalie Allaman-Pillet; Amar Abderrahmani; Mourad Ferdaoussi; Raphaël Roduit; Fabienne Maurer; Jacques S. Beckmann; Jette S. Kastrup; Michael Gajhede; Christophe Bonny

Islet‐brain 1 (IB1 or JIP‐1) is a scaffold protein that interacts with components of the c‐Jun N‐terminal kinase (JNK) signal‐transduction pathway. IB1 is expressed at high levels in neurons and in pancreatic β‐cells, where it controls expression of several insulin‐secretory components and secretion. IB1 has been shown to homodimerize, but neither the molecular mechanisms nor the function of dimerization have yet been characterized. Here, we show that IB1 homodimerizes through a novel and unique set of Src homology 3 (SH3)–SH3 interactions. X‐ray crystallography studies show that the dimer interface covers a region usually engaged in PxxP‐mediated ligand recognition, even though the IB1 SH3 domain lacks this motif. The highly stable IB1 homodimer can be significantly destabilized in vitro by three individual point mutations directed against key residues involved in dimerization. Each mutation reduces IB1‐dependent basal JNK activity in 293T cells. Impaired dimerization also results in a reduction in glucose transporter type 2 expression and in glucose‐dependent insulin secretion in pancreatic β‐cells. Taken together, these results indicate that IB1 homodimerization through its SH3 domain has pleiotropic effects including regulation of the insulin secretion process.


Journal of Biomolecular Screening | 2006

Homogeneous and Nonradioactive High-Throughput Screening Platform for the Characterization of Kinase Inhibitors in Cell Lysates

Sylvie Guenat; Nathalie Rouleau; Christelle Bielmann; Julie Bédard; Fabienne Maurer; Nathalie Allaman-Pillet; Pascal Nicod; Martina Bielefeld-Sévigny; Jacques S. Beckmann; Christophe Bonny; Roger Bossé; Raphaël Roduit

Protein kinases are directly implicated in many human diseases; therefore, kinase inhibitors show great promises as new therapeutic drugs. In an effort to facilitate the screening and the characterization of kinase inhibitors, a novel application of the AlphaScreen technology was developed to monitor JNK activity from (1) purified kinase preparations and (2) endogenous kinase from cell lysates preactivated with different cytokines. The authors confirmed that both adenosine triphosphate (ATP) competitive as well as peptide-based JNK inhibitors were able to block the activity of both recombinant and HepG2 endogenous JNK activity. Using the same luminescence technique adapted for binding studies, the authors characterized peptide inhibitor mechanisms by measuring the binding affinity of the inhibitors for JNK. Because of the versatility of the technology, this cell-based JNK kinase assay could be adapted to other kinases and would represent a powerful tool to evaluate endogenous kinase activity and test a large number of potential inhibitors in a more physiologically relevant environment.


Ophthalmic Genetics | 2013

The Bcl-2/Bcl-XL inhibitor ABT-737 promotes death of retinoblastoma cancer cells.

Nathalie Allaman-Pillet; Anne Oberson; Francis L. Munier; Daniel F. Schorderet

Purpose: Retinoblastoma is a malignant tumor that usually develops in early childhood. During retinoblastoma spreading, RB1 gene inactivation is followed by additional genomic modifications which progressively lead to resistance of tumor cells to death. Drugs that act at downstream levels of death signaling pathways should therefore be interesting in killing retinoblastoma cells. ABT-737, a BH3 mimetic molecule effective at the mitochondrial level, has been shown to induce apoptosis in different human tumoral cell lines as well as in primary patient-derived cells, and in a mouse xenograph model. Methods: In this report, we analyzed the pro-death effect of ABT-737 on two human retinoblastoma cell lines, Y79 and WERI-Rb, as well as on the mouse photoreceptor cell line 661W. Results: We observed that ABT-737 was very effective as a single agent in inducing human WERI-Rb cells apoptosis without affecting the mouse 661W photoreceptor cells. However human Y79 cells were resistant to ABT-737, as a probable consequence of the absence of Bax. The high sensitivity of WERI-Rb to ABT-737 can be increased by downregulating Mcl-1 using the proteasome inhibitor MG-132. Preliminary analysis in primary mouse retinoblastoma tumoral cell lines predicts high sensitivity to ABT-737. Conclusion: Our data suggest that ABT-737 or related compounds could be a highly effective drug in the treatment of some retinoblastomas.


Molecular Cancer Research | 2015

BIRO1, a Cell-Permeable BH3 Peptide, Promotes Mitochondrial Fragmentation and Death of Retinoblastoma Cells

Nathalie Allaman-Pillet; Anne Oberson; Daniel F. Schorderet

Retinoblastoma is the most common pediatric intraocular neoplasm. While retinoblastoma development requires the inactivation of both alleles of the retinoblastoma tumor suppressor gene (RB1) in the developing retina, additional genomic changes are involved in tumor progression, which progressively lead to resistance of tumor cells to death. Therapeutics acting at very downstream levels of death signaling pathways should therefore be interesting in killing retinoblastoma cells. The BH3-only proteins promote apoptosis by modulating the interaction between the pro- and antiapoptotic members of the BCL2 protein family, and this effect can be recapitulated by the BH3 domains. This report analyzes the effect of various BH3 peptides, corresponding to different BH3-only proteins, on two retinoblastoma cell lines, Y79 and WERI-Rb, as well as on the photoreceptor cell line 661W. The BH3 peptide BIRO1, derived from the BCL2L11 death domain, was very effective in promoting Y79 and WERI-Rb cell death without affecting the 661W photoreceptor cells. This cell death was efficient even in absence of BAX and was shown to be caspase independent. While ROS production or AIF release was not detected from mitochondria of treated cells, BIRO1 initiated mitochondria fragmentation in a short period of time following treatment. Implications: The BIRO1 peptide is highly effective at killing retinoblastoma cells and has potential as a peptidomimetic. Mol Cancer Res; 13(1); 86–97. ©2014 AACR.


Scientific Reports | 2016

Identifying mutations in Tunisian families with retinal dystrophy

Imen Habibi; A. Chebil; Yosra Falfoul; Nathalie Allaman-Pillet; Fedra Kort; Daniel F. Schorderet; Leila El Matri

Retinal dystrophies (RD) are a rare genetic disorder with high genetic heterogeneity. This study aimed at identifying disease-causing variants in fifteen consanguineous Tunisian families. Full ophthalmic examination was performed. Index patients were subjected to IROme analysis or whole exome sequencing followed by homozygosity mapping. All detected variations were confirmed by direct Sanger sequencing. Mutation analysis in our patients revealed two compound heterozygous mutations p.(R91W);(V172D) in RPE65, and five novel homozygous mutations: p.R765C in CNGB1, p.H337R in PDE6B, splice site variant c.1129-2A > G and c.678_681delGAAG in FAM161A and c.1133 + 3_1133 + 6delAAGT in CERKL. The latter mutation impacts pre-mRNA splicing of CERKL. The other changes detected were six previously reported mutations in CNGB3 (p.R203*), ABCA4 (p.W782*), NR2E3 (p.R311Q), RPE65 (p.H182Y), PROM1 (c.1354dupT) and EYS (c.5928-2A > G). Segregation analysis in each family showed that all affected individuals were homozygotes and unaffected individuals were either heterozygote carriers or homozygous wild type allele. These results confirm the involvement of a large number of genes in RD in the Tunisian population.Retinal dystrophies (RD) are a rare genetic disorder with high genetic heterogeneity. This study aimed at identifying disease-causing variants in fifteen consanguineous Tunisian families. Full ophthalmic examination was performed. Index patients were subjected to IROme analysis or whole exome sequencing followed by homozygosity mapping. All detected variations were confirmed by direct Sanger sequencing. Mutation analysis in our patients revealed two compound heterozygous mutations p.(R91W);(V172D) in RPE65, and five novel homozygous mutations: p.R765C in CNGB1, p.H337R in PDE6B, splice site variant c.1129-2A > G and c.678_681delGAAG in FAM161A and c.1133 + 3_1133 + 6delAAGT in CERKL. The latter mutation impacts pre-mRNA splicing of CERKL. The other changes detected were six previously reported mutations in CNGB3 (p.R203*), ABCA4 (p.W782*), NR2E3 (p.R311Q), RPE65 (p.H182Y), PROM1 (c.1354dupT) and EYS (c.5928-2A > G). Segregation analysis in each family showed that all affected individuals were homozygotes and unaffected individuals were either heterozygote carriers or homozygous wild type allele. These results confirm the involvement of a large number of genes in RD in the Tunisian population.


Oncotarget | 2017

Bigh3 silencing increases retinoblastoma tumor growth in the murine SV40-TAg-Rb model.

Nathalie Allaman-Pillet; Anne Oberson; Daniel F. Schorderet

BIGH3, a secreted protein of the extracellular matrix interacts with collagen and integrins on the cell surface. BIGH3 can have opposing functions in cancer, acting either as tumor suppressor or promoter by enhancing tumor progression and angiogenesis. In the eye, BIGH3 is expressed in the cornea and the retinal pigment epithelium and could impact on the development of retinoblastoma, the most common paediatric intraocular neoplasm. Retinoblastoma initiation requires the inactivation of both alleles of the RB1 tumor suppressor gene in the developing retina and tumor progression involves additional genomic changes. To determine whether BIGH3 affects retinoblastoma development, we generated a retinoblastoma mouse model with disruption of the Bigh3 genomic locus. Bigh3 silencing in these mice resulted in enhanced tumor development in the retina. A decrease in apoptosis is involved in the initial events of tumorigenesis, followed by an increased activity of the pro-survival ERK pathway as well as an upregulation of cyclin-dependent kinases (CDKs). Taken together, these data suggest that BIGH3 acts as a tumor suppressor in the retina.


Experimental Eye Research | 2015

Tgfbi/Bigh3 silencing activates ERK in mouse retina

Nathalie Allaman-Pillet; Anne Oberson; Mauro Bustamante; Andrea Tasinato; Edith Hummler; Daniel F. Schorderet

BIGH3 is a secreted protein, part of the extracellular matrix where it interacts with collagen and integrins on the cell surface. BIGH3 can play opposing roles in cancer, acting as either tumor suppressor or promoter, and its mutations lead to different forms of corneal dystrophy. Although many studies have been carried out, little is known about the physiological role of BIGH3. Using the cre-loxP system, we generated a mouse model with disruption of the Bigh3 genomic locus. Bigh3 silencing did not result in any apparent phenotype modifications, the mice remained viable and fertile. We were able to determine the presence of BIGH3 in the retinal pigment epithelium (RPE). In the absence of BIGH3, a transient decrease in the apoptotic process involved in retina maturation was observed, leading to a transient increase in the INL thickness at P15. This phenomenon was accompanied by an increased activity of the pro-survival ERK pathway.


Scientific Reports | 2017

Corrigendum: Identifying mutations in Tunisian families with retinal dystrophy

Imen Habibi; A. Chebil; Yosra Falfoul; Nathalie Allaman-Pillet; Fedra Kort; Daniel F. Schorderet; Leila El Matri

Scientific Reports 6: Article number: 37455; published online: 22 November 2016; updated: 04 May 2017 This Article contains errors. The position of the mutation p.(R91W); (V172D) was incorrectly calculated, taking as a starting point the beginning of cDNA rather than the start codon. The correct position of the mutation is c.


Diabetes | 2004

Intracellular Stress Signaling Pathways Activated During Human Islet Preparation and Following Acute Cytokine Exposure

Saida Abdelli; Jeff Ansite; Raphaël Roduit; Tiziana Borsello; Ippei Matsumoto; Toshiya Sawada; Nathalie Allaman-Pillet; Hugues Henry; Jacques S. Beckmann; Bernhard J. Hering; Christophe Bonny


Biochimica et Biophysica Acta | 2007

Cell-permeable peptides induce dose- and length-dependent cytotoxic effects.

Alessandra K Cardozo; Valérie Büchillier; Marc Mathieu; Jianhua Chen; Fernanda Ortis; Laurence Ladrière; Nathalie Allaman-Pillet; Olivier Poirot; Stephan Kellenberger; Jacques S. Beckmann; Decio L. Eizirik; Christophe Bonny; Fabienne Maurer

Collaboration


Dive into the Nathalie Allaman-Pillet's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sylvie Guenat

University Hospital of Lausanne

View shared research outputs
Top Co-Authors

Avatar

Imen Habibi

École Polytechnique Fédérale de Lausanne

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge