63 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. 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
4. 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
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. A Quantitative Approach to Characterize MR Contrasts with Histology.
- Author
-
Yaël Balbastre, Michel E. Vandenberghe, Anne-Sophie Hérard, Pauline Gipchtein, Caroline Jan, Anselme L. Perrier, Philippe Hantraye, Romina Aron-Badin, Jean-François Mangin, and Thierry Delzescaux
- Published
- 2015
- Full Text
- View/download PDF
12. Propagation of Distinct α-Synuclein Strains Within Human Reconstructed Neuronal Network and Associated Neuronal Dysfunctions
- Author
-
Simona Gribaudo, Luc Bousset, Josquin Courte, Ronald Melki, Jean-Michel Peyrin, Anselme L. Perrier, Institut du Fer à Moulin (IFM - Inserm U1270 - SU), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Service MIRCEN (MIRCEN), Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie François JACOB (JACOB), Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Laboratoire des Maladies Neurodégénératives - UMR 9199 (LMN), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie François JACOB (JACOB), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Centre National de la Recherche Scientifique (CNRS), Neuroscience Paris Seine (NPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie Paris Seine (IBPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), CEA- Saclay (CEA), Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut des cellules souches pour le traitement et l'étude des maladies monogéniques (I-STEM), and Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay-Généthon
- Subjects
[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] - Abstract
International audience; Abstract Cell motility is critical for tumor malignancy. Metabolism being an obligatory step in shaping cell behavior, we looked for metabolic weaknesses shared by motile cells across the diverse genetic contexts of patients’ glioblastoma. Computational analyses of single-cell transcriptomes from thirty patients’ tumors isolated cells with high motile potential and highlighted their metabolic specificities. These cells were characterized by enhanced mitochondrial load and oxidative stress coupled with mobilization of the cysteine metabolism enzyme 3-Mercaptopyruvate sulfurtransferase (MPST). Functional assays with patients’ tumor-derived cells and -tissue organoids, and genetic and pharmacological manipulations confirmed that the cells depend on enhanced ROS production and MPST activity for their motility. MPST action involved protection of protein cysteine residues from damaging hyperoxidation. Its knockdown translated in reduced tumor burden, and a robust increase in mice survival. Starting from cell-by-cell analyses of the patients’ tumors, our work unravels metabolic dependencies of cell malignancy maintained across heterogeneous genomic landscapes.
- Published
- 2022
- Full Text
- View/download PDF
13. Propagation of Distinct α-Synuclein Strains Within Human Reconstructed Neuronal Network and Associated Neuronal Dysfunctions
- Author
-
Simona, Gribaudo, Luc, Bousset, Josquin, Courte, Ronald, Melki, Jean-Michel, Peyrin, and Anselme L, Perrier
- Subjects
Neurons ,Synucleinopathies ,Prions ,alpha-Synuclein ,Humans ,Parkinson Disease - Abstract
Aggregated alpha-synuclein (α-Syn) in neurons is a hallmark of Parkinson's disease (PD) and other synucleinopathies. Recent advances (1) in the production and purification of synthetic assemblies of α-Syn, (2) in the design and production of microfluidic devices allowing the construction of oriented and compartmentalized neuronal network on a chip, and (3) in the differentiation of human pluripotent stem cells (hPSCs) into specific neuronal subtypes now allow the study of cellular and molecular determinants of the prion-like properties of α-Syn in vitro. Here, we described the methods we used to reconstruct a cortico-cortical human neuronal network in microfluidic devices and how to take advantage of this cellular model to characterize (1) the prion-like properties of different α-Syn strains and (2) the neuronal dysfunctions and the alterations associated with the exposure to α-Syn strains or the nucleation of endogenous α-Syn protein in vitro.
- Published
- 2022
14. I07 Selective lowering of mutant-huntingtin by antisense oligonucleotides (ASOs) in human cortical neurons derived from patient-specific induced pluripotent stem cells (HD-IPSC)
- Author
-
Laurent Cotter, Romane A Lahaye, Wei Chou Tseng, Yuanjing Liu, Aude Lemesle, Sophie Lenoir, Elena Dale, Frédéric Saudou, and Anselme L Perrier
- Published
- 2022
- Full Text
- View/download PDF
15. Modeling and Targeting Neuroglial Interactions with Human Pluripotent Stem Cell Models
- Author
-
Julie Bigarreau, Nathalie Rouach, Anselme L. Perrier, Franck Mouthon, Mathieu Charvériat, Theranexus [Lyon], Centre interdisciplinaire de recherche en biologie (CIRB), Labex MemoLife, École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Collège de France (CdF (institution))-Ecole Superieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris sciences et lettres (PSL)-École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut des cellules souches pour le traitement et l'étude des maladies monogéniques (I-STEM), and Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay-Généthon
- Subjects
neuroglial interactions ,QH301-705.5 ,Induced Pluripotent Stem Cells ,neurons ,microglia ,Catalysis ,Inorganic Chemistry ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Humans ,human pluripotent stem cells ,Physical and Theoretical Chemistry ,Biology (General) ,Molecular Biology ,QD1-999 ,Spectroscopy ,Organic Chemistry ,astrocytes ,Cell Differentiation ,Neurodegenerative Diseases ,General Medicine ,Computer Science Applications ,Chemistry ,Neurodevelopmental Disorders ,pathological modeling ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,Neuroglia - Abstract
International audience; Generation of relevant and robust models for neurological disorders is of main importance for both target identification and drug discovery. The non-cell autonomous effects of glial cells on neurons have been described in a broad range of neurodegenerative and neurodevelopmental disorders, pointing to neuroglial interactions as novel alternative targets for therapeutics development. Interestingly, the recent breakthrough discovery of human induced pluripotent stem cells (hiPSCs) has opened a new road for studying neurological and neurodevelopmental disorders “in a dish”. Here, we provide an overview of the generation and modeling of both neuronal and glial cells from human iPSCs and a brief synthesis of recent work investigating neuroglial interactions using hiPSCs in a pathophysiological context.
- Published
- 2022
- Full Text
- View/download PDF
16. Translating cell therapies for neurodegenerative diseases: Huntington's disease as a model disorder
- Author
-
Anne E. Rosser, Monica E. Busse, William P. Gray, Romina Aron Badin, Anselme L. Perrier, Vicki Wheelock, Emanuele Cozzi, Unai Perpiña Martin, Cristina Salado-Manzano, Laura J. Mills, Cheney Drew, Steven A. Goldman, Josep M. Canals, and Leslie M. Thompson
- Subjects
Huntington's Disease ,IMMUNE REJECTION ,Cell- and Tissue-Based Therapy ,Neurodegenerative ,Huntington's chorea ,Regenerative Medicine ,Medical and Health Sciences ,clinical translation ,POSITRON-EMISSION-TOMOGRAPHY ,Rare Diseases ,PARKINSONS-DISEASE ,Corea de Huntington ,stem cells ,NEURAL TRANSPLANTATION ,Genetics ,Animals ,Humans ,FETAL STRIATAL TRANSPLANTATION ,Huntington's ,Neurology & Neurosurgery ,5.2 Cellular and gene therapies ,Psychology and Cognitive Sciences ,Malalties neurodegeneratives ,neurodegeneration ,Neurosciences ,Brain ,Neurodegenerative Diseases ,DOPAMINERGIC-NEURONS ,RANDOMIZED-TRIALS ,Brain Disorders ,Huntington Disease ,Orphan Drug ,Neurological ,FUNCTIONAL RECOVERY ,Neurology (clinical) ,cell therapy ,Development of treatments and therapeutic interventions ,STEM-CELLS ,CLINICAL-TRIALS ,Huntington’s disease ,Biotechnology - Abstract
There has been substantial progress in the development of regenerative medicine strategies for CNS disorders over the last decade, with progression to early clinical studies for some conditions. However, there are multiple challenges along the translational pipeline, many of which are common across diseases and pertinent to multiple donor cell types. These include defining the point at which the preclinical data are sufficiently compelling to permit progression to the first clinical studies; scaling-up, characterization, quality control and validation of the cell product; design, validation and approval of the surgical device; and operative procedures for safe and effective delivery of cell product to the brain. Furthermore, clinical trials that incorporate principles of efficient design and disease-specific outcomes are urgently needed (particularly for those undertaken in rare diseases, where relatively small cohorts are an additional limiting factor), and all processes must be adaptable in a dynamic regulatory environment. Here we set out the challenges associated with the clinical translation of cell therapy, using Huntington's disease as a specific example, and suggest potential strategies to address these challenges. Huntington's disease presents a clear unmet need, but, importantly, it is an autosomal dominant condition with a readily available gene test, full genetic penetrance and a wide range of associated animal models, which together mean that it is a powerful condition in which to develop principles and test experimental therapeutics. We propose that solving these challenges in Huntington's disease would provide a road map for many other neurological conditions. This white paper represents a consensus opinion emerging from a series of meetings of the international translational platforms Stem Cells for Huntington's Disease and the European Huntington's Disease Network Advanced Therapies Working Group, established to identify the challenges of cell therapy, share experience, develop guidance, and highlight future directions, with the aim to expedite progress towards therapies for clinical benefit in Huntington's disease.Rosser et al. discuss the challenges associated with clinical translation of cell therapies, using Huntington's disease as the primary example. They suggest strategies to address these challenges, based on the consensus view emerging from international workshops and discussion within the platform 'Stem Cells for Huntington's Disease'.
- Published
- 2022
- Full Text
- View/download PDF
17. Cell Therapy for Huntington's Disease: Learning from Failure
- Author
-
Anne Elizabeth Rosser, Josep M. Canals, Monica Busse, Anselme L. Perrier, Steven A. Goldman, Vicki L. Wheelock, Romina Aron Badin, William P. Gray, and Leslie M. Thompson
- Subjects
Learning from failure ,business.industry ,Cell- and Tissue-Based Therapy ,MEDLINE ,Bioinformatics ,medicine.disease ,Cell therapy ,Huntington Disease ,Text mining ,Neurology ,Huntington's disease ,Humans ,Medicine ,Neurology (clinical) ,business - Published
- 2021
- Full Text
- View/download PDF
18. MHC matching fails to prevent long-term rejection of iPSC-derived neurons in non-human primates
- Author
-
Susannah Williams, Alberto Nassi, Philippe Hantraye, Aurore Bugi, Marie Michael, Sophie Lecourtois, Caroline Jan, Emanuele Cozzi, Antoine Blancher, Anselme L. Perrier, Romina Aron Badin, Marta Vadori, Service MIRCEN (MIRCEN), Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie François JACOB (JACOB), Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Laboratoire des Maladies Neurodégénératives - UMR 9199 (LMN), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie François JACOB (JACOB), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Centre National de la Recherche Scientifique (CNRS), Institut des cellules souches pour le traitement et l'étude des maladies monogéniques (I-STEM), Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay-Généthon, Consorzio per la Ricerca sul Trapianto d'Organ = Consortium for Research in Organ Transplantation (CORIT), Azienda Ospedale Università di Padova = Hospital-University of Padua (AOUP), Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université de Toulouse (UT)-Université de Toulouse (UT)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Agence Nationale pour la Recherche HD-SCT French National Research Agency (ANR) [ANR-2010-RFSC-003], ANR-11-INBS-0011,NeurATRIS,Infrastructure de Recherche Translationnelle pour les Biothérapies en Neurosciences(2011), ANR-10-LABX-0073,REVIVE,Stem Cells in Regenerative Biology and Medicine(2010), European Project: 602245,EC:FP7:HEALTH,FP7-HEALTH-2013-INNOVATION-1,REPAIR-HD(2013), Institut de Biologie François JACOB (JACOB), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Institut de Biologie François JACOB (JACOB), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Généthon-Université d'Évry-Val-d'Essonne (UEVE), Université Fédérale Toulouse Midi-Pyrénées, University Hospital of Padua, Consortium for Research in Organ Transplantation [Padua, Italy] (CORIT), Université Paris-Saclay-Institut de Biologie François JACOB (JACOB), Centre National de la Recherche Scientifique (CNRS)-Service MIRCEN (MIRCEN), Transplantation Immunology Unit [Padua, Italy] (Department of Transfusion Medicine), University of Padua–Ospedale Giustinianeo [Padua, Italy], Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay, Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Généthon, Hantraye, Philippe, Infrastructures - Infrastructure de Recherche Translationnelle pour les Biothérapies en Neurosciences - - NeurATRIS2011 - ANR-11-INBS-0011 - INBS - VALID, Laboratoires d'excellence - Stem Cells in Regenerative Biology and Medicine - - REVIVE2010 - ANR-10-LABX-0073 - LABX - VALID, and Human pluripotent stem cell differentiation, safety and preparation for therapeutic transplantation in Huntington’s disease - REPAIR-HD - - EC:FP7:HEALTH2013-10-01 - 2017-09-30 - 602245 - VALID
- Subjects
0301 basic medicine ,Cytotoxicity, Immunologic ,Graft Rejection ,Cytotoxicity ,medicine.medical_treatment ,[SDV]Life Sciences [q-bio] ,Nude ,MESH: Major Histocompatibility Complex / immunology ,General Physics and Astronomy ,Histocompatibility Testing ,Disease ,MESH: Cytotoxicity, Immunologic / immunology ,Cell therapy ,Major Histocompatibility Complex ,0302 clinical medicine ,Immunologic ,MESH: Animals ,lcsh:Science ,Induced pluripotent stem cell ,MESH: Rats, Nude ,Neurons ,Multidisciplinary ,biology ,Immune evasion ,Immunogenicity ,Immunosuppression ,Cell Differentiation ,Huntington's disease ,MESH: Huntington Disease / immunology ,MESH: Induced Pluripotent Stem Cells / transplantation ,MESH: Transplantation, Autologous ,3. Good health ,[SDV] Life Sciences [q-bio] ,MESH: Primates ,Induced pluripotent stem cells ,Huntington Disease ,MESH: Neurons / immunology ,Animals ,Disease Models, Animal ,Humans ,Induced Pluripotent Stem Cells ,Primates ,Rats, Nude ,Transplantation, Autologous ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,MESH: Cell Differentiation / immunology ,MESH: Huntington Disease / therapy ,Autologous ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,Science ,MESH: Histocompatibility Testing ,Major histocompatibility complex ,General Biochemistry, Genetics and Molecular Biology ,Article ,MESH: Induced Pluripotent Stem Cells / cytology ,03 medical and health sciences ,medicine ,[INFO.INFO-DL]Computer Science [cs]/Digital Libraries [cs.DL] ,MESH: Neurons / transplantation ,Transplantation ,MESH: Humans ,Animal ,business.industry ,MESH: Neurons / cytology ,General Chemistry ,Rats ,030104 developmental biology ,MESH: Graft Rejection / immunology ,Disease Models ,biology.protein ,lcsh:Q ,MESH: Disease Models, Animal ,MESH: Induced Pluripotent Stem Cells / immunology ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Cell therapy products (CTP) derived from pluripotent stem cells (iPSCs) may constitute a renewable, specifically differentiated source of cells to potentially cure patients with neurodegenerative disorders. However, the immunogenicity of CTP remains a major issue for therapeutic approaches based on transplantation of non-autologous stem cell-derived neural grafts. Despite its considerable side-effects, long-term immunosuppression, appears indispensable to mitigate neuro-inflammation and prevent rejection of allogeneic CTP. Matching iPSC donors’ and patients’ HLA haplotypes has been proposed as a way to access CTP with enhanced immunological compatibility, ultimately reducing the need for immunosuppression. In the present work, we challenge this paradigm by grafting autologous, MHC-matched and mis-matched neuronal grafts in a primate model of Huntington’s disease. Unlike previous reports in unlesioned hosts, we show that in the absence of immunosuppression MHC matching alone is insufficient to grant long-term survival of neuronal grafts in the lesioned brain., 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
19. CTIP2-regulated reduction in PKA-dependent DARPP32 phosphorylation in human medium spiny neurons: \ud implications for Huntington’s disease
- Author
-
Emma Dyke, Zongze Li, Marija Fjodorova, Simon Philip Brooks, Morgane Louessard, Anselme L. Perrier, Meng Li, and Daniel C. De La Fuente
- Subjects
0301 basic medicine ,Dopamine and cAMP-Regulated Phosphoprotein 32 ,Huntingtin ,neural differentiation ,Human Embryonic Stem Cells ,Biology ,Medium spiny neuron ,medium spiny neuron ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Report ,Genetics ,medicine ,Humans ,Protein phosphorylation ,Receptors, AMPA ,Phosphorylation ,Protein kinase A ,lcsh:QH301-705.5 ,Transcription factor ,Gene Editing ,Neurons ,lcsh:R5-920 ,Tumor Suppressor Proteins ,DARPP32 ,Neurodegeneration ,Cell Differentiation ,Cell Biology ,Huntington disease ,medicine.disease ,Cyclic AMP-Dependent Protein Kinases ,Corpus Striatum ,Cell biology ,Repressor Proteins ,Oxidative Stress ,030104 developmental biology ,lcsh:Biology (General) ,CTIP2 ,CRISPR-Cas Systems ,Transcriptome ,lcsh:Medicine (General) ,030217 neurology & neurosurgery ,Homeostasis ,Signal Transduction ,Developmental Biology - 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., Highlights • CTIP2 protects human MSNs against oxidative stress-induced cell death • PKA-regulated DARPP32 and GLUR1 phosphorylation is reduced in CTIP2-deficient MSNs • HD MSNs share CTIP2-dependent downregulation of protein phosphorylation by PKA, 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.
- Published
- 2019
20. Longitudinal characterization of cognitive and motor deficits in an excitotoxic lesion model of striatal dysfunction in non-human primates
- Author
-
Sébastien Goutal, Thierry Delzescaux, Romina Aron Badin, Julien Valette, Martine Guillermier, Nadja Van Camp, Sonia Lavisse, Pauline Gipchtein, Sophie Lecourtois, Anselme L. Perrier, Caroline Jan, Philippe Hantraye, Leopold Eymin, Susannah Williams, Laboratoire des Maladies Neurodégénératives - UMR 9199 (LMN), Service MIRCEN (MIRCEN), Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie François JACOB (JACOB), Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie François JACOB (JACOB), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Centre National de la Recherche Scientifique (CNRS), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut des cellules souches pour le traitement et l'étude des maladies monogéniques (I-STEM), Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Généthon, Institut National de la Santé et de la Recherche Médicale (INSERM), ANR-07-PRIB-0016,TK-safe,'L'interrupteur de secours': Assurer la sureté de produits de thérapie cellulaire grace à l' ablation sélective du greffon par le gène suicide TK(2007), ANR-11-INBS-0011,NeurATRIS,Infrastructure de Recherche Translationnelle pour les Biothérapies en Neurosciences(2011), ANR-10-RFCS-0003,HD-SCT,Evaluation préclinique d'une thérapie cellulaire de la maladie de Huntington fondée sur l'utilisation de cellules souches pluripotentes(2010), ANR-10-LABX-0073,REVIVE,Stem Cells in Regenerative Biology and Medicine(2010), European Project: 602245,EC:FP7:HEALTH,FP7-HEALTH-2013-INNOVATION-1,REPAIR-HD(2013), Centre National de la Recherche Scientifique (CNRS)-Service MIRCEN (MIRCEN), Université Paris-Saclay-Institut de Biologie François JACOB (JACOB), and Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Institut de Biologie François JACOB (JACOB)
- Subjects
Male ,0301 basic medicine ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Motor Disorders ,Caudate nucleus ,PET imaging ,Substantia nigra ,Striatum ,lcsh:RC321-571 ,Lesion ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Animals ,Medicine ,Cognitive Dysfunction ,Animal model ,Longitudinal Studies ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Movement disorder ,[SDV.NEU.PC]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Psychology and behavior ,business.industry ,Putamen ,[SCCO.NEUR]Cognitive science/Neuroscience ,Huntington's disease ,Quinolinic Acid ,Non-human primate ,Corpus Striatum ,Striatal dysfunction ,Dorsolateral prefrontal cortex ,Disease Models, Animal ,Macaca fascicularis ,Huntington Disease ,030104 developmental biology ,Globus pallidus ,medicine.anatomical_structure ,Cognitive impairment ,Neurology ,chemistry ,nervous system ,medicine.symptom ,business ,Neuroscience ,030217 neurology & neurosurgery ,Quinolinic acid - Abstract
International audience; As research progresses in the understanding of the molecular and cellular mechanisms underlying neurode-generative 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, transla-tional 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 pharma-cotherapy approaches in restoring the functionality of the striatal circuitry.
- Published
- 2019
- Full Text
- View/download PDF
21. Quantification of Total and Mutant Huntingtin Protein Levels in Biospecimens Using a Novel alphaLISA Assay
- Author
-
Catriona McLean, Glenda M. Halliday, Åsa Petersén, Deniz Kirik, Rachel Y. Cheong, Barbara Baldo, Anselme L. Perrier, Ravi Vijayvargia, Muhammad Umar Sajjad, Ihn Sik Seong, and Julie Bigarreau
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Huntingtin ,animal diseases ,Mutant ,polyglutamines ,Novel Tools and Methods ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Huntington's disease ,Neural Stem Cells ,mental disorders ,medicine ,Huntingtin Protein ,Animals ,Humans ,Induced pluripotent stem cell ,Methods/New Tools ,030304 developmental biology ,AlphaLISA ,Immunoassay ,0303 health sciences ,medicine.diagnostic_test ,Chemistry ,General Neuroscience ,HEK 293 cells ,Reproducibility of Results ,General Medicine ,medicine.disease ,Neural stem cell ,3. Good health ,Cell biology ,nervous system diseases ,Disease Models, Animal ,HEK293 Cells ,Huntington Disease ,nervous system ,7.2 ,Mutation ,030217 neurology & neurosurgery ,Huntington’s disease - Abstract
The neurodegenerative Huntington’s disease (HD) is caused by a polyglutamine (polyQ) amplification in the huntingtin protein (HTT). Currently there is no effective therapy available for HD; however, several efforts are directed to develop and optimize HTT-lowering methods to improve HD phenotypes. To validate these approaches, there is an immediate need for reliable, sensitive, and easily accessible methods to quantify HTT expression. Using the AlphaLISA platform, we developed two novel sensitive and robust assays for quantification of HTT in biological samples using commercially available antibodies. The first, a polyQ-independent assay, measures the total pool of HTT, while the second, a polyQ-dependent assay, preferentially detects the mutant form of HTT. Using purified HTT protein standards and brain homogenates from an HD mouse model, we determine a lower limit of quantification of 1 and 3 pmand optimal reproducibility with CV values lower than 7% for intra- and 20% for interassay. In addition, we used the assays to quantify HTT in neural stem cells generated from patient-derived induced pluripotent stem cellsin vitroand in human brain tissue lysates. Finally, we could detect changes in HTT levels in a mouse model where mutant HTT was conditionally deleted in neural tissue, verifying the potential to monitor the outcome of HTT-lowering strategies. This analytical platform is ideal for high-throughput screens and thus has an added value for the HD community as a tool to optimize novel therapeutic approaches aimed at modulating HTT protein levels.
- Published
- 2018
22. B16 Astrocytes derived from patient specific human pluripotent stem cells: a valuable biological resource for target identification in hd?
- Author
-
Åsa Petersén, Johana Tournois, Marie Michael, Gilles Bonvento, Margot Jarrige, Anselme L. Perrier, Barbara Baldo, Morgane Louessard, Deniz Kirik, Gurvan Mahé, and Julie Bigarreau
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Cell type ,nervous system ,biology ,High-throughput screening ,Lentivirus ,Immunocytochemistry ,Glutamate receptor ,Extracellular ,Gene silencing ,biology.organism_classification ,Induced pluripotent stem cell ,Cell biology - Abstract
Several studies have outlined a non-cell-autonomous contribution in Huntington Disease (HD). Mutant-HTT (mut-HTT) expressed solely in astrocytes is sufficient for recapitulating the neurological and peripheral symptoms of HD in mice. Our objective is to identify molecules that can rescue cellular dysfunctions mediated by mut-HTT in astrocytes derived from human pluripotent stem cells (hPSC). Herein, we describe a scalable and reproducible protocol to differentiate HD-hPSC into astrocytes. The cellular identity of these hPSC-derived glial cells was characterized by qPCR and immunocytochemistry (GFAP, S100B, Connexin-43, GLT-1). Astrocytes are the cell type primarily responsible for rapid removal of glutamate from the extracellular space. This process is required for the survival and normal function of neurons. We therefore measure the capacity of the hPSC-derived glial cells we generated to glutamate uptake in a sodium-dependent manner. Assessment of previously described dysfunctions in HD-hPSC-derived astrocytes is ongoing. Next, we addressed the issue of the quantification in HD-hPSC neural derivatives of the level of mut-HTT. We adapted a mut-HTT-targeting AlphaLISA assay for use in 384-well plate format for high throughput screening. This assay is CAG dependent and can measure mut-HTT in a broad variety of human samples. We demonstrated the specificity of this assay using allele-specific silencing of the mut-HTT (lentivirus and siRNA) and comparing WT and HD-hPSC-derived samples. This assay displays a good signal-to-background ratio and is sensitive enough to reliably detect mut in as few as 5000 HD cells. The feasibility of performing drug screening on neuronal or glial cells derived from HD-hPSC using this assay is currently tested.
- Published
- 2018
- Full Text
- View/download PDF
23. I07 Allele specific gene editing for huntington’s disease mediated by the KAMICAS9 self-inactivating CRISPR/CAS9 system
- Author
-
Nicole Déglon, Gabriel Vachey, Anselme L. Perrier, and Maria Rey
- Subjects
Genetics ,congenital, hereditary, and neonatal diseases and abnormalities ,Exon ,Huntingtin ,Genome editing ,Cas9 ,mental disorders ,Mutant ,HEK 293 cells ,Intron ,CRISPR ,Biology ,nervous system diseases - Abstract
Huntington’s disease (HD) is a fatal neurodegenerative disorder caused by CAG expansion in the huntingtin (HTT) gene. Considering that the mutation is a toxic gain-of-function, a promising approach would be to decrease the expression level of the mutant HTT. This can be achieved with genome-editing technologies, in particular the recently characterized CRISPR/Cas9 system. In a previous work, we described the kamiCas9, a self-inactivating CRISPR/Cas9 system designed for the transient expression of the Cas9 protein. We demonstrated the high editing efficiency of this system both in in vitro and in vivo HD models with an important reduction of the off-target frequency. However, a selective editing of mutant HTT, using an allele-specific approach, represents the safest way to preserve WT HTT expression and functions. We thus developed more complex strategies to discriminate mutant and wild-type HTT genes by using single-guide RNA targeting sequences containing Single Nucleotide Polymorphism (SNP) in the HTT gene. A first in vitro screening, allow us to discriminate the two best candidates to trigger the cleavage of the mutant HTT respectively in the promoter and intron 1. Through this approach the exon 1 of the mutant HTT, which is the region containing the CAG expansion, could be selective removed. These strategies have then been validated in human embryonic kidney 293T (HEK- 293T) cells and are currently tested in HD mouse models. These results demonstrate the potential of the self-inactivating CRISPR/Cas9 editing for applications in the context of neurodegenerative diseases and a proof of principle of allele specific disruption of the human HTT gene.
- Published
- 2018
- Full Text
- View/download PDF
24. A40 Modulation of DARPP32 homeostasis by htt protein in derivatives of disease-specific and control human pluripotent stem cells
- Author
-
Margot Jarrige, Julie Bigarreau, Gabriel Vachey, Morgane Louessard, Nicole Déglon, Anselme L. Perrier, and Michel Cailleret
- Subjects
Gene isoform ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.diagnostic_test ,Body movement ,Biology ,Medium spiny neuron ,Cell biology ,Small hairpin RNA ,nervous system ,Western blot ,RNA interference ,Phosphoprotein ,mental disorders ,medicine ,Induced pluripotent stem cell - Abstract
In Huntington disease (HD), the most affected cells are the GABA-releasing medium spiny neurons (MSN) of the striatum, the subcortical brain structure that controls body movement. DARPP32 (dopamine- and 3’,5’-cyclic adenosine monophosphate-regulated phosphoprotein, 32 kDa) is a class defining protein marker for striatal MSNs and a central mediator of dopamine signalling and other first messengers in these cells. DARPP-32 is expressed in 97% of the MSNs, several cortical layers, and cerebellar Purkinje cells. This protein has the capacity to function as either a kinase or a phosphatase inhibitor, depending on the phosphorylation state of key amino acid residues. DARPP32 is highly down-regulated in the striatum of HD patient and in the majority of mouse models of HD. The rational of our work is to use derivatives of several types of HD and WT-human Pluripotent Stem Cell (hPSC) lines challenged or not with HTT-targeting RNA interference (shRNA lentiviral vectors) or HTT-targeting CRISPR-Cas9 complex to decipher possible wt-HTT dosage, mut-HTT dominant-negative, mut-HTT de novo toxic or CAG repeat length effects on DARPP32 homeostasis. Using lentiviral vectors, we investigated the role of HTT isoforms on DARPP-32 protein levels measured by Western Blot and immunocytochemistry. We observed changes in DARPP32 and phopsho-DARPP32 protein level in hPSC derivatives treated with shHTT lentivirus. Next, we generated a series of CRISPER-CAS9 HTT-gene edited clones of HD-iPSC and WT-iPSC. Either or both allele of HTT gene was inactivated in those cells and the MSN derived from those clones. Exploration of DARPP32 protein levels in undifferentiated cells and MSN derived from these clones is ongoing. This study should help understand the role of HTT isoforms on human MSN homeostasis.
- Published
- 2018
- Full Text
- View/download PDF
25. I21 Functional assessment of grafted human embryonic stem cells-derived progenitors in a rat model of huntington’s disease
- Author
-
Marija Fjodorova, Anne Elizabeth Rosser, Meng Li, Marie Michael, Ana Garcia, Aurore Bugi, Stephen B. Dunnett, Anselme L. Perrier, Ngoc-Nga Vinh, and Mariah Jillian Lelos
- Subjects
Striatum ,Biology ,Medium spiny neuron ,medicine.disease ,Embryonic stem cell ,Transplantation ,Cell therapy ,chemistry.chemical_compound ,chemistry ,Huntington's disease ,medicine ,Progenitor cell ,Neuroscience ,Quinolinic acid - Abstract
Cell replacement therapies in Huntington’s disease (HD) aim to repair the nervous system by reintroducing the previously degenerated medium spiny neurons (MSNs) of the striatum and thereby restoring efferent and afferent projections. Despite significant advances in the generation of efficient protocols to differentiate pluripotent cells into authentic MSNs little is known about their functional efficacy. This study aimed to evaluate the survival, safety, integration, connectivity and functional efficacy of a hESC-derived cell therapy product (CTP) that was designed to be enriched for MSNs progenitors. Human CTP was stereotaxically transplanted into the striatum of a rat quinolinic acid model of HD. Rodents are treated with cyclosporine to prevent rejection and are being assessed on a range of behavioural tests sensitive to both motor and non-motor impairments over a period of 27 weeks to allow graft differentiation and development. While this is an on-going experiment, the following has been established with the current hESC-derived MSN CTP: (1) it consistently survives following transplantation into the rodent brain (as evidenced by bioluminescence imaging) and expresses markers of MSN differentiation (e.g., Darpp-32, CTIP2); (2) is safe (i.e., no evidence of neural overgrowth); (3) it is capable of forming functional synapses with the host brain and (4) and there is preliminary evidence of reduced motor bias in HD rats grafted with the hESC-derived CTP.
- Published
- 2018
- Full Text
- View/download PDF
26. I24 MHC matching fails to prevent long-term rejection of ipsc-derived neurons in non-human primate
- Author
-
Anselme L. Perrier, Marie Michael, Romina Aron Badin, Marta Vadori, Sophie Lecourtois, Philippe Hantraye, Caroline Jan, Emanuele Cozzi, Susannah Williams, Alberto Nassi, and Aurore Bugi
- Subjects
biology ,business.industry ,medicine.medical_treatment ,Immunosuppression ,Human leukocyte antigen ,Major histocompatibility complex ,Cell therapy ,Transplantation ,Immunology ,MHC class I ,biology.protein ,Medicine ,Stem cell ,business ,CD8 - Abstract
Securing scalable sources of cell therapy products with enhanced or full immunological compatibility could be achieved by matching haplotypes of iPSC donors (HLA homozygous lines). Recent experiments have shown that major histocompatibility complex (MHC) matching could be a solution for allogeneic stem cell transplantation in the retina#1 and in the striatum#2 of non-lesioned non-human primates (NHP) without immunosuppressive medication for 6 and 4 months post-transplantation, respectively. Here we present a study in which we challenged the efficacy of MHC matching in long-term allograft rejection in a NHP model of Huntington’s disease. We performed a comparative assessment of the immunogenicity of autologous, haplotype-matched and two-haplotype mismatched neuronal grafts in the excitotoxically-lesioned striatum of NHP. First, blood cells from different NHPs homozygous for MHC Class I&II were used to produce several iPSC lines that were subsequently differentiated into striatal cells. Next we assessed their potential immunogenicity at 3 and/or 6 months after intra-striatal grafting in haplotype mis-matched, matched and autologous NHP recipients. Our results suggest that, unlike autologous neuronal grafts, allogenic and haplotype-matched grafts elicit a local infiltration of CD8+ T cells, CD68+ macrophages cells and an increase in local Iba1 and HLA-DR staining. Serum levels of antibodies against all 3 types of grafted cells and their ability to trigger complement dependent cytotoxicity (CDC) in vitro were measured longitudinally and no humoral response or CDC activity were elicited by the graft at any time after transplantation. In the specific context of transplantation in the brain, our pre-clinical data suggest that, HLA matching alone is insufficient to grant long-term graft survival but could be a cost effective compromise to reduce peripheral immunosuppression and its side effects in the clinical setting.
- Published
- 2018
- Full Text
- View/download PDF
27. Differentiation of nonhuman primate pluripotent stem cells into functional keratinocytes
- Author
-
Cécile Martinat, Yolande Masson, Jennifer Allouche, Sophie Domingues, Aurore Marteyn, Gilles Lemaitre, Christine Baldeschi, Marc Peschanski, and Anselme L. Perrier
- Subjects
Keratinocytes ,Pluripotent Stem Cells ,Skin graft ,0301 basic medicine ,medicine.medical_treatment ,Short Report ,Medicine (miscellaneous) ,Human skin ,Context (language use) ,Biology ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,lcsh:Biochemistry ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,lcsh:QD415-436 ,Induced pluripotent stem cell ,lcsh:R5-920 ,Epidermis (botany) ,Cell Differentiation ,Haplorhini ,Cell Biology ,Embryonic stem cell ,Cell biology ,030104 developmental biology ,Nonhuman primate model ,Molecular Medicine ,Skin grafting ,Stem cell ,lcsh:Medicine (General) ,030217 neurology & neurosurgery - 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. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0741-9) contains supplementary material, which is available to authorized users.
- Published
- 2017
- Full Text
- View/download PDF
28. Human ESC-Derived Dopamine Neurons Show Similar Preclinical Efficacy and Potency to Fetal Neurons when Grafted in a Rat Model of Parkinson’s Disease
- Author
-
Philippe Hantraye, Bengt Mattsson, Shane Grealish, Elsa Diguet, Andreas Heuer, Malin Parmar, Agnete Kirkeby, Yann Bramoullé, Anselme L. Perrier, Nadja Van Camp, and Anders Björklund
- Subjects
Male ,Parkinson's disease ,Time Factors ,Cell Survival ,Substantia nigra ,Biology ,Neurotransmission ,Synaptic Transmission ,Midbrain ,Rats, Nude ,Fetus ,Dopamine ,Mesencephalon ,Genetics ,medicine ,Potency ,Animals ,Humans ,Embryonic Stem Cells ,Otx Transcription Factors ,Dopaminergic Neurons ,Parkinson Disease ,Cell Biology ,Anatomy ,medicine.disease ,Embryonic stem cell ,Axons ,3. Good health ,Neostriatum ,Substantia Nigra ,Disease Models, Animal ,nervous system ,Molecular Medicine ,Clinical Progress ,Neuroscience ,medicine.drug - Abstract
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., Graphical Abstract, Highlights • Transplants of hESC-DA survive long term and restore DA neurotransmission in vivo • The functional potency of hESC-DA is similar to human fetal midbrain DA neurons • hESC-DA are capable of long-distance, target-specific innervation of the host brain • The axonal outgrowth capacity of hESC-DA meets the requirements for use in humans, Grealish et al. provide preclinical evidence that hESC-derived dopamine neurons are functionally equivalent to those derived from fetal tissue, supporting continued development of hESC-derived cells as a clinical approach for the treatment of Parkinson’s disease.
- Published
- 2014
29. 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
30. 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
31. 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
Cerebral Cortex ,Gene Editing ,Neurons ,Huntingtin Protein ,Base Sequence ,Induced Pluripotent Stem Cells ,lentiviral vectors ,self-inactivating system ,Kinetics ,Mice ,KamiCas9 ,HEK293 Cells ,lcsh:Biology (General) ,Central Nervous System Diseases ,Astrocytes ,Animals ,Astrocytes/cytology ,Astrocytes/metabolism ,CRISPR-Cas Systems/genetics ,Cells, Cultured ,Central Nervous System Diseases/genetics ,Cerebral Cortex/cytology ,Humans ,Huntingtin Protein/genetics ,Induced Pluripotent Stem Cells/cytology ,Induced Pluripotent Stem Cells/metabolism ,Neurons/cytology ,Neurons/metabolism ,CRISPR/Cas9 ,Huntington’s disease ,gene editing ,neurodegenerative diseases ,CRISPR-Cas Systems ,lcsh: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
- 2016
32. Human Pluripotent Stem Cell-derived Cortical Neurons for High Throughput Medication Screening in Autism: A Proof of Concept Study in SHANK3 Haploinsufficiency Syndrome
- Author
-
Aurélie Poulet, Alexandra Benchoua, Richard Delorme, Laure Chatrousse, Francis Cogé, Delphine Héron, Frédérique Rodet-Amsellem, Millan Mark, Claire Boissart, Thomas Bourgeron, Hélène Darville, Anselme L. Perrier, Caroline Nava, Julie Pernelle, Margot Jarrige, Marc Peschanski, Institut des cellules souches pour le traitement et l'étude des maladies monogéniques (I-STEM), Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Généthon, French Biodiversity Agency, Service psychiatrique de l'enfant et de l'adolescent [CHU Hôpital Robert Debré], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Robert Debré, Hydrosystèmes et Bioprocédés (UR HBAN), Centre national du machinisme agricole, du génie rural, des eaux et forêts (CEMAGREF), CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Groupe de Recherche Clinique : Déficience Intellectuelle et Autisme (GRC), Université Pierre et Marie Curie - Paris 6 (UPMC), Institut du Cerveau et de la Moëlle Epinière = Brain and Spine Institute (ICM), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Division of General Internal Medicine (DGIM), Université de Genève = University of Geneva (UNIGE), Institut de Recherche Servier, SERVIER, Génétique humaine et fonctions cognitives - Human Genetics and Cognitive Functions (GHFC (UMR_3571 / U-Pasteur_1)), Institut Pasteur [Paris] (IP)-Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Institut Pasteur [Paris] (IP), AP-HP Hôpital universitaire Robert-Debré [Paris], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), This study has been in part funded by grants from 'Investissements d'Avenir' – (ANR-11-INBS-0009 - INGESTEM – and ANR-11-INBS-0011 - NeurATRIS), the French National Research Agency ANR (ANR-13-SAMA-0006, SynDivAutism), the Laboratory of Excellence GENMED (ANR-10-LABX-0013), the Bettencourt-Schueller Foundation, the Cognacq Jay Foundation, and the Fondamental Foundation. This study used samples from the NINDS Human Genetics Resource Center DNA and Cell Line Repository, as well as clinical data. NINDS Repository sample numbers corresponding to the samples used are GM 1869., The authors are thankful to I-Stem's HTS platform staff for constant technical support. We thank the cell bank of Pitié-Salpétrière hospital and the Clinical Investigation Center of Robert Debré hospital for assistance with patient recruitment, information, sampling and fibroblast preparation. H.D. received a PhD grant from Servier's Laboratories. I-Stem is part of the Biotherapies Institute for Rare Diseases (BIRD) supported by the Association Française contre les Myopathies (AFM-Téléthon)., ANR-11-INBS-0009,INGESTEM,INFRASTRUCTURE NATIONALE D'INGENIERIE DES CELLULES SOUCHES PLURIPOTENTES(2011), ANR-11-INBS-0011,NeurATRIS,Infrastructure de Recherche Translationnelle pour les Biothérapies en Neurosciences(2011), ANR-13-SAMA-0006,SynDivAutism,Diversité Synaptique dans l'autisme(2013), ANR-10-LABX-0013,GENMED,Medical Genomics(2010), Service de Génétique Cytogénétique et Embryologie [CHU Pitié-Salpêtrière], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), d'Eggis, Gilles, Infrastructures - INFRASTRUCTURE NATIONALE D'INGENIERIE DES CELLULES SOUCHES PLURIPOTENTES - - INGESTEM2011 - ANR-11-INBS-0009 - INBS - VALID, Infrastructures - Infrastructure de Recherche Translationnelle pour les Biothérapies en Neurosciences - - NeurATRIS2011 - ANR-11-INBS-0011 - INBS - VALID, Santé Mentale et Addictions - Diversité Synaptique dans l'autisme - - SynDivAutism2013 - ANR-13-SAMA-0006 - SAMENTA - VALID, Medical Genomics - - GENMED2010 - ANR-10-LABX-0013 - LABX - VALID, Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM), Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-Hôpital Robert Debré, CHU Pitié-Salpêtrière [APHP], Service de Génétique et Cytogénétique [CHU Pitié-Salpêtrière], Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-CHU Pitié-Salpêtrière [APHP], Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-CHU Pitié-Salpêtrière [APHP], University of Geneva [Switzerland], Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS)-Institut Pasteur [Paris], Institut Pasteur [Paris], ANR-11-INBS-0009/11-INBS-0009,INGESTEM,INFRASTRUCTURE NATIONALE D'INGENIERIE DES CELLULES SOUCHES PLURIPOTENTES(2011), ANR-11-INBS-0011/11-INBS-0011,NeurATRIS,Infrastructure de Recherche Translationnelle pour les Biothérapies en Neurosciences(2011), ANR: PREFI-10-LABX-13/10-LABX-0013,GENMED,Medical Genomics(2010), and Institut Pasteur [Paris]-Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Male ,GAS, Global Assessment Scale ,0301 basic medicine ,VPA, valproic acid ,Autism Spectrum Disorder ,Cellular differentiation ,Autism ,Human Embryonic Stem Cells ,Drug repurposing ,MESH: Neurons ,lcsh:Medicine ,hESC, human embryonic stem cells ,Haploinsufficiency ,Severity of Illness Index ,RPM, Raven's progressive matrices ,MESH: Nerve Tissue Proteins ,MESH: Neuronal Plasticity ,Induced pluripotent stem cell ,SHANK3 ,Cells, Cultured ,ASD, autism spectrum disorders ,Neurons ,ADOS, Autism Diagnosis Observational Scale ,Genetics ,MESH: Autism Spectrum Disorder ,lcsh:R5-920 ,Neuronal Plasticity ,PPVT, Peabody Picture Vocabulary Test ,PSC, pluripotent stem cells ,Cell Differentiation ,General Medicine ,Neural stem cell ,3. Good health ,Drug repositioning ,Phenotype ,Autism spectrum disorder ,MESH: Pluripotent Stem Cells ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,MESH: Haploinsufficiency ,lcsh:Medicine (General) ,Research Paper ,MESH: Cells, Cultured ,Pluripotent Stem Cells ,MESH: Cell Differentiation ,[SDV.SP.MED] Life Sciences [q-bio]/Pharmaceutical sciences/Medication ,Nerve Tissue Proteins ,ADHD, Attention Deficit Hyperactive Disorder ,Lithium ,SHANK3, SH3 and multiple ankyrin repeat domains 3 ,MESH: Phenotype ,General Biochemistry, Genetics and Molecular Biology ,iPSC, induced pluripotent stem cells ,03 medical and health sciences ,[SDV.SP.MED]Life Sciences [q-bio]/Pharmaceutical sciences/Medication ,High throughput screening ,MESH: Severity of Illness Index ,medicine ,Humans ,NSC, neural stem cells ,RNA, Messenger ,[SDV.NEU] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,HTS, high throughput screening ,MESH: RNA, Messenger ,Valproate ,MESH: Humans ,MESH: Lithium ,business.industry ,Valproic Acid ,MESH: Transcriptome ,lcsh:R ,MESH: Human Embryonic Stem Cells ,medicine.disease ,FDA, Food and Drug Administration ,Embryonic stem cell ,MESH: Male ,DMSO, dimethylsulfoxyde ,PPIA, Peptidylprolyl Isomerase A ,030104 developmental biology ,SRS, Social Responsiveness Scale ,Transcriptome ,business ,Neuroscience ,MESH: Valproic Acid - Abstract
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., Highlights • Human neurons were used to screen for compounds correcting symptoms associated with SHANK3 haploinsufficiency syndrome. • Screening criteria were the ability to increase SHANK3 expression and to increase glutamatergic transmission. • Selected hit compounds were then validated using neurons differentiated from individuals with SHANK3 disrupting mutations. • Lithium was selected and delivered to one of SHANK3 patient showing encouraging positive clinical outcomes after one year. The clinical heterogeneity between individuals affected by autism makes it difficult to anticipate the effectiveness of a treatment. Furthermore, clinical practice lacks biological tools to help make such decisions. Here we use neurons, produced from pluripotent stem cells derived from patients affected by SHANK3 haploinsufficiency syndrome, to test the efficiency of therapeutic compounds. We screened the biological activity of more than 200 compounds on SHANK3 expression. Lithium was ultimately selected and delivered to one patient with a SHANK3-disruptive mutation. This resulted in a positive outcome, as determined by improved autistic core symptoms, thus supporting the usefulness of this type of predictive approach.
- Published
- 2016
- Full Text
- View/download PDF
33. How Can Human Pluripotent Stem Cells Help Decipher and Cure Huntington’s Disease?
- Author
-
Anselme L. Perrier and Marc Peschanski
- Subjects
Pluripotent Stem Cells ,Disease ,Biology ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,Huntington's disease ,medicine ,Genetics ,Humans ,Induced pluripotent stem cell ,030304 developmental biology ,0303 health sciences ,business.industry ,Drug discovery ,Genetic Therapy ,Cell Biology ,medicine.disease ,3. Good health ,Biotechnology ,Huntington Disease ,DECIPHER ,Molecular Medicine ,business ,Neuroscience ,030217 neurology & neurosurgery ,Stem Cell Transplantation - Abstract
Pluripotent stem cell (PSC) technologies are becoming a key asset for deciphering pathological cascades and for developing new treatments against many neurodegenerative disorders, including Huntington’s 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.
- Published
- 2012
- Full Text
- View/download PDF
34. Human Pluripotent Stem Cell Therapy for Huntington’s Disease: Technical, Immunological, and Safety Challenges
- Author
-
Marc Peschanski, Anselme L. Perrier, Camille Nicoleau, and Pedro Viegas
- Subjects
Pharmacology ,0303 health sciences ,business.industry ,Neurogenesis ,medicine.disease ,Embryonic stem cell ,Neural stem cell ,3. Good health ,Cell therapy ,Transplantation ,03 medical and health sciences ,0302 clinical medicine ,Huntington's disease ,medicine ,Pharmacology (medical) ,Neurology (clinical) ,Induced pluripotent stem cell ,business ,Neuroscience ,Reprogramming ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Intra-striatal transplantation of homotypic fetal tissue at the time of peak striatal neurogenesis can provide some functional benefit to patients suffering from Huntington’s disease. Currently, the only approach shown to slow down the course of this condition is replacement of the neurons primarily targeted in this disorder, although it has been transient and has only worked with a limited number of patients. Otherwise, this dominantly inherited neurodegenerative disease inevitably results in the progressive decline of motricity, cognition, and behavior, and leads to death within 15 to 20 years of onset. However, fetal neural cell therapy of Huntington’s disease, as with a similar approach in Parkinson’s disease, is marred with both technical and biological hurdles related to the source of grafting material. This heavily restricts the number of patients who can be treated. A substitute cell source is therefore needed, but must perform at least as well as fetal neural graft in terms of brain recovery and reconstruction, while overcoming its major obstacles. Human pluripotent stem cells (embryonic in origin or induced from adult cells through genetic reprogramming) have the potential to meet those challenges. In this review, the therapeutic potential in view of 4 major issues is identified during fetal cell therapy clinical trials: 1) logistics of graft procurement, 2) quality control of the cell preparation, 3) immunogenicity of the graft, and 4) safety of the procedure. Electronic supplementary material The online version of this article (doi:10.1007/s13311-011-0079-4) contains supplementary material, which is available to authorized users.
- Published
- 2011
- Full Text
- View/download PDF
35. Improvement of Culture Conditions of Human Embryoid Bodies Using a Controlled Perfused and Dialyzed Bioreactor System
- Author
-
Marc Peschanski, Laetitia Aubry, Johana Tournois, Julien Côme, Anselme L. Perrier, Xavier Nissan, Michel Cailleret, and Mathilde Girard
- Subjects
Embryonic Induction ,Neurons ,Tissue Engineering ,Cell Survival ,Stem Cells ,Cell Culture Techniques ,Biomedical Engineering ,Medicine (miscellaneous) ,Environment controlled ,Cell Differentiation ,Bioengineering ,Equipment Design ,Embryoid body ,Hydrogen-Ion Concentration ,Biology ,Embryo, Mammalian ,Flow Cytometry ,Embryonic stem cell ,Cell Line ,Bioreactors ,Bioreactor ,Humans ,Cell Division ,Biomedical engineering - Abstract
In parallel to the active search for therapeutic and industrial applications of human embryonic stem cells (hESCs), designing automated means of producing those cells is a timely goal. Slow-turning lateral vessels (STLVs) with low shear stress have shown promise for expanding the cells at the embryoid body stage. We have improved this technology by developing two complementary systems, allowing continuous optimization of the culture conditions. First, perfused STLV bioreactors were set up, to provide continuous delivery of culture medium to the cells growing in the rotating chamber. This allowed the external control of the culture medium, and consequently optimized oxygenation, pH, nutrient supply, and waste elimination. Second, a dialysis chamber was adapted. This led to a further enhanced controlled environment and a decrease in the quantity of adjunct products (e.g., growth factors) necessary to the cells inside the bioreactor chamber. hESC aggregation and initial differentiation-taking neural induction as an example-were compared between the perfused and dialyzed STLV system and static cultures. Perfused and dialyzed STLV bioreactors promoted formation of embryoid bodies that were differentiated more rapidly and were homogeneously synchronized in a statistically significant manner.
- Published
- 2008
- Full Text
- View/download PDF
36. 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
37. A Quantitative Approach to Characterize MR Contrasts with Histology
- Author
-
Yaël Balbastre, Pauline Gipchtein, Caroline Jan, Romina Aron-Badin, Thierry Delzescaux, Anne-Sophie Hérard, Philippe Hantraye, Michel E. Vandenberghe, Anselme L. Perrier, and Jean-François Mangin
- Subjects
Ground truth ,Modalities ,medicine.diagnostic_test ,Computer science ,business.industry ,Histology ,Pattern recognition ,Gold standard (test) ,computer.software_genre ,Discriminative model ,Positron emission tomography ,Voxel ,Null distribution ,medicine ,Artificial intelligence ,business ,computer - Abstract
Immunohistochemistry is widely used as a gold standard to inspect tissues, characterize their structure and detect pathological alterations. As such, the joint analysis of histological images and other imaging modalities (MRI, PET) is of major interest to interpret these physical signals and establish their correspondence with the biological constitution of the tissues. However, it is challenging to provide a meaningful characterization of the signal specificity. In this paper, we propose an integrated method to quantitatively evaluate the discriminative power of imaging modalities. This method was validated using a macaque brain dataset containing: 3 immunohistochemically stained and 1 histochemically stained series, 1 photographic volume and 1 in vivo T2 weighted MRI. First, biological regions of interest (ROIs) were automatically delineated from histological sections stained for markers of interest and mapped on the target non-specific modalities through co-registration. These non-overlapping ROIs were considered ground truth for later classification. Voxels were evenly split in training and testing sets for a logistic regression model. The statistical significance of resulting accuracy scores was evaluated through null distribution simulations. Such an approach could be of major interest to assess relevant biological characteristics from various imaging modalities.
- Published
- 2016
- Full Text
- View/download PDF
38. 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
39. Derivation of midbrain dopamine neurons from human embryonic stem cells
- Author
-
Viviane Tabar, Anselme L. Perrier, Tiziano Barberi, Lorenz Studer, Norbert Topf, Maria E. Rubio, Juan Bruses, and Neil L. Harrison
- Subjects
Primates ,Nervous system ,Cell type ,Dopamine ,Cellular differentiation ,Cell Culture Techniques ,Biology ,Cell Line ,Mice ,Mesencephalon ,medicine ,Animals ,Humans ,Neurons ,Multidisciplinary ,Stem Cells ,Cell Differentiation ,Anatomy ,Biological Sciences ,Embryo, Mammalian ,Embryonic stem cell ,Neural stem cell ,Phenotype ,medicine.anatomical_structure ,nervous system ,Neuron ,Stromal Cells ,Stem cell ,Neuroscience ,Developmental biology - Abstract
Human embryonic stem (hES) cells are defined by their extensive self-renewal capacity and their potential to differentiate into any cell type of the human body. The challenge in using hES cells for developmental biology and regenerative medicine has been to direct the wide differentiation potential toward the derivation of a specific cell fate. Within the nervous system, hES cells have been shown to differentiate in vitro into neural progenitor cells, neurons, and astrocytes. However, to our knowledge, the selective derivation of any given neuron subtype has not yet been demonstrated. Here, we describe conditions to direct hES cells into neurons of midbrain dopaminergic identity. Neuroectodermal differentiation was triggered on stromal feeder cells followed by regional specification by means of the sequential application of defined patterning molecules that direct in vivo midbrain development. Progression toward a midbrain dopamine (DA) neuron fate was monitored by the sequential expression of key transcription factors, including Pax2, Pax5, and engrailed-1 (En1), measurements of DA release, the presence of tetrodotoxin-sensitive action potentials, and the electron-microscopic visualization of tyrosinehydroxylase-positive synaptic terminals. High-yield DA neuron derivation was confirmed from three independent hES and two monkey embryonic stem cell lines. The availability of unlimited numbers of midbrain DA neurons is a first step toward exploring the potential of hES cells in preclinical models of Parkinson's disease. This experimental system also provides a powerful tool to probe the molecular mechanisms that control the development and function of human midbrain DA neurons.
- Published
- 2004
- Full Text
- View/download PDF
40. 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
41. Regenerative medicine in Huntington's disease: current status on fetal grafts and prospects for the use of pluripotent stem cell
- Author
-
Anne-Catherine Bachoud-Lévi and Anselme L. Perrier
- Subjects
Pluripotent Stem Cells ,medicine.medical_specialty ,Disease ,Bioinformatics ,Regenerative Medicine ,Regenerative medicine ,Cell therapy ,Therapeutic approach ,Neuroblast ,Huntington's disease ,Neural Stem Cells ,Fetal Tissue Transplantation ,medicine ,Disease Transmission, Infectious ,Animals ,Humans ,Induced pluripotent stem cell ,business.industry ,medicine.disease ,Surgery ,Transplantation ,Huntington Disease ,Neurology ,Blood Group Incompatibility ,Neurology (clinical) ,business ,Stem Cell Transplantation - Abstract
Huntington's disease is currently incurable, but cell therapy is seen as a promising alternative treatment. We analyze the safety and efficacy of the intrastriatal transplantation of human fetal neuroblasts from ganglionic eminences in patients with Huntington's disease. A few rare surgical incidents were reported, but the main difficulty associated with this therapeutic approach is the occurrence of recipient alloimmunization against the graft and the lack of availability, standardization and quality control for the fetus-derived products required for cell therapy. Some patients showed sustained cognitive improvement over periods of more than six years, and motor improvements for more than four years. Grafting outcomes are variable even within individual transplantation centers. The reasons for this variability are poorly understood, highlighting the need for further research in this specific area. With the perspective of additional trials in the future, we review here the development of human pluripotent stem cell-derived cell therapy products for HD, and their advantages and disadvantages with respect to fetal cells.
- Published
- 2014
42. Two Distinct Proteins Are Associated with Tetrameric Acetylcholinesterase on the Cell Surface
- Author
-
Terrone L. Rosenberry, Suzanne Bon, Samuel R. Pickett, Robert Haas, Eric Krejci, Jean Massoulié, Anselme L. Perrier, Xavier Cousin, William L. Roberts, Nicola Boschetti, and Jean-Marc Chatel
- Subjects
Protein subunit ,Blotting, Western ,Molecular Sequence Data ,Biology ,Biochemistry ,Mice ,chemistry.chemical_compound ,Biopolymers ,Tetramer ,Complementary DNA ,Tumor Cells, Cultured ,Animals ,Humans ,RNA, Antisense ,Amino Acid Sequence ,RNA, Messenger ,Cloning, Molecular ,Molecular Biology ,Peptide sequence ,chemistry.chemical_classification ,Sequence Homology, Amino Acid ,cDNA library ,Brain ,Membrane Proteins ,Proteins ,Cell Biology ,Acetylcholinesterase ,Molecular biology ,Amino acid ,chemistry ,Membrane protein ,Cattle - Abstract
In mammalian brain, acetylcholinesterase (AChE) exists mostly as a tetramer of 70-kDa catalytic subunits that are linked through disulfide bonds to a hydrophobic subunit P of approximately 20 kDa. To characterize P, we reduced the disulfide bonds in purified bovine brain AChE and sequenced tryptic fragments from bands in the 20-kDa region. We obtained sequences belonging to at least two distinct proteins: the P protein and another protein that was not disulfide-linked to catalytic subunits. Both proteins were recognized in Western blots by antisera raised against specific peptides. We cloned cDNA encoding the second protein in a cDNA library from bovine substantia nigra and obtained rat and human homologs. We call this protein mCutA because of its homology to a bacterial protein (CutA). We could not demonstrate a direct interaction between mCutA and AChE in vitro in transfected cells. However, in a mouse neuroblastoma cell line that produced membrane-bound AChE as an amphiphilic tetramer, the expression of mCutA antisense mRNA eliminated cell surface AChE and decreased the level of amphiphilic tetramer in cell extracts. mCutA therefore appears necessary for the localization of AChE at the cell surface; it may be part of a multicomponent complex that anchors AChE in membranes, together with the hydrophobic P protein.
- Published
- 2000
- Full Text
- View/download PDF
43. Des cellules souches embryonnaires humaines pour la thérapie cellulaire de la maladie de Huntington
- Author
-
Laetitia Aubry, Anselme L. Perrier, and Marc Peschanski
- Subjects
Central nervous system ,General Medicine ,Biology ,medicine.disease ,Embryonic stem cell ,Molecular biology ,General Biochemistry, Genetics and Molecular Biology ,Cell therapy ,Central nervous system disease ,medicine.anatomical_structure ,Degenerative disease ,Basal ganglia ,medicine ,Stem cell - Published
- 2009
- Full Text
- View/download PDF
44. Les cellules souches embryonnaires humaines révèlent l’existence d’une région hautement instable du génome
- Author
-
Annelise Bennaceur-Griscelli, Cécile Bas, Olivier Feraud, Gérard Tachdjian, Nathalie Lefort, Anselme L. Perrier, Maxime Feyeux, and Marc Peschanski
- Subjects
General Medicine ,Biology ,General Biochemistry, Genetics and Molecular Biology - Published
- 2009
- Full Text
- View/download PDF
45. Des cellules souches embryonnaires humaines pour le traitement de la maladie de Parkinson ?
- Author
-
Anselme L. Perrier
- Subjects
Central nervous system disease ,Degenerative disease ,medicine ,General Medicine ,Stem cell ,Biology ,medicine.disease ,Embryonic stem cell ,Molecular biology ,General Biochemistry, Genetics and Molecular Biology - Published
- 2005
- Full Text
- View/download PDF
46. Fabrication of precisely aligned microwire and microchannel structures: Toward heat stimulation of guided neurites in neuronal cultures
- Author
-
Ka My Dang, Bastian Haberkorn, Simona Gribaudo, Jan Schnitker, Philipp Rinklin, Stefan Weigel, Michael Daenen, Bernhard Wolfrum, Jorne Carolus, Anselme L. Perrier, K. Zobel, Andreas Offenhäusser, and Harald Luksch
- Subjects
Microchannel ,Fabrication ,Materials science ,Neurite ,010401 analytical chemistry ,Nanotechnology ,02 engineering and technology ,Surfaces and Interfaces ,021001 nanoscience & nanotechnology ,Condensed Matter Physics ,01 natural sciences ,0104 chemical sciences ,Surfaces, Coatings and Films ,Electronic, Optical and Magnetic Materials ,Thermal stimulation ,Etching (microfabrication) ,Materials Chemistry ,Electrical and Electronic Engineering ,0210 nano-technology ,Layer (electronics) - Abstract
Microwire arrays are a powerful tool for the exertion of localized thermal stress on cellular networks. Combining microwire arrays with a set of orthogonal axon-guiding microchannels on-chip allows for the positioning of neurites, as well as control over their polarity. In this paper, we present a new fabrication approach, based on standard clean room fabrication and sacrificial layer etching, for the integration of microwire arrays into neurite guiding structures. The system permits the application of strong temperature gradients, enabling localized thermal stimulation inside microchannels.
- Published
- 2017
- Full Text
- View/download PDF
47. 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
48. 327. Genetic Editing for Huntington's Disease
- Author
-
Nicole Déglon, Cécile Meunier, Maria Rey, Virginie Zimmer, Gabriel Vachey, Guillaume Perriard, Renaud Du Pasquier, Catherine Pythoud, Lucas Hergott, Tim Beltraminelli, Anselme L. Perrier, Luc Pellerin, Mathieu Canales, and Thomas Dequesne
- Subjects
Pharmacology ,Genetics ,congenital, hereditary, and neonatal diseases and abnormalities ,Cas9 ,Mutant ,Biology ,medicine.disease ,Cell biology ,Viral vector ,chemistry.chemical_compound ,Exon ,nervous system ,chemistry ,Huntington's disease ,mental disorders ,Drug Discovery ,medicine ,Molecular Medicine ,CRISPR ,Molecular Biology ,Gene ,DNA - Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a pathological CAG expansion at the 3’ end of the first exon of the huntingtin gene (HTT). Currently, there is no efficient treatment for HD. Editing of the mutant HTT gene with the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) system represents a new and promising approach. Recognition of the HTT target sequence by a single-guide RNA sequences (sgRNA) and the Cas9 protein is inducing DNA double-strand breaks (DSB), which activate endogenous cellular repair pathways. Non-homologous end joining (NHEJ) will introduce small insertion/deletion (indel) that alter the reading frame of HTT gene while homologous directed repair (HDR) is activated in the presence of a DNA template. To validate the approach and optimize the delivery of the CRISPR system with viral vectors, we first targeted artificial sequences containing fluorescent reporter genes in HEK 293T cells. An efficient gene disruption was measured and associated with a loss of fluorescence in neurons, astrocytes, in vitro and in vivo. Furthermore, we developed multiple strategies to disrupt the mutant HTT gene. Quantification demonstrated a high rate of indels, leading to a strong reduction of HTT protein in HEK 293T cells, mouse cortical neurons and human iPS-derived neurons. Blocking HTT expression in vitro HD models is improving several physiopathological parameters. We are currently evaluating the impact of allele or non-allele specific mutant HTT editing in human neurons from HD patients. Altogether, these data demonstrate the potential of the CRISPR technology as therapeutic strategy for HD.
- Published
- 2016
- Full Text
- View/download PDF
49. Derivation of striatal neurons from human stem cells
- Author
-
Pedro, Viegas, Camille, Nicoleau, and Anselme L, Perrier
- Subjects
Neurons ,Stem Cells ,Humans ,Corpus Striatum - Abstract
Huntington's disease cell therapy is for the moment the only therapeutic approach for this devastating neurodegenerative disorder that has demonstrated significant and long-lasting functional benefits in patient. Logistical and biological difficulties associated with the use of human fetal tissue however dramatically reduce the number of patients eligible to this therapy. During the past decade, the exploration of alternative cellular sources, conducted in parallel to the clinical trials, has gradually put forward human pluripotent stem cells as prime candidate for Huntington's disease cell therapy. Protocols for the differentiation of such cells into therapeutically relevant striatal neuron precursors require thorough understanding of the molecular determinant that controls the development of the anterior and ventral part of the forebrain from which the striatum arises. Key secreted molecules that play pivotal roles in the development of these regions in mice have been successfully used to direct the specification of neural derivatives of human pluripotent stem cells. Assessment of the therapeutic potential of resulting striatal grafts has made significant progress in the last 4 years. The proof of principle that human embryonic stem cell derivatives can achieve some degrees of functional striatal repair in a mouse model of Huntington's disease is now established. As in other neurodegenerative diseases such as Parkinson's disease, recent developments have raised hopes for stem cell-based therapy of Huntington's disease.
- Published
- 2012
50. Early transcriptional changes linked to naturally occurring Huntington's disease mutations in neural derivatives of human embryonic stem cells
- Author
-
Lesley Jones, Sophie Aubert, Nicole Déglon, Aurore Bugi, Peter Giles, Marta Ruiz, Fany Bourgois-Rocha, Anselme L. Perrier, Nathalie Lefort, Nicholas D. Allen, Marc Peschanski, Amanda Redfern, Caroline Bonnefond, and Maxime Feyeux
- Subjects
Huntingtin ,Transcription, Genetic ,Mutant ,Nerve Tissue Proteins ,Biology ,medicine.disease_cause ,Models, Biological ,Cell Line ,Huntington's disease ,Neural Stem Cells ,Genetics ,medicine ,Huntingtin Protein ,Humans ,Molecular Biology ,Genetics (clinical) ,Embryonic Stem Cells ,Regulation of gene expression ,Neurons ,Mutation ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Reproducibility of Results ,General Medicine ,medicine.disease ,Neural stem cell ,Gene expression profiling ,Huntington Disease ,Gene Expression Regulation ,Transcriptome - Abstract
Huntington's 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.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.