Network


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

Hotspot


Dive into the research topics where Anselme L. Perrier is active.

Publication


Featured researches published by Anselme L. Perrier.


Nature Biotechnology | 2008

Human embryonic stem cells reveal recurrent genomic instability at 20q11.21.

Nathalie Lefort; Maxime Feyeux; Cécile Bas; Olivier Féraud; Annelise Bennaceur-Griscelli; Gérard Tachdjian; Marc Peschanski; Anselme L. Perrier

By analyzing five human embryonic stem (hES) cell lines over long-term culture, we identified a recurrent genomic instability in the human genome. An amplification of 2.5–4.6 Mb at 20q11.21, encompassing ∼23 genes in common, was detected in four cell lines of different origins. This amplification, which has been associated with oncogenic transformation, may provide a selective advantage to hES cells in culture.


Cell Stem Cell | 2014

Human ESC-Derived Dopamine Neurons Show Similar Preclinical Efficacy and Potency to Fetal Neurons when Grafted in a Rat Model of Parkinson’s Disease

Shane Grealish; Elsa Diguet; Agnete Kirkeby; Bengt Mattsson; Andreas Heuer; Yann Bramoullé; Nadja Van Camp; Anselme L. Perrier; Philippe Hantraye; Anders Björklund; Malin Parmar

Summary Considerable progress has been made in generating fully functional and transplantable dopamine neurons from human embryonic stem cells (hESCs). Before these cells can be used for cell replacement therapy in Parkinson’s disease (PD), it is important to verify their functional properties and efficacy in animal models. Here we provide a comprehensive preclinical assessment of hESC-derived midbrain dopamine neurons in a rat model of PD. We show long-term survival and functionality using clinically relevant MRI and PET imaging techniques and demonstrate efficacy in restoration of motor function with a potency comparable to that seen with human fetal dopamine neurons. Furthermore, we show that hESC-derived dopamine neurons can project sufficiently long distances for use in humans, fully regenerate midbrain-to-forebrain projections, and innervate correct target structures. This provides strong preclinical support for clinical translation of hESC-derived dopamine neurons using approaches similar to those established with fetal cells for the treatment of Parkinson’s disease.


Stroke | 2010

The Postischemic Environment Differentially Impacts Teratoma or Tumor Formation After Transplantation of Human Embryonic Stem Cell-Derived Neural Progenitors

Christine Seminatore; Jérôme Polentes; Ditte Ellman; Nataliya Kozubenko; Valérie Itier; Samir Tine; Laurent Tritschler; Marion Brenot; Emmanuelle Guidou; Johanna Blondeau; Mickael Lhuillier; Aurore Bugi; Laetitia Aubry; Pavla Jendelová; Eva Syková; Anselme L. Perrier; Bente Finsen; Brigitte Onteniente

Background and Purpose— Risk of tumorigenesis is a major obstacle to human embryonic and induced pluripotent stem cell therapy. Likely linked to the stage of differentiation of the cells at the time of implantation, formation of teratoma/tumors can also be influenced by factors released by the host tissue. We have analyzed the relative effects of the stage of differentiation and the postischemic environment on the formation of adverse structures by transplanted human embryonic stem cell-derived neural progenitors. Methods— Four differentiation stages were identified on the basis of quantitative polymerase chain reaction expression of pluripotency, proliferation, and differentiation markers. Neural progenitors were transplanted at these 4 stages into rats with no, small, or large middle cerebral artery occlusion lesions. The fate of each transplant was compared with their pretransplantation status 1 to 4 months posttransplantation. Results— The influence of the postischemic environment was limited to graft survival and occurrence of nonneuroectodermal structures after transplantation of very immature neural progenitors. Both effects were lost with differentiation. We identified a particular stage of differentiation characterized in vitro by a rebound of proliferative activity that produced highly proliferative grafts susceptible to threaten surrounding host tissues. Conclusion— The effects of the ischemic environment on the formation of teratoma by transplanted human embryonic stem cell-derived neural progenitors are limited to early differentiation stages that will likely not be used for stem cell therapy. In contrast, hyperproliferation observed at later stages of differentiation corresponds to an intrinsic activity that should be monitored to avoid tumorigenesis.


Stem Cells | 2013

Embryonic stem cells neural differentiation qualifies the role of Wnt/β-Catenin signals in human telencephalic specification and regionalization

Camille Nicoleau; Christine Varela; Caroline Bonnefond; Yves Maury; Aurore Bugi; Laetitia Aubry; Pedro Viegas; Fany Bourgois-Rocha; Marc Peschanski; Anselme L. Perrier

Wnt‐ligands are among key morphogens that mediate patterning of the anterior territories of the developing brain in mammals. We qualified the role of Wnt‐signals in regional specification and subregional organization of the human telencephalon using human pluripotent stem cells (hPSCs). One step neural conversion of hPSCs using SMAD inhibitors leads to progenitors with a default rostral identity. It provides an ideal biological substrate for investigating the role of Wnt signaling in both anteroposterior and dorso‐ventral processes. Challenging hPSC‐neural derivatives with Wnt‐antagonists, alone or combined with sonic hedgehog (Shh), we found that Wnt‐inhibition promote both telencephalic specification and ventral patterning of telencephalic neural precursors in a dose‐dependent manner. Using optimal Wnt‐antagonist and Shh‐agonist signals we produced human ventral‐telencephalic precursors, committed to differentiation into striatal projection neurons both in vitro and in vivo after homotypic transplantation in quinolinate‐lesioned rats. This study indicates that sequentially organized Wnt‐signals play a key role in the development of human ventral telencephalic territories from which the striatum arise. In addition, the optimized production of hPSC‐derived striatal cells described here offers a relevant biological resource for exploring and curing Huntington disease. Stem Cells 2013;31:1763‐1774


Stem Cells | 2013

High throughput screening for inhibitors of REST in neural derivatives of human embryonic stem cells reveals a chemical compound that promotes expression of neuronal genes

Jérémie Charbord; Pauline Poydenot; Caroline Bonnefond; Maxime Feyeux; Fabrice Casagrande; Benjamin Brinon; Laetitia Francelle; Gwenaelle Auregan; Martine Guillermier; Michel Cailleret; Pedro Viegas; Camille Nicoleau; Cécile Martinat; Emmanuel Brouillet; Marc Peschanski; Marc Lechuga; Anselme L. Perrier

Decreased expression of neuronal genes such as brain‐derived neurotrophic factor (BDNF) is associated with several neurological disorders. One molecular mechanism associated with Huntington disease (HD) is a discrete increase in the nuclear activity of the transcriptional repressor REST/NRSF binding to repressor element‐1 (RE1) sequences. High‐throughput screening of a library of 6,984 compounds with luciferase‐assay measuring REST activity in neural derivatives of human embryonic stem cells led to identify two benzoimidazole‐5‐carboxamide derivatives that inhibited REST silencing in a RE1‐dependent manner. The most potent compound, X5050, targeted REST degradation, but neither REST expression, RNA splicing nor binding to RE1 sequence. Differential transcriptomic analysis revealed the upregulation of neuronal genes targeted by REST in wild‐type neural cells treated with X5050. This activity was confirmed in neural cells produced from human induced pluripotent stem cells derived from a HD patient. Acute intraventricular delivery of X5050 increased the expressions of BDNF and several other REST‐regulated genes in the prefrontal cortex of mice with quinolinate‐induced striatal lesions. This study demonstrates that the use of pluripotent stem cell derivatives can represent a crucial step toward the identification of pharmacological compounds with therapeutic potential in neurological affections involving decreased expression of neuronal genes associated to increased REST activity, such as Huntington disease. Stem Cells 2013;31:1816‐1828


Human Molecular Genetics | 2012

Early transcriptional changes linked to naturally occurring Huntington's disease mutations in neural derivatives of human embryonic stem cells

Maxime Feyeux; Fany Bourgois-Rocha; Amanda Redfern; Peter Giles; Nathalie Lefort; Sophie Aubert; Caroline Bonnefond; Aurore Bugi; Marta Ruiz; Nicole Déglon; Lesley Jones; Marc Peschanski; Nicholas Denby Allen; Anselme L. Perrier

Huntingtons disease (HD) is characterized by a late clinical onset despite ubiquitous expression of the mutant gene at all developmental stages. How mutant huntingtin impacts on signalling pathways in the pre-symptomatic period has remained essentially unexplored in humans due to a lack of appropriate models. Using multiple human embryonic stem cell lines derived from blastocysts diagnosed as carrying the mutant huntingtin gene by pre-implantation genetic diagnosis, we explored early developmental changes in gene expression using differential transcriptomics, combined with gain and loss of function strategies. We demonstrated a down-regulation of the HTT gene itself in HD neural cells and identified three genes, the expression of which differs significantly in HD cells when compared with wild-type controls, namely CHCHD2, TRIM4 and PKIB. Similar dysregulation had been observed previously for CHCDH2 and TRIM4 in blood cells from patients. CHCHD2 is involved in mitochondrial function and PKIB in protein kinase A-dependent pathway regulation, which suggests that these functions may be precociously impacted in HD.


PLOS ONE | 2014

Allele-specific silencing of mutant huntingtin in rodent brain and human stem cells.

Valerie Drouet; Marta Ruiz; Diana Zala; Maxime Feyeux; Gwennaelle Auregan; Karine Cambon; Laetitia Troquier; Johann Carpentier; Sophie Aubert; Nicolas Merienne; Fany Bourgois-Rocha; Raymonde Hassig; Maria Rey; Noelle Dufour; Frédéric Saudou; Anselme L. Perrier; Philippe Hantraye; Nicole Déglon

Huntingtons disease (HD) is an autosomal dominant neurodegenerative disorder resulting from polyglutamine expansion in the huntingtin (HTT) protein and for which there is no cure. Although suppression of both wild type and mutant HTT expression by RNA interference is a promising therapeutic strategy, a selective silencing of mutant HTT represents the safest approach preserving WT HTT expression and functions. We developed small hairpin RNAs (shRNAs) targeting single nucleotide polymorphisms (SNP) present in the HTT gene to selectively target the disease HTT isoform. Most of these shRNAs silenced, efficiently and selectively, mutant HTT in vitro. Lentiviral-mediated infection with the shRNAs led to selective degradation of mutant HTT mRNA and prevented the apparition of neuropathology in HD rats striatum expressing mutant HTT containing the various SNPs. In transgenic BACHD mice, the mutant HTT allele was also silenced by this approach, further demonstrating the potential for allele-specific silencing. Finally, the allele-specific silencing of mutant HTT in human embryonic stem cells was accompanied by functional recovery of the vesicular transport of BDNF along microtubules. These findings provide evidence of the therapeutic potential of allele-specific RNA interference for HD.


American Journal of Human Genetics | 2008

Evolutionary Forces Shape the Human RFPL1,2,3 Genes toward a Role in Neocortex Development

Jérôme Bonnefont; Sergey Igorievich Nikolaev; Anselme L. Perrier; Song Guo; Laetitia Cartier; Silvia Sorce; Térèse Laforge; Laetitia Aubry; Philipp Khaitovich; Marc Peschanski; Karl-Heinz Krause

The size and organization of the brain neocortex has dramatically changed during primate evolution. This is probably due to the emergence of novel genes after duplication events, evolutionary changes in gene expression, and/or acceleration in protein evolution. Here, we describe a human Ret finger protein-like (hRFPL)1,2,3 gene cluster on chromosome 22, which is transactivated by the corticogenic transcription factor Pax6. High hRFPL1,2,3 transcript levels were detected at the onset of neurogenesis in differentiating human embryonic stem cells and in the developing human neocortex, whereas the unique murine RFPL gene is expressed in liver but not in neural tissue. Study of the evolutionary history of the RFPL gene family revealed that the RFPL1,2,3 gene ancestor emerged after the Euarchonta-Glires split. Subsequent duplication events led to the presence of multiple RFPL1,2,3 genes in Catarrhini ( approximately 34 mya) resulting in an increase in gene copy number in the hominoid lineage. In Catarrhini, RFPL1,2,3 expression profile diverged toward the neocortex and cerebellum over the liver. Importantly, humans showed a striking increase in cortical RFPL1,2,3 expression in comparison to their cerebellum, and to chimpanzee and macaque neocortex. Acceleration in RFPL-protein evolution was also observed with signs of positive selection in the RFPL1,2,3 cluster and two neofunctionalization events (acquisition of a specific RFPL-Defining Motif in all RFPLs and of a N-terminal 29 amino-acid sequence in catarrhinian RFPL1,2,3). Thus, we propose that the recent emergence and multiplication of the RFPL1,2,3 genes contribute to changes in primate neocortex size and/or organization.


Cell Stem Cell | 2012

How Can Human Pluripotent Stem Cells Help Decipher and Cure Huntington’s Disease?

Anselme L. Perrier; Marc Peschanski

Pluripotent stem cell (PSC) technologies are becoming a key asset for deciphering pathological cascades and for developing new treatments against many neurodegenerative disorders, including Huntingtons disease (HD). This perspective discusses the challenges and opportunities facing the use of PSCs for treating HD, focusing on four major applications: namely, the use of PSCs as a substitute source of human striatal cells for current HD cell therapy, as a cellular model of HD for the validation of human-specific gene therapies, for deciphering molecular mechanisms underlying HD, and in drug discovery.


EBioMedicine | 2016

Human Pluripotent Stem Cell-derived Cortical Neurons for High Throughput Medication Screening in Autism: A Proof of Concept Study in SHANK3 Haploinsufficiency Syndrome

Hélène Darville; Aurélie Poulet; Frédérique Rodet-Amsellem; Laure Chatrousse; Julie Pernelle; Claire Boissart; Delphine Héron; Caroline Nava; Anselme L. Perrier; Margot Jarrige; Francis Cogé; Mark J. Millan; Thomas Bourgeron; Marc Peschanski; Richard Delorme; Alexandra Benchoua

Autism spectrum disorders affect millions of individuals worldwide, but their heterogeneity complicates therapeutic intervention that is essentially symptomatic. A versatile yet relevant model to rationally screen among hundreds of therapeutic options would help improving clinical practice. Here we investigated whether neurons differentiated from pluripotent stem cells can provide such a tool using SHANK3 haploinsufficiency as a proof of principle. A library of compounds was screened for potential to increase SHANK3 mRNA content in neurons differentiated from control human embryonic stem cells. Using induced pluripotent stem cell technology, active compounds were then evaluated for efficacy in correcting dysfunctional networks of neurons differentiated from individuals with deleterious point mutations of SHANK3. Among 202 compounds tested, lithium and valproic acid showed the best efficacy at corrected SHANK3 haploinsufficiency associated phenotypes in cellulo. Lithium pharmacotherapy was subsequently provided to one patient and, after one year, an encouraging decrease in autism severity was observed. This demonstrated that pluripotent stem cell-derived neurons provide a novel cellular paradigm exploitable in the search for specific disease-modifying treatments.

Collaboration


Dive into the Anselme L. Perrier's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria Rey

University of Lausanne

View shared research outputs
Top Co-Authors

Avatar

Philippe Hantraye

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge