19 results on '"Anselme L. Perrier"'
Search Results
2. Huntingtin lowering impairs the maturation and synchronized synaptic activity of human cortical neuronal networks derived from induced pluripotent stem cells
- Author
-
Mathilde Louçã, Donya El Akrouti, Aude Lemesle, Morgane Louessard, Noëlle Dufour, Chloé Baroin, Aurore de la Fouchardière, Laurent Cotter, Hélène Jean-Jacques, Virginie Redeker, and Anselme L. Perrier
- Subjects
Neuronal network ,HTT lowering ,Huntington disease ,Huntingtin ,Pluripotent stem cell ,Synapse ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Despite growing descriptions of wild-type Huntingtin (wt-HTT) roles in both adult brain function and, more recently, development, several clinical trials are exploring HTT-lowering approaches that target both wt-HTT and the mutant isoform (mut-HTT) responsible for Huntington's disease (HD). This non-selective targeting is based on the autosomal dominant inheritance of HD, supporting the idea that mut-HTT exerts its harmful effects through a toxic gain-of-function or a dominant-negative mechanism. However, the precise amount of wt-HTT needed for healthy neurons in adults and during development remains unclear. In this study, we address this question by examining how wt-HTT loss affects human neuronal network formation, synaptic maturation, and homeostasis in vitro. Our findings establish a role of wt-HTT in the maturation of dendritic arborization and the acquisition of network-wide synchronized activity by human cortical neuronal networks modeled in vitro. Interestingly, the network synchronization defects only became apparent when more than two-thirds of the wt-HTT protein was depleted. Our study underscores the critical need to precisely understand wt-HTT role in neuronal health. It also emphasizes the potential risks of excessive wt-HTT loss associated with non-selective therapeutic approaches targeting both wt- and mut-HTT isoforms in HD patients.
- Published
- 2024
- Full Text
- View/download PDF
3. CTIP2-Regulated Reduction in PKA-Dependent DARPP32 Phosphorylation in Human Medium Spiny Neurons: Implications for Huntington Disease
- Author
-
Marija Fjodorova, Morgane Louessard, Zongze Li, Daniel C. De La Fuente, Emma Dyke, Simon P. Brooks, Anselme L. Perrier, and Meng Li
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: The mechanisms underlying the selective degeneration of medium spiny neurons (MSNs) in Huntington disease (HD) remain largely unknown. CTIP2, a transcription factor expressed by all MSNs, is implicated in HD pathogenesis because of its interactions with mutant huntingtin. Here, we report a key role for CTIP2 in protein phosphorylation via governing protein kinase A (PKA) signaling in human striatal neurons. Transcriptomic analysis of CTIP2-deficient MSNs implicates CTIP2 target genes at the heart of cAMP-Ca2+ signal integration in the PKA pathway. These findings are further supported by experimental evidence of a substantial reduction in phosphorylation of DARPP32 and GLUR1, two PKA targets in CTIP2-deficient MSNs. Moreover, we show that CTIP2-dependent dysregulation of protein phosphorylation is shared by HD hPSC-derived MSNs and striatal tissues of two HD mouse models. This study therefore establishes an essential role for CTIP2 in human MSN homeostasis and provides mechanistic and potential therapeutic insight into striatal neurodegeneration. : In this article, Fjodorova and colleagues provide mechanistic and potential therapeutic insight into striatal neurodegeneration caused by CTIP2 hypofunction. CTIP2 plays an essential role in human MSN homeostasis via regulating PKA-dependent DARPP32 phosphorylation and protecting MSNs from oxidative stress-induced cell death. Protein phosphorylation deficits occur in human and mouse Huntington disease MSNs due to CTIP2- and mHTT-co-regulated molecular signaling abnormalities. Keywords: CTIP2, DARPP32, Huntington disease, medium spiny neuron, neural differentiation
- Published
- 2019
- Full Text
- View/download PDF
4. MHC matching fails to prevent long-term rejection of iPSC-derived neurons in non-human primates
- Author
-
Romina Aron Badin, Aurore Bugi, Susannah Williams, Marta Vadori, Marie Michael, Caroline Jan, Alberto Nassi, Sophie Lecourtois, Antoine Blancher, Emanuele Cozzi, Philippe Hantraye, and Anselme L. Perrier
- Subjects
Science - Abstract
Matching iPSC donors’ and patients’ HLA haplotypes has been proposed as a way to generate cell therapy products with enhanced immunological compatibility. Here the authors show that MHC matching alone is insufficient to grant long-term survival of neuronal grafts in the lesioned brain of non-human primates.
- Published
- 2019
- Full Text
- View/download PDF
5. Propagation of α-Synuclein Strains within Human Reconstructed Neuronal Network
- Author
-
Simona Gribaudo, Philippe Tixador, Luc Bousset, Alexis Fenyi, Patricia Lino, Ronald Melki, Jean-Michel Peyrin, and Anselme L. Perrier
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Reappraisal of neuropathological studies suggests that pathological hallmarks of Alzheimer’s disease and Parkinson’s disease (PD) spread progressively along predictable neuronal pathways in the human brain through unknown mechanisms. Although there is much evidence supporting the prion-like propagation and amplification of α-synuclein (α-Syn) in vitro and in rodent models, whether this scenario occurs in the human brain remains to be substantiated. Here we reconstructed in microfluidic devices corticocortical neuronal networks using human induced pluripotent stem cells derived from a healthy donor. We provide unique experimental evidence that different strains of human α-Syn disseminate in “wild-type” human neuronal networks in a prion-like manner. We show that two distinct α-Syn strains we named fibrils and ribbons are transported, traffic between neurons, and trigger to different extents, in a dose- and structure-dependent manner, the progressive accumulation of PD-like pathological hallmarks. We further demonstrate that seeded aggregation of endogenous soluble α-Syn affects synaptic integrity and mitochondria morphology. : In this article, Gribaudo and colleagues report the internalization and trafficking of exogenous α-synuclein fibrils and ribbons through fluidically isolated cortical networks derived from wild-type human iPSCs. These assemblies trigger via seeding mechanisms the accumulation of endogenous phosphorylated α-synuclein, in a structure-, dose-, and time-dependent manner. Phosphorylated α-Syn structures resist degradation and accumulate in the cytoplasm of neurons, affecting Ca2+ homeostasis and mitochondria morphology. Keywords: human pluripotent stem cells, Parkinson's disease, microfluidic, prion-like, nucleation, synuclein, Lewy body, human cortical neuron, neuronal dysfunction
- Published
- 2019
- Full Text
- View/download PDF
6. Human Induced Pluripotent Stem Cell-Derived Astrocytes Are Differentially Activated by Multiple Sclerosis-Associated Cytokines
- Author
-
Sylvain Perriot, Amandine Mathias, Guillaume Perriard, Mathieu Canales, Nils Jonkmans, Nicolas Merienne, Cécile Meunier, Lina El Kassar, Anselme L. Perrier, David-Axel Laplaud, Myriam Schluep, Nicole Déglon, and Renaud Du Pasquier
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Recent studies highlighted the importance of astrocytes in neuroinflammatory diseases, interacting closely with other CNS cells but also with the immune system. However, due to the difficulty in obtaining human astrocytes, their role in these pathologies is still poorly characterized. Here, we develop a serum-free protocol to differentiate human induced pluripotent stem cells (hiPSCs) into astrocytes. Gene expression and functional assays show that our protocol consistently yields a highly enriched population of resting mature astrocytes across the 13 hiPSC lines differentiated. Using this model, we first highlight the importance of serum-free media for astrocyte culture to generate resting astrocytes. Second, we assess the astrocytic response to IL-1β, TNF-α, and IL-6, all cytokines important in neuroinflammation, such as multiple sclerosis. Our study reveals very specific profiles of reactive astrocytes depending on the triggering stimulus. This model provides ideal conditions for in-depth and unbiased characterization of astrocyte reactivity in neuroinflammatory conditions. : Perriot et al. describe a new protocol to differentiate both resting and reactive astrocytes from human induced pluripotent stem cells in serum-free conditions. They show that astrocytes exhibit highly contrasted responses to serum and to cytokines involved in multiple sclerosis, establishing that this model is suitable to study neuroinflammation. Keywords: induced pluripotent stem cells, differentiation, astrocytes, multiple sclerosis, neuroinflammation
- Published
- 2018
- Full Text
- View/download PDF
7. Differentiation of nonhuman primate pluripotent stem cells into functional keratinocytes
- Author
-
Sophie Domingues, Yolande Masson, Aurore Marteyn, Jennifer Allouche, Anselme L. Perrier, Marc Peschanski, Cecile Martinat, Christine Baldeschi, and Gilles Lemaître
- Subjects
Keratinocytes ,Pluripotent stem cells ,Nonhuman primate model ,Skin graft ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Epidermal grafting using cells derived from pluripotent stem cells will change the face of this side of regenerative cutaneous medicine. To date, the safety of the graft would be the major unmet deal in order to implement long-term skin grafting. In this context, experiments on large animals appear unavoidable to assess this question and possible rejection. Cellular tools for large animal models should be constructed. Methods In this study, we generated monkey pluripotent stem cell-derived keratinocytes and evaluated their capacities to reconstruct an epidermis, in vitro as well as in vivo. Results Monkey pluripotent stem cells were differentiated efficiently into keratinocytes able to reconstruct fully epidermis presenting a low level of major histocompatibility complex class-I antigens, opening the way for autologous or allogeneic epidermal long-term grafting. Conclusions Functional keratinocytes generated from nonhuman primate embryonic stem cells and induced pluripotent stem cells reproduce an in-vitro and in-vivo stratified epidermis. These monkey skin grafts will be considered to model autologous or allogeneic epidermal grafting using either embryonic stem cells or induced pluripotent stem cells. This graft model will allow us to further investigate the safety, efficacy and immunogenicity of nonhuman primate PSC-derived epidermis in the perspective of human skin cell therapy.
- Published
- 2017
- Full Text
- View/download PDF
8. The Self-Inactivating KamiCas9 System for the Editing of CNS Disease Genes
- Author
-
Nicolas Merienne, Gabriel Vachey, Lucie de Longprez, Cécile Meunier, Virginie Zimmer, Guillaume Perriard, Mathieu Canales, Amandine Mathias, Lucas Herrgott, Tim Beltraminelli, Axelle Maulet, Thomas Dequesne, Catherine Pythoud, Maria Rey, Luc Pellerin, Emmanuel Brouillet, Anselme L. Perrier, Renaud du Pasquier, and Nicole Déglon
- Subjects
CRISPR/Cas9 ,gene editing ,self-inactivating system ,KamiCas9 ,neurodegenerative diseases ,Huntington’s disease ,lentiviral vectors ,Biology (General) ,QH301-705.5 - Abstract
Neurodegenerative disorders are a major public health problem because of the high frequency of these diseases. Genome editing with the CRISPR/Cas9 system is making it possible to modify the sequence of genes linked to these disorders. We designed the KamiCas9 self-inactivating editing system to achieve transient expression of the Cas9 protein and high editing efficiency. In the first application, the gene responsible for Huntington’s disease (HD) was targeted in adult mouse neuronal and glial cells. Mutant huntingtin (HTT) was efficiently inactivated in mouse models of HD, leading to an improvement in key markers of the disease. Sequencing of potential off-targets with the constitutive Cas9 system in differentiated human iPSC revealed a very low incidence with only one site above background level. This off-target frequency was significantly reduced with the KamiCas9 system. These results demonstrate the potential of the self-inactivating CRISPR/Cas9 editing for applications in the context of neurodegenerative diseases.
- Published
- 2017
- Full Text
- View/download PDF
9. Preclinical Evaluation of a Lentiviral Vector for Huntingtin Silencing
- Author
-
Karine Cambon, Virginie Zimmer, Sylvain Martineau, Marie-Claude Gaillard, Margot Jarrige, Aurore Bugi, Jana Miniarikova, Maria Rey, Raymonde Hassig, Noelle Dufour, Gwenaelle Auregan, Philippe Hantraye, Anselme L. Perrier, and Nicole Déglon
- Subjects
Huntington ,RNA interference ,biosafety ,off-targets ,gene expression ,iPSCs ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Huntington’s disease (HD) is an autosomal dominant neurodegenerative disorder resulting from a polyglutamine expansion in the huntingtin (HTT) protein. There is currently no cure for this disease, but recent studies suggest that RNAi to downregulate the expression of both normal and mutant HTT is a promising therapeutic approach. We previously developed a small hairpin RNA (shRNA), vectorized in an HIV-1-derived lentiviral vector (LV), that reduced pathology in an HD rodent model. Here, we modified this vector for preclinical development by using a tat-independent third-generation LV (pCCL) backbone and removing the original reporter genes. We demonstrate that this novel vector efficiently downregulated HTT expression in vitro in striatal neurons derived from induced pluripotent stem cells (iPSCs) of HD patients. It reduced two major pathological HD hallmarks while triggering a minimal inflammatory response, up to 6 weeks after injection, when administered by stereotaxic surgery in the striatum of an in vivo rodent HD model. Further assessment of this shRNA vector in vitro showed proper processing by the endogenous silencing machinery, and we analyzed gene expression changes to identify potential off-targets. These preclinical data suggest that this new shRNA vector fulfills primary biosafety and efficiency requirements for further development in the clinic as a cure for HD.
- Published
- 2017
- Full Text
- View/download PDF
10. Longitudinal characterization of cognitive and motor deficits in an excitotoxic lesion model of striatal dysfunction in non-human primates
- Author
-
Sonia Lavisse, Susannah Williams, Sophie Lecourtois, Nadja van Camp, Martine Guillermier, Pauline Gipchtein, Caroline Jan, Sébastien Goutal, Leopold Eymin, Julien Valette, Thierry Delzescaux, Anselme L. Perrier, Philippe Hantraye, and Romina Aron Badin
- Subjects
Animal model ,Non-human primate ,Cognitive impairment ,Movement disorder ,PET imaging ,Striatal dysfunction ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
As research progresses in the understanding of the molecular and cellular mechanisms underlying neurodegenerative diseases like Huntington's disease (HD) and expands towards preclinical work for the development of new therapies, highly relevant animal models are increasingly needed to test new hypotheses and to validate new therapeutic approaches. In this light, we characterized an excitotoxic lesion model of striatal dysfunction in non-human primates (NHPs) using cognitive and motor behaviour assessment as well as functional imaging and post-mortem anatomical analyses.NHPs received intra-striatal stereotaxic injections of quinolinic acid bilaterally in the caudate nucleus and unilaterally in the left sensorimotor putamen. Post-operative MRI scans showed atrophy of the caudate nucleus and a large ventricular enlargement in all 6 NHPs that correlated with post-mortem measurements. Behavioral analysis showed deficits in 2 analogues of the Wisconsin card sorting test (perseverative behavior) and in an executive task, while no deficits were observed in a visual recognition or an episodic memory task at 6 months following surgery. Spontaneous locomotor activity was decreased after lesion and the incidence of apomorphine-induced dyskinesias was significantly increased at 3 and 6 months following lesion. Positron emission tomography scans obtained at end-point showed a major deficit in glucose metabolism and D2 receptor density limited to the lesioned striatum of all NHPs compared to controls. Post-mortem analyses revealed a significant loss of medium-sized spiny neurons in the striatum, a loss of neurons and fibers in the globus pallidus, a unilateral decrease in dopaminergic neurons of the substantia nigra and a loss of neurons in the motor and dorsolateral prefrontal cortex.Overall, we show that this robust NHP model presents specific behavioral (learning, execution and retention of cognitive tests) and metabolic functional deficits that, to the best of our knowledge, are currently not mimicked in any available large animal model of striatal dysfunction. Moreover, we used non-invasive, translational techniques like behavior and imaging to quantify such deficits and found that they correlate to a significant cell loss in the striatum and its main input and output structures. This model can thus significantly contribute to the pre-clinical longitudinal evaluation of the ability of new therapeutic cell, gene or pharmacotherapy approaches in restoring the functionality of the striatal circuitry.
- Published
- 2019
- Full Text
- View/download PDF
11. Embryonic stem cells neural differentiation qualifies the role of Wnt/β-Catenin signals in human telencephalic specification and regionalization
- Author
-
Aurore Bugi, Christine Varela, Laetitia Aubry, Camille Nicoleau, Yves Maury, Fany Bourgois-Rocha, Pedro Viegas, Anselme L. Perrier, Marc Peschanski, and Caroline Bonnefond
- Subjects
Telencephalon ,Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Animals ,Humans ,Hedgehog Proteins ,Progenitor cell ,Sonic hedgehog ,Induced pluripotent stem cell ,Wnt Signaling Pathway ,Embryonic Stem Cells ,030304 developmental biology ,Body Patterning ,Neurons ,0303 health sciences ,Wnt signaling pathway ,Cell Differentiation ,Cell Biology ,Anatomy ,Embryonic stem cell ,Rats ,Transplantation ,Huntington Disease ,Organ Specificity ,biology.protein ,Molecular Medicine ,Stem cell ,Neuroscience ,Developmental biology ,Heterocyclic Compounds, 3-Ring ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
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.
- Published
- 2013
- Full Text
- View/download PDF
12. 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
- Author
-
Camille Nicoleau, Michel Cailleret, Marc Lechuga, Benjamin Brinon, Martine Guillermier, Emmanuel Brouillet, Caroline Bonnefond, Laetitia Francelle, Maxime Feyeux, Pedro Viegas, Gwenaëlle Auregan, Cécile Martinat, Marc Peschanski, Elena Cattaneo, Anselme L. Perrier, Jérémie Charbord, Pauline Poydenot, and Fabrice Casagrande
- Subjects
Male ,Repressor ,Biology ,Cell Line ,Small Molecule Libraries ,Transcriptome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,Genes, Reporter ,Neurosphere ,Animals ,Humans ,Gene silencing ,Luciferases ,Induced pluripotent stem cell ,Embryonic Stem Cells ,030304 developmental biology ,Neurons ,0303 health sciences ,Cell Biology ,Embryonic stem cell ,Molecular biology ,Neural stem cell ,High-Throughput Screening Assays ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,Repressor Proteins ,Disease Models, Animal ,Huntington Disease ,Gene Expression Regulation ,Molecular Medicine ,Stem cell ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
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.
- Published
- 2013
- Full Text
- View/download PDF
13. Evolutionary Forces Shape the Human RFPL1,2,3 Genes toward a Role in Neocortex Development
- Author
-
Térèse Laforge, Karl-Heinz Krause, Jérôme Bonnefont, Stylianos E. Antonarakis, Anselme L. Perrier, Laetitia Aubry, Song Guo, Sergey Nikolaev, Marc Peschanski, Laetitia Cartier, Silvia Sorce, and Philipp Khaitovich
- Subjects
Liver/metabolism ,Pan troglodytes ,Amino Acid Motifs ,Neocortex ,ddc:616.07 ,Biology ,Article ,Evolution, Molecular ,Molecular evolution ,Gene duplication ,Gene cluster ,medicine ,Genetics ,Animals ,Humans ,Gene family ,Genetics(clinical) ,Gene ,Embryonic Stem Cells ,Genetics (clinical) ,Embryonic Stem Cells/cytology ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Neocortex/embryology/metabolism ,medicine.anatomical_structure ,Liver ,Hela Cells ,Macaca ,Neofunctionalization ,Carrier Proteins/biosynthesis ,PAX6 ,Carrier Proteins ,HeLa Cells - Abstract
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.
- Published
- 2008
- Full Text
- View/download PDF
14. Dominant-Negative Effects of Adult-Onset Huntingtin Mutations Alter the Division of Human Embryonic Stem Cells-Derived Neural Cells
- Author
-
Carla Lopes, Sophie Aubert, Fany Bourgois-Rocha, Monia Barnat, Anselme L. Perrier, Nicole Déglon, Sandrine Humbert, and Ana Cristina Rego
- Subjects
0301 basic medicine ,Huntingtin ,Human Embryonic Stem Cells ,lcsh:Medicine ,Cell Cycle Proteins ,Biochemistry ,Spindle pole body ,0302 clinical medicine ,Neural Stem Cells ,Nuclear Matrix-Associated Proteins ,Animal Cells ,Medicine and Health Sciences ,Cell Cycle and Cell Division ,Age of Onset ,RNA, Small Interfering ,Induced pluripotent stem cell ,lcsh:Science ,Cells, Cultured ,Genes, Dominant ,Neurons ,Genetics ,Huntingtin Protein ,Multidisciplinary ,Stem Cells ,Microtubule Motors ,Antigens, Nuclear ,Neurodegenerative Diseases ,Dynactin Complex ,Neural stem cell ,Cell biology ,Protein Transport ,Huntington Disease ,Neurology ,Cell Processes ,Genetic Diseases ,RNA Interference ,Cellular Types ,Microtubule-Associated Proteins ,Subcellular Fractions ,Research Article ,Adult ,Pluripotent Stem Cells ,congenital, hereditary, and neonatal diseases and abnormalities ,Motor Proteins ,Nerve Tissue Proteins ,Spindle Apparatus ,Biology ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Developmental Neuroscience ,Molecular Motors ,mental disorders ,Humans ,Alleles ,Metaphase ,Clinical Genetics ,Autosomal Dominant Diseases ,lcsh:R ,Dyneins ,Biology and Life Sciences ,Proteins ,Cell Biology ,Embryonic stem cell ,Spindle apparatus ,nervous system diseases ,Cytoskeletal Proteins ,030104 developmental biology ,nervous system ,Cellular Neuroscience ,Mutation ,Dynactin ,lcsh:Q ,Peptides ,Trinucleotide Repeat Expansion ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Mutations of the huntingtin protein (HTT) gene underlie both adult-onset and juvenile forms of Huntington's disease (HD). HTT modulates mitotic spindle orientation and cell fate in mouse cortical progenitors from the ventricular zone. Using human embryonic stem cells (hESC) characterized as carrying mutations associated with adult-onset disease during pre-implantation genetic diagnosis, we investigated the influence of human HTT and of an adult-onset HD mutation on mitotic spindle orientation in human neural stem cells (NSCs) derived from hESCs. The RNAi-mediated silencing of both HTT alleles in neural stem cells derived from hESCs disrupted spindle orientation and led to the mislocalization of dynein, the p150Glued subunit of dynactin and the large nuclear mitotic apparatus (NuMA) protein. We also investigated the effect of the adult-onset HD mutation on the role of HTT during spindle orientation in NSCs derived from HD-hESCs. By combining SNP-targeting allele-specific silencing and gain-of-function approaches, we showed that a 46-glutamine expansion in human HTT was sufficient for a dominant-negative effect on spindle orientation and changes in the distribution within the spindle pole and the cell cortex of dynein, p150Glued and NuMA in neural cells. Thus, neural derivatives of disease-specific human pluripotent stem cells constitute a relevant biological resource for exploring the impact of adult-onset HD mutations of the HTT gene on the division of neural progenitors, with potential applications in HD drug discovery targeting HTT-dynein-p150Glued complex interactions.
- Published
- 2016
15. Expression of PRiMA in the mouse brain: membrane anchoring and accumulation of 'tailed' acetylcholinesterase
- Author
-
Sonia Khérif, Noël A. Perrier, Anselme L. Perrier, Jacques Mallet, Sylvie Dumas, and Jean Massoulié
- Subjects
Male ,Molecular Sequence Data ,Nerve Tissue Proteins ,In situ hybridization ,Biology ,Choline O-Acetyltransferase ,Mice ,chemistry.chemical_compound ,Postsynaptic potential ,Animals ,Humans ,RNA, Messenger ,Cholinergic neuron ,In Situ Hybridization ,Cholinesterase ,Mice, Inbred BALB C ,Reverse Transcriptase Polymerase Chain Reaction ,General Neuroscience ,Receptor, Muscarinic M1 ,Brain ,Membrane Proteins ,Acetylcholinesterase ,Molecular biology ,Choline acetyltransferase ,humanities ,Alternative Splicing ,Membrane protein ,chemistry ,biology.protein ,Autoradiography ,Cholinergic - Abstract
We analysed the expression of PRiMA (proline-rich membrane anchor), the membrane anchor of acetylcholinesterase (AChE), by in situ hybridization in the mouse brain. We compared the pattern of PRiMA transcripts with that of AChE transcripts, as well as those of choline acetyltransferase and M1 muscarinic receptors which are considered pre- and postsynaptic cholinergic markers. We also analysed cholinesterase activity and its molecular forms in several brain structures. The results suggest that PRiMA expression is predominantly or exclusively related to the cholinergic system and that anchoring of cholinesterases to cell membranes by PRiMA represents a limiting factor for production of the AChE tailed splice variant (AChET)-PRiMA complex, which represents the major AChE component in the brain. This enzyme species is mostly associated with cholinergic neurons because the pattern of PRiMA mRNA expression largely coincides with that of ChAT. We also show that, in both mouse and human, PRiMA proteins exist as two alternative splice variants which differ in their cytoplasmic regions.
- Published
- 2003
- Full Text
- View/download PDF
16. 174 Differentiation of non-human primate pluripotent stem cells into keratinocytes
- Author
-
Antoine Marteyn, Manoubia Saidani, Marc Peschanski, Cécile Martinat, Anselme L. Perrier, Yolande Masson, Gilles Lemaitre, Sophie Domingues, Jennifer Allouche, and Christine Baldeschi
- Subjects
Endothelial stem cell ,Non human primate ,Cell Biology ,Dermatology ,Embryoid body ,Biology ,Stem cell ,Induced pluripotent stem cell ,Molecular Biology ,Biochemistry ,Cell biology - Published
- 2016
- Full Text
- View/download PDF
17. Human embryonic stem cells and genomic instability
- Author
-
Christine Varela, Anselme L. Perrier, Yacine Laâbi, Marc Peschanski, and Nathalie Lefort
- Subjects
Genetics ,Genome instability ,Embryology ,Biomedical Engineering ,Karyotype ,Computational biology ,Biology ,Embryonic stem cell ,Regenerative medicine ,Genomic Instability ,Genomic Stability ,Humans ,Induced pluripotent stem cell ,Embryonic Stem Cells - Abstract
Owing to their original properties, pluripotent human embryonic stem cells (hESCs) and their progenies are highly valuable not only for regenerative medicine, but also as tools to study development and pathologies or as cellular substrates to screen and test new drugs. However, ensuring their genomic integrity is one important prerequisite for both research and therapeutic applications. Until recently, several studies about the genomic stability of cultured hESCs had described chromosomal or else large genomic alterations detectable with conventional karyotypic methods. In the past year, several laboratories have reported many small genomic alterations, in the megabase-sized range, using more sensitive karyotyping methods, showing that hESCs are prone to acquire focal genomic abnormalities in culture. As these alterations were found to be nonrandom, these findings strongly advocate for high-resolution monitoring of human pluripotent stem cell lines, especially when intended to be used for clinical applications
- Published
- 2009
18. PRiMA: the membrane anchor of acetylcholinesterase in the brain
- Author
-
Anselme L, Perrier, Jean, Massoulié, and Eric, Krejci
- Subjects
DNA, Complementary ,Proline ,Xenopus ,Cell Membrane ,Molecular Sequence Data ,Brain ,Membrane Proteins ,Muscle Proteins ,Nerve Tissue Proteins ,Protein Structure, Tertiary ,Mice ,Butyrylcholinesterase ,Acetylcholinesterase ,Oocytes ,Tumor Cells, Cultured ,Animals ,Collagen ,Muscle, Skeletal - Abstract
As a tetramer, acetylcholinesterase (AChE) is anchored to the basal lamina of the neuromuscular junction and to the membrane of neuronal synapses. We have previously shown that collagen Q (ColQ) anchors AChE at the neuromuscular junction. We have now cloned the gene PRiMA (proline-rich membrane anchor) encoding the AChE anchor in mammalian brain. We show that PRiMA is able to organize AChE into tetramers and to anchor them at the surface of transfected cells. Furthermore, we demonstrate that AChE is actually anchored in neural cell membranes through its interaction with PRiMA. Finally, we propose that only PRiMA anchors AChE in mammalian brain and muscle cell membranes.
- Published
- 2002
19. Allele-specific silencing of mutant huntingtin in rodent brain and human stem cells.
- Author
-
Valérie Drouet, Marta Ruiz, Diana Zala, Maxime Feyeux, Gwennaëlle Auregan, Karine Cambon, Laetitia Troquier, Johann Carpentier, Sophie Aubert, Nicolas Merienne, Fany Bourgois-Rocha, Raymonde Hassig, Maria Rey, Noëlle Dufour, Frédéric Saudou, Anselme L Perrier, Philippe Hantraye, and Nicole Déglon
- Subjects
Medicine ,Science - Abstract
Huntington's 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 rat's 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.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.