Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 21
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Brain Commun ; 5(3): fcad158, 2023.
Article de Anglais | MEDLINE | ID: mdl-37274831

RÉSUMÉ

Frontotemporal dementia (FTD) is the second most prevalent type of early-onset dementia and up to 40% of cases are familial forms. One of the genes mutated in patients is CHMP2B, which encodes a protein found in a complex important for maturation of late endosomes, an essential process for recycling membrane proteins through the endolysosomal system. Here, we have generated a CHMP2B-mutated human embryonic stem cell line using genome editing with the purpose to create a human in vitro FTD disease model. To date, most studies have focused on neuronal alterations; however, we present a new co-culture system in which neurons and astrocytes are independently generated from human embryonic stem cells and combined in co-cultures. With this approach, we have identified alterations in the endolysosomal system of FTD astrocytes, a higher capacity of astrocytes to uptake and respond to glutamate, and a neuronal network hyperactivity as well as excessive synchronization. Overall, our data indicates that astrocyte alterations precede neuronal impairments and could potentially trigger neuronal network changes, indicating the important and specific role of astrocytes in disease development.

2.
Mol Cancer Ther ; 22(2): 274-286, 2023 02 01.
Article de Anglais | MEDLINE | ID: mdl-36508391

RÉSUMÉ

Direct cellular reprogramming has recently gained attention of cancer researchers for the possibility to convert undifferentiated cancer cells into more differentiated, postmitotic cell types. While a few studies have attempted reprogramming of glioblastoma (GBM) cells toward a neuronal fate, this approach has not yet been used to induce differentiation into other lineages and in vivo data on reduction in tumorigenicity are limited. Here, we employ cellular reprogramming to induce astrocytic differentiation as a therapeutic approach in GBM. To this end, we overexpressed key transcriptional regulators of astroglial development in human GBM and GBM stem cell lines. Treated cells undergo a remarkable shift in structure, acquiring an astrocyte-like morphology with star-shaped bodies and radial branched processes. Differentiated cells express typical glial markers and show a marked decrease in their proliferative state. In addition, forced differentiation induces astrocytic functions such as induced calcium transients and ability to respond to inflammatory stimuli. Most importantly, forced differentiation substantially reduces tumorigenicity of GBM cells in an in vivo xenotransplantation model. The current study capitalizes on cellular plasticity with a novel application in cancer. We take advantage of the similarity between neural developmental processes and cancer hierarchy to mitigate, if not completely abolish, the malignant nature of tumor cells and pave the way for new intervention strategies.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Humains , Glioblastome/traitement médicamenteux , Astrocytes , Facteurs de transcription/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Lignée cellulaire tumorale , Différenciation cellulaire , Cellules souches tumorales/métabolisme
3.
Front Cell Dev Biol ; 10: 1039636, 2022.
Article de Anglais | MEDLINE | ID: mdl-36313554

RÉSUMÉ

Primitive, neonatal and adult erythroid cells have been previously shown to have an active pentose phosphate pathway (PPP) that fuels various processes. However, it is unclear whether the PPP plays a role during the emergence of erythroid progenitors from hemogenic endothelium (HE). In this study, we explored PPP and its genetic regulation in developmental erythropoiesis. We induced hematopoietic differentiation of human induced pluripotent stem cells (hiPSCs) to obtain HE cells. These cells were treated with lentiviral vectors harboring shRNAs against FOXO1, or with inhibitors against the PPP, NRF2 or AKT. Erythroid differentiation, proliferation and frequency were evaluated by flow cytometry. Gene expression was assessed by qPCR or by analysis of available RNAseq data. We found that PPP is indispensable for the erythroid differentiation of HE cells and it partially fuels nucleotide biosynthesis. Moreover, we showed that NRF2 and AKT are essential, while FOXO1 is detrimental, for HE-derived erythroid differentiation. In contrast, blocking FOXO1 expression did not affect erythroid differentiation of cord-blood HSPCs. Mechanistically, FOXO1 inhibition in HE cells led to an increase in the non-oxidative branch of the PPP. During developmental erythropoiesis, the gradual decrease in FOXO1 activates the PPP and fuels nucleotide biosynthesis and cell proliferation.

4.
Stem Cell Reports ; 17(7): 1620-1635, 2022 07 12.
Article de Anglais | MEDLINE | ID: mdl-35750047

RÉSUMÉ

Astrocytes are emerging key players in neurological disorders. However, their role in disease etiology is poorly understood owing to inaccessibility of primary human astrocytes. Pluripotent stem cell-derived cells fail to mimic age and due to their clonal origin do not mimic genetic heterogeneity of patients. In contrast, direct conversion constitutes an attractive approach to generate human astrocytes that capture age and genetic diversity. We describe efficient direct conversion of human fibroblasts to functional induced astrocytes (iAs). Expression of the minimal combination Sox9 and Nfib generates iAs with molecular, phenotypic, and functional properties resembling primary human astrocytes. iAs could be obtained by conversion of fibroblasts covering the entire human lifespan. Importantly, iAs supported function of induced neurons obtained through direct conversion from the same fibroblast population. Fibroblast-derived iAs will become a useful tool to elucidate the biology of astrocytes and complement current in vitro models for studies of late-onset neurological disorders.


Sujet(s)
Astrocytes , Cellules souches pluripotentes , Astrocytes/métabolisme , Cellules cultivées , Fibroblastes/métabolisme , Humains , Neurones , Cellules souches pluripotentes/métabolisme
5.
Methods Mol Biol ; 2549: 409-425, 2022.
Article de Anglais | MEDLINE | ID: mdl-33755903

RÉSUMÉ

The discovery that the CRISPR/Cas9 system could be used for genome editing purposes represented a major breakthrough in the field. This advancement has notably facilitated the introduction or correction of disease-specific mutations in healthy or disease stem cell lines respectively; therefore, easing disease modeling studies in combination with differentiation protocols. For many years, variability in the genetic background of different stem cell lines has been a major burden to specifically identify phenotypes arising uniquely from the presence of the mutation and not from differences in other genomic regions.Here, we provide a complete protocol to introduce random indels in human wild type pluripotent stem cells using CRISPR/Cas9 in order to generate clonal lines with potential pathogenic alterations in any gene of interest. In this protocol, we use transfection of a ribonucleoprotein complex to diminish the risk of off-target effects, and select clonal lines with promising indels to obtain disease induced pluripotent stem cell lines.


Sujet(s)
Cellules souches pluripotentes induites , Cellules souches pluripotentes , Systèmes CRISPR-Cas , Édition de gène/méthodes , Génome humain , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes/métabolisme , /génétique , /métabolisme , Ribonucléoprotéines/génétique , Ribonucléoprotéines/métabolisme
6.
EMBO Rep ; 23(2): e54384, 2022 02 03.
Article de Anglais | MEDLINE | ID: mdl-34914165

RÉSUMÉ

During embryonic development, hematopoiesis occurs through primitive and definitive waves, giving rise to distinct blood lineages. Hematopoietic stem cells (HSCs) emerge from hemogenic endothelial (HE) cells, through endothelial-to-hematopoietic transition (EHT). In the adult, HSC quiescence, maintenance, and differentiation are closely linked to changes in metabolism. However, metabolic processes underlying the emergence of HSCs from HE cells remain unclear. Here, we show that the emergence of blood is regulated by multiple metabolic pathways that induce or modulate the differentiation toward specific hematopoietic lineages during human EHT. In both in vitro and in vivo settings, steering pyruvate use toward glycolysis or OXPHOS differentially skews the hematopoietic output of HE cells toward either an erythroid fate with primitive phenotype, or a definitive lymphoid fate, respectively. We demonstrate that glycolysis-mediated differentiation of HE toward primitive erythroid hematopoiesis is dependent on the epigenetic regulator LSD1. In contrast, OXPHOS-mediated differentiation of HE toward definitive hematopoiesis is dependent on cholesterol metabolism. Our findings reveal that during EHT, metabolism is a major regulator of primitive versus definitive hematopoietic differentiation.


Sujet(s)
Hémangioblastes , Différenciation cellulaire , Lignage cellulaire/génétique , Femelle , Hémangioblastes/métabolisme , Hématopoïèse/génétique , Cellules souches hématopoïétiques/métabolisme , Humains , Grossesse , Pyruvates/métabolisme
7.
Cell Reprogram ; 23(4): 206-220, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34388027

RÉSUMÉ

In the last years, the use of pluripotent stem cells in studies of human biology has grown exponentially. These cells represent an infinite source for differentiation into several human cell types facilitating the investigation on biological processes, functionality of cells, or diseases mechanisms in relevant human models. In the neurobiology field, pluripotent stem cells have been extensively used to generate the main neuronal and glial cells of the brain. Traditionally, protocols following developmental cues have been applied to pluripotent stem cells to drive differentiation toward different cell lineages; however, these protocols give rise to populations with mixed identities. Interestingly, new protocols applying overexpression of lineage-specific transcription factors (TFs) have emerged and facilitated the generation of highly pure populations of specific subtypes of neurons and glial cells in an easy, reproducible, and rapid manner. In this study, we review protocols based on this strategy to generate excitatory, inhibitory, dopaminergic, and motor neurons as well as astrocytes, oligodendrocytes, and microglia. In addition, we will discuss the main applications for cells generated with these protocols, including disease modeling, drug screening, and mechanistic studies. Finally, we will discuss the advantages and disadvantages of TF-based protocols and present our view of the future in this field.


Sujet(s)
Différenciation cellulaire , Lignage cellulaire , Neurones/cytologie , Cellules souches pluripotentes/cytologie , Facteurs de transcription/métabolisme , Humains , Neurones/métabolisme , Cellules souches pluripotentes/métabolisme
8.
Methods Mol Biol ; 2352: 133-148, 2021.
Article de Anglais | MEDLINE | ID: mdl-34324185

RÉSUMÉ

Astrocytes are essential cells for normal brain functionality and have recently emerged as key players in many neurological diseases. However, the limited availability of human primary astrocytes for cell culture studies hinders our understanding of their physiology and precise role in disease development and progression. Here, we describe a detailed step-by-step protocol to rapidly and efficiently generate functionally mature induced astrocytes (iAs) from human embryonic and induced pluripotent stem cells (hES/iPSCs). Astrocyte induction is accomplished by ectopic lentiviral expression of two gliogenic transcription factors, Sox9 and Nfib. iAs exhibit morphology features as well as gene and protein expression similar to human mature astrocytes and display important astrocytic functions, such as glutamate uptake, propagation of calcium waves, expression of various cytokines after stimulation, and support of synapse formation and function, making them suitable models for studying the role of astrocytes in health and disease. Moreover, we describe a procedure for cryopreservation of iAs for long-term storage or shipping. Finally, we provide the required information needed to set up cocultures with human induced neurons (iNs, also described in this book), generated from hES/iPSCs, to generate cocultures, allowing studies on astrocyte-neuron interactions and providing new insights in astrocyte-associated disease mechanisms.


Sujet(s)
Astrocytes/cytologie , Astrocytes/métabolisme , Reprogrammation cellulaire/génétique , Neurones/cytologie , Neurones/métabolisme , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/métabolisme , Facteurs de transcription/génétique , Techniques de culture cellulaire , Cellules cultivées , Techniques de reprogrammation cellulaire , Techniques de coculture , Vecteurs génétiques/administration et posologie , Vecteurs génétiques/génétique , Humains , Immunohistochimie
9.
Methods Mol Biol ; 2352: 237-251, 2021.
Article de Anglais | MEDLINE | ID: mdl-34324191

RÉSUMÉ

Recent advances in genome editing have brought new hopes for personalized and precision medicine but have also dramatically facilitated disease modeling studies. Combined with reprogramming approaches, stem cells and differentiation toward neural lineages, genome engineering holds great potential for regenerative approaches and to model neurological disorders. The use of patient-specific induced pluripotent stem cells combined with neural differentiation allows studying the effect of specific mutations in different brain cells. New genome editing tools such as CRISPR/Cas9 represent a step further by facilitating the introduction or correction of specific mutations within the same cell line, thus eliminating variability due to differences in the genetic background. Here, we present a step-by-step protocol from design to generation of human pluripotent stem cell lines with specific mutations introduced or corrected with CRISPR/Cas9 gene editing that can be used in combination with transcription factor-based protocols to dissect underlying mechanisms of neurological disorders.


Sujet(s)
Systèmes CRISPR-Cas , Différenciation cellulaire , Édition de gène , Maladies du système nerveux/étiologie , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/métabolisme , Différenciation cellulaire/génétique , Clonage moléculaire , Humains , Modèles biologiques , Maladies du système nerveux/métabolisme , , Transfection
10.
Front Cell Neurosci ; 15: 602888, 2021.
Article de Anglais | MEDLINE | ID: mdl-33679325

RÉSUMÉ

Recent advancements in cell engineering have succeeded in manipulating cell identity with the targeted overexpression of specific cell fate determining transcription factors in a process named transcriptional programming. Neurogenin2 (NGN2) is sufficient to instruct pluripotent stem cells (PSCs) to acquire a neuronal identity when delivered with an integrating system, which arises some safety concerns for clinical applications. A non-integrating system based on modified messenger RNA (mmRNA) delivery method, represents a valuable alternative to lentiviral-based approaches. The ability of NGN2 mmRNA to instruct PSC fate change has not been thoroughly investigated yet. Here we aimed at understanding whether the use of an NGN2 mmRNA-based approach combined with a miniaturized system, which allows a higher transfection efficiency in a cost-effective system, is able to drive human induced PSCs (hiPSCs) toward the neuronal lineage. We show that NGN2 mRNA alone is able to induce cell fate conversion. Surprisingly, the outcome cell population accounts for multiple phenotypes along the neural development trajectory. We found that this mixed population is mainly constituted by neural stem cells (45% ± 18 PAX6 positive cells) and neurons (38% ± 8 ßIIITUBULIN positive cells) only when NGN2 is delivered as mmRNA. On the other hand, when the delivery system is lentiviral-based, both providing a constant expression of NGN2 or only a transient pulse, the outcome differentiated population is formed by a clear majority of neurons (88% ± 1 ßIIITUBULIN positive cells). Altogether, our data confirm the ability of NGN2 to induce neuralization in hiPSCs and opens a new point of view in respect to the delivery system method when it comes to transcriptional programming applications.

11.
Int J Mol Sci ; 21(21)2020 Oct 22.
Article de Anglais | MEDLINE | ID: mdl-33105639

RÉSUMÉ

Sanfilippo syndrome or mucopolysaccharidosis III is a lysosomal storage disorder caused by mutations in genes responsible for the degradation of heparan sulfate, a glycosaminoglycan located in the extracellular membrane. Undegraded heparan sulfate molecules accumulate within lysosomes leading to cellular dysfunction and pathology in several organs, with severe central nervous system degeneration as the main phenotypical feature. The exact molecular and cellular mechanisms by which impaired degradation and storage lead to cellular dysfunction and neuronal degeneration are still not fully understood. Here, we compile the knowledge on this issue and review all available animal and cellular models that can be used to contribute to increase our understanding of Sanfilippo syndrome disease mechanisms. Moreover, we provide an update in advances regarding the different and most successful therapeutic approaches that are currently under study to treat Sanfilippo syndrome patients and discuss the potential of new tools such as induced pluripotent stem cells to be used for disease modeling and therapy development.


Sujet(s)
Héparitine sulfate/métabolisme , Mucopolysaccharidose de type III/étiologie , Mucopolysaccharidose de type III/thérapie , Acetyltransferases/génétique , Animaux , Modèles animaux de maladie humaine , Thérapie enzymatique substitutive/méthodes , Thérapie génétique , Humains , Hydrolases/génétique , Mucopolysaccharidose de type III/anatomopathologie , Mutation , Transplantation de cellules souches
12.
Int J Mol Sci ; 21(9)2020 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-32366037

RÉSUMÉ

Leigh syndrome (LS) is the most frequent infantile mitochondrial disorder (MD) and is characterized by neurodegeneration and astrogliosis in the basal ganglia or the brain stem. At present, there is no cure or treatment for this disease, partly due to scarcity of LS models. Current models generally fail to recapitulate important traits of the disease. Therefore, there is an urgent need to develop new human in vitro models. Establishment of induced pluripotent stem cells (iPSCs) followed by differentiation into neurons is a powerful tool to obtain an in vitro model for LS. Here, we describe the generation and characterization of iPSCs, neural stem cells (NSCs) and iPSC-derived neurons harboring the mtDNA mutation m.13513G>A in heteroplasmy. We have performed mitochondrial characterization, analysis of electrophysiological properties and calcium imaging of LS neurons. Here, we show a clearly compromised oxidative phosphorylation (OXPHOS) function in LS patient neurons. This is also the first report of electrophysiological studies performed on iPSC-derived neurons harboring an mtDNA mutation, which revealed that, in spite of having identical electrical properties, diseased neurons manifested mitochondrial dysfunction together with a diminished calcium buffering capacity. This could lead to an overload of cytoplasmic calcium concentration and the consequent cell death observed in patients. Importantly, our results highlight the importance of calcium homeostasis in LS pathology.


Sujet(s)
Calcium/métabolisme , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Maladie de Leigh/métabolisme , Consommation d'oxygène/physiologie , Technique de Western , Prolifération cellulaire/physiologie , Cellules cultivées , Électrophysiologie , Technique d'immunofluorescence , Humains , Acide lactique/métabolisme , Maladie de Leigh/anatomopathologie , Mitochondries/métabolisme , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Neurones/cytologie , Neurones/métabolisme , Consommation d'oxygène/génétique
13.
J Clin Med ; 9(3)2020 Feb 28.
Article de Anglais | MEDLINE | ID: mdl-32121121

RÉSUMÉ

Sanfilippo syndrome type C (mucopolysaccharidosis IIIC) is an early-onset neurodegenerative lysosomal storage disorder, which is currently untreatable. The vast majority of studies focusing on disease mechanisms of Sanfilippo syndrome were performed on non-neural cells or mouse models, which present obvious limitations. Induced pluripotent stem cells (iPSCs) are an efficient way to model human diseases in vitro. Recently developed transcription factor-based differentiation protocols allow fast and efficient conversion of iPSCs into the cell type of interest. By applying these protocols, we have generated new neuronal and astrocytic models of Sanfilippo syndrome using our previously established disease iPSC lines. Moreover, our neuronal model exhibits disease-specific molecular phenotypes, such as increase in lysosomes and heparan sulfate. Lastly, we tested an experimental, siRNA-based treatment previously shown to be successful in patients' fibroblasts and demonstrated its lack of efficacy in neurons. Our findings highlight the need to use relevant human cellular models to test therapeutic interventions and shows the applicability of our neuronal and astrocytic models of Sanfilippo syndrome for future studies on disease mechanisms and drug development.

16.
Methods Mol Biol ; 1919: 73-88, 2019.
Article de Anglais | MEDLINE | ID: mdl-30656622

RÉSUMÉ

Recent progress in stem cell biology and epigenetic reprogramming has opened up previously unimaginable possibilities to study and develop regenerative approaches for neurological disorders. Human neurons and glial cells can be generated by differentiation of embryonic and neural stem cells and from somatic cells through reprogramming to pluripotency (followed by differentiation) as well as by direct conversion. All of these cells have the potential to be used for studying and treating neurological disorders. However, before considering using human neural cells derived from these sources for modelling or regenerative purposes, they need to be verified in terms of functionality and similarity to endogenous cells in the central nervous system (CNS).In this chapter, we describe how to assess functionality of neurons and astrocytes derived from stem cells and through direct reprogramming, using calcium imaging and electrophysiology.


Sujet(s)
Astrocytes/physiologie , Signalisation calcique , Calcium/métabolisme , Phénomènes électrophysiologiques , Imagerie moléculaire , Neurones/physiologie , Astrocytes/cytologie , Marqueurs biologiques , Différenciation cellulaire , Reprogrammation cellulaire , Technique d'immunofluorescence , Humains , Neurones/cytologie , Synapses/métabolisme
17.
Nat Methods ; 16(1): 134, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30514884

RÉSUMÉ

In the version of Supplementary Fig. 1 originally published with this paper, some images in panel e were accidental duplicates of images in panel b. This error has been corrected in the online integrated supplementary information and in the Supplementary Information PDF.

18.
Nat Methods ; 15(9): 693-696, 2018 09.
Article de Anglais | MEDLINE | ID: mdl-30127505

RÉSUMÉ

The derivation of astrocytes from human pluripotent stem cells is currently slow and inefficient. We demonstrate that overexpression of the transcription factors SOX9 and NFIB in human pluripotent stem cells rapidly and efficiently yields homogeneous populations of induced astrocytes. In our study these cells exhibited molecular and functional properties resembling those of adult human astrocytes and were deemed suitable for disease modeling. Our method provides new possibilities for the study of human astrocytes in health and disease.


Sujet(s)
Astrocytes/cytologie , Différenciation cellulaire , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/métabolisme , Facteur de transcription SOX-9/métabolisme , Humains , Facteurs nucléaires-I/métabolisme
19.
Sci Rep ; 5: 13654, 2015 Sep 08.
Article de Anglais | MEDLINE | ID: mdl-26347037

RÉSUMÉ

Sanfilippo syndrome is a rare lysosomal storage disorder caused by an impaired degradation of heparan sulfate (HS). It presents severe and progressive neurodegeneration and currently there is no effective treatment. Substrate reduction therapy (SRT) may be a useful option for neurological disorders of this kind, and several approaches have been tested to date. Here we use different siRNAs targeting EXTL2 and EXTL3 genes, which are important for HS synthesis, as SRT in Sanfilippo C patients' fibroblasts in order to decrease glycosaminoglycan (GAG) storage inside the lysosomes. The results show a high inhibition of the EXTL gene mRNAs (around 90%), a decrease in GAG synthesis after three days (30-60%) and a decrease in GAG storage after 14 days (up to 24%). Moreover, immunocytochemistry analyses showed a clear reversion of the phenotype after treatment. The in vitro inhibition of HS synthesis genes using siRNAs shown here is a first step in the development of a future therapeutic option for Sanfilippo C syndrome.


Sujet(s)
Protéines membranaires/génétique , Mucopolysaccharidose de type III/génétique , N-acetylglucosaminyltransferase/génétique , Interférence par ARN , ARN messager/génétique , Petit ARN interférent/génétique , Fibroblastes/métabolisme , Expression des gènes , Glycosaminoglycanes/métabolisme , Héparitine sulfate/métabolisme , Humains , Immunohistochimie , Protéines membranaires/métabolisme , Mucopolysaccharidose de type III/métabolisme , Mucopolysaccharidose de type III/thérapie , N-acetylglucosaminyltransferase/métabolisme , Transfection
20.
Stem Cell Reports ; 5(4): 546-57, 2015 Oct 13.
Article de Anglais | MEDLINE | ID: mdl-26411903

RÉSUMÉ

Induced pluripotent stem cell (iPSC) technology has been successfully used to recapitulate phenotypic traits of several human diseases in vitro. Patient-specific iPSC-based disease models are also expected to reveal early functional phenotypes, although this remains to be proved. Here, we generated iPSC lines from two patients with Sanfilippo type C syndrome, a lysosomal storage disorder with inheritable progressive neurodegeneration. Mature neurons obtained from patient-specific iPSC lines recapitulated the main known phenotypes of the disease, not present in genetically corrected patient-specific iPSC-derived cultures. Moreover, neuronal networks organized in vitro from mature patient-derived neurons showed early defects in neuronal activity, network-wide degradation, and altered effective connectivity. Our findings establish the importance of iPSC-based technology to identify early functional phenotypes, which can in turn shed light on the pathological mechanisms occurring in Sanfilippo syndrome. This technology also has the potential to provide valuable readouts to screen compounds, which can prevent the onset of neurodegeneration.


Sujet(s)
Cellules souches pluripotentes induites/anatomopathologie , Mucopolysaccharidose de type III/anatomopathologie , Réseau nerveux/anatomopathologie , Neurones/anatomopathologie , Techniques de culture cellulaire , Cellules cultivées , Humains , Neurogenèse
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...