Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 5 de 5
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Proc Natl Acad Sci U S A ; 120(34): e2301301120, 2023 08 22.
Article de Anglais | MEDLINE | ID: mdl-37585469

RÉSUMÉ

The auditory organ of Corti is comprised of only two major cell types-the mechanosensory hair cells and their associated supporting cells-both specified from a single pool of prosensory progenitors in the cochlear duct. Here, we show that competence to respond to Atoh1, a transcriptional master regulator necessary and sufficient for induction of mechanosensory hair cells, is established in the prosensory progenitors between E12.0 and 13.5. The transition to the competent state is rapid and is associated with extensive remodeling of the epigenetic landscape controlled by the SoxC group of transcription factors. Conditional loss of Sox4 and Sox11-the two homologous family members transiently expressed in the inner ear at the time of competence establishment-blocks the ability of prosensory progenitors to differentiate as hair cells. Mechanistically, we show that Sox4 binds to and establishes accessibility of early sensory lineage-specific regulatory elements, including ones associated with Atoh1 and its direct downstream targets. Consistent with these observations, overexpression of Sox4 or Sox11 prior to developmental establishment of competence precociously induces hair cell differentiation in the cochlear progenitors. Further, reintroducing Sox4 or Sox11 expression restores the ability of postnatal supporting cells to differentiate as hair cells in vitro and in vivo. Our findings demonstrate the pivotal role of SoxC family members as agents of epigenetic and transcriptional changes necessary for establishing competence for sensory receptor differentiation in the inner ear.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice , Facteurs de transcription SOX-C , Animaux , Facteurs de transcription SOX-C/génétique , Facteurs de transcription SOX-C/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Cochlée/métabolisme , Cellules ciliées auditives/métabolisme , Différenciation cellulaire , Facteurs de transcription/métabolisme , Épigenèse génétique , Organe spiral , Régulation de l'expression des gènes au cours du développement , Mammifères/métabolisme
2.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Article de Anglais | MEDLINE | ID: mdl-34266958

RÉSUMÉ

During embryonic development, hierarchical cascades of transcription factors interact with lineage-specific chromatin structures to control the sequential steps in the differentiation of specialized cell types. While examples of transcription factor cascades have been well documented, the mechanisms underlying developmental changes in accessibility of cell type-specific enhancers remain poorly understood. Here, we show that the transcriptional "master regulator" ATOH1-which is necessary for the differentiation of two distinct mechanoreceptor cell types, hair cells in the inner ear and Merkel cells of the epidermis-is unable to access much of its target enhancer network in the progenitor populations of either cell type when it first appears, imposing a block to further differentiation. This block is overcome by a feed-forward mechanism in which ATOH1 first stimulates expression of POU4F3, which subsequently acts as a pioneer factor to provide access to closed ATOH1 enhancers, allowing hair cell and Merkel cell differentiation to proceed. Our analysis also indicates the presence of both shared and divergent ATOH1/POU4F3-dependent enhancer networks in hair cells and Merkel cells. These cells share a deep developmental lineage relationship, deriving from their common epidermal origin, and suggesting that this feed-forward mechanism preceded the evolutionary divergence of these very different mechanoreceptive cell types.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Cellules ciliées auditives/métabolisme , Protéines à homéodomaine/métabolisme , Mécanorécepteurs/métabolisme , Facteur de transcription Brn-3C/métabolisme , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Différenciation cellulaire , Cochlée/métabolisme , Éléments activateurs (génétique) , Épigenèse génétique , Cellules ciliées auditives/cytologie , Protéines à homéodomaine/génétique , Humains , Cellules de Merkel/métabolisme , Souris , Facteur de transcription Brn-3C/génétique
3.
Dev Cell ; 56(17): 2471-2485.e5, 2021 09 13.
Article de Anglais | MEDLINE | ID: mdl-34331868

RÉSUMÉ

Adult mammalian tissues such as heart, brain, retina, and the sensory structures of the inner ear do not effectively regenerate, although a latent capacity for regeneration exists at embryonic and perinatal times. We explored the epigenetic basis for this latent regenerative potential in the mouse inner ear and its rapid loss during maturation. In perinatal supporting cells, whose fate is maintained by Notch-mediated lateral inhibition, the hair cell enhancer network is epigenetically primed (H3K4me1) but silenced (active H3K27 de-acetylation and trimethylation). Blocking Notch signaling during the perinatal period of plasticity rapidly eliminates epigenetic silencing and allows supporting cells to transdifferentiate into hair cells. Importantly, H3K4me1 priming of the hair cell enhancers in supporting cells is removed during the first post-natal week, coinciding with the loss of transdifferentiation potential. We hypothesize that enhancer decommissioning during cochlear maturation contributes to the failure of hair cell regeneration in the mature organ of Corti.


Sujet(s)
Différenciation cellulaire/physiologie , Cellules ciliées auditives/métabolisme , Récepteurs Notch/métabolisme , Régénération/physiologie , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Transdifférenciation cellulaire/génétique , Transdifférenciation cellulaire/physiologie , Épigenèse génétique/génétique , Épigenèse génétique/physiologie , Cellules ciliées auditives/cytologie , Souris transgéniques , Séquences d'acides nucléiques régulatrices/génétique
4.
Elife ; 92020 06 30.
Article de Anglais | MEDLINE | ID: mdl-32602462

RÉSUMÉ

The mechanoreceptive sensory hair cells in the inner ear are selectively vulnerable to numerous genetic and environmental insults. In mammals, hair cells lack regenerative capacity, and their death leads to permanent hearing loss and vestibular dysfunction. Their paucity and inaccessibility has limited the search for otoprotective and regenerative strategies. Growing hair cells in vitro would provide a route to overcome this experimental bottleneck. We report a combination of four transcription factors (Six1, Atoh1, Pou4f3, and Gfi1) that can convert mouse embryonic fibroblasts, adult tail-tip fibroblasts and postnatal supporting cells into induced hair cell-like cells (iHCs). iHCs exhibit hair cell-like morphology, transcriptomic and epigenetic profiles, electrophysiological properties, mechanosensory channel expression, and vulnerability to ototoxin in a high-content phenotypic screening system. Thus, direct reprogramming provides a platform to identify causes and treatments for hair cell loss, and may help identify future gene therapy approaches for restoring hearing.


Worldwide, hearing loss is the most common loss of sensation. Most cases of hearing loss are due to the death of specialized hair cells found deep inside the ear. These hair cells convert sounds into nerve impulses which can be understood by the brain. Hair cells naturally degrade as part of aging and can be damaged by other factors including loud noises, and otherwise therapeutic drugs, such as those used in chemotherapy for cancer. In humans and other mammals, once hair cells are lost they cannot be replaced. Hair cells have often been studied using mice, but the small number of hair cells in their ears, and their location deep inside the skull, makes it particularly difficult to study them in this way. Scientists are seeking ways to grow hair cells in the laboratory to make it easier to understand how they work and the factors that contribute to their damage and loss. Different cell types in the body are formed in response to specific combinations of biological signals. Currently, scientists do not have an efficient way to grow hair cells in the laboratory, because the correct signals needed to create them are not known. Menendez et al. have now identified four proteins which, when activated, convert fibroblasts, a common type of cell, into hair cells similar to those in the ear. These proteins are called Six1, Atoh1, Pou4f3 and Gfi1. Menendez et al. termed the resulting cells induced hair cells, or iHCs for short, and analyzed these cells to identify those characteristics that are similar to normal hair cells, as well as their differences. Importantly, the iHCs were found to be damaged by the same chemicals that specifically harm normal hair cells, suggesting they are useful test subjects. The ability to create hair cells in the laboratory using more easily available cells has many uses. These cells can help to understand the normal function of hair cells and how they become damaged. They can also be used to test new drugs to assess their success in preventing or reversing hearing loss. These findings may also lead to genetic solutions to curing hearing loss.


Sujet(s)
Lignage cellulaire , Fibroblastes/physiologie , Cellules ciliées auditives internes/physiologie , Cellules de soutien du labyrinthe/physiologie , Souris/physiologie , Animaux , Souris transgéniques , Queue , Facteurs de transcription/métabolisme
5.
Proc Natl Acad Sci U S A ; 117(24): 13552-13561, 2020 06 16.
Article de Anglais | MEDLINE | ID: mdl-32482884

RÉSUMÉ

Precise control of organ growth and patterning is executed through a balanced regulation of progenitor self-renewal and differentiation. In the auditory sensory epithelium-the organ of Corti-progenitor cells exit the cell cycle in a coordinated wave between E12.5 and E14.5 before the initiation of sensory receptor cell differentiation, making it a unique system for studying the molecular mechanisms controlling the switch between proliferation and differentiation. Here we identify the Yap/Tead complex as a key regulator of the self-renewal gene network in organ of Corti progenitor cells. We show that Tead transcription factors bind directly to the putative regulatory elements of many stemness- and cell cycle-related genes. We also show that the Tead coactivator protein, Yap, is degraded specifically in the Sox2-positive domain of the cochlear duct, resulting in down-regulation of Tead gene targets. Further, conditional loss of the Yap gene in the inner ear results in the formation of significantly smaller auditory and vestibular sensory epithelia, while conditional overexpression of a constitutively active version of Yap, Yap5SA, is sufficient to prevent cell cycle exit and to prolong sensory tissue growth. We also show that viral gene delivery of Yap5SA in the postnatal inner ear sensory epithelia in vivo drives cell cycle reentry after hair cell loss. Taken together, these data highlight the key role of the Yap/Tead transcription factor complex in maintaining inner ear progenitors during development, and suggest new strategies to induce sensory cell regeneration.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Protéines du cycle cellulaire/métabolisme , Auto-renouvellement cellulaire , Organe spiral/embryologie , Organe spiral/métabolisme , Cellules souches/cytologie , Facteurs de transcription/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Animaux , Cycle cellulaire , Protéines du cycle cellulaire/génétique , Différenciation cellulaire , Régulation de l'expression des gènes au cours du développement , Cellules ciliées auditives , Souris , Organe spiral/cytologie , Liaison aux protéines , Facteurs de transcription SOX-B1/génétique , Facteurs de transcription SOX-B1/métabolisme , Cellules souches/métabolisme , Facteurs de transcription/génétique , Protéines de signalisation YAP
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE