Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
STAR Protoc ; 5(3): 103280, 2024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39213150

RÉSUMÉ

The generation of human pluripotent stem cell (hPSC)-derived brain organoids is continuously refined, enhancing their reproducibility and complexity. Here, we present a guided differentiation protocol for generating cortical forebrain organoids and cortico-pericyte (CP)assembloids composed of a robust outer radial glia (oRG) population and an expanded outer subventricular zone (oSVZ). We describe the steps to generate hPSC-derived cortical organoids (COs), cortical pericytes, and CP assembloids. Moreover, we outline the procedures to characterize the organoids by immunostaining and to perform single-cell dissociation. For complete details on the use and execution of this protocol, please refer to Walsh et al.1.

3.
Cell Rep ; 43(4): 114031, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38583153

RÉSUMÉ

Outer radial glia (oRG) emerge as cortical progenitor cells that support the development of an enlarged outer subventricular zone (oSVZ) and the expansion of the neocortex. The in vitro generation of oRG is essential to investigate the underlying mechanisms of human neocortical development and expansion. By activating the STAT3 signaling pathway using leukemia inhibitory factor (LIF), which is not expressed in guided cortical organoids, we define a cortical organoid differentiation method from human pluripotent stem cells (hPSCs) that recapitulates the expansion of a progenitor pool into the oSVZ. The oSVZ comprises progenitor cells expressing specific oRG markers such as GFAP, LIFR, and HOPX, closely matching human fetal oRG. Finally, incorporating neural crest-derived LIF-producing cortical pericytes into cortical organoids recapitulates the effects of LIF treatment. These data indicate that increasing the cellular complexity of the organoid microenvironment promotes the emergence of oRG and supports a platform to study oRG in hPSC-derived brain organoids routinely.


Sujet(s)
Différenciation cellulaire , Ventricules latéraux , Facteur inhibiteur de la leucémie , Organoïdes , Cellules souches pluripotentes , Humains , Organoïdes/métabolisme , Organoïdes/cytologie , Facteur inhibiteur de la leucémie/métabolisme , Facteur inhibiteur de la leucémie/pharmacologie , Cellules souches pluripotentes/métabolisme , Cellules souches pluripotentes/cytologie , Ventricules latéraux/cytologie , Ventricules latéraux/métabolisme , Facteur de transcription STAT-3/métabolisme , Névroglie/métabolisme , Névroglie/cytologie , Transduction du signal
4.
Genes Dev ; 38(1-2): 70-94, 2024 02 13.
Article de Anglais | MEDLINE | ID: mdl-38316520

RÉSUMÉ

Since genome instability can drive cancer initiation and progression, cells have evolved highly effective and ubiquitous DNA damage response (DDR) programs. However, some cells (for example, in skin) are normally exposed to high levels of DNA-damaging agents. Whether such high-risk cells possess lineage-specific mechanisms that tailor DNA repair to the tissue remains largely unknown. Using melanoma as a model, we show here that the microphthalmia-associated transcription factor MITF, a lineage addition oncogene that coordinates many aspects of melanocyte and melanoma biology, plays a nontranscriptional role in shaping the DDR. On exposure to DNA-damaging agents, MITF is phosphorylated at S325, and its interactome is dramatically remodeled; most transcription cofactors dissociate, and instead MITF interacts with the MRE11-RAD50-NBS1 (MRN) complex. Consequently, cells with high MITF levels accumulate stalled replication forks and display defects in homologous recombination-mediated repair associated with impaired MRN recruitment to DNA damage. In agreement with this, high MITF levels are associated with increased single-nucleotide and copy number variant burdens in melanoma. Significantly, the SUMOylation-defective MITF-E318K melanoma predisposition mutation recapitulates the effects of DNA-PKcs-phosphorylated MITF. Our data suggest that a nontranscriptional function of a lineage-restricted transcription factor contributes to a tissue-specialized modulation of the DDR that can impact cancer initiation.


Sujet(s)
Mélanome , Humains , Mélanome/génétique , Facteur de transcription associé à la microphtalmie/génétique , Altération de l'ADN , Instabilité du génome/génétique , ADN
5.
Nat Biotechnol ; 2024 Jan 02.
Article de Anglais | MEDLINE | ID: mdl-38168993

RÉSUMÉ

The maturation of human pluripotent stem cell (hPSC)-derived neurons mimics the protracted timing of human brain development, extending over months to years for reaching adult-like function. Prolonged in vitro maturation presents a major challenge to stem cell-based applications in modeling and treating neurological disease. Therefore, we designed a high-content imaging assay based on morphological and functional readouts in hPSC-derived cortical neurons which identified multiple compounds that drive neuronal maturation including inhibitors of lysine-specific demethylase 1 and disruptor of telomerase-like 1 and activators of calcium-dependent transcription. A cocktail of four factors, GSK2879552, EPZ-5676, N-methyl-D-aspartate and Bay K 8644, collectively termed GENtoniK, triggered maturation across all parameters tested, including synaptic density, electrophysiology and transcriptomics. Maturation effects were further validated in cortical organoids, spinal motoneurons and non-neural lineages including melanocytes and pancreatic ß-cells. The effects on maturation observed across a broad range of hPSC-derived cell types indicate that some of the mechanisms controlling the timing of human maturation might be shared across lineages.

6.
Mol Oncol ; 18(4): 793-796, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38282579

RÉSUMÉ

When we think about cancer, the link to development might not immediately spring to mind. Yet, many foundational concepts in cancer biology trace their roots back to developmental processes. Several defining traits of cancer were indeed initially observed and studied within developing embryos. As our comprehension of embryonic mechanisms deepens, it not only illuminates how and why cancer cells hijack these processes but also spearheads the emergence of innovative technologies for modeling and comprehending tumor biology. Among these technologies are stem cell-based models, made feasible through our grasp of fundamental mechanisms related to embryonic development. The intersection between cancer and stem cell research is evolving into a tangible synergy that extends beyond the concepts of cancer stem cells and cell-of-origin, offering novel tools to unravel the mechanisms of cancer initiation and progression.


Sujet(s)
Tumeurs , Cellules souches tumorales , Femelle , Grossesse , Humains , Différenciation cellulaire , Développement embryonnaire , Biologie du développement
7.
Nature ; 626(8000): 881-890, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38297124

RÉSUMÉ

The pace of human brain development is highly protracted compared with most other species1-7. The maturation of cortical neurons is particularly slow, taking months to years to develop adult functions3-5. Remarkably, such protracted timing is retained in cortical neurons derived from human pluripotent stem cells (hPSCs) during in vitro differentiation or upon transplantation into the mouse brain4,8,9. Those findings suggest the presence of a cell-intrinsic clock setting the pace of neuronal maturation, although the molecular nature of this clock remains unknown. Here we identify an epigenetic developmental programme that sets the timing of human neuronal maturation. First, we developed a hPSC-based approach to synchronize the birth of cortical neurons in vitro which enabled us to define an atlas of morphological, functional and molecular maturation. We observed a slow unfolding of maturation programmes, limited by the retention of specific epigenetic factors. Loss of function of several of those factors in cortical neurons enables precocious maturation. Transient inhibition of EZH2, EHMT1 and EHMT2 or DOT1L, at progenitor stage primes newly born neurons to rapidly acquire mature properties upon differentiation. Thus our findings reveal that the rate at which human neurons mature is set well before neurogenesis through the establishment of an epigenetic barrier in progenitor cells. Mechanistically, this barrier holds transcriptional maturation programmes in a poised state that is gradually released to ensure the prolonged timeline of human cortical neuron maturation.


Sujet(s)
Épigenèse génétique , Régulation de l'expression des gènes au cours du développement , Cellules souches embryonnaires humaines , Cellules souches neurales , Neurogenèse , Neurones , Adulte , Animaux , Humains , Souris , Antigènes d'histocompatibilité/métabolisme , Histone-lysine N-methyltransferase/antagonistes et inhibiteurs , Histone-lysine N-methyltransferase/métabolisme , Cellules souches embryonnaires humaines/cytologie , Cellules souches embryonnaires humaines/métabolisme , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Neurogenèse/génétique , Neurones/cytologie , Neurones/métabolisme , Facteurs temps , Transcription génétique
8.
Adv Mater ; 36(3): e2307747, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37990971

RÉSUMÉ

Current treatments for modulating the glial-mediated inflammatory response after spinal cord injury (SCI) have limited ability to improve recovery. This is quite likely due to the lack of a selective therapeutic approach acting on microgliosis and astrocytosis, the glia components most involved after trauma, while maximizing efficacy and minimizing side effects. A new nanogel that can selectively release active compounds in microglial cells and astrocytes is developed and characterized. The degree of selectivity and subcellular distribution of the nanogel is evaluated by applying an innovative super-resolution microscopy technique, expansion microscopy. Two different administration schemes are then tested in a SCI mouse model: in an early phase, the nanogel loaded with Rolipram, an anti-inflammatory drug, achieves significant improvement in the animal's motor performance due to the increased recruitment of microglia and macrophages that are able to localize the lesion. Treatment in the late phase, however, gives opposite results, with worse motor recovery because of the widespread degeneration. These findings demonstrate that the nanovector can be selective and functional in the treatment of the glial component in different phases of SCI. They also open a new therapeutic scenario for tackling glia-mediated inflammation after neurodegenerative events in the central nervous system.


Sujet(s)
Polyéthylène glycols , Polyéthylèneimine , Traumatismes de la moelle épinière , Souris , Animaux , Nanogels/usage thérapeutique , Traumatismes de la moelle épinière/anatomopathologie , Névroglie/anatomopathologie , Microglie
9.
Nat Commun ; 14(1): 3192, 2023 06 02.
Article de Anglais | MEDLINE | ID: mdl-37268606

RÉSUMÉ

Melanoma exhibits numerous transcriptional cell states including neural crest-like cells as well as pigmented melanocytic cells. How these different cell states relate to distinct tumorigenic phenotypes remains unclear. Here, we use a zebrafish melanoma model to identify a transcriptional program linking the melanocytic cell state to a dependence on lipid droplets, the specialized organelle responsible for lipid storage. Single-cell RNA-sequencing of these tumors show a concordance between genes regulating pigmentation and those involved in lipid and oxidative metabolism. This state is conserved across human melanoma cell lines and patient tumors. This melanocytic state demonstrates increased fatty acid uptake, an increased number of lipid droplets, and dependence upon fatty acid oxidative metabolism. Genetic and pharmacologic suppression of lipid droplet production is sufficient to disrupt cell cycle progression and slow melanoma growth in vivo. Because the melanocytic cell state is linked to poor outcomes in patients, these data indicate a metabolic vulnerability in melanoma that depends on the lipid droplet organelle.


Sujet(s)
Gouttelettes lipidiques , Mélanome , Animaux , Humains , Gouttelettes lipidiques/métabolisme , Danio zébré/génétique , Mélanome/anatomopathologie , Mélanocytes/métabolisme , Acides gras/métabolisme , Métabolisme lipidique/génétique
10.
bioRxiv ; 2023 Apr 21.
Article de Anglais | MEDLINE | ID: mdl-37131595

RÉSUMÉ

Since genome instability can drive cancer initiation and progression, cells have evolved highly effective and ubiquitous DNA Damage Response (DDR) programs. However, some cells, in skin for example, are normally exposed to high levels of DNA damaging agents. Whether such high-risk cells possess lineage-specific mechanisms that tailor DNA repair to the tissue remains largely unknown. Here we show, using melanoma as a model, that the microphthalmia-associated transcription factor MITF, a lineage addition oncogene that coordinates many aspects of melanocyte and melanoma biology, plays a non-transcriptional role in shaping the DDR. On exposure to DNA damaging agents, MITF is phosphorylated by ATM/DNA-PKcs, and unexpectedly its interactome is dramatically remodelled; most transcription (co)factors dissociate, and instead MITF interacts with the MRE11-RAD50-NBS1 (MRN) complex. Consequently, cells with high MITF levels accumulate stalled replication forks, and display defects in homologous recombination-mediated repair associated with impaired MRN recruitment to DNA damage. In agreement, high MITF levels are associated with increased SNV burden in melanoma. Significantly, the SUMOylation-defective MITF-E318K melanoma predisposition mutation recapitulates the effects of ATM/DNA-PKcs-phosphorylated MITF. Our data suggest that a non-transcriptional function of a lineage-restricted transcription factor contributes to a tissue-specialised modulation of the DDR that can impact cancer initiation.

11.
Cell Stem Cell ; 30(3): 264-282.e9, 2023 03 02.
Article de Anglais | MEDLINE | ID: mdl-36868194

RÉSUMÉ

The enteric nervous system (ENS) is derived from both the vagal and sacral component of the neural crest (NC). Here, we present the derivation of sacral ENS precursors from human PSCs via timed exposure to FGF, WNT, and GDF11, which enables posterior patterning and transition from posterior trunk to sacral NC identity, respectively. Using a SOX2::H2B-tdTomato/T::H2B-GFP dual reporter hPSC line, we demonstrate that both trunk and sacral NC emerge from a double-positive neuro-mesodermal progenitor (NMP). Vagal and sacral NC precursors yield distinct neuronal subtypes and migratory behaviors in vitro and in vivo. Remarkably, xenografting of both vagal and sacral NC lineages is required to rescue a mouse model of total aganglionosis, suggesting opportunities in the treatment of severe forms of Hirschsprung's disease.


Sujet(s)
Maladie de Hirschsprung , Animaux , Humains , Souris , Protéines morphogénétiques osseuses , Modèles animaux de maladie humaine , Facteurs de croissance et de différenciation , Hétérogreffes , Histone , Crête neurale
12.
bioRxiv ; 2023 Feb 17.
Article de Anglais | MEDLINE | ID: mdl-36824730

RÉSUMÉ

Mammalian outer radial glia (oRG) emerge as cortical progenitor cells that directly support the development of an enlarged outer subventricular zone (oSVZ) and, in turn, the expansion of the neocortex. The in vitro generation of oRG is essential to model and investigate the underlying mechanisms of human neocortical development and expansion. By activating the STAT3 pathway using LIF, which is not produced in guided cortical organoids, we developed a cerebral organoid differentiation method from human pluripotent stem cells (hPSCs) that recapitulates the expansion of a progenitor pool into the oSVZ. The structured oSVZ is composed of progenitor cells expressing specific oRG markers such as GFAP, LIFR, HOPX , which closely matches human oRG in vivo . In this microenvironment, cortical neurons showed faster maturation with enhanced metabolic and functional activity. Incorporation of hPSC-derived brain vascular LIF- producing pericytes in cerebral organoids mimicked the effects of LIF treatment. These data indicate that the cellular complexity of the cortical microenvironment, including cell-types of the brain vasculature, favors the appearance of oRG and provides a platform to routinely study oRG in hPSC-derived brain organoids.

13.
Nature ; 604(7905): 354-361, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35355015

RÉSUMÉ

Oncogenic alterations to DNA are not transforming in all cellular contexts1,2. This may be due to pre-existing transcriptional programmes in the cell of origin. Here we define anatomic position as a major determinant of why cells respond to specific oncogenes. Cutaneous melanoma arises throughout the body, whereas the acral subtype arises on the palms of the hands, soles of the feet or under the nails3. We sequenced the DNA of cutaneous and acral melanomas from a large cohort of human patients and found a specific enrichment for BRAF mutations in cutaneous melanoma and enrichment for CRKL amplifications in acral melanoma. We modelled these changes in transgenic zebrafish models and found that CRKL-driven tumours formed predominantly in the fins of the fish. The fins are the evolutionary precursors to tetrapod limbs, indicating that melanocytes in these acral locations may be uniquely susceptible to CRKL. RNA profiling of these fin and limb melanocytes, when compared with body melanocytes, revealed a positional identity gene programme typified by posterior HOX13 genes. This positional gene programme synergized with CRKL to amplify insulin-like growth factor (IGF) signalling and drive tumours at acral sites. Abrogation of this CRKL-driven programme eliminated the anatomic specificity of acral melanoma. These data suggest that the anatomic position of the cell of origin endows it with a unique transcriptional state that makes it susceptible to only certain oncogenic insults.


Sujet(s)
Mélanome , Tumeurs cutanées , Animaux , Animal génétiquement modifié , Carcinogenèse/génétique , Pied , Main , Humains , Mélanome/anatomopathologie , Ongles , Oncogènes/génétique , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie , Transcription génétique , Danio zébré/génétique ,
14.
Science ; 373(6559): eabc1048, 2021 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-34516843

RÉSUMÉ

Oncogenes only transform cells under certain cellular contexts, a phenomenon called oncogenic competence. Using a combination of a human pluripotent stem cell­derived cancer model along with zebrafish transgenesis, we demonstrate that the transforming ability of BRAFV600E along with additional mutations depends on the intrinsic transcriptional program present in the cell of origin. In both systems, melanocytes are less responsive to mutations, whereas both neural crest and melanoblast populations are readily transformed. Profiling reveals that progenitors have higher expression of chromatin-modifying enzymes such as ATAD2, a melanoma competence factor that forms a complex with SOX10 and allows for expression of downstream oncogenic and neural crest programs. These data suggest that oncogenic competence is mediated by regulation of developmental chromatin factors, which then allow for proper response to those oncogenes.


Sujet(s)
Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Chromatine/métabolisme , Mélanome/génétique , Mélanome/anatomopathologie , Crête neurale/anatomopathologie , ATPases associated with diverse cellular activities/génétique , ATPases associated with diverse cellular activities/métabolisme , Animaux , Animal génétiquement modifié , Chromatine/génétique , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Humains , Mélanocytes/métabolisme , Mélanocytes/anatomopathologie , Souris , Tumeurs expérimentales , Cellules souches tumorales/anatomopathologie , Crête neurale/métabolisme , Cellules souches pluripotentes/anatomopathologie , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes B-raf/métabolisme , Facteurs de transcription SOX-E/génétique , Facteurs de transcription SOX-E/métabolisme , Transcription génétique , Danio zébré
15.
Nat Commun ; 12(1): 5056, 2021 08 20.
Article de Anglais | MEDLINE | ID: mdl-34417458

RÉSUMÉ

Melanoma cells rely on developmental programs during tumor initiation and progression. Here we show that the embryonic stem cell (ESC) factor Sall4 is re-expressed in the Tyr::NrasQ61K; Cdkn2a-/- melanoma model and that its expression is necessary for primary melanoma formation. Surprisingly, while Sall4 loss prevents tumor formation, it promotes micrometastases to distant organs in this melanoma-prone mouse model. Transcriptional profiling and in vitro assays using human melanoma cells demonstrate that SALL4 loss induces a phenotype switch and the acquisition of an invasive phenotype. We show that SALL4 negatively regulates invasiveness through interaction with the histone deacetylase (HDAC) 2 and direct co-binding to a set of invasiveness genes. Consequently, SALL4 knock down, as well as HDAC inhibition, promote the expression of an invasive signature, while inhibition of histone acetylation partially reverts the invasiveness program induced by SALL4 loss. Thus, SALL4 appears to regulate phenotype switching in melanoma through an HDAC2-mediated mechanism.


Sujet(s)
Épigenèse génétique , Mélanome/génétique , Mélanome/anatomopathologie , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie , Facteur de croissance des cellules souches/métabolisme , Facteurs de transcription/métabolisme , Acétylation , Animaux , Séquence nucléotidique , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Adhérence cellulaire/génétique , Lignée cellulaire tumorale , Lignage cellulaire , Prolifération cellulaire , Protéines de liaison à l'ADN/métabolisme , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Histone Deacetylase 2/métabolisme , Histone/métabolisme , Humains , Mélanocytes/métabolisme , Mélanocytes/anatomopathologie , Souris nude , Souris transgéniques , Invasion tumorale , Micrométastase tumorale , Liaison aux protéines , Charge tumorale
16.
Cell Stem Cell ; 24(4): 637-653.e9, 2019 04 04.
Article de Anglais | MEDLINE | ID: mdl-30951662

RÉSUMÉ

Increasing evidence suggests that cancer cells highjack developmental programs for disease initiation and progression. Melanoma arises from melanocytes that originate during development from neural crest stem cells (NCSCs). Here, we identified the transcription factor Yin Yang 1 (Yy1) as an NCSCs regulator. Conditional deletion of Yy1 in NCSCs resulted in stage-dependent hypoplasia of all major neural crest derivatives due to decreased proliferation and increased cell death. Moreover, conditional ablation of one Yy1 allele in a melanoma mouse model prevented tumorigenesis, indicating a particular susceptibility of melanoma cells to reduced Yy1 levels. Combined RNA sequencing (RNA-seq), chromatin immunoprecipitation (ChIP)-seq, and untargeted metabolomics demonstrated that YY1 governs multiple metabolic pathways and protein synthesis in both NCSCs and melanoma. In addition to directly regulating a metabolic gene set, YY1 can act upstream of MITF/c-MYC as part of a gene regulatory network controlling metabolism. Thus, both NCSC development and melanoma formation depend on an intricate YY1-controlled metabolic program.


Sujet(s)
Mélanome/métabolisme , Mélanome/anatomopathologie , Crête neurale/cytologie , Crête neurale/métabolisme , Facteur de transcription YY1/métabolisme , Animaux , Lignée cellulaire tumorale , Humains , Souris , Souris knockout , Souris transgéniques , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Facteur de transcription YY1/déficit
17.
Cell Stem Cell ; 21(5): 679-693.e6, 2017 Nov 02.
Article de Anglais | MEDLINE | ID: mdl-29033351

RÉSUMÉ

To identify the cells at the origin of melanoma, we combined single-cell lineage-tracing and transcriptomics approaches with time-lapse imaging. A mouse model that recapitulates key histopathological features of human melanomagenesis was created by inducing a BRafV600E-driven melanomagenic program in tail interfollicular melanocytes. Most targeted mature, melanin-producing melanocytes expanded clonally within the epidermis before losing their differentiated features through transcriptional reprogramming and eventually invading the dermis. Tumors did not form within interscales, which contain both mature and dormant amelanotic melanocytes. The hair follicle bulge, which contains melanocyte stem cells, was also refractory to melanomagenesis. These studies identify varying tumor susceptibilities within the melanocytic lineage, highlighting pigment-producing cells as the melanoma cell of origin, and indicate that regional variation in tumor predisposition is dictated by microenvironmental cues rather than intrinsic differences in cellular origin. Critically, this work provides in vivo evidence that differentiated somatic cells can be reprogrammed into cancer initiating cells.


Sujet(s)
Dédifférenciation cellulaire , Mélanocytes/anatomopathologie , Mélanome/génétique , Mutation/génétique , Protéines proto-oncogènes B-raf/génétique , Tumeurs cutanées/génétique , Pigmentation de la peau , Animaux , Marqueurs biologiques/métabolisme , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Prolifération cellulaire , Derme/anatomopathologie , Follicule pileux/anatomopathologie , Humains , Mélanocytes/métabolisme , Mélanome/anatomopathologie , Souris , Invasion tumorale , Tumeurs cutanées/anatomopathologie , Niche de cellules souches , Queue , Transcriptome/génétique ,
18.
J Neuroinflammation ; 12: 62, 2015 Apr 02.
Article de Anglais | MEDLINE | ID: mdl-25889302

RÉSUMÉ

BACKGROUND: Oligodendrocytes are myelinating cells of the central nervous system which support functionally, structurally, and metabolically neurons. Mature oligodendrocytes are generally believed to be mere targets of destruction in the context of neuroinflammation and tissue damage, but their real degree of in vivo plasticity has become a matter of debate. We thus investigated the in vivo dynamic, actin-related response of these cells under different kinds of demyelinating stress. METHODS: We used a novel mouse model (oLucR) expressing luciferase in myelin oligodendrocyte glycoprotein-positive oligodendrocytes under the control of a ß-actin promoter. Activity of this promoter served as surrogate for dynamics of the cytoskeleton gene transcription through recording of in vivo bioluminescence following diphtheria toxin-induced oligodendrocyte death and autoimmune demyelination. Cytoskeletal gene expression was quantified from mature oligodendrocytes directly isolated from transgenic animals through cell sorting. RESULTS: Experimental demyelinating setups augmented oligodendrocyte-specific in vivo bioluminescence. These changes in luciferase signal were confirmed by further ex vivo analysis of the central nervous system tissue from oLucR mice. Increase in bioluminescence upon autoimmune inflammation was parallel to an oligodendrocyte-specific increased transcription of ß-tubulin. CONCLUSIONS: Mature oligodendrocytes acutely increase their cytoskeletal plasticity in vivo during demyelination. They are therefore not passive players under demyelinating conditions but can rather react dynamically to external insults.


Sujet(s)
Maladies du système nerveux central/anatomopathologie , Système nerveux central/métabolisme , Cytosquelette/métabolisme , Glycoprotéine MOG/métabolisme , Oligodendroglie/métabolisme , Animaux , Protéines de liaison au calcium/génétique , Protéines de liaison au calcium/métabolisme , Cytokines/métabolisme , Toxine diphtérique/pharmacologie , Modèles animaux de maladie humaine , Encéphalomyélite auto-immune expérimentale/induit chimiquement , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Régulation de l'expression des gènes/génétique , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Techniques in vitro , Souris , Souris transgéniques , Protéines des microfilaments/génétique , Protéines des microfilaments/métabolisme , Glycoprotéine MOG/génétique
19.
Cell Stem Cell ; 16(3): 314-22, 2015 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-25748934

RÉSUMÉ

The neural crest (NC) is an embryonic stem/progenitor cell population that generates a diverse array of cell lineages, including peripheral neurons, myelinating Schwann cells, and melanocytes, among others. However, there is a long-standing controversy as to whether this broad developmental perspective reflects in vivo multipotency of individual NC cells or whether the NC is comprised of a heterogeneous mixture of lineage-restricted progenitors. Here, we resolve this controversy by performing in vivo fate mapping of single trunk NC cells both at premigratory and migratory stages using the R26R-Confetti mouse model. By combining quantitative clonal analyses with definitive markers of differentiation, we demonstrate that the vast majority of individual NC cells are multipotent, with only few clones contributing to single derivatives. Intriguingly, multipotency is maintained in migratory NC cells. Thus, our findings provide definitive evidence for the in vivo multipotency of both premigratory and migrating NC cells in the mouse.


Sujet(s)
Antigènes de différenciation/métabolisme , Lignage cellulaire/physiologie , Mouvement cellulaire/physiologie , Cellules souches multipotentes/métabolisme , Crête neurale/embryologie , Crête neurale/métabolisme , Animaux , Souris , Souris transgéniques , Cellules souches multipotentes/cytologie , Crête neurale/cytologie
20.
J Neurosci ; 34(17): 6112-22, 2014 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-24760871

RÉSUMÉ

Schwann cells, the myelinating glia of the peripheral nervous system (PNS), originate from multipotent neural crest cells that also give rise to other cells, including neurons, melanocytes, chondrocytes, and smooth muscle cells. The transcription factor Sox10 is required for peripheral glia specification. However, all neural crest cells express Sox10 and the mechanisms directing neural crest cells into a specific lineage are poorly understood. We show here that histone deacetylases 1 and 2 (HDAC1/2) are essential for the specification of neural crest cells into Schwann cell precursors and satellite glia, which express the early determinants of their lineage myelin protein zero (P0) and/or fatty acid binding protein 7 (Fabp7). In neural crest cells, HDAC1/2 induced expression of the transcription factor Pax3 by binding and activating the Pax3 promoter. In turn, Pax3 was required to maintain high Sox10 levels and to trigger expression of Fabp7. In addition, HDAC1/2 were bound to the P0 promoter and activated P0 transcription. Consistently, in vivo genetic deletion of HDAC1/2 in mouse neural crest cells led to strongly decreased Sox10 expression, no detectable Pax3, virtually no satellite glia, and no Schwann cell precursors in dorsal root ganglia and peripheral nerves. Similarly, in vivo ablation of Pax3 in the mouse neural crest resulted in strongly reduced expression of Sox10 and Fabp7. Therefore, by controlling the expression of Pax3 and the concerted action of Pax3 and Sox10 on their target genes, HDAC1/2 direct the specification of neural crest cells into peripheral glia.


Sujet(s)
Différenciation cellulaire/physiologie , Histone Deacetylase 1/métabolisme , Histone Deacetylase 2/métabolisme , Crête neurale/métabolisme , Cellules souches neurales/métabolisme , Oligodendroglie/métabolisme , Cellules de Schwann/métabolisme , Animaux , Régulation de l'expression des gènes au cours du développement , Histone Deacetylase 1/génétique , Histone Deacetylase 2/génétique , Souris , Crête neurale/cytologie , Cellules souches neurales/cytologie , Oligodendroglie/cytologie , Facteur de transcription PAX3 , Facteurs de transcription PAX/génétique , Facteurs de transcription PAX/métabolisme , Facteurs de transcription SOX-E/génétique , Facteurs de transcription SOX-E/métabolisme , Cellules de Schwann/cytologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE